Sunteți pe pagina 1din 81

Physiol Rev 90: 675–754, 2010;

doi:10.1152/physrev.00012.2009.

The Ventilatory Response to Hypoxia in Mammals:


Mechanisms, Measurement, and Analysis

LUC J. TEPPEMA AND ALBERT DAHAN

Department of Anesthesiology, Leiden University Medical Center, Leiden, The Netherlands

I. Introduction 676
II. The Hypoxic Ventilatory Response in Fetal and Neonatal Mammals 677
A. Fetal breathing movements 677
B. Hypoxia inhibits fetal breathing movements 677
C. Maturation of the HVR in neonatal animals 679
D. Developmental plasticity of the HVR 681
E. The HVR in term and preterm babies 684
III. Peripheral and Central Mechanisms Mediating the Hypoxic Ventilatory Response in Adult Mammals 685
A. Carotid body mechanisms in oxygen sensing 685
B. Hypoxia activates the transcription factor hypoxia-inducible factor-1 690
C. Central effects of hypoxia 692
D. Hypoxic ventilatory decline in the adult mammal 698
IV. Genetic Influence on the Ventilatory Response to Hypoxia 703
A. The genome and HVR 703
B. Mutations and HVR 704
V. Pathophysiological Influences on the Hypoxic Ventilatory Response 705
A. Biological variability 705
B. Ageing 706
C. Gender and pregnancy 706
D. Influence of CO2 on the HVR: O2-CO2 interaction 707
E. Carotid body resection and the HVR 710
F. Chronic hypoxia and the HVR 711
G. Chronic intermittent hypoxia and the HVR 720
VI. Measuring, Expressing, and Modeling the Hypoxic Ventilatory Response 725
A. Measuring the HVR 725
B. Expressing the steady-state HVR 731
C. Modeling the HVR 731
VII. Comments and Future Challenges 732

Teppema LJ, Dahan A. The Ventilatory Response to Hypoxia in Mammals: Mechanisms, Measurement, and
Analysis. Physiol Rev 90: 675–754, 2010; doi:10.1152/physrev.00012.2009.—The respiratory response to hypoxia
in mammals develops from an inhibition of breathing movements in utero into a sustained increase in
ventilation in the adult. This ventilatory response to hypoxia (HVR) in mammals is the subject of this review.
The period immediately after birth contains a critical time window in which environmental factors can cause
long-term changes in the structural and functional properties of the respiratory system, resulting in an altered
HVR phenotype. Both neonatal chronic and chronic intermittent hypoxia, but also chronic hyperoxia, can
induce such plastic changes, the nature of which depends on the time pattern and duration of the exposure
(acute or chronic, episodic or not, etc.). At adult age, exposure to chronic hypoxic paradigms induces
adjustments in the HVR that seem reversible when the respiratory system is fully matured. These changes are
orchestrated by transcription factors of which hypoxia-inducible factor 1 has been identified as the master
regulator. We discuss the mechanisms underlying the HVR and its adaptations to chronic changes in ambient
oxygen concentration, with emphasis on the carotid bodies that contain oxygen sensors and initiate the
response, and on the contribution of central neurotransmitters and brain stem regions. We also briefly
summarize the techniques used in small animals and in humans to measure the HVR and discuss the specific
difficulties encountered in its measurement and analysis.

www.prv.org 0031-9333/10 $18.00 Copyright © 2010 the American Physiological Society 675
676 LUC J. TEPPEMA AND ALBERT DAHAN

I. INTRODUCTION response is maintained) but without a(n) (appreciable)


decrease in metabolism.
The independent discovery of oxygen by the Swedish Fetal animals respond differently to hypoxia. They
apothecary Carl Wilhelm Sheele in 1772 and the English display breathing movements in utero that can be de-
scientist Joseph Priestley in 1774 is considered one of the tected in an early stage of gestation (for example, ⬃40
greatest scientific discoveries (for a historical overview, days in lambs with a term duration of ⬃147 days and ⬃10
see Ref. 698). To date, 235 years later, it is a well estab- wk in humans). When the fetus is exposed to a low
lished fact that aerobic metabolism in mammals relies on maternal PaO2 or umbilical cord occlusion, breathing
sufficient oxygen availability in the mitochondria where movements are reduced in frequency and can be com-
its reduction to water is coupled to the generation of ATP pletely abolished (54, 137, 170). This review describes the
molecules. To be completed, this process of oxidative mechanisms underlying both the fetal and neonatal
phosphorylation requires efficient transport of electrons breathing responses to hypoxia and summarizes how the
between five major protein complexes (mitochondrial neonatal response gradually matures into the adult phe-
electron transport chain, attached to the inner mitochon- notype. The influence of the genotype on the HVR is also
drial membrane). The total amount of oxygen in the body briefly discussed.
is low (⬃1.5 liter in total, ⬃50 ml of which is stored in the Both in neonates and in the adult, the initial increase
tissues), so an undisturbed and complete oxidative phos- in ventilation in hypoxia is initiated by the carotid bodies,
phorylation requires a continuous fresh supply of oxygen located at the carotid bifurcations and containing oxygen-
from the environment. Thus an insufficient supply of ox- sensitive cells that increase their activity in hypoxia (278,
ygen may compromise the electron transport chain result- 280). The carotid bodies send their afferent input to the
ing not only in a decrease in ATP production but also in an brain stem where further processing by the respiratory
increased generation of reactive oxygen species (ROS), centers takes place. During the last two decades, consid-
such as superoxide anion. ROS can cause structural dam- erable progression has been made in elucidating the
age to nucleic acids, proteins, and lipids. At the same mechanism of oxygen sensing in the carotid bodies, the
time, cytoplasmic signaling-transduction cascades can di- factors that modulate it, and the way in which the brain
rectly be influenced by ROS. Mobilization of antioxidant stem processes the afferent traffic from the carotid bod-
defense mechanisms, such as superoxide dismutase that ies, and in this review we present a state of the art of
catalyzes the conversion of superoxide anion into hydro- these areas.
gen peroxide and catalase that accelerates the conversion Studying the mechanisms underlying respiratory ad-
of H2O2 into water and oxygen, helps to restore the cel- justments (rise in ventilation, increase in the HVR) to
lular redox balance. chronic hypoxia and chronic intermittent hypoxia in ani-
Apart from cellular antioxidant enzymes, the body mals (mainly rats) has revealed a fascinating picture of
has several means to compensate for an acute decrease in numerous plastic morphological, biochemical, and func-
oxygen availability (acute hypoxia). Animals relying on tional changes in the carotid bodies and brain stem that
pulmonary ventilation for their oxygen supply increase are initiated and maintained by transcription factors that
their ventilation to improve the uptake of oxygen. This regulate the expression of many genes. Both during neo-
ventilatory response to hypoxia (HVR) in mammals, i.e., natal development and in adulthood, the respiratory sys-
the increase in pulmonary ventilation that results from a tems of rats and other species display plasticity, i.e., the
decrease in the partial pressure of oxygen in the arterial ability to learn from previous experience and to alter the
blood (PaO2) is the subject of this review. Another means phenotype of several reflex responses. One developmen-
that mammals can utilize to preserve oxygen homeostasis tal period is of particular interest: immediately after birth,
in acute hypoxia is to optimize and decrease the rate of within the period of postnatal maturation, there is a crit-
aerobic metabolism, thus reducing oxygen demand and at ical period (“critical time window”) during which environ-
the same time increasing ATP production from anaerobic mental factors can modulate structural and functional
metabolism (glycolysis). properties of the respiratory system. These plastic
Neonatal and adult animals use the options of chang- changes can be permanent and may be irreversible in
ing ventilation and/or metabolism in a different way. adulthood and possibly even in adolescence (115). An
Apart from an initial fast and transient rise in ventilation, example is chronic exposure to hyperoxia during this
neonates, when exposed to acute hypoxia lasting 20 –30 period: it leads to degenerative changes in the carotid
min, display only a minor increase in ventilation but an bodies and to a reduced HVR that may persist until
appreciable reduction in metabolism, thereby optimizing adulthood (189, 220, 838). Adaptations of the HVR to
the ratio ventilation over oxygen consumption. During (intermittent) chronic hypoxic paradigms in this criti-
maturation into adulthood, this composite response grad- cal neonatal time window will be compared with those
ually develops into one with a predominant and sustained in adulthood and placed in the context of plastic
increase in ventilation (while the biphasic character of the changes in the carotid bodies and brain stem that result

Physiol Rev • VOL 90 • APRIL 2010 • www.prv.org


THE VENTILATORY RESPONSE TO HYPOXIA IN MAMMALS 677

from changes in gene expression. Of critical impor- levels of ⬃30% leads to adaptations such as spinal inhibi-
tance in interpreting these studies is to take into ac- tion, a reduction in rapid eye movements, facilitation of
count the specific methods that are used to measure the high-voltage electrocortical electroencephalogram (EEG)
HVR, the environmental and physiological context in activity, inhibition of FBM, a fall in metabolic rate, and a
which they are preformed, and the analytical methods rise in cerebral blood flow (CBF) (85, 88, 137, 170, 317,
that are applied to quantify and compare data. We 408, 588). In the lamb, FBM account for ⬃30% of overall
briefly summarize the techniques used in small animals oxygen consumption (670). Anemia, reduced uterine
and in humans and discuss the specific difficulties that blood flow, and/or umbilical cord occlusion also lead to
are encountered in measurement and analysis. FBM inhibition (74, 87, 328, 830). Hypoxic inhibition of
FBM also occurs in fetal rat (391), rhesus monkeys (488),
II. THE HYPOXIC VENTILATORY RESPONSE IN and given the relative low FBM incidence in growth-
FETAL AND NEONATAL MAMMALS retarded fetuses, presumably also in humans (54, 251).
The mechanism of hypoxic inhibition of FBM is complex
and may be causally related to one or more of the above
A. Fetal Breathing Movements adaptations. Below we discuss some major factors influ-
encing hypoxic inhibition of FBM with emphasis on those
Fetal breathing movements (FBM) are coordinated that may still contribute to shaping the HVR in newborns
rhythmical contractions of respiratory muscles that inter- and even adults.
mittently reduce intrathoracic pressure and cause amni-
otic fluid movements within the trachea (302). FBM are 1. CBF and metabolism and hypoxic inhibition
crucial for fetal lung growth and maturation and are ob- of FBM
served in all mammalian species studied thus far (49, 85,
302, 357, 391, 651, 725, 814). The most commonly ob- The fetal brain exhibits a relatively large rate of
served FBM are rapid irregular movements and can be oxygen consumption that is tightly linked to oxygen de-
detected in sheep (mean term duration 147 days) from livery (588). In the fetus, hypoxia not only leads to de-
⬃40 days gestation and in humans from ⬃10 wk (88, 137, creases in neuronal activity and metabolic rate, but also
170, 302). Early in gestation, until about day 115, fetal to a rise in CBF, and both these responses have been
lambs show continuous FBM that are sometimes inter- linked to the inhibition of FBM (588).
rupted by apneas; in the third semester, EEG activity
starts to differentiate between states of low- and high-
2. Pontine and thalamic areas and hypoxic inhibition
voltage cortical activity associated with rapid eye move-
of FBM
ments (active sleep) and active movements of neck and
limb muscles, respectively. From this developmental stage un- In fetal lambs, brain stem transection at the (mid)
til term, FBM are limited to active sleep (low voltage collicular level or electrolytic lesions somewhat more
cortical activity; reviewed in Refs. 85, 170, 302, 317, 357, caudally in the upper lateral pons in the vicinity of the
625, 814). This association of FBM and electrocortical trigeminal sensory nuclei involving parts of the medial
activity is disrupted after brain transection rostral from parabrachial and Kölliker Fuse nuclei has two important
the caudal hypothalamus; when this transection is per- consequences: 1) it leads to continuous breathing in nor-
formed at upper pontine level, i.e., a region close to the moxia and to a dissociation between EEG activity and
inferior colliculus in the vicinity of the principal sensory
breathing, and 2) it converts the inhibitory response to
and motor nuclei of the trigeminal nerve, FBM become
hypoxia in the intact condition into a stimulatory one
almost continuous (170, 171, 271, 363, 396, 523, 814, 815).
after transection (170, 171, 271, 363, 396, 523, 814, 815).
Sleep state should be considered as a factor modulating
Transection more rostrally through the caudal hypothal-
fetal breathing rather than providing a primary inducing
amus did not affect hypoxia-induced inhibition of FBM,
role: in fetal lambs, periodic FBM precede the electrocor-
indicating that an area caudally from it is able to elicit this
tical differentiation by ⬃1 wk, while at term the animal
response independently from higher centers (170).
starts to breathe continuously without changes in EEG
In fetal lambs, hypoxia facilitates a high-voltage elec-
pattern (651).
trocortical EEG pattern (80). FBM and low-voltage elec-
trocortical activity are closely linked, so brain areas in-
B. Hypoxia Inhibits Fetal Breathing Movements fluencing behavioral state may modulate hypoxic inhibi-
tion of FBM. One such area is the parafascicular nucleus
In fetal sheep, hypoxia caused by (gradually) expos- of the thalamus. Lesions in this area abolish hypoxia-in-
ing ewes to a hypoxic gas mixture (mostly with CO2 duced inhibition of FBM while local electrical stimulation
added to maintain isocapnia) to achieve fetal saturation effectively decreases respiratory frequency (397–399).

Physiol Rev • VOL 90 • APRIL 2010 • www.prv.org


678 LUC J. TEPPEMA AND ALBERT DAHAN

3. Peripheral chemoreceptors and hypoxic inhibition 5. Neuromodulators and receptors in hypoxic


of FBM inhibition of FBM

In fetal sheep, the carotid bodies are active and A) ADRENERGIC MECHANISMS. Hypoxia-induced inhibition
respond to hypoxia with excitation (79). Thus the ques- of FBM may be mediated via adrenergic receptors. In fetal
tion arises whether they may have a counteractive role sheep, hypoxia activates catecholaminergic neurons in
in hypoxia or, alternatively, stimulate brain regions that subcoeruleus- and Kölliker-Fuse regions in the dorsolat-
are directly involved in inhibiting FBM. Carotid sinus eral pons, some of which appear to have direct connec-
nerve and aortic transection do not prevent hypoxic tions with the phrenic motoneuron pool in the cervical
inhibition of FBM but delay the onset of the response, cord (551). In both intact and brain stem transected fetal
suggesting a supportive but not indispensable influence lambs, systemic clonidine reduced the incidence of FBM,
of the peripheral chemoreceptors (406). After midcol- and this effect was blocked by an ␣2-adrenergic antago-
licular transection, hypoxia induced an increase in nist (32). Prolonged reduction in uterine blood flow in-
FBM in fetal lambs, and this response still existed after creased plasma norepinephrine concentration in fetal
subsequent carotid body denervation (171). After ca- lambs (328), while ventricular administration of clonidine
rotid sinus nerve transection, some fetal sheep showed reduced the incidence of breathing and a specific ␣2-
continuous breathing in normoxia and even FBM in adrenergic receptor antagonist abolished hypoxic inhibi-
hypoxia; peripheral chemodenervation alone neither tion of FBM (31). Systemic administration of both ␣1- and
changed the incidence of FBM in normoxia nor the ␣2-adrenergic antagonists had similar effects (270).
hypoxia-induced inhibition of breathing (523). In an- B) ADENOSINE A1 AND A2 RECEPTORS. An important mole-
other study, hypoxic stimulation of FBM after transec- cule in the coupling of cerebral blood flow to metabolism
tion depended on carotid sinus nerve integrity, suggest- is adenosine. Via adenosine A2 receptors, this molecule
ing that in the intact condition (i.e., without any lesion) induces ⬃50% of the hypoxia-induced rise in CBF, while
a stimulatory influence of the carotid bodies may be A1 receptors mediate hypoxic depression of cerebral me-
overridden (363, 396). In general, the emerging picture tabolism (83, 479, 588).
is that at least in fetal lambs intact carotid bodies are Experimental evidence indicates that adenosine is an
not required to produce hypoxic inhibition of FBM, important player in the hypoxia-induced reduction in
while their role in stimulating breathing in hypoxia FBM incidence. In fetal lambs, systemic adenosine caused
after brain stem transection is somewhat controversial. a decrease in FBM incidence similar to hypoxia, while
adenosine receptor antagonists had the reverse effects
4. Placental factors and hypoxic inhibition of FBM (76, 125, 405). The posteromedial thalamus displays in-
creased adenosine production during hypoxia and con-
By itself, the fact that umbilical cord occlusion tains a high density of adenosine A2A receptors that me-
leads to hypoxia in the fetus and decreases the inci- diate FBM inhibition (402, 404, 830, 862). Intra-arterial
dence of FBM (328, 830) does not exclude placental infusion of a specific A2A but not A1 receptor antagonist
factors as the source of FBM inhibition during maternal abolished both the hypoxic inhibition of rapid eye and
hypoxemia. Theoretically, a reduced release by the pla- breathing movements, while A1 antagonism only elimi-
centa of stimulatory agents could contribute to the nated the FBM response (403). Anemic hypoxia upregu-
reduction of FBM, but to date, such factors have not lates pontine adenosine A1 receptors in the brain stem of
been identified. The placenta produces inhibitory sub- fetal lambs, while the same receptor type in the rostroven-
stances such as prostaglandin E2 (PGE2), adenosine, trolateral medulla is also involved in the hypoxic FBM
and several peptides (16, 704, 815), while indomethacin response (76, 396), possibly by directly depressing local
and adenosine antagonists induce continuous breathing metabolism (84, 479).
and reverse hypoxic inhibition of FBM, respectively The way in which adenosine causes hypoxic inhibi-
(125, 170, 403, 404, 407). After prolonged reductions in tion of FBM may be quite complex. Following lesion of
uterine blood flow, plasma PGE2 concentration in fetal the upper lateral pons in the vicinity of the trigeminal
lambs and the decrease in incidence of FBM showed sensory nucleus (see above), normoxic respiratory fre-
parallel time courses (328). However, because sub- quency substantially decreases (363). Thus this region
stances released from the placenta during maternal may exert a tonic excitatory effect on ventrolateral med-
hypoxemia did not reduce FBM when the fetal PO2 was ullary respiratory neurons in normoxia, possibly by hy-
kept normoxic (413), we suggest that in this condition perpolarizing postinspiratory neurons. If during hypoxia
hypoxic FBM inhibition is caused by local factors in the adenosine, by a presynaptic action, inhibits these pontine
hypoxic fetal brain that also inhibit respiratory move- neurons, postinspiratory neurons may now depolarize,
ments during reduced uterine blood flow/umbilical cord thus inhibiting respiratory motor output. This would be
occlusion. consistent with in vitro data from neonatal rat and mice

Physiol Rev • VOL 90 • APRIL 2010 • www.prv.org


THE VENTILATORY RESPONSE TO HYPOXIA IN MAMMALS 679

showing hypoxia-induced depolarization of postinspira- signal may be contaminated by airway resistance effects
tory neurons (see references in Ref. 73). that may lead to erroneous estimation of tidal volume
In summary, ample evidence thus far indicates in- (218, 219, 356, 477, 751).
volvement of adrenergic and purinergic, adenosine A1 as The absence of blood gas data (in most cases) com-
well as A2A mechanisms in the hypoxic inhibition of FBM, bined with the impossibility to control end-tidal gases
but further studies are necessary to further characterize renders a reliable comparison of HVRs between treat-
and localize other transmitters and receptor subtypes ments difficult. By sheer necessity, the ambient O2 frac-
involved. tion is often taken as the independent variable, but its
relationship with the actual stimulus is uncertain and may
6. Some further considerations on hypoxia and vary. If in a chronic paradigm normoxic ventilation in-
fetal breathing creases, arterial PO2 will be higher and PCO2 lower for any
inspired PO2 level resulting in less carotid body stimula-
In most of the above studies, hypoxia was tested in tion; thus, in such cases, it is inaccurate to quantify the
fetal lambs in the last gestational semester. In midgesta- HVR as the magnitude of the change in ventilation per
tion, hypoxic inhibition of FBM is milder if not absent, given decrease in ambient PO2. In addition, a lower initial
indicating that it develops over the second half of preg- PCO2 may tend to further reduce at least the early phase of
nancy (496). the HVR, owing to O2-CO2 interaction in the carotid bod-
Does the low fetal arterial PO2 (⬃25 Torr) imply a ies (see sect. V). Without addition of CO2, hypoxic tests
tonic reduction of FBM? This seems unlikely, since nei- may be poikilocapnic depending on changes in ventilation
ther fetal lambs (125–130 days gestation) nor normal hu- and/or metabolism. In the ideal case, preferably three or
man fetuses show significant increases in the incidence of four measurement points well within the curvilinear part
FBM during maternal hyperoxia (54, 78, 88, 654 and ref- of the exponential ventilation-PO2 relationship should be
erences therein), although at least in the lamb it cannot be chosen, avoiding levels at which maximal ventilation is
excluded that after ⬎130 days of gestation a response of reached (39). A further point concerns the influence of the
continuous breathing to hyperoxia starts to develop (24). vigilance state on the HVR (421, 472, 729). Particularly in
In humans, it is only in growth-retarded fetuses that hy- neonates, EEG recordings are challenging and often not
peroxia results in a higher FBM frequency (54, 251). performed, so vigilance state-dependent changes in sen-
In fetal lambs, prolonged hypoxia (⬎12 h) initially sitivity cannot be accounted for. On the other hand, the
leads to a reduction in FBM frequency, but after a period influence of the latter factor may be limited because
of 12–20 h it returns to normal (86, 328, 401, 496, 831). sleep-wake cycles are not clearly defined in early life.
How this reversal occurs is unknown, but it has been Balbir et al. (29) proposed a noninvasive method for
related to a downregulation of adenosine receptors and sleep-wake assessment in neonatal mice, providing a po-
plasma glucose levels, respectively (328, 405). tential means to account for (changes in) brain state. These
Several alternative mechanisms to explain the hy- authors found a sound correlation between high electromyo-
poxic inhibition of FBM have been suggested, of which gram (EMG) activity (associated with the awake state) and
acidemia (647), increased blood flow through the ductus coordinated limb/head movements and suggested that this
venosus (217), and decreased mitochondrial ATP produc- behavioral index could be useful in avoiding the stress re-
tion (407) warrant further investigation. The cellular basis lated to EMG instrumentation in newborn animals (29).
of the response is unknown. These are only a few of the considerations to take into
account when comparing HVRs between treatments.
C. Maturation of the HVR in Neonatal Animals
2. Maturation of the HVR in neonatal animals
1. Measuring the HVR in small animals
In neonatal animals, the HVR appears as a biphasic
The most frequently used techniques to study breath- response consisting of a short initial rise in ventilation
ing in awake mice and rats are modifications of body (augmentation phase) rapidly followed by a secondary
plethysmography described by Drorbaugh and Fenn (193). roll-off mostly to a level below control (depressing phase;
Their barometric method is based on the principle that Ref. 73). Postnatal maturation of the HVR was already
when an animal in a closed chamber inhales a tidal vol- described in older studies and in various species such as
ume, this volume is warmed to body temperature, satu- lambs (e.g., Ref. 102), cats (89, 90, 301, 487, 653), piglets
rated with water vapor giving rise to pressure changes in (212), and rats (208, 453, 454). In all species, the relative
the chamber. As discussed by others, both closed and magnitudes of both phases are subject to maturation.
open systems are subject to limitations (e.g., related to Generally, shortly after birth, the initial rise in ventilation
pressure changes due to warming, humidification, and air is (much) smaller than the secondary roll-off, rapidly
leaks) and particularly in neonatal animals the pressure resulting in a lower baseline ventilation compared with

Physiol Rev • VOL 90 • APRIL 2010 • www.prv.org


680 LUC J. TEPPEMA AND ALBERT DAHAN

normoxia. However, when maturation proceeds, the ini- body weight and therefore could be limited or absent in
tial rise becomes larger, the secondary depression be- adults of larger species (243, 531). The processes under-
comes smaller, and eventually a sustained rise in ventila- lying maturation of the HVR are briefly reviewed below by
tion remains. Age-matched comparisons between species considering the augmentation and depressing phases sep-
are complicated by the different stimulus paradigms em- arately. In section IIIC, we briefly discuss a potential neu-
ployed. For example, deeper hypoxia may result in a ronal circuit that may be involved in the thermogenic
greater initial increase and a larger decrease in ventilation component of the HVR.
in the depressing phase (see Ref. 73 in which data from A) AUGMENTATION PHASE OF THE HVR. In newborn lambs,
humans, rat, cat, monkey, and sheep are shown). In ad- piglets, and rats, the relatively rapid initial rise in ventila-
dition, relatively mature species at birth have a better tion in acute hypoxia, designated as the augmentation
sustained neonatal HVR than those born less mature, e.g., phase (73), is abolished or greatly attenuated after carotid
sheep (mean term duration 147 days) are more mature sinus nerve transection, indicating that the carotid bodies
than rat (term duration 18 –23 days) and cat (term dura- mediate the augmentation phase (103, 166, 249, 473, 474,
tion 58 – 64 days; Refs. 73, 529). In 2- to 17-day-old piglets 533, 740). Shortly after birth, in newborn lambs, the PaO2
(term duration 112–120 days), the secondary roll-off threshold for hypoxic activation of the carotid body is
seems to be limited to the phrenic motoneuron pool, since reset from the low fetal PO2 (25 Torr) to ⬃55 Torr (79, 82).
upper airways muscle EMGs showed a sustained increase This is followed by a gradual increase in hypoxic sensi-
in activity in the presence of a biphasic response of the tivity of the carotid sinus nerve, a shift of the hyperbolic
diaphragm (490). response curve to higher PO2 values, a gradual appearance
In most neonates, a vital component of the hypoxic of O2-CO2 interaction, and an improved ability to maintain
response is a decrease in metabolism (hypoxic hypome- elevated discharge levels during prolonged hypoxia (116,
tabolism; Refs. 90, 529, 531, 532). Newborns of many 188, 487, 533, 601). The time course of these developmen-
species demonstrate a drop in oxygen consumption (V̇O2) tal changes (a few days to 3– 4 wk) varies between spe-
that in relatively mature species tends to be smaller. All cies, and the relative maturity at birth determines the time
species investigated show an increase in the ratio expired point at which adultlike responses are reached (33, 103,
ventilation (V̇E)/V̇O2 (i.e., the ratio expired ventilation over 208, 487, 601, 826). Processes underlying maturation of
O2 consumption), indicating a hyperventilatory response, the augmentation phase appear to be a complex fine
and this occurs irrespective of the magnitude of the HVR tuning between afferent nerve innervation of the carotid
(529, 531, 532). Thus ventilation may decrease in hypoxia, bodies involving trophic factors such as brain-derived
but still the outcome will be hyperventilation because of neuorotrophic factor and all elements of the of hypoxic
a larger drop in V̇O2. The fall in metabolism is due to a stimulus-transduction cascade including membrane chan-
reduction in nonshivering thermogenesis by brown adi- nels, neurotransmitters, receptors, and enzymes (see sect.
pose tissue, rather than to the hypoxia-induced (limited) III) and are reviewed elsewhere (26, 94, 191, 258, 611).
decrease in body temperature (526, 529, 531, 532). Resting These maturational processes must keep pace with
metabolism is an important determinant of the hypoxic developmental changes in the central nervous system, for
drop in V̇O2. Generally, animals with higher metabolic rate example, in the nuclei of the solitary tracts (NTS), the first
(e.g., small animals) demonstrate larger decreases in V̇O2, relay station of incoming carotid body afferent input.
but there are exceptions (243, 526, 531). In a cold envi- These processes involve changes in the activity of tran-
ronment, requiring more thermogenesis to generate heat, scription factors, formation of anatomical connections
a given animal will show much larger hypoxia-induced within the neuronal respiratory circuitry, and alterations
decreases in V̇O2 and ventilation (or smaller rise in venti- in neurotransmitter contents and receptor densities to set
lation) than at an ambient temperature close to thermo- the balance between excitatory and inhibitory neuro-
neutrality (526, 531). Consequently, measurements of the transmission. For example, during maturation, the density
HVR per se are more meaningful when combined with V̇O2 and expression of the glutamate receptor ionotropic N-
measurements and supplemented with end-tidal CO2 val- methyl-D-aspartate 1 (NR1) subunit of the NMDA (N-methyl-
ues. Furthermore, comparisons between treatments D-aspartic acid) receptor in the rat NTS increases, which
should preferably be made at equal ambient temperature leads to the upregulation of transcription factors and
(526, 531). other proteins that are involved in the hypoxic response
When maturation proceeds towards the adult stage, (564, 702). Another example of a developmental change in
hyperpnea replaces hypometabolism as the predominant the NTS is the downregulation of platelet-derived growth
feature of the hypoxic response, but in many adult spe- factor (PDGF) during early maturation (702; PDGF exerts
cies, depending on the ambient temperature, the drop in an inhibitory influence on ion currents through NMDA
metabolism can remain an important element of it (243, receptors, see also sect. IIID). Also in the rat, Wong-Riley
260, 482, 526, 531, 675). The magnitude of the fall in and Liu (847) have shown shifts in the expression of
metabolism is inversely related to resting V̇O2 relative to GABA receptor subunits in the NTS and Pre-Bötzinger

Physiol Rev • VOL 90 • APRIL 2010 • www.prv.org


THE VENTILATORY RESPONSE TO HYPOXIA IN MAMMALS 681

complex around postnatal day 12 (P12) that were paral- response, focal cooling of regions in the locus coeruleus
leled by ventilatory adjustments at the end of the second prevented the (secondary) fall in respiratory motor out-
postnatal week. Between P12 and P16, the HVR showed a put induced by adenosine and hypoxia, respectively (522).
sharp decline to reach a minimum at P13, whereafter it In 3-day-old piglets, the hypoxia-induced increase in tidal
gradually increased to a final magnitude on P20 –P21. volume waned between the second and fifth minute of
Body temperature during hypoxia gradually rose during hypoxia, but this gradual waning could be prevented by
the first three postnatal weeks to also reach the normoxic the adenosine antagonist 8-phenyltheophylline; 3-wk-old
value at P20 –P21 (847), consistent with a lesser contribu- animals, however, did not show this secondary decrease
tion of the thermogenic component to the hypoxic re- in tidal volume, whilst the adenosine antagonist did not
sponse at older age (see sect. IIC2). Maturation of the have any influence on the sustained rise in ventilation
mammalian respiratory network is reviewed in Refer- (212). In piglets 2–7 days of age, aminophylline prevented
ences 115, 186, 846. the secondary decrease in phrenic activity during hypoxia
(121). In summary, the secondary roll-off in neonates may
3. Depressing phase of the HVR be partly a manifestation of similar mechanisms that in-
duce hypoxic inhibition of FBM in the fetus on which a
The secondary roll-off in neonates may be associated postnatal influence of the peripheral chemoreceptors is
with pulmonary mechanics, a time-dependent decrease in superimposed. Note, however, that in many of the above
carotid body activity or sensitivity, impaired central inte- neonatal studies, the end-tidal PCO2 was uncontrolled. If in
gration of afferent carotid sinus nerve input, decreased small animals the HVR is measured at temperatures below
metabolism, increased CBF and, similar to the inhibition thermoneutrality (which is often the case), a fall in me-
of FBM, active inhibition by pontine and mesencephalic tabolism could well contribute to the secondary roll-off.
structures involving inhibitory neurotransmitters (re- In these cases, measurements of metabolism and body
viewed in Ref. 73). Muscle fatigue is unlikely to play a temperature would be very helpful (260). Other factors
significant role, since the phrenic nerve response in anes- such as the vigilance state, the severity of the hypoxic
thetized neonatal animals does also show a biphasic pat- stimulus, and other experimental conditions will also in-
tern, indicating that the secondary roll-off is a neurogenic fluence the magnitude of the depressing phase in the
mechanism (121, 249, 522). neonatal period.
In 5- to 10-day-old rabbit pups, brain stem transection
at the midbrain-pontine junction converted the hypoxia-
D. Developmental Plasticity of the HVR
induced depression in controls into sustained stimulation
in test animals (491). Focal cooling in the locus coeruleus
A definition of developmental plasticity of respira-
reversibly abolished the hypoxia-induced depression in 3-
tory control was provided by Carroll (115) as “long-term
to 8-day-old anesthetized lambs (522).
alterations in the structure or function of the respiratory
Available evidence also points to a role of the carotid
control neural network caused by experience during pre-
bodies in the depressing phase. The temporal response of
or postnatal formation of the respiratory control system.”
the sinus nerve discharge in kittens differed among stud-
Overwhelming evidence has identified the neonatal
ies, declining in some (116, 487) but sustained in others
environmental PO2 as a major factor influencing normal
(81). In 2- to 3-day-old lambs, a sudden hyperoxic stimulus
HVR development. Hypoxia and/or hyperoxia around the
during a hypoxic exposure had a larger effect after 3 min
time of birth and the first few weeks thereafter can dis-
than after 15 min, suggesting waning peripheral chemore-
turb normal development of the HVR that may persist
ceptor activity or impaired central integration (117). Ca-
until adulthood. The effects of perinatal chronic or inter-
rotid body denervation attenuated but not completely
mittent hypercapnia were less frequently studied; in the
abolished most of the secondary roll-off in 0- to 13-day-old
rat, neither the neonatal nor the adult HVR seems to be
rats (249), and in newborn lambs the subcoeruleus area
affected by neonatal hypercapnic exposures (43, 646).
showed increased activation in hypoxia after carotid body
denervation only (95).
1. Effects of neonatal chronic intermittent hypoxia
In neonates, adenosine, serotonin, prostaglandin,
GABA, opioids, catecholamines acting at ␣2-receptors, Term and preterm infants in particular are repeatedly
and other neuromodulators may all be involved in the exposed to hypoxia due to their irregular breathing pat-
depressing phase (e.g., Refs. 212, 340, 400, 462, 740; see tern with many apneas. Animal studies have now made
further references in Refs. 73, 285, 702). The strongest clear that within a critical time window of neonatal de-
case was made for adenosine, which, similar to the fetus, velopmental plasticity, chronic intermittent hypoxia can
in neonatal sheep reduces hypoxic ventilation both via induce life-long alterations in the control of breathing
adenosine A1A, but mainly adenosine A2A receptors (400). with potential pathophysiological consequences (617,
In newborn lambs showing a biphasic phrenic hypoxic 633, 827). In this context, the critical time window of

Physiol Rev • VOL 90 • APRIL 2010 • www.prv.org


682 LUC J. TEPPEMA AND ALBERT DAHAN

neonatal plasticity was defined as a period within the These distinct effects of various specific neonatal
developmental stage in which the organism is uniquely chronic intermittent hypoxia protocols on the augmenta-
sensitive to environmental perturbations that may alter tion phase of the HVR and mechanisms underlying neo-
the ultimate configuration of the neuronal network and natal respiratory plasticity are reviewed elsewhere (45,
associated functions (115, 632). In terms of respiratory 115, 515, 631, 632, 827). The effects on the depressing
control, it may imply that exposure to low or abnormally phase have not been studied extensively, but at least in
high oxygen (or CO2) tensions within this window can the rat, the secondary roll-off seems to be attenuated and
lead to life-long perturbations in respiratory control. related to increased nitric oxide synthesis in the caudal
The effects of repeated exposures to hypoxia can be brain stem (286).
quite complex and critically depend on the total duration We attribute the above divergence in study outcomes
of an intermittent stimulus, its frequency, the overall num- to the considerable variations in the applied stimulus
ber of cycles, and postnatal age which all appear to inter- level, cycle length, the amount of hypoxic episodes, and
act (633, 827). One reason for this interaction is that the the number of exposure days. Differences in methodology
HVR is build up of a series of sequential events with (e.g., plethysmography vs. pneumotachography) and de-
different lags, time constants, gains, and recovery times. velopmental critical time windows between species will
For example, hypoxia augments carotid body activity, also play a role (115). Interpretation problems due to
increases the discharge frequency in the carotid sinus chronic intermittent hypoxia-induced changes in nor-
nerve that results in the release of neurotransmitters in moxic ventilation and the employment of different meth-
the NTS, in turn leading to the release of neurotransmit- ods to normalize data are illustrated by the work of Ling
ters in the ventrolateral medulla etc., eventually causing a et al. (449) and Reeves et al. (633) in adult and neonatal
time-dependent recruitment of motoneurons. When a new animals, respectively. The poikilocapnic nature of the
stimulus is offered while one of the processes initiated by experiments is also a confounding factor. As shown in
the previous stimulus is still in its recovery time or even infants, kittens, and dogs, hypoxic hypometabolism can
(partly or fully) active, it is conceivable that the effects of be accompanied by a (sustained) fall in PaCO2 in the
both stimuli on motor output will differ (827). depressing phase despite the decrease in ventilation (93,
To investigate the effect of chronic intermittent hyp- 489, 528, 648, 652).
oxia on developmental plasticity of the HVR, a variety of The problem of measuring in a closed loop configu-
paradigms with different stimulus magnitude, length, and ration can be overcome by working with anesthetized
total duration and number of exposures were used in animals or ex vivo carotid body preparations. Rat pups
several species such as mice (197), rabbits (789), piglets subjected to chronic intermittent hypoxia soon after birth
(828), and rats (reviewed in Ref. 286). In the rat studies, (15 s in 5% O2 followed by 5 min in 21%, 9 episodes/h
the age of the animals varied from 0 to as many as 30 days during 16 h) showed an augmented ex vivo hypoxic ca-
and more, well beyond the critical time window for de- rotid body sensitivity that persisted until adulthood and
velopmental plasticity (115, 286). The results of these was associated with hyperplasia of glomus cells (585).
studies in different species are equivocal, varying from a Carotid bodies from neonates are much more sensitive to
reduced (45, 497, 633, 635, 828 and references therein), chronic intermittent hypoxia than those of adult animals
unaltered (197, 676) to an enhanced (367, 585, 598) HVR and the effects in neonatal animals are permanent (585).
and/or carotid body sensitivity. In the rat, blunting of the Recently, Pawar et al. (586) showed that in neonatal rats
HVR was reported to persist at 1 mo after cessation of the chronic intermittent hypoxia induced an augmented basal
stimulus paradigm (635). As shown in neonatal piglets, release of endothelin-1 (ET-1) in normoxia, upregulation
the background PCO2 during the hypoxic episodes may of ETA receptor mRNA, and an enhanced carotid body
have an important influence: a poikilocapnic paradigm sensitivity to ET-1. In another study, a chronic intermit-
(10% O2) blunted the HVR, but hypercapnic chronic inter- tent paradigm led to decreases in both the HVR and
mittent hypoxia (6% CO2, 10% O2) caused an increase isocapnic hypoxic phrenic response (635), which would
(828, 829). In instrumented anesthetized piglets, 11 peri- be more in line with the effect of chronic hypoxia. The
ods of recurrent hypoxia blunted the phrenic nerve re- paradigm consisted of alternating room air with 10% O2
sponse to acute hypoxia in young (2–10 days) but not every 90 s (12 h/day, during 30 days), raising the question
older (2–3 and 8 –10 wk) animals (509; note however that whether after a short period of low oxygen, 90 s is suffi-
in all recovery periods, these animals breathed 100% O2 cient to completely restore tissue oxygenation to normal,
which may have an effect by itself, see below). In most possibly resulting in chronic hypoxia rather than chronic
(but not all) studies, normoxic ventilation increased (599, intermittent hypoxia.
631– 633) and, depending on the animal’s age at the onset In conclusion, recent data from ex vivo carotid bod-
of the hypoxic stimulus, this effect was permanent (the ies indicate an increase in sensitivity by neonatal chronic
first two postnatal weeks were most efficient in this re- intermittent hypoxia. Technical limitations and method-
spect; Refs. 632, 633, reviewed in Ref. 45). ological differences are probably responsible for the fact

Physiol Rev • VOL 90 • APRIL 2010 • www.prv.org


THE VENTILATORY RESPONSE TO HYPOXIA IN MAMMALS 683

that as yet these important results could not be consis- currents that would tend to increase the excitability of
tently confirmed at the level of minute ventilation. Stan- type I cells were also reported (315, 316, 726, 727).
dardizing protocols, uniform quantification of HVR data Neonatal chronic hypoxia will also affect or delay
(combined with metabolic data), and blood gas measure- normal maturation of the central respiratory neuronal
ments may help to reach consensus on the effects of network. For example, exposing newborn rat pups to 6
neonatal chronic intermittent hypoxia. Intermittent expo- days of chronic hypoxia resulted in a decreased norepi-
sures in which hypoxic episodes follow each other so nephrine turnover in the caudal part of the noradrenergic
closely in time that episodic reoxygenation is incomplete A2 region in the NTS (an important projection area of
or does not occur at all may be interpreted as chronic (see carotid body afferents) at the age of 8 wk, suggesting
also ref 827). long-term effects of the neonatal challenge (721). NMDA
receptors in pons and caudal medulla of rats raised in 10%
for 4 –5 wk from birth were downregulated (248). After
2. Effects of neonatal chronic hypoxia
they were raised in 13–15% O2 from birth to 5–10 wk
Cats, sheep, and rats subjected to hypoxia (10 –15% postnatally, anesthetized rats showed normal (i.e., similar
O2) from birth until 24 h before they were tested at ages to age-matched normoxic controls) isocapnic hypoxic si-
varying from 5 days to 10 wk showed a markedly reduced nus nerve responses, but their HVR remained blunted
HVR, mainly caused by a depression of the augmentation (207). Adult anesthetized rats, kept in 10% O2 during their
phase (39, 42, 42, 45, 207, 567, 703 and references therein). first postnatal week, had a normal phrenic response to
In rats and sheep exposed to neonatal hypoxia for 1–2 isocapnic hypoxia, but their HVR was reduced (43). The
wk, blunting of the HVR persisted for at least 5–12 wk (39, authors of the latter two studies suggested that the sus-
476, 567, 703), which in the rat was sex specific (only ob- tained reduction in HVR after neonatal CH may originate
served in males; Refs. 42, 476). The effect of neonatal hyp- downstream from the phrenic motoneuron pool. Abnor-
oxia on normoxic ventilation is variable: in some studies, a mal and/or retarded lung growth may then provide a
potential explanation (493). Normal maturation of the
long-lasting increase was observed (527, 566, 567) while it
HVR is also associated with an age-dependent attenuation
was absent in others (42, 476, 703). At the age of 12 wk, both
of the, centrally governed, depressing phase (see above).
male and female rats displayed an impaired ventilatory ac-
In rat puppies, chronic hypoxia from birth delays this
climatization to chronic hypoxia (2 wk in 10% O2) when they
decrease (207). Other adaptations observed after neonatal
were raised in hypoxia until postnatal day 10 (476).
chronic hypoxia in rats are an increase in hematocrit,
Initially, in the first weeks of maturation the blunted
decrease in metabolism, and an enlarged alveolar gas
HVR in chronically hypoxic neonates may be due to a
exchange surface perhaps tending to compensate for a
delayed carotid body resetting and/or to an attenuation of
reduced HVR (527, 531 and references therein). Exposing
the maturation of the O2-CO2 interaction (301, 303, 437,
newborn piglets to hypobaric hypoxia during 3 days
730, 826; further references in Ref. 115). Single fibers of caused pulmonary hypertension and prevented the rapid
the carotid sinus nerve from rat puppies that were raised remodeling of the cytoskeleton of pulmonary arterial smooth
in 9% O2 from the first or second postnatal day until 3– 4 muscle cells that normally contributes to the postnatal de-
wk later, showed a 50% lower peak response to severe crease in pulmonary vascular resistance (298).
hypoxia than controls (186). Both at the ages of 3 and 8 Taken together, neonatal chronic hypoxia-induced blunt-
wk, rats exposed to hypoxia for 6 days in their neonatal ing of the HVR has multifactorial causes involving plastic
period had increased carotid body dopamine and norepi- changes in the carotid bodies, the central nervous system,
nephrine levels (721). Glomus cells from neonatal rats and possibly also lung mechanics and pulmonary vessels.
reared in 12% O2 during 1–3 wk demonstrated a reduced
rise in intracellular [Ca2⫹] in hypoxia compared with cells
3. Effects of postnatal chronic hyperoxia
from normoxic aged-matched controls, and this impaired
maturation partially recovered 1 wk upon return to nor- Earlier experiments in fetal sheep suggested that
moxia (731). Rat puppies born and raised in 10% O2 for brief prenatal hyperoxia would hasten resetting of the
9 –14 days had a substantially reduced HVR and possessed carotid bodies explaining the reported increased carotid
type I cells that displayed a reduced expression of maxi-K body O2 sensitivity in these animals (82). This contrasts
channels and failed to depolarize in hypoxia (854). In with many studies showing a reduced HVR after chronic
neonatal and juvenile type I cells from rats cultured in 6% neonatal hyperoxia. For example, kittens breathing 30%
O2 for 12 days, maxi-K channels were downregulated O2 during their first 13 days showed a reduced early-phase
(354). These studies suggest that the neonatal chronic HVR right away after this exposure (301). In recent years,
hypoxia-induced HVR reduction is at least partly due to an much attention was focused on the effects of immediate
impaired carotid body function. Not all data, however, postnatal chronic hyperoxia on the HVR on the longer
support this because changes in sodium and potassium term, extending to the adult age (reviewed in Refs. 39, 45,

Physiol Rev • VOL 90 • APRIL 2010 • www.prv.org


684 LUC J. TEPPEMA AND ALBERT DAHAN

115). Here we will briefly highlight some main findings. Most recordings are from quiet sleep when breathing is
The effect of neonatal hyperoxia on adult normoxic base- predominantly under metabolic control and somewhat
line ventilation appears variable, with some studies show- less unstable than in active sleep, with less frequent
ing an increase and others reporting no change (refer- arousals. Control of end-tidal gases is unusual so that the
ences in Ref. 45). The consequences for the HVR and magnitude of the HVR reflects the closed-loop properties
hypoxic responses of the phrenic and carotid sinus nerves of the respiratory system.
at the adult age can be summarized as follows: 1) chronic With the exception of very small premature infants
neonatal hyperoxia causes a decrease in hypoxic sensi- showing a sustained “fetal-like” decrease in ventilation
tivity at all these three integration levels that persists into (15), most term and preterm infants exhibit a biphasic
adulthood (71, 246, 449). 2) The critical time window to HVR upon exposure to 15% O2 during 5 min, much similar
induce this effect, which is dose dependent and can al- to that in neonatal animals. The secondary fall appears to
ready be induced by 30% O2, lies within the first 2 wk; be due mainly to a fall in frequency while in most cases
longer exposures cause no additional depression (40, 41, tidal volume shows a sustained increase, sometimes after
71). 3) The attenuation of the HVR can be limited by an initial overshoot (138, 139, 489, 552, 637, 648, 651, 652).
alternating the chronic hyperoxia with periodic hypercap- With some exceptions (e.g., Refs. 139, 723) early in devel-
nia or by replacing chronic by intermittent hyperoxia, opment the depressing phase of the HVR is associated
suggesting that intermittently increasing carotid body ac- with a sustained decrease in end-tidal PCO2 (489, 637, 648),
tivity reduces the effect (43). 4) Central (carotid sinus to similar to neonatal animals, and this is possibly related to
phrenic nerve) integration remains unimpaired, suggest- a fall in metabolism (147, 332). During maturation, the
ing that the depressing effect originates from the carotid decrease in frequency gradually becomes less dramatic,
bodies (450). 5) The carotid bodies show degeneration, and with advancing age, tidal volume tends to remain
hypoplasia, changes in membrane properties, and a re- elevated (489, 648, 651).
duced number of unmyelinated fibers that altogether lead The normal developmental time course of the HVR in
to reduced O2 sensitivity of glomus cells and the carotid humans appears difficult to determine. Breathing in term
body in vitro (189, 220, 622, 838). 6) Petrosal ganglion and especially preterm infants is characterized by irregu-
neurons also show degenerative changes (loss of tyrosine larities and apneas (625) that have been related to imma-
hydroxylase expression; Ref. 220). ture brain stem function and connections (318) and also
A recent study focused on the time course of alter- to a relatively low PaO2 leading to brisk hypoxic respon-
ations in hypoxic responses of glomus cells and carotid ses, hypocapnia, and central apneas (552). Consequently,
sinus nerve afferents from rats exposed to chronic hyper- when occurring within the critical developmental time
oxia starting on postnatal day 7 and lasting for 1, 3, 5, 8, window, both the resulting chronic and intermittent hyp-
and 14 days, respectively. On postnatal day 8 (so after 1 oxia or a combination may hinder a normal development
day of hyperoxia), the secretory response of glomus cells of the HVR. Despite the quite different term durations in
and the nerve response to hypoxia were increased, but rodents and humans, the animal data raise the question as
beyond exposure days 3–5, both responses were reduced, to whether oxygen therapy in the neonatal period may
and this reduced sensitivity persisted until at least 1 wk have unwanted side effects with regard to the develop-
after exposure (187). Are these effects reversible once ment of normal O2 and CO2 reflex sensitivities. By itself,
animals have reached the adult age? Bisgard et al. (72) preterm birth exposes the newborn to an environment
reported that sustained hypoxia at adult age (3–5 mo) that is hyperoxic relative to postconceptional age.
only sensitized the carotid sinus nerve when the chronic Whether this is related to the lower HVR of preterm
neonatal hyperoxia did not outlast 2 wk. However, an- infants compared with age-matched controls remains to
other study showed recovery from the deleterious effects be investigated. The higher prevalence of the sudden
of chronic hyperoxia during the first month after birth at infant death syndrome (SIDS) in preterm compared with
the age of 4 mo (247). An antioxidant rich diet during the term babies has been related to relative immaturity of the
neonatal hyperoxia did not prevent an impaired HVR at cardiorespiratory neuronal circuitry resulting in failure of
adult age (44), not supporting ROS-mediated toxicity. Fu- ventilatory and arousal responses (518).
ture studies are warranted to further examine the influ- Of practical interest when studying the HVR in both
ence of dose and duration of neonatal chronic hyperoxia neonatal animals and infants is its dependency on the
and how an impaired HVR at adult age might be restored. vigilance state (317, 319, 472, 648, 651, 729). Most HVR
data were collected in quiet sleep, but potential influences
of arousal are not always taken into consideration (648).
E. The HVR in Term and Preterm Babies In a longitudinal study in healthy term infants, the HVR
was reported to mature between 2–5 wk and 5– 6 mo of
In most cases, the HVR in young babies is measured age in quiet but not active sleep, especially in children
by exposing them to 15% O2, seldom with addition of CO2. that did not arouse by the stimulus (15% O2 during 5 min

Physiol Rev • VOL 90 • APRIL 2010 • www.prv.org


THE VENTILATORY RESPONSE TO HYPOXIA IN MAMMALS 685

or 85% SpO2, see Fig. 1). Even at the age of 5– 6 mo, highest blood flow-to-metabolism ratio in the body and,
ventilation did not exceed prehypoxic control after a both anatomically and biochemically, are extremely com-
5-min hypoxic challenge, and this was considered an im- plex organs (278, 280 and references therein). Not only do
mature HVR. However, taking into account that the pro- they possess cells specialized in oxygen sensing, but in
tocol was clearly poikilocapnic and that the HVR was fact, they appear to be polymodal and sensitive to a
identical to that after 2–3 mo and very similar to the variety of stimuli such as pressure, hypercapnia, acidosis,
poikilocapnic HVR in adult humans (cf. Fig. 1) leads us to hyperkalemia, hyposmolality, circulating hormones, hy-
suggest that maturation of the HVR in term infants may be perthermia, hypoglycemia, and numerous pharmacologi-
complete at ⬃2 mo. Direct assessments of carotid body cal agents (reviewed in Refs. 416, 558).
sensitivity with brief exposures to 100% O2 (Dejours test; The two main cell types in the carotid bodies are
Ref. 13) or by offering alternating breaths containing glomus (type I) cells, having many similarities with sen-
room air and 16% O2 (108, 843) pointed to full maturation sory neurons and thought to be oxygen sensitive and
of the peripheral chemoreflex within the first few weeks sustentacular (type II) cells with a glialike appearance
and possibly even the first 48 h after birth (13; note, (278). Sensory neurons with cell bodies in the petrosal
however, that data obtained with Dejours tests should be ganglia send peripheral axons to the carotid bodies that
interpreted with caution because the peripheral chemo- run with the carotid sinus nerve, a side-branch of the
receptors may still display considerable activity in hyper- glossopharyngeal nerve, and have nerve endings closely
oxia). That in term infants the O2-CO2 interaction has oriented to type I cells. Central axons of these sensory
been reported to emerge at an age somewhere between neurons enter the brain stem with the glossopharyngeal
day 2 and week 8 (no testing after ⬃2 wk) is not incon- nerve and terminate in caudal subnuclei of the NTS that
sistent with this picture (723). have extensive connections with the brain stem respira-
tory network (references in Refs. 232, 335).
Hypoxia is followed by release of neurotransmitters
III. PERIPHERAL AND CENTRAL MECHANISMS
by type I cells that act on the afferent endings of the
MEDIATING THE HYPOXIC VENTILATORY
carotid sinus nerve to increase impulse traffic to the brain
RESPONSE IN ADULT MAMMALS
stem (278). Neurotransmitter release is the result of a rise
in intracellular [Ca2⫹] following depolarization of the
A. Carotid Body Mechanisms in Oxygen Sensing membrane potential of type I cells. The mechanisms un-
derlying the entire signal-transduction cascade in type I
The ventilatory response to hypoxia is initiated by cells from hypoxia to the release of neurotransmitters
the carotid bodies, with the aortic bodies playing only a have been the focus of numerous studies over the last two
secondary role, perhaps with the exception of conditions decades. This has resulted in the acceptance, albeit not
wherein the carotid bodies are functionally eliminated without controversies, of a framework in which hypoxia
(278). The carotid bodies, bilaterally located in the carotid leads to inhibition of ion currents through potassium
bifurcations at the port of the brain circulation, have the channels in the cell membrane resulting in membrane
V

*
.
V

** *
**

FIG. 1. Poikilocapnic hypoxic ventilatory responses in young babies and adults. A: maturation of the ventilatory response to 15% O2 in 15 term
infants between the ages of 2–3 wk (■), 2–3 mo (E), and 5– 6 mo (Œ). *Significantly different with levels after 2–3 mo. **Significantly different when
compared with 5– 6 mo. Measurements were performed in quiet sleep, and the children did not arouse from the stimulus. B: poikilocapnic hypoxic
ventilatory responses in 10 healthy young adults upon a step decrease to and end-tidal PO2 of ⬃44 Torr. Note that both after 2 and 5 mo the
poikilocapnic responses are qualitatively similar to the adult. [Adapted from Richardson et al. (648) and Steinback and Poulin (728).]

Physiol Rev • VOL 90 • APRIL 2010 • www.prv.org


686 LUC J. TEPPEMA AND ALBERT DAHAN

depolarization, influx of extracellular Ca2⫹ via voltage- our insight into the way type I cells couple changes in
gated (mainly L and P/Q type) Ca2⫹ channels, and release PaO2 to alterations in potassium channel activity.
of neurotransmitters (278, 465, 467, 469, 662, 835, see Fig. 2).
Overall, the main focuses of attention were the following: 1. Hypoxia decreases the open probability of
1) the element(s) that initiate(s) the cascade, in other potassium channels
words the identity of the intrinsic O2 sensor(s); 2) the
It is now widely accepted that hypoxia decreases the
cause(s) by which the resting membrane potential chan-
open probability of potassium channels. A first report in
ges, the identity of potassium channels involved in this,
1998 showing hypoxic inhibition of voltage-gated potas-
and the link of these channels with the intrinsic O2 sen-
sium channels in rabbit type I cells (466) was followed by
sor(s); and 3) the identity of the neurotransmitters that
numerous other studies, but here we only highlight major
are released by type I cells and excite afferent nerve
findings. Three main categories of O2-sensitive potassium
endings. With regard to the identity of the contributing
channels in type I cells have been described. 1) Channels
potassium channels and excitatory neurotransmitters, that produce voltage-gated K⫹ currents (Kv channels), of
there seems to be a reasonable consensus, although some which several isoforms have been suggested to be O2
aspects remain controversial. These issues are covered by sensitive (reviewed in Ref. 470). 2) Large-conductance K⫹
several recent reviews and are briefly referred to below channels that are activated by both voltage and intracel-
but not discussed in detail. Recent studies on the identity lular Ca2⫹ (KCa2⫹, maxi-K, or BK channels; reviewed in
of the O2 sensor(s), and their link with potassium chan- Ref. 594). 3) Voltage-independent two-pore domain back-
nels, to be discussed below in more detail, have improved ground (K2P) channels that contribute to the resting mem-
brane potential and of which several isoforms are de-
tected in glomus cells (861): K2P3.1 (TASK-1), K2P4.1
(TRAAK), K2P5.1 (TASK-2), and K2P9.1 (TASK-3 ). TASK-1
(TWIK-1-related acid-sensitive K⫹ channel; TWIK channel,
tandem pore domain weak inward rectifying potassium
channel) is the most likely candidate to operate as O2-
sensitive background K⫹ channel (reviewed in Refs. 98,
196, 788). Different species appear to have distinct types
of oxygen-sensitive potassium channels. For example, in
rabbits, Kv isoforms are the predominant types, but in the
rat carotid body, significant roles of both maxi-K and
TASK-1 have been demonstrated (98, 278, 594 for further
references).

2. Carotid body neurotransmitters


The carotid bodies contain a variety of neurotrans-
mitters that are either stored in vesicles or appear as
by-products of enzymatic processes in the cytosol where
they can influence the signal-transduction cascade (426,
558). The first category comprises the biogenic amines
acetylcholine, dopamine, norepinephrine, serotonin, and
histamine; (neuro)peptides such as substance P, endothe-
lins, and angiotensin II; and amino acids such as GABA
and the purine ATP (see Refs. 394 and 438 for histamine
and Ref. 426 for further references). Nitric oxide (NO) and
carbon monoxide (CO) are important representatives of
the second category, synthesized with the aid of the
FIG. 2. Stimulus transduction cascade in oxygen-sensitive cells in heme-containing enzymes NO synthase and heme oxygen-
the carotid body in hypoxia. A decrease in oxygen tension in oxygen- ase, respectively. For an extensive discussion of the role
sensitive carotid body type I cells is detected by primary oxygen sensors of all these neurotransmitters and modulators, the reader
that rapidly communicate with potassium channels (mitochondria could
also sense changes in PO2 and communicate with potassium channels). is referred to several excellent reviews (425, 426, 558, 621,
This leads to closure of these channels, which results in membrane 701, 868). With regard to the overall stimulus-transduction
depolarization, influx of Ca2⫹ via voltage-dependent Ca2⫹ channels, and cascade, there seems to be consensus, albeit not without
subsequently to a release of neurotransmitters, whereby acetylcholine
and ATP are thought to excite the afferent nerve endings of fibers that controversy, that hypoxia induces the release of acetyl-
run in the carotid sinus nerve. NTS, nucleus of the solitary tract. choline and ATP and that these neurotransmitters are

Physiol Rev • VOL 90 • APRIL 2010 • www.prv.org


THE VENTILATORY RESPONSE TO HYPOXIA IN MAMMALS 687

responsible for the excitation of the afferent nerve end- hibited by low PO2 (593, 853). In carotid body cells iso-
ings (350, 559, 644, 645, 799; see Refs. 558 and 868 for lated from neonatal (8 –11 day) rats, substrates of HO-2
further references). (heme and NADPH) activated native maxi-K channels.
Since increased HO-2 activity generates CO, that by itself
3. O2 sensing in the carotid bodies activates maxi-K channels, it was suggested that the en-
zyme acts as an O2 sensor (844). How CO specifically
Over the last decades two main hypothesis of O2
interacts with maxi-K channels is unknown.
sensing have been at the foreground: 1) ion channels, and
At an age ⬍3 mo, mice deficient in HO-2 (HO-2⫺/⫺)
potassium channels in particular, as the initiators of the
show a carotid body phenotype typical for oxidative
transduction cascade, referred to as the “membrane hy-
stress with enlargement of the organ and downregulation
pothesis”; and 2) heme proteins as oxygen sensors, re-
of maxi-K channels and a marked upregulation of tyrosine
ferred to as the “metabolic” or “mitochondrial” hypothesis
hydroxylase, suggesting that HO-2 could be a potential O2
(278, 279, 430, 620).
sensor (574, 854). Type I cells isolated from HO-2⫺/⫺ mice
The role of potassium channels in type I cell mem-
have a normal catecholamine secretory response to hyp-
brane depolarization is now well established (see above).
oxia, which may suggest that HO-2 is not a key O2 sensor
In excised membrane patches, this hypoxia-induced de-
and maxi-K channels are not required for a normal secre-
polarization is still present, but in the course of time a
tory response to occur (574). Carotid sinus nerve record-
significant rundown occurs, indicating that O2 sensing
ings were not performed in this study, so it remains to be
may be a membrane-delimited process requiring cytosolic
seen whether a normal secretory response also predicts a
factors to be preserved for longer time periods (800, 842).
normal nerve response. Although at the level of type I
Whether or not potassium channels possess structural
cells the release of catecholamines is considered a mea-
components required for direct oxygen sensing, many of
sure of type I cell activation (e.g., Ref. 574), changes in
them appear to be redox sensitive (392, 409, 468, 603), but
their release do not always mirror alterations of carotid
it is unlikely that hypoxia inhibits the current through
sinus nerve activity (100, 185, 350, 351 and references
these channels by oxidizing or reducing functional chan-
therein). It is interesting that HO-2⫺/⫺ mice showed a
nel protein components (reviewed in Ref. 282). Generally,
reduced poikilocapnic HVR despite the fact that, com-
reductants and oxidants decrease and increase, respec-
pared with wild types, they were challenged with a much
tively, the open probability of rapidly inactivating O2-
more severe hypoxic stimulus because of their lower
sensitive potassium channels in particular, but the effects on
initial arterial PO2 (4). So it seems reasonable to conclude
maxi-K channels are more variable (references in Ref. 282).
that to date the identity of HO-2 as an oxygen sensor
For several decades, mitochondrial cytochromes
remains unsettled. Finally, concerning the participation of
were considered potential O2 sensors providing the link
maxi-K channels in oxygen sensing, it is worth noting that
between type I cell metabolism and afferent nerve activ-
these channels tend to be activated at a membrane poten-
ity, but to date, their exact role is still a subject of discus-
tial more positive than that seen in type I cells. Therefore,
sion (recently reviewed in Refs. 122, 295, 296, 415, 465,
in order for the maxi-K channel to be the initial sensor, it
837). Generally, with a few exceptions, inhibitors of mi-
would have to be open at or near the glomus cell resting
tochondrial complex proteins inhibit currents through
membrane potential.
potassium channels, depolarize type I cells, and can oc-
clude the hypoxic response (references below). However,
inhibition of the cytosolic heme-containing enzymes 2. NADPH oxidase
NADPH oxidase, heme oxygenase 2, and NO synthase
Acker and co-workers (2, 3) suggested a role of the
may also have these effects and may thus also serve as
membrane-bound extra-mitochondrial enzyme complex
candidate O2 sensors.
NADPH oxidase using NADPH as an electron donor that
produces O⫺ 2 in proportion to PO2 and proposed that ROS
1. Heme oxygenase 2
would activate nearby potassium channels in normoxia
After demonstrating the presence of heme oxygenase but less so in hypoxia when NADPH oxidase activity
2 (HO-2) in rat and cat carotid bodies, Prabhakar et al. would be reduced (reviewed in Ref. 3). Several NADPH
616) showed that blockade of HO-2 increased afferent oxidase isoforms were identified in carotid body cells
carotid sinus nerve discharge in the cat that was revers- including those containing the subunits gp91phox and
ible with CO. Recently, Kemp, Peers, and co-workers p47phox (references in Ref. 3). Later, NADPH oxidase 4
(844) showed that HO-2 coimmunoprecipitated with het- was identified as one of these isoforms in glomus cells
erologously expressed maxi-K channels in human embry- (184, 276). Diphenyliodonium (DPI), an unspecific NADPH
onic kidney cells and that blocking the enzyme with small oxidase inhibitor, increased basal carotid sinus nerve activ-
interference RNA abolished channel inhibition by low PO2 ity in the rat and inhibited the hypoxia-induced increase in
(844). In rat carotid body cells, maxi-K channels are in- nerve activity (146). In an in vitro perfused carotid body

Physiol Rev • VOL 90 • APRIL 2010 • www.prv.org


688 LUC J. TEPPEMA AND ALBERT DAHAN

preparation from the rat, hypoxia reduced a cytochrome ing blood flow and/or exerting direct effects on type I cells
b558 isoform whilst the specific NADPH oxidase inhibitor (references in Refs. 109, 426, 619).
4-(-2-aminoethyl)-benzenesulfonyl fluoride (AEBSF) increased
carotid sinus nerve activity and occluded the hypoxic 4. Mitochondrial complex proteins and
response (424). Transfection of TASK-1 channels in hu- complex-derived ROS
man embryonic kidney (HEK) cells endogenously ex-
pressing NADPH oxidase 4 resulted in colocalization of The role of ROS in oxygen sensing is the subject of a
both in the plasma membrane; overexpression of the long debate with many controversial data and different
enzyme enhanced the hypoxia-induced TASK-1 current views. Partly, the lack of consensus is caused by the
inhibition in these cells, but eliminating the enzyme abol- difficulty of measuring ROS, by local differences in their
ished the hypoxic response (439). production in various microdomains of the cell (cell mem-
In the HEK cell model, the concept of ROS-mediated brane, cytosol, nucleus, mitochondria), and by differ-
TASK-1 activation was recently challenged because it ap- ences between cell types (see Refs. 276, 282 for refer-
peared to be determined by O2 binding to the heme moi- ences).
ety of the enzyme rather than by the production of ROS Generally, inhibitors and uncouplers of the electron
(582). Other evidence did not support a crucial role for transport chain inhibit background potassium channels
NADPH oxidase. For example, in both rat and rabbit and cause membrane depolarization, voltage-gated Ca2⫹
carotid body cells, specific inhibitors of the enzyme did entry, and neurosecretion. They also stimulate the carotid
not affect catcholamine release in normoxia or its in- bodies and, provided the inhibition of electron flux is
crease in hypoxia; in addition, DPI seemed to have effects complete, occlude the hypoxic response (851). Originally
independent from NADPH oxidase inhibition (561). Com- it was thought that hypoxia would reduce cytochrome c
pared with wild types, adult mice deficient in gp91phox do activity of complex IV of the electron transport chain,
not show altered responses of ventilation, carotid sinus which then would cause mitochondrial depolarization
nerve activity, potassium currents, or intracellular [Ca2⫹] and Ca2⫹ release (194). Later the rise in intracellular
to hypoxia (306, 310, 667). Very interesting results were [Ca2⫹] appeared to depend on extracellular Ca2⫹, and the
obtained from p47phox⫺/⫺ mice: 1) type I cells from role of mitochondria as oxygen sensors lost support (99,
knockout mice did not show the hypoxia-induced in- 465, 802). In addition, several blockers of the electron
crease in [ROS] that was observed in cells from wild transport chain (rotenone in particular) have nonspecific
types, but displayed an enhanced depression of potassium effects on potassium channels (573).
currents and a substantial greater rise intracellular A modulating role of mitochondrial ROS, and in this
[Ca2⫹]; and 2) both the HVR and hypoxic carotid sinus context the influence of cellular redox state on oxygen
nerve responses in knockout mice were substantially sensing was investigated by Gonzalez et al. (281). The
greater than in wild types (184, 310, 678). From these cellular redox state was assessed by measuring the ratios
observations it was concluded that hypoxia induces a rise of reduced (GSH) to oxidized glutathione (GSSG), from
in [ROS] in type I cells with NADPH oxidase as their which the redox potential (EGSH) can be calculated. EGSH
source and that these ROS modulate (increase) the open is considered the main cellular buffer of ROS and repre-
probability of (Kv) potassium channels, rather than being sentative for the general redox environment of cells (281).
directly involved in oxygen sensing. Briefly, Gonzalez et al. (281) found the following. 1) In
calf and rabbit carotid body cells, hypoxia did not reduce
3. NO synthase EGSH. 2) The antioxidant and GSH precursor N-acetylcys-
teine increased EGSH in normoxia in these cells without
At least two NO synthase (NOS) isoforms have been eliciting an increased catecholamine release. 3) N-acetyl-
identified in the carotid bodies: 1) NOS-1 (neuronal NOS) cysteine also did not alter the enhanced neurotransmitter
located in sensory and autonomic nerve fibers innervating release induced by hypoxia and high extracellular [K⫹].
vessels and lobules of glomus cells and 2) endothelial 4) In rat carotid body cells, specific inhibition of mito-
NOS (NOS-3; references in Refs. 109, 426). Aspecific NOS chondrial complexes I, II, III, and IV all reduced EGSH,
inhibition increases basal carotid sinus nerve discharge and, with the exception of the irreversible complex II
and enhances hypoxic carotid body sensitivity and the inhibitor 3-nitropropionate, decreased cellular ATP levels
HVR, whilst NO donors do the opposite (290, 619 for and increased catecholamine release. N-acetylcysteine pre-
further references). Whether NOS is activated or inhibited vented these changes in redox state without influencing
during hypoxia is controversial (references in Ref. 426). the effects on [ATP] and neurosecretion. 5) The mito-
NOS-1⫺/⫺ mice show an enhanced hypoxic carotid body chondrial uncoupler dinitrophenole activated rat carotid
sensitivity and HVR, supporting an inhibitory role of NO body cells without affecting [ATP] and EGSH (275, 281,
(389). NO may modulate carotid body sensitivity by alter- 282, 679).

Physiol Rev • VOL 90 • APRIL 2010 • www.prv.org


THE VENTILATORY RESPONSE TO HYPOXIA IN MAMMALS 689

The conclusion from these studies by Gonzalez et al. drial hypopolarization (194), has a similar stimulatory
(281) is that oxygen sensing in carotid body cells occurs effect on type I cells as mitochondrial complex inhibitors
independently from changes in cellular redox state. In and uncouplers which cause mitochondrial depolariza-
fact, this was supported at the level of afferent nerve tion (851). Despite this, the idea of heme-containing oxy-
discharges in superfused rat carotid bodies (666). How- gen sensors with different O2 affinities in distinct mi-
ever, given the fact that N-acetylcysteine restores the crodomains of the cell, enabling it to adapt to changes in
redox state but does not prevent ROS production per se, cellular PO2 profile (“oxygen redirection”), is interesting
the question now arises whether mitochondrial ROS may (837 for references).
play any (indirect) role, for example, by influencing ATP In rat PC12 cells, hypoxia produced a selective and
production. As explained in insightful reviews by Gonza- fast downregulation of complex I mRNA. This suggested
lez and co-workers (276, 282), there is much controversy that, apart from being involved in the flow of electrons
as to whether hypoxia decreases or increases mitochon- through the electron transport chain and as such may
drial [ROS]. Briefly, these authors provided reasonable indirectly contribute to rapid changes in [ATP], complex I
arguments why, unlike some other cell lines, type I cells may play an important role in adapting mitochondrial
most likely do not increase mitochondrial ROS produc- respiration to prolonged periods of hypoxia (606). In
tion in acute hypoxia and why the limited observed rise in chronic intermittent hypoxia, carotid body complex I ac-
cellular [ROS] in type I cells is most likely due to in- tivity also appeared to be reduced (596).
creased NADPH activity (310). Yamamoto et al. (860) also Mice heterozygous for succinate dehydrogenase sub-
observed an increase in [ROS]. Maintenance of the redox unit D (SDHD⫹/⫺), one of the anchoring proteins of com-
state in the face of increased ROS production via this plex II (607), show mild carotid body hypertrophy and
pathway would occur by an increased production of hyperplasia and an enhanced catecholamine release by
NADPH (reducing equivalent) along the hexose mono- type I cells in normoxia. These features are associated
phosphate pathway (276, 310). Mitochondrial inhibitors with a decrease in K⫹ conductance (due to a a decreased
with divergent effects on local ROS production all had Ca2⫹ sensitivity of maxi-K channels) and a persistent
2⫹
stimulating effects on type I cells and occluded the hyp- Ca influx; the hypoxia-induced CA release was enhanced
oxic response to hypoxia, suggesting no crucial role of in these animals, but not significantly (607). Thus these
mitochondrial ROS on O2 sensing (851). mice do not seem to support a crucial role of complex II
An absence of a direct involvement of ROS and the in O2 sensing, and this may fit the finding that the irre-
cellular redox state in oxygen sensing does not preclude versible complex II inhibitor 3-nitropropionate did not
a modulating effect of oxidants and reductants. In the alter the catecholamine release and ATP content of rat
model proposed by He et al. (310) and Dinger et al. (184), carotid body cells (275). However, carotid body-sinus
ROS, generated by NADPH oxidase activity, facilitates nerve preparations from SDHD⫹/⫺ mice were not studied.
potassium channels. In rat type I cells, inhibition of the Human carriers of a missense mutation within the SDHD
cytochrome P-450 enzyme system using NAD(P)H as elec- gene show an altitude-related phenotype (20). Thus it
tron donor prevents hypoxia-induced inhibition of potas- remains to be seen whether there may exist a relation
sium channels (304). As suggested by Kline et al. (389), a between complex II activity and oxygen sensing (for fur-
rise in [ROS] could decrease the bioavailability of NO, ther discussion and references, see sect. IV).
increasing hypoxic sensitivity. Mice deficient in neuronal
NOS have an augmented HVR (389). 6. Mitochondria and the link between metabolism and
potassium channel activity
5. Individual mitochondrial complexes
Failure to show an indispensable role of mitochon-
To date, the identification of one single mitochon- drial ROS in O2 sensing, and the fact that it has been
drial complex protein as the oxygen sensor in type I cells elusive to identify one of the electron transport chain
has been elusive. Light absorption photometry has iden- complexes as the O2 sensor that initiates the entire trans-
tified a cytochrome, a592, as a unique component of ca- duction cascade, does not preclude a mitochondrial role
rotid body cytochrome-c oxidase (complex IV) with a because O2 sensing could be a direct or indirect function
redox sensitivity that depends on the PO2 in a nonlinear of mitochondrial energy metabolism and ATP synthesis
fashion (3, 733). By inducing a shortcut of electron flow (851). Mitochondrial complex inhibitors and uncouplers
within complex IV, this low O2 affinity cytochrome would have variable effects on [ROS], but two effects that they
contribute to mitochondrial depolarization and intracel- have in common are that they decrease ATP levels and
lular Ca2⫹ regulation. However, the intracellular [Ca2⫹] activate type I cells (276, 851), perhaps with the exception
response to hypoxia depends on extracellular, not intra- of specific complex II blockers (275).
cellular, calcium stores (99, 465, 802), whilst the ATP Recently, cellular energy status was linked to cell
synthase inhibitor oligomycin, despite causing mitochon- activity in type I cells via AMP-activated protein kinase

Physiol Rev • VOL 90 • APRIL 2010 • www.prv.org


690 LUC J. TEPPEMA AND ALBERT DAHAN

(AMPK), which is considered the master regulator of Refs. 440, 870 and references cited therein). Interestingly,
glucose and fatty acid metabolism (417 and 786 for refer- in humans, low plasma glucose was reported to double
ences). AMPK is a cellular energy sensor and sensitive to the HVR, but this could be indirectly mediated by coun-
the cellular AMP-to-ATP ratio. Metabolic stress including terregulatory hormones (825).
hypoxia increases this ratio and activates AMPK, with the In summary, various heme proteins, such as NADPH
result that catabolic processes are upregulated and ana- oxidase, HO-2, NOS, and mitochondrial complex proteins
bolic processes inhibited (417, 786). Recently, Evans et al. may operate in parallel or sequentially as oxygen sensors.
(223) proposed an AMPK-mediated mechanism of O2 The oxygen sensing machinery is already operative in
sensing in type I cells, and they provided substantial mild hypoxia, in a PO2 range in which mitochondrial com-
experimental evidence to support their model. Their main plex proteins, due to their low P50, will not yet be com-
findings were as follows. 1) In rat type I cells, they iden- promised (427, 621). This suggests that more than one O2
tified an AMPK isoform that is located close enough to the sensor must be operative (380, 621, 800). Across (but also
cell membrane to participate in a “membrane delimited” pro- within) animal species, various classes of potassium
cess. 2) In type I cells, the AMPK mimetic agent AICAR channels play a crucial role in the membrane depolariza-
(5-aminoimmidazole-4-carboxamide riboside) inhibited tion of type I cells in hypoxia by reducing their current.
TASK-1 and maxi-K channels and caused depolarization There are multiple ways in which these channels can be
and increased Ca2⫹ influx whilst also increasing sensory inhibited. One of these, changes in the cellular AMP/ATP
afferent discharge from an isolated carotid body prepara- ratio, has recently been identified as a very sensitive
tion. 3) These effects could be inhibited by the AMPK manner to couple cell metabolism to potassium channel
antagonist compound C that also reduced the effect of activity and appears to operate already well before mito-
hypoxia on type I cells (223, 852, 855). This model is chondrial complex proteins are compromised (415). Type
attractive not only because it provides a link between I cells have an extremely complex biochemical content
cellular metabolism and potassium channel inhibition but and release numerous substances during hypoxia, of
also because it may reconcile the “membrane” and “mito- which ACh and ATP are thought to be the principle excita-
chondrial” hypotheses of O2 sensing (852). tory neurotransmitters acting at the afferent endings of
Other, more direct, links between metabolism and petrosal ganglion neurons that innervate the carotid bodies.
TASK-1 channel inhibition are also possible. In rat type I
cells, MgATP and other Mg nucleotides have a powerful
stimulatory action on background potassium channels B. Hypoxia Activates the Transcription Factor
(800). Mitochondrial complex inhibitors cause a rise in Hypoxia-Inducible Factor-1
[Mg]i reflecting a fall in [MgATP]i, and the channels were
suggested to be coupled to (or possess) a low-affinity Mg Chronic hypoxia induces expression of genes encod-
nucleotide sensor (800). A direct, depolarizing effect of ing factors that 1) promote tissue oxygenation (e.g., an-
adenosine via A2A receptors resulting in a rise in [Ca2⫹] in giogenesis); 2) sensitize oxygen-sensing mechanisms and
type I cells has also been reported (856). Depletion of facilitate central processing, thus augmenting the HVR;
MgATP reflects increased production of AMP, providing 3) increase the blood O2 transport capacity (erythropoie-
pathways to increase [adenosine] and activate AMPK, tin production); and 4) adapt cellular metabolism by fa-
respectively (800). cilitating glucose uptake (upregulation of glucose trans-
The idea of carotid bodies as metabolic sensors is not porters) and promoting anaerobic metabolism whilst re-
new. For example, in an in vitro preparation, the cat pressing but also optimizing oxidative phosphorylation.
carotid body is excited by inhibition of glycolysis (560). This altered expression of these and many other genes is
Recent studies have identified type I cells in isolation, initiated by increased activity of numerous transcription
both in coculture with petrosal ganglion neurons and in factors (reviewed in Refs. 101, 133, 149, 381, 661, 693), one
fresh carotid body slices, as direct glucose sensors that of which appears to orchestrate this master plan of gene
depolarize and release neurotransmitters upon lowering expression: hypoxia-inducible factor-1 (HIF-1), discov-
of glucose levels (254, 580, 870). However, a direct effect ered by Semenza and co-workers (see Ref. 689).
of glucose on type I cells is not uncontroversial because in HIF-1 is a heterodimer consisting of the constitu-
the rat, low glucose increased spontaneous ventilation, an tively expressed nuclear subunits HIF-1␤ and HIF-1␣. The
effect that depended on the integrity of the carotid sinus latter subunit, HIF-1␣, undergoes (cytoplasmic) posttran-
nerve, while in isolated carotid body-sinus nerve prepara- scriptional modification under the influence of the PO2
tions it failed to have any effect on nerve discharge and (Fig. 3; Ref. 690). HIF-1 induces many target genes encod-
the release of catecholamines and ATP, respectively (64, ing proteins that influence O2 supply, metabolism, tran-
141). Several studies have clearly established the role of scription factors, and apoptotic and enzymatic processes
the carotid bodies in glucose homeostasis, which can be (101, 133, 371, 693). In euoxic conditions, HIF-1␣ is both
modulated by GABAergic mechanisms in the NTS (see synthesized and rapidly degraded, but in hypoxia it is

Physiol Rev • VOL 90 • APRIL 2010 • www.prv.org


THE VENTILATORY RESPONSE TO HYPOXIA IN MAMMALS 691

FIG. 3. Influence of prolyl hydroxylases on hypoxia inducible factor (HIF) stability. In normoxia, with sufficient oxygen availability, prolyl
hydroxylases [PHD; PHD2 is the isoform that is considered the primary HIF prolyl hydroxylase in normoxia and utilizes iron and ascorbic acid (asc)
as co-factors] hydroxylate crucial proline residues in the degradation domain of HIF-1␣. This enables the von Hippel-Lindau protein (pVHL) to bind
to these residues and ubiquitylate (ubi) the protein for proteasomal degradation. As a consequence, the nucleus contains low levels of HIF-1␣ and
thus also of HIF-1, the heterodimer of HIF-1␣ and HIF-1␤. In hypoxia, PHD2 activity is limited by low PO2. Functional activity of HIF-1␣ is regulated
by factor inhibiting HIF (FIH) which, in an oxygen-dependent way, hydroxylates a crucial asparagine residue in the COOH-terminal domain (not
shown). In hypoxia, HIF-1␣ is stabilized, activated, and translocated to the nucleus. An important additional step in HIF activation in hypoxia,
correlated with the induction of target genes, is the activation of the coactivators p300 and cAMP response element binding protein (CREB) which
combine with the heterodimer. This complex binds to the hypoxia response element (HRE) of target genes while additional coactivators are also
recruited. ␣KG, ␣-ketoglutarate; succ, succinate; ROS, reactive oxygen species.

rescued from proteasomal degradation enabling it to normoxia (63, further references in Ref. 369). PHD2 is a
translocate to the nucleus where it dimerizes with the dioxygenase that uses molecular oxygen and ␣-ketogluta-
constitutively expressed HIF-1␤ to form the transcription rate as substrates, generates CO2 and succinate as by-
factor HIF-1. The dimer binds to the hypoxia response products, and utilizes Fe2⫹ and ascorbic acid as cofactors.
elements of target genes where it recruits coactivator Prolyl hydroxylation of HIF 1-␣ is required for binding of
proteins to promote transcription. Structure and proper- the von Hippel-Lindau tumor suppression protein pVHL
ties of HIF-1 and its subunits can be found in References which targets the complex for ubiquitin-mediated degra-
689 – 691. HIF-1␣ has two paralogs: 1) HIF-2␣, which is dation by interacting with a multiprotein ubiquitin ligase
also regulated by O2, and, when dimerized with HIF-1␤, complex thus creating a recognition site for proteasomal
promotes the transcription of both overlapping and dis- degradation (Fig. 3; Refs. 352, 353, 499). The current view
tinct target genes (references in Ref. 694); and 2) HIF-3␣ is that during hypoxia the activity of PHD2 is reduced by
with a largely unexplored function (369, 694). In many if a combined effect of substrate (oxygen) limitation and
not all body cells, HIF-1 is upregulated during hypoxia. inhibition of the Fe2⫹ catalytic center by ROS possibly
There are many pathways of posttranscriptional modifica- produced by complex III of the mitochondrial electron
tion of HIF-1␣ (reviewed in Refs. 133, 369, 692, 694). Here we transport chain (55, 122, 123).
only briefly discuss the controversial role of ROS.
2. HIF-1 stabilization via decreased [ROS]
1. HIF-1␣ stabilization via increased [ROS]
An opposite role of ROS in HIF-1 stabilization was
HIF-1␣ contains an oxygen-dependent degradation proposed by Huang et al. (341; see Ref. 276 for further
domain with highly conserved specific proline residues references). In human Hela cells, they showed increased
(Pro402 and Pro563 in mice, Pro402 and Pro564 in humans) stability of the HIF-1␣ subunit in hypoxia and a rapid
that are hydroxylated in normoxic conditions. This hy- breakdown in normoxia. In hepatoma 3B (Hep3B) cells,
droxylation is catalyzed by enzymes containing a prolyl hypoxia also induced HIF-1␣ stabilization and an in-
hydroxylase domain (PHD). Currently three PHD family creased expression of erythropoietin mRNA, and both
members are known, PHD1, PHD2, and PHD3, of which effects were dose-dependently blocked by preexposure to
PHD2 is considered the primary HIF prolyl hydroxylase in hydrogen peroxide. Oxidizing HIF-1␣ inhibited HIF-1

Physiol Rev • VOL 90 • APRIL 2010 • www.prv.org


692 LUC J. TEPPEMA AND ALBERT DAHAN

DNA binding, and this could be reversed by reducing increase in potassium conductance and hyperpolarization
agents, possibly indicating that hypoxia stabilizes HIF-1␣ may ensue acting to conserve energy. Some neurons,
by reducing specific cysteine sulfhydryls in the molecule, however, depolarize in hypoxia and if some other condi-
whilst hypoxia and hydrogen peroxide would reverse this tions are also met may be classified as central O2 sensors
(341 and references therein). (see sect. IIIC2). Severe, permanent hypoxia is generally
What is the scenario in glomus cells? In carotid bodies followed by intracellular acidosis, influx of Ca2⫹ and Na⫹,
from rats exposed to chronic intermittent hypoxia, HIF-1␣ efflux of K⫹, persisting depolarization, further decreases
was upregulated and complex I activity was reduced, whilst in intracellular [ATP], and eventually cell death (435).
[ROS] and hypoxic carotid body sensitivity were increased Hypoxia in the brain (central hypoxia) affects breathing in
(597, 600). HIF⫹/⫺ mice did not display these adaptations, a way that depends on the integrity and activity of the
and together these findings would suggest stabilization of carotid bodies. In addition, several brain regions are
HIF-1␣ by increased [ROS] resulting in the augmented HVR claimed to contain oxygen sensors that may play a role in
(600; see also sect. VG). However, with the assumption of the HVR in some circumstances.
enhanced ROS production by mitochondrial complex inhib- Shortly after carotid body denervation, cats anesthe-
itors, the finding that in type I cells from rat, complex tized with chloralose (urethane) show decreases in ven-
inhibitors significantly reduced HIF-1␣ upregulation induced tilation and phrenic nerve activity with both hypoxemic
by superfusion with a hypoxic solution for ⬃45 min, would and CO hypoxia (314, 361, 507). With severe hypoxia, the
rather support the notion of HIF stabilization with reduced phrenic neurogram displays a gasplike pattern (507). An-
[ROS] (22). This model could also provide a potential expla- imals anesthetized with thiamylal, however, failed to
nation for the increase in HVR in humans after chronic show ventilatory depression immediately after carotid
treatment with antioxidants (321, 608). Notably, the antiox- body excision, even with severe hypoxia (710). Carotid
idant N-acetylcysteine not only augmented the HVR in young body-denervated rats anesthetized with urethane do also
subjects but also increased the production of erythropoietin, show a clear-cut hypoxia-induced decrease in respiratory
indicating upregulation of HIF-1 (321). Reduction of the motor activity (436). A depressant response of CNS hyp-
isocapnic HVR by low-dose anesthetics or the carbonic an- oxia is also manifest in chloralose-urethane anesthetized
hydrase inhibitor acetazolamide could be reversed by an cats subjected to isolated brain stem perfusion with hyp-
antioxidant cocktail consisting of ␣-tocopherol and ascorbic oxic blood, even when the PCO2 of the peripheral blood is
acid (777, 780, 781). This reversal occurred in the setting of kept hypercapnic (794, 823). In anesthetized cats sub-
an acute experiment, so presumably it was unlikely to be jected to carotid body denervation or artificial brain stem
mediated via increased stabilization of HIF-1␣. It was spec- perfusion, hypoxic ventilatory depression may be caused
ulated that the reversal could be due to independent, inhib- by both a fall in medullary PCO2, the metabolic stress of
iting effects of the antioxidants on potassium channels, al- lactic acidosis and/or decrease in [ATP], as well as inhib-
though other explanations are possible (777, 780, 781). The itory neurotransmitters (62, 436, 540, 543). The absence of
stimulating effect of chronic ascorbic acid on the HVR is a depression in thiamylal-anesthetized animals (710)
hard to reconcile with a single role of this antioxidant as might suggest involvement of a GABAergic mechanism.
cofactor of PHD2. An effort to eliminate the other cofactor Artificial brain stem perfusion via one vertebral artery in
of PHD2, Fe2⫹, by infusing the iron chelator desferrioxam- the cat supplies the pons, medulla, and cerebellum with
ine (DFO) for 8 h did not alter the HVR in humans (639). This “central blood” (59) so central hypoxic depression in this
could be due to failure of DFO to penetrate into type I cells, preparation must originate from one or more of these
and this is supported by data from in vitro perfused carotid regions, presumably pons and/or medulla.
body from the rat (168). Finally, in humans, iron infusion did At least in cat and rat, the effect of (severe) CNS
not influence the augmentation of the HVR that is normally hypoxia on breathing differs between the anesthetized
observed after chronic hypoxia (see sect. VF), and this could be and awake states. Awake carotid body denervated cats
due to the inability of iron to reach the interior of type I cells respond to mild hypoxia (FIO2 ⬃14%) with mild to mod-
(711). erate hypoventilation (237, 261, 510) or virtually no
change (464). Severe hypoxia, however, results in a pro-
nounced tachypnea and decrease in tidal volume resulting
C. Central Effects of Hypoxia
in either a reported decrease (261) or increase (510),
respectively, in alveolar PCO2. In unanesthetized animals,
1. General effects of central nervous system hypoxia
a transition into unconsciousness during severe hypoxia
on breathing
did not result in any change in breathing pattern, suggest-
In the central nervous system (CNS), moderate hyp- ing the diencephalon as the source of the tachypnea (510).
oxia causes a generalized reduction in neuronal excitabil- Immediately after carotid body denervation, the awake
ity and decrease in intracellular [ATP]. Depending on the rat may show some hypoxic respiratory depression (492),
duration and severity an initial transient alkalosis, an but the most common response is a limited rise in venti-

Physiol Rev • VOL 90 • APRIL 2010 • www.prv.org


THE VENTILATORY RESPONSE TO HYPOXIA IN MAMMALS 693

lation (570, 664, 665), raising the question whether the In slice preparations, the locus coeruleus in the dorsolat-
carotid body-denervated rat is a suitable model to study eral pons contains many neurons that are inhibited by
the isolated effect of central hypoxia. This is illustrated by hypoxia and a smaller percentage that is depolarized, but
the fact that acute hyperoxia leads to an increase in it is uncertain whether this represents intrinsic O2 sensi-
ventilation in this model, indicating a depressant effect of tivity (545, 864).
central hypoxia (570). With prolonged central hypoxia, Both stimulation of the carotid bodies and central
awake goats and dogs undergoing selective carotid body hypoxia activates neurons in the RVLM, and in the latter
perfusion with normocapnic-normoxic blood showed an case, this activation is resistant to local glutamate recep-
increase in ventilation (165, 836) and an absence of respi- tor antagonism and denervation of the carotid bodies. So
ratory depression (706), respectively. Sleeping dogs in- these cells may be intrinsically O2 sensitive, and this no-
creased their ventilation with central hypoxia, but after tion was corroborated by the finding that in slice prepa-
carotid body denervation there was no response (151). rations RVLM neurons retained their depolarizing re-
Soon after carotid body denervation, ponies showed no sponse to hypoxia or cyanide even with synaptic block-
hypoxic ventilatory depression; however, this does not ade (741, 742, 744, 745). Nolan and Waldrop (553)
preclude an inhibitory effect of low central PO2 during reported activation of ⬃64% of neurons studied in the
chronic hypoxia because acute hyperoxia induced hyper- same RVLM area when exposing anesthetized spontane-
ventilation in these animals (239). ously breathing rats to hypoxia. Many of these neurons
Finally, note that the depressant effect of central displayed a temporal discharge pattern related to the
hypoxia in anesthetized cats with denervated carotid bod- cardiac and/or respiratory cycle; a similar percentage was
ies is different mechanistically from HVD in intact cats, activated after bilateral transection of both the vagus and
with the latter being initiated by the carotid bodies (see carotid sinus nerves and cervical sympathetic and aortic
also section VC). depressor nerves (553). Brain slices including this RVLM
A) CENTRAL O2 SENSORS AND HVR. Before a neuron that area and from a region projecting to the spinal cord
depolarizes in hypoxia can be classified as intrinsically contained neurons the majority (⬃70 – 80%) of which was
sensitive to O2, at least four criteria must be met. 1) The excited by low PO2 even after synaptic blockade; however,
depolarization should occur independently from synaptic a substantial amount (⬃20%) was inhibited (553). O2 sen-
events. 2) Activation of these neurons is independent sitivity in the RVLM is not a unique property of spinally
from input from the arterial chemoreceptors. 3) Within projecting barosensitive neurons because it seems to be
the range of physiological O2 levels, the sensitivity of widely distributed in this area with many neurons either
these neurons should be much greater than that of other excited or inhibited by hypoxia (378). Taken together, the
neurons. 4) At least under certain physiological condi- caudal hypothalamus, PAG, and RVLM might act as an
tions, stimulation by low oxygen of these neurons should accessory circuit to amplify ventilatory and cardiovascu-
be able to elicit an appropriate (cardio)respiratory re- lar responses to hypoxia (410).
sponse. Below we summarize which regions belonging to Located just rostral to the C1 region of the RVLM, the
the respiratory neuronal network are claimed to possess pre-Bötzinger complex contains oxygen-sensitive cells.
neurons that are intrinsically sensitive to oxygen. Local application of NaCN into this area augmented in-
I) Location of central O2 sensors. Exposing anesthe- spiratory output including gasping in the phrenic neuro-
tized cats to 10% O2 excited ⬃20% of neurons studied in gram (718). Dissociated cell cultures with neurons from
the caudal hypothalamus, independently from carotid si- both the C1 region and pre-Bötzinger complex contained
nus nerve integrity (183). Ninety percent of these cells cells of which ⬃60% responded to NaCN with membrane
had a cardiovascular and/or respiratory rhythm, even af- depolarization and an increase in firing frequency, but
ter carotid body denervation (183). In brain slice prepa- ⬃40% with inhibition, and these responses were resistant
rations, ⬃80% of caudal hypothalamic neurons responded to synaptic blockade (500). Both RVLM regions express
to hypoxia independently from synaptic blockade (182, HO-2 (501), an enzyme that in the carotid bodies may be
329). O2-sensitive neurons in the caudal hypothalamus involved in oxygen sensing (844; see also sect. IIIC1). The
were also found in the rat (330, 671). Oxygen-sensitive expression of HO-2 in these regions is limited to oxygen-
cells in the hypothalamus may also reside rostrally in sensitive neurons; blocking enzyme activity abolishes the re-
parvocellular cells of the paraventricular nucleus (385). sponse of these neurons to hypoxic hypoxia and NaCN (153).
Located in an area from which O2-sensitive neurons Oxygen-sensitive cells were also found in the NTS. In
in the caudal hypothalamus can be antidromically acti- coronal slices from the level of the obex, 75% of the
vated, the pontine periaqueductal gray (PAG) contains neurons tested responded to hypoxia with hyperpolariza-
many neurons intrinsically sensitive to O2 (410, 671). De- tion, decreased input resistance, and decreased activity,
pending on the stimulus location, PAG activation elicits while 25% showed increased activity (583). Hypoxia in
cardiovascular responses that are mediated via the ros- these slices also decreased postsynaptic potentials elic-
troventrolateral medulla (RVLM; references in Ref. 410). ited by electric stimulation of afferents in the solitary

Physiol Rev • VOL 90 • APRIL 2010 • www.prv.org


694 LUC J. TEPPEMA AND ALBERT DAHAN

Physiol Rev • VOL 90 • APRIL 2010 • www.prv.org


THE VENTILATORY RESPONSE TO HYPOXIA IN MAMMALS 695

tract, suggesting hypoxic gating of incoming afferent in- In cats, ponies, and rats, the respiratory system dis-
put (583). O2-sensitive neurons in the NTS were also plays a remarkable plasticity in restoring the HVR after
found by Nolan and Waldrop (553). denervation of the carotid bodies (70, 492, 664, 710). Rats
Finally, the hypoglosssal nucleus also contains neu- displayed a complete recovery of the HVR 90 days after
rons that depolarize upon exposure to hypoxia (297). carotid body denervation which was temporally related to
The mechanism of oxygen sensing in brain stem and changes in tyrosine hydroxylase expression in various
hypothalamic neurons is largely unknown. Does a uni- brain stem areas containing central O2 sensors, e.g., the
form O2 sensor operate that would facilitate identification A2/C2 region in the caudal NTS and the A1/C1 region in
and localization of these neurons? Some authors have the RVLM (665, 665). Complete HVR recovery on a com-
proposed a lack or paucity of ATP-sensitive potassium parable time scale was also observed in rats after lesion-
channels as a potential basis of oxygen sensing (e.g., Ref. ing glutamate receptor-containing neurons in the NTS,
864), which could make sense because hypoxia decreases initially resulting in a 70% reduction of the response (130).
intracellular [ATP] in the brain (435, 436). Also, several It is tempting to speculate that tyrosine hydroxylase-con-
other potassium channel species have been suggested to taining central O2 sensors could serve as a “back-up”
be involved in central oxygen sensing, as well as various mechanism to detect low oxygen levels in cases where
calcium and sodium channels (329, 745, 864; reviewed in peripheral chemoreceptors are functionally eliminated,
Ref. 544). Recently, D’Agostino et al. (153) reported that but further studies are necessary to confirm this.
excitation by hypoxia of neurons in the C1 region and Diencephalic structures play a role in the severe
pre-Bötzinger complex of rats critically depended on the hypoxia-induced tachypnea in awake cats with dener-
activity of HO-2. vated carotid bodies (510). Disinhibition of cortical-hypo-
thalamic interactions by low PO2 contribute to this phe-
2. Role of central O2 sensors in the HVR? nomenon. Because the caudal hypothalamus contains
neurons excited by O2 independently from the carotid
The contribution of central oxygen sensors to the bodies and provide the RVLM with an excitatory input,
HVR is unknown. Does the absence of an increase in these O2 sensors could contribute to it (331, 813). Direct
ventilation shortly after carotid body denervation as ob- stimulation of the caudal hypothalamus increases respi-
served in cats and dogs imply a trivial contribution of ratory frequency (813).
central O2 chemoreceptors to the HVR of an intact organ-
ism? Taking into account that the carotid body may pro- 3. CNS neurotransmitters and HVR
vide oxygen-sensitive neurons in the RVLM with a tonic
excitatory input (741, 744), a preparation with denervated A) GLUTAMATE, GABA, GLYCINE, ADENOSINE, AND ATP. The
carotid bodies would not be the most suitable model to entire respiratory network participates in the HVR, so it is
get more insight into this issue. Could the increase in not surprising that it can be influenced by numerous
ventilation with isolated central hypoxia in goats and neurotransmitters/modulators and receptor agonists/an-
dogs with intact, separately perfused carotid bodies be tagonists. Respiratory midbrain and brain stem regions,
due to a facilitating peripheral chemoreceptor input, ex- each with a separate contribution to the HVR, contain a
plaining, for example, the elimination of the response in variety of neurotransmitters (recently reviewed in Ref.
dogs after carotid body denervation (151)? Selectively 732). In Figure 4, the major medullary and pontine regions
perfusing the carotid bodies with either hypoxic or hyper- are shown that participate in the HVR. Classical media-
oxic blood to subsequently measure the effects of central tors of the HVR are GABA, glutamate, and adenosine, and
hypoxia could give some clue. In anesthetized cats and their temporal profiles and roles were reviewed previ-
awake goats, the reverse paradigm, i.e., varying the pe- ously (104, 327, 379, 502, 540, 649, 752). Generally, gluta-
ripheral PO2 while keeping central PO2 constant at differ- mate is considered an excitatory neurotransmitter in the
ent levels, did not reveal an influence of brain stem PO2 on HVR, while GABA, glycine, and adenosine are inhibitory
the ventilatory response induced by carotid body hypoxia (18, 104, 327, 379, 565). This is probably an oversimplifi-
(i.e., no peripheral-central O2 interaction; Refs. 165, 794). cation, because one particular transmitter or modulator

FIG. 4. Major brain stem regions involved in the ventilatory response to hypoxia. Major cell groups involved in the ventilatory response to
hypoxia are shown in coronal sections between rostrocaudal levels of the rat brain stem from Bregma ⫺9.16 to ⫺14.60 mm. 1, A1 noradrenergic
region; 2, caudal ventral respiratory group; 3, A2 noradrenergic region; 4, raphe pallidus; 5, raphe obscurus; 6, medial subnucleus of the nucleus of
the solitary tract; 7, commissural subnucleus of the nucleus of the solitary tract; 8, rostral ventral respiratory group; 9, C1/A1 adrenergic/
noradrenergic region; 10, C2 adrenergic region; 11, C1 adrenergic region; 12, pre-Bötzinger complex; 13, Bötzinger complex; 14, raphe magnus; 15,
retrotrapezoid nucleus; 16, A5 noradrenergic region; 17, locus coeruleus; 18, parabrachial nucleus, medial part; 19, Kölliker-Fuse nucleus. Py,
pyramidal tract; scp, superior cerebellar peduncle; Sol, nucleus of the solitary tract; V, motor nucleus of the trigeminal nerve; VII, facial nucleus.
[Sections redrawn from Paxinos and Watson (587).]

Physiol Rev • VOL 90 • APRIL 2010 • www.prv.org


696 LUC J. TEPPEMA AND ALBERT DAHAN

can have distinct effects on the HVR, depending on the chronic hypoxia may be related to (temporal) upregula-
dose, route of administration, and site of application in tion of local tyrosine hydroxylase (references in sect. VD).
the CNS. For example, in rats, at an ambient temperature In the caudal NTS, norepinephrine can influence both
of 30°C, the GABA agonist muscimol reduces the HVR gluatamatergic and GABAergic transmission via presyn-
when microdialyzed into the NTS but augments it when aptic receptors and may thus have variable effects on the
applied in raphe magnus (539, 772). Recently, a modula- HVR (873 and references cited therein). Neurons in the A1
tory (facilitatory) role for ATP has been described, in and C1 groups are activated in acute hypoxia but do not
which it is released in ventral medullary surface areas in seem to have an altered catecholamine turnover in
the secondary phase of the HVR and possibly acts in the chronic hypoxia (221, 722, 782). The contribution of A6
Pre-Bötzinger complex to increase respiratory frequency neurons will be discussed in the subsection below on the
via activation of metabotropic (P2Y) receptors (284, 471). role of central NO.
Below we summarize the most relevant data, mainly col- Serotonergic neurons in the brain stem are mainly
lected over the last decade, concerning the important role concentrated in medullary, pontine, and mesencephalic
of monoamines (with emphasis on serotonin) and NO in raphe nuclei (references in Ref. 322) and may be of spe-
the HVR. Figure 4 can be used as a reference to envisage cial interest for the HVR for several reasons. First, they
the different brain stem regions where these transmitters have connections with respiratory motoneurons and are
exert their action. able to increase their excitability (322, and references
B) MONOAMINES AND THE HVR. The absence of a signifi- cited therein). Second, they are crucial for homeostatic
cant effect of catecholamine depletion on the HVR may thermoregulation. About 50% of 5-HT neurons in raphe
suggest that norepinephrine and/or dopamine do not pallidus/obscurus increase their activity upon environ-
function as primary neurotransmitters in hypoxia (503). mental and/or hypothalamic cooling, and there is growing
However, monoamines are widely distributed in the brain evidence that raphe neurons modulate the hypothermic
and as such may be expected to contribute in a complex component of the HVR (323, 324). Third, they mediate
way. Mice lacking the gene for the dopamine transporter episodic hypoxia-induced long-term facilitation (LTF) of
(DAT⫺/⫺) show a reduced poikilocapnic HVR (805), but breathing by causing an enhanced release of 5-HT into the
data from these mice are difficult to interpret because phrenic motor nucleus (230, 481; for a discussion on LTF,
both carotid body and central dopamine levels will be see sect. VG; note that LTF can also be evoked by electri-
increased. The same problem is encountered in those cal simulation of raphe obscurus neurons, Ref. 512). And
studies using dopaminergic blockers that act both periph- finally, many raphe neurons also possess intrinsic CO2
erally and centrally. As is also the case for serotonin (see sensitivity (322), so at least in the secondary phase of
below), the influence of central dopamine on the HVR is poikilocapnic hypoxia they will certainly have an influ-
not limited to the ventilatory component of the response ence on ventilatory output.
but also concerns the hypometabolic/hypothermic part of Activation of neurons in the nucleus raphe magnus
it. For example, infusion of haloperidol in the third cere- attenuated the processing of afferent input from the ca-
bral ventricle of rats (aimed at exerting effects in the rotid bodies by the NTS in anesthetized rats (602). Micro-
hypothalamus) reduced the drop in hypoxia-induced body dialysis of the GABA receptor agonist muscimol into ra-
temperature without affecting the ventilatory component phe magnus of conscious rats augmented the poikilocap-
(35). Dopamine 1 receptors in the anteroventral preoptic nic HVR measured at an environmental temperature of
region of the hypothalamus in particular seem to be in- 30°C (within in the thermoneutral zone for rats) but re-
volved in this response (36). duced it at 24.5–26.5°C; muscimol reduced body temper-
Several regions belonging to the central neuronal ature at 24.5–26.5°C and increased the anapyrexic com-
respiratory network contain noradrenergic/adrenergic ponent of the HVR without influencing these parameters
neurons, notably the A2 noradrenergic cell group in the at 30°C (772). In the same species, lesions of raphe mag-
caudal NTS where carotid body afferents terminate, the A5 nus neurons with ibotenic acid (unspecific for serotonergic
noradrenergic cell group in the ventrolateral pons, the A6 neurons) increased the hypoxia-induced rise in ventilation
group in the locus coeruleus in the dorsolateral pons, and measured at an ambient temperature of 25°C without
the A1/C1 noradrenergic cell groups in the ventrolateral affecting the anapyrexic response (256). However, micro-
medulla (255, 803). All these regions may contribute to the injection of a specific 5-HT1A receptor antagonist into
magnitude and/or shaping of the HVR. For example, Coles raphe magnus decreased the poikilocapnic HVR (557).
et al. (140) reported that lesions in the A5 group reduce or Unless it is speculated that the applied antagonist (WAY-
abolish the reduction of respiratory frequency following 100635) acted on presynaptic inhibitory receptors (auto-
hypoxia. The A2 group, containing neurons responsive to receptors) on serotonergic neurons, the latter finding
dopamine, is considered to play a modulatory role in the seems difficult to reconcile with the stimulatory effects of
ventilatory acclimatization to chronic hypoxia; the in- local lesions and muscimol application on the ventilatory
crease in CNS gain (CNS gain is defined in sect. VF4) in component of the HVR.

Physiol Rev • VOL 90 • APRIL 2010 • www.prv.org


THE VENTILATORY RESPONSE TO HYPOXIA IN MAMMALS 697

To address the role of central serotonin in respiratory gic neurons in the HVR. A few general conclusions, how-
and thermoregulatory control, Richerson and co-workers ever, seem justified. First, in rodents, serotonergic neu-
(324) utilized conditional knockout mice almost entirely rons in medullary raphe are crucial for the homeostatic
devoid of central 5-HT neurons, designated as Lmxlbf/f/p. control of body temperature and participate in both the
These mice are deficient in the capacity to generate heat, ventilatory and hypometabolic/anapyretic components of
resulting in a rapid lowering of body temperature when the HVR. Second, serotonergic neurons in raphe nuclei
placed in a cold environment; ventilatory CO2 sensitivity possessing connections with respiratory (premotor) neu-
is also greatly reduced in these animals (324). Although rons are activated in hypoxia. Third, activation of seroto-
they had the expected temperature-dependent hypoxia- nergic neurons in raphe magnus tend to dampen the rise
induced changes in ventilation and metabolism (i.e., lowering in ventilation and to augment the fall in body temperature,
the environmental temperature decreased the ventilatory but the magnitude of these effects depends on the ambi-
component of the HVR but increased the anapyrexic re- ent temperature. And, finally, the role of raphe serotoner-
sponse), these animals had a reduced ventilatory and gic neurons in hypoxia-induced LTF of breathing is well
V̇E/V̇O2 response compared with wild types when the HVR established (discussed in sect. VG1).
was measured at 25°C. At an ambient temperature of C) CENTRAL NO AND HVR. I) NTS. Carotid body stimula-
30°C, no difference in HVR between both genotypes was tion in hypoxia is followed by release of glutamate and
visible, whilst intracerebroventricular administration of production of NO in the caudal (commissural) NTS. When
serotonin restored the impaired ventilatory response to injected in the NTS, the NO donor sodium nitroprusside
CO2 but did not alter the (normal) HVR (323). Whether augments the HVR while the nonselective NOS inhibitor
Lmxlbf/f/pmice would fail to show episodic hypoxia-in- NG-monomethyl-L-arginine (L-NMMA) reduces it. These
duced LTF of breathing (see sect. v) remains to be exam- observations in unanesthetized rats, together with the
ined. Hodges and co-workers (323, 324) suggested that finding that the increased production of NO is inhibited by
serotonergic neurons are not directly involved in the HVR NMDA receptor antagonism, led to the suggestion that NO
per se but may be critically important to thermogenesis. and glutamate operate in the NTS in a positive feedback
Mice showing overwhelming amounts of serotonin in cycle to increase the HVR (562; see also Ref. 257). Gluta-
brain extracellular fluid are represented by a genotype mate augments the HVR by activating NMDA receptors in
lacking expression of the serotonin transporter (5-HTT). projection areas of carotid body afferents in the NTS that
5-HTT knockout mice showed a reduced activity of brain are rich in NO (130, 286, 288, 446; reviewed in Ref. 287). In
serotonergic neurons and decreased 5-HT1A receptor the conscious rat, injection of both the nonselective NOS
binding (444). At room temperature, these animals had a inhibitor NG-nitro-L-arginine methyl ester (L-NAME) and
larger ventilation and higher metabolism but normal body the selective neuronal inhibitor of the enzyme Nw-propyl-
temperature. When challenged with 10% O2, they re- L-arginine in the caudal NTS reduced the increase in re-
sponded with a greater increase in ventilation and larger spiratory frequency induced by carotid body stimulation
fall in metabolism than wild types, but the change in the with KCN (291).
V̇E/V̇O2 ratio was equal in both genotypes (444). Lipton et al. (451) proposed an involvement of NO in
A role of serotonergic neurons in medullary raphe in the HVR via endogenous production of S-nitrothiosols
thermoregulation is consistent with the finding that dis- within the NTS. Hemoglobin desaturation goes along with
inhibition of raphe pallidus neurons (by means of local formation of S-nitrothiosols from gluthathione and cys-
injection of the GABA antagonist bicuculline) increased teine and Hb-bound NO producing S-nitroso-gluthathione
brown adipose sympathetic nerve activity, responsible for (GSNO) and S-nitroso-L-cysteine (L-CSNO), respectively.
thermogenesis in cold stress (482). In the rat, an increased GSNO is converted into CGSNO (S-nitroso-cysteinyl gly-
activity of brown adipose sympathetic nerve activity cine), and this requires the enzyme ␥-glutamylpeptidase
(thought to underlie the anapyrexic response to hypoxia) (␥-GT; Ref. 451 and references therein). Local injection of
can be induced in several ways: cold stress (i.e., skin all three nitrothiosols into the NTS mimicked the HVR in
cooling), activation of neurons in preoptic thalamic re- conscious rats including its recovery phase (short-term
gions, and disinhibition of raphe pallidus neurons and potentiation) after removal of the hypoxic stimulus, and
also of neurons in the commissural subnucleus of the NTS the same was observed after injection of plasma from
(482). Interestingly, carotid body activation with NaCN or deoxygenated but not oxygenated blood (451). The effect
hypoxia completely inhibits the increase in sympathetic of CGSNO was inhibited by pretreatment with a blocker
outflow to brown adipose tissue by these interventions. of ␥-GT, suggesting that conversion of GSNO into CGSNO
This suggests that the carotid body not only plays a pivotal is an essential step in producing the effect of the former.
role in the hypoxia-induced rise in ventilation but also in the L-CSNO is less stable in vivo, and thus Lipton et al. (451)
anapyrexic response (further references in Ref. 482). hypothesized that ␥-GT would be required for a normal
From all of the above data, it is not easy to distill a HVR to occur and found the short-term potentiation to be
clear and detailed picture of the role of central serotoner- abolished in ␥-GT-deficient mice. Apparently, however,

Physiol Rev • VOL 90 • APRIL 2010 • www.prv.org


698 LUC J. TEPPEMA AND ALBERT DAHAN

the hypoxia-induced rise in ventilation was not eliminated (including those applying ventricular administrations), so
in ␥-GT⫺/⫺ mice, but their report did not show the mag- perhaps inhibitor concentrations at target sites within the
nitude of this initial increase (451). From the fact that in brain were too small to produce significant effects (al-
the rat denervation of the carotid bodies reduces the HVR though blood pressure clearly increased; Ref. 347).
with at least 70% and virtually eliminates it in other spe-
cies, the quantitative contribution of S-nitrothiosols can
be estimated to be small or negligible unless carotid body- D. Hypoxic Ventilatory Decline in the
activated NOS within the NTS would play a major role in Adult Mammal
producing these molecules during hypoxia. On the other
hand, the reported increase of the HVR in humans by In adult mammals, the ventilatory response to sus-
N-acetylcysteine (321) could be related to generation of tained (15–30 min) hypoxia (end-tidal PO2 50 –55 Torr and
S-nitrothiosols with the aid of Hb-bound NO (578), rather SaO2 ⬃80%) is biphasic (Figs. 5 and 6; Refs. 162, 204, 370,
than to the antioxidant properties of this molecule. 383, 464, 752, 806). During moderate isocapnic hypoxia
II) Pons and locus coeruleus. In the pontine reticular (i.e., arterial PO2 ⬃50 Torr) lasting ⬎3–5 min, ventilation
formation, NO promotes the release of acetylcholine shows a slow decline from its peak (75–150% above nor-
which induces sleep associated with rapid eye move- moxic resting values) to reach a new steady-state level
ments and respiratory depression (442). The dorsolateral (25– 40% of normoxic values) within 15–20 min (162, 204,
pons uses NO to promote the termination of inspiration 236, 383). The slow decline was formerly known as “hyp-
during lung inflation (pneumotaxic mechanism). In awake oxic ventilatory depression”; current names include hyp-
rats, electrolytic bilateral lesions in the locus coeruleus oxic ventilatory decline (HVD), hypoxic ventilatory roll-
substantially augmented the HVR (224). In addition, off, and secondary roll-off of the HVR.
whereas acute intracerebroventricular administration of Although consistently found across mammalian spe-
the nonselective NOS inhibitor L-NAME almost com- cies, dogs and goats are the exception with little or no
pletely inhibited the HVR in control animals, it did so by HVD during exposure to moderate hypoxia (113, 151,
only ⬃50% in those with the bilateral lesions, suggesting a 546). However, HVD is dependent on various factors, such
substantial contribution of the locus coeruleus to the as age, sex, CNS arousal state, (type of) anesthesia,
inhibition in intact animals (224). The locus coeruleus breathing route, and hypoxic test (12, 111, 498, 677, 773).
contains the A6 cell group that is considered the major For example, HVD is most pronounced under isocapnic
noradrenergic cell group in the brain stem (references in experimental conditions, while the poikilocapnic HVD is
Ref. 803). Microinjection of L-NAME in the locus coer- much smaller in magnitude (12).
uleus had an impressive inhibitory effect on the HVR, In the awake state, the ventilatory response to sus-
indicating that the “permissive” effect of NO may be due tained isocapnic hypoxia has specific characteristics.
to inhibition of, presumably noradrenergic, neurons in 1) The magnitude of HVD is proportionally related to the
this area (225). It is unclear why in another study in rats size of the initial hyperventilatory response to acute hyp-
chronic injection (7 days) of small amounts of L-NAME oxia (162, 263, 266, 383). 2) Bilateral carotid body resec-
into the right cerebral ventricle did not modify the HVR tion or chemodenervation results in the loss of the V̇I
unless it is hypothesized that chronic NO shortage may be (inspired ventilation) response to acute hypoxia and ab-
compensated by upregulation of NOS (688). Notwithstand- sence of HVD (Fig. 5; Refs. 383, 464). 3) HVD persists
ing this latter study, all data taken together clearly identify beyond the initial period of hypoxic exposure. The V̇I
the locus coeruleus as an important inhibitory modulator of response to acute hypoxia following 20 min of moderate
the HVR, possibly analogous to an active inhibitory role of isocapnic hypoxia and 5 min of air breathing is depressed
lateral pontine regions in the hypoxic inhibition of breathing by ⬎50%. This delayed recovery persists for up to 1 h (162,
in newborn lambs (522; see also sect. II). 205). 4) Suppression of the peripheral drive with low-dose
III) Other regions. Microinjection of the nonselective dopamine yields no HVD during 20-min hypoxia. And,
NOS inhibitor L-NMMA into the RVLM reduced the HVR in despite the presence of initial central hypoxia, a subse-
rats but not the hypothermic response (173). The same quent V̇I response to acute hypoxia develops fully (Fig. 6;
inhibitor had a similar effect when injected into the nu- Ref. 162). 5) The magnitude of the fall in ventilation upon
cleus raphe magnus (556). A possible role of brain stem the relief of hypoxia is smaller than the V̇I response gener-
NO in the acclimatization to chronic hypoxia has been ated at the onset of hypoxia (204, 382). 6) Awake humans
briefly mentioned in section VF. and awake cats display similar response characteristics with
In humans, the acute ventilatory response to hypoxia respect to characteristics 1–3 and 5 (Fig. 5; Refs. 204, 464).
does not seem to be influenced by systemic administra- These observations (most importantly characteristic
tion of L-NMMA (347). The intravenous dose applied in 4) suggest that in conscious mammals the peripheral
this study (⬃62 ␮g/kg), however, was much smaller than chemoreceptors play a pivotal role in the development of
that mostly used for NOS inhibitors in animal studies HVD. The issue remains whether HVD is due to adapta-

Physiol Rev • VOL 90 • APRIL 2010 • www.prv.org


THE VENTILATORY RESPONSE TO HYPOXIA IN MAMMALS 699

FIG. 5. Ventilatory effects of hypoxia in cats and humans. A: ventilatory responses to sustained (25–30 min) isocapnic hypoxia in the awake
cat (mean response of 7 cats) [adapted from Long et al. (461)] and awake chemodenervated cat. B: mean response of 3 cats. [Adapted from Long
et al. (464).] C: isocapnic (open symbols) and poikilocapnic (closed symbols) hypoxic ventilatory response (HVR) in 10 young subjects. [Adapted
from Steinback and Poulin (728).] D: isocapnic HVR in awake humans after bilateral carotid body resection (mean of 7 subjects). The data are
derived from otherwise healthy patients that underwent a bilateral carotid body resection after development of familial carotid body tumors. As
reported in Reference 227, the hypercapnic response in these subjects could be adequately described by a single slow (central) component,
indicating a minor or no significant contribution of a fast (peripheral) component to the response.

tion of the peripheral chemoreceptors (i.e., an exclusive ance towards a net inhibitory effect on V̇I (162, 266, 810).
peripheral mechanism) or to the way the afferent input At the termination of hypoxia, changes in neurotransmit-
from the carotid bodies is processed by the CNS (657). ter turnover are not immediate and, therefore, the depres-
The hypothesis that HVD is related to peripheral chemo- sant effects of hypoxia wane slowly. In the awake state, a
receptor adaptation is supported by characteristics 1, 2, small part of HVD is related to an increase in brain blood
and 5 and is further corroborated by the observation that flow and the resultant washout of acid metabolites/H⫹
hypoxic sensitivity declines during the development of from the brain. Suzuki et al. (746) showed a decrease in
HVD (37). However, in the anesthetized cat, the periph- the gradient between jugular venous and arterial PCO2 of
eral chemoreceptors do not adapt during hypoxic expo- ⬃2 Torr during 15 min of moderate isocapnic hypoxia.
sure in terms of ventilation and afferent nerve activity, Assuming that jugular venous PCO2 approximates brain
indicating that an exclusive peripheral mechanism is un- tissue PCO2, ⬃10 –20% of measured HVD is related to an
likely (17, 58, 807). Furthermore, characteristics 3 and 4 increase in brain blood flow in the adult awake human.
indicate an effect of HVD on HVR beyond hypoxic expo- Finally, at deeper levels of hypoxia, substrate insuffi-
sure when peripheral chemoreceptor activity in terms of ciency and depressed cellular metabolism may become
the V̇I response to CO2 has normalized (57, 267, 461). The the major origin of HVD (435, 540).
current hypothesis is that, at least in the awake mammal, Various mechanisms could explain the decrease in
a large part of HVD is due to modulation of glutamatergic excitatory neuronal output during sustained hypoxia. For
excitatory activity within the NTS (arising from afferent example, as suggested by Kazemi and Hoop (379), periph-
carotid body input) shifting the excitatory/inhibitory bal- eral chemoreceptor afferents cause the enhancement of

Physiol Rev • VOL 90 • APRIL 2010 • www.prv.org


700 LUC J. TEPPEMA AND ALBERT DAHAN

may originate from the paraventricular nucleus (PVN) of


the hypothalamus. It is known that arginine vasopressin-
containing neurons in the PVN and supraoptic nucleus are
stimulated by hypoxia (709 and references therein), but it
remains to be seen whether NTS neurons could be di-
rectly inhibited during hypoxia via this pathway.
Other inhibitory modulators possibly involved in
HVD include dopamine, adenosine, serotonin, endogenous opi-
oid peptides, lactic acid, NO, and Ca2⫹ (Tables 1 and 2 for
references). A relatively new observation is that of Gozal et al.
(289) who examined the role of the platelet-derived growth
factor (PDGF) isoforms PDGF-AA (with two A chains and
acting on the PDGF-␣ receptor) and PDGF-BB (with two B
chains and acting on the PDGF-␤ receptor) in the NTS during
hypoxia. The NTS and other brain stem respiratory nuclei show
FIG. 6. Dopamine and hypoxic ventilatory decline (HVD) in hu- an abundant expression of the PDGF-␤ but not PDGF-␣ recep-
mans. Ventilatory response to 20-min isocapnic hypoxia (SpO2 ⫽ 80%), tor. In contrast to PDGF-AA, the isoform containing two B
5-min normoxia and subsequently a second hypoxic episode (SpO2 ⫽ chains and its receptor PDGF-␤ appeared to play a functional
80%, lasting 5 min). Closed circles, control response showing the HVD
from early to late hypoxia and a marked reduced response to a second role in the development of HVD. Awake mice with a heterozy-
hypoxic exposure; open circles, low-dose dopamine infusion (gray bar: gous mutation of the PDGF-␤ receptor had an intact V̇I re-
3 ␮g 䡠 kg⫺1 䡠 min⫺1) during sustained hypoxia blunts the initial hyperven- sponse to acute hypoxia but a diminished HVD, an effect which
tilatory response and prevents development of HVD. The ventilatory
response to the second hypoxic exposure develops fully. Data are the was unaffected by the injection of a PDGF-␤ receptor antago-
means of 16 subjects. [Adapted from Dahan et al. (162).] nist. However, wild-type animals showed a reduced HVD after
the antagonist. Many NTS neurons in wild-type mice (and in
rats) showed increased expression of PDGF-B chain mRNA
glutamatergic excitation in areas of the brain stem in- and protein (release) in hypoxia. These data suggest an inhib-
volved in hypoxic ventilatory control. Glutamate serves as itory role for PDGF-BB and its receptor on ventilation during
the precursor of GABA, and conversion of glutamate into sustained hypoxia, possibly by inhibiting NMDA receptors
GABA may enhance GABAergic activity with conse- within the NTS (289).
quently a net inhibitory effect on V̇I. In the awake rat, There is strong evidence that HVD development dur-
Tabata et al. (752) measured an increased GABA concen- ing anesthesia is uncoupled from peripheral chemorecep-
tration in the extracellular fluid in the NTS during HVD tor activity (657). For example, in anesthetized cats sub-
development, which was absent in animals after bilateral jected to artificial brain stem perfusion, central hypoxia
carotid body resection. Both the reduction in O2 availabil- with the carotid bodies kept normoxic results in a decline
ity and intracellular acidosis increase GABA levels in the in ventilation with dynamics similar to that observed after
brain with kinetics that are fast enough to account for central changes in PCO2 (823). Comparable observations
HVD (increased synthesis from glutamate through the are made in carotid body perfusion experiments and anes-
enzyme glutamic acid decarboxylase and decreased thetized chemodenervated animals (361, 538, 543; Table 2).
breakdown by the enzyme GABA-␣-oxyglutarate transam- Similarly, in humans exposed to halothane, the initial
inase; references in Ref. 540). Via this pathway, the NTS hyperventilatory response is greatly reduced while the
itself could be the source of increased GABA during hyp- absolute magnitude of the HVD is similar to that observed
oxia (540, 752). However, other sources of GABA are also in the awake state (160). Furthermore, the ventilatory
possible because the NTS contains many terminals immu- response to a step out of sustained hypoxia (the off-
noreactive to GABA that may inhibit glutamatergic inputs response) shows a marked undershoot and is similar in
(e.g., Ref. 673 and references therein). GABAergic neu- magnitude to the initial hyperventilatory response (the
rons in the NTS send inhibitory efferents to the caudal on-response). These symmetric on- and off-responses in
ventrolateral medulla and also to the locus coeruleus, humans exposed to halothane indicate that in this condi-
providing other possible pathways involved in HVD (7, tion peripheral sensitivity, although reduced under of low-
795). Other regions such as the hypothalamus may also dose halothane, remains unchanged during HVD. Similar
participate in HVD via a GABAergic mechanism. For ex- observations were made in humans injected with the
ample, Lemus et al. (440) depicted an interesting circuitry intravenous anesthetic propofol (549). These data sharply
participating in the glycemic response of carotid body contrast with data in awake humans and animals that do
stimulation involving antidiuretic hormone (ADH)-secret- not show HVD development after carotid body resection or
ing neurons in the supraoptic nucleus and ADH receptors when the peripheral drive is reduced by dopamine (Fig. 6;
on the membrane of GABAergic neurons in the NTS that Refs. 162, 383, 464).

Physiol Rev • VOL 90 • APRIL 2010 • www.prv.org


THE VENTILATORY RESPONSE TO HYPOXIA IN MAMMALS 701

TABLE 1. Effect of pharmacological interventions on the development of HVD in adult humans

Reference
Target Agent Arousal State Effect Comments Nos.

␣2-Adrenergic receptor Clonidine Sedated AHR⫽ HVD⫽ In line with the observation that 236
the magnitude of HVD is
related to AHR
Adenosine receptor Aminophylline (adenosine Awake AHR⫽ HVD2 Data suggest a role for 203
antagonist) adenosine in the development
of HVD
265
Dipyridamole (adenosine Awake AHR1 HVD1 Pretreatment with aminophylline 859
reuptake inhibitor) abolished the dipyridamole
effect; data indicate a role for
adenosine in HVD
development
Calcium channel Verapamil (Ca2⫹ channel Awake AHR⫽ HVD⫽ Data suggest no involvement of 459
blocker) calcium ions in HVD
development
D2 receptor Low-dose dopamine Awake AHR2 HVD2 Dopamine acts peripherally at 162
this dose; the reduced HVD
prevents depression of a
subsequent V̇I response to
acute hypoxia
Awake AHR2 HVD⫽ 38
Awake AHR2 HVD2 821
Haloperidol (central and Awake AHR1 HVD⫽ Effect contrasts findings in 589
peripheral D2 antagonist) awake cats (see Refs. 769 and
460)
Domperidone (peripheral D2 Awake AHR1 HVD⫽ 38, 235
antagonist)
Lactic acid Dichloroacetate (blocks Awake AHR⫽ HVD⫽ Data suggest that brain lactic 264
lactate production) acidosis is unlikely involved in
HVD development
Opioid receptor Naloxone (opioid receptor Awake AHR⫽ HVD⫽ Data suggest no involvement of 370
antagonist) endogenous opioid system in
HVD development
Alfentanil (␮-opioid receptor Awake AHR2 HVD⫽ Data suggest that exogenous 120
agonist) opioids potentiate HVD
Morphine (␮-opioid receptor Awake AHR2 HVD⫽ 680
agonist)
Intrathecal morphine Awake AHR2 HVD⫽ 25
5-HT receptor Methysergide (5-HT receptor Awake AHR⫽ HVD⫽ Data suggest no involvement of 459
antagonist) serotonin in HVD development
Somatostatin receptor Somatostatin Awake AHR2 HVD2 Effect possibly via disinhibition 231
of GABAergic inhibitory
system
Carotid body Almitrine Awake AHR1 HVD1 In line with the observation that 266
the magnitude of HVD is
related to AHR
Central nervous system Low-dose inhalational Lightly anesthetized AHR2 HVD⫽ Data suggest that the link 798
anesthetics (halothane, between AHR and HVD is lost
isoflurane, enflurane) during anesthesia
535
235
Propofol (intravenous Lightly anesthetized AHR2 HVD1 Propofol may have increased 549
anesthetic) HVD via its action at the
GABA receptor complex;
propofol had no effect on
intermittent hypoxic
ventilation

AHR, acute hypoxic ventilatory response; HVD, hypoxic ventilatory decline; V̇I, inspired ventilation.

During anesthesia, when the peripheral drive is either state, it becomes the major contributor during anesthesia.
absent or severely reduced, it seems appropriate to as- In the anesthetized cat, brain blood flow-related HVD
cribe the development of HVD to an effect arising selec- does explain the similarities in ventilatory time constants
tively from within the CNS. One possibility is that while of central CO2 steps and hypoxic steps, respectively, re-
CBF-induced HVD plays only a minor role in the awake stricted to the brain stem (823). It would also explain the

Physiol Rev • VOL 90 • APRIL 2010 • www.prv.org


TABLE 2. Effect of pharmacological interventions on the development of HVD in awake and anesthetized adult animals 702
Reference
Target Agent Animal Arousal State Effect Comments Nos.

Adenosine receptor Aminophylline (adenosine Cat Anesthetized (chloralose- HVD2 The carotid sinus nerves and vagi 361
antagonist) urethane) of the animals were bilaterally
denervated
Theophylline iv and icv Cat Anesthetized (sodium pentothal) iv: AHR1 Data from these interventions 866
(adenosine antagonist) suggest involvement of
adenosine in development of
HVD
iv: HVD2
icv: AHR⫽
icv: HVD2
D2 receptor Haloperidol (central and Cat Awake AHR⫽ HVD2 HVD was abolished by 769
peripheral D2 antagonist) haloperidol
460
463
Cat Anesthetized (chloralose- AHR⫽ HVD2 Identical results as in the awake 769
urethane) cat; carotid sinus nerve activity
remained constant during HVD
development
Domperidone (peripheral D2 Cat Awake AHR⫽ HVD⫽ Data from these interventions 769
antagonist) suggest that central but not
peripheral dopaminergic

Physiol Rev • VOL


mechanisms are directly
involved in HVD
GABA receptor Bicuculline (GABA antagonist) Rat Awake HVD2 GABA antagonists reduced HVD 752
microinjected in the NTS in intact animals only; in
carotid body denervated
animals, no HVD was present

90 • APRIL 2010 •
and no HVD developed upon
injection of GABA antagonists
during hypoxia
Bicuculline iv (GABA antagonist) Cat Anesthetized (chloralose) HVD2 Animals were glomectomized; 508
phrenic nerve response to
LUC J. TEPPEMA AND ALBERT DAHAN

www.prv.org
progressive CO was measured
Lactic acid Dichloroacetate (prevents lactate Cat Anesthetized (chloralose) HVD2 Animals were peripherally 543
production) chemodenervated; progressive
hypoxia induced by inhalation
of CO; data suggest a role for
lactic acid in HVD generation
Nitric oxide NG-nitro-L-Arginine (a nonspecific Rat Awake HVD1 Control animals did not display 290
synthase (NOS) NOS blocker) HVD
S-methyl-L-thiocitrtrulline (a
selective neural NOS inhibitor)
Opioid receptor Naloxone (opioid receptor Cat Anesthetized (chloralose) HVD2 Animals were peripherally 541
antagonist) chemodenervated; ventilatory
response to progressive CO
was measured
Morphine (␮-opioid receptor Cat Anesthetzied (chloralose- HVD2 Steady-state response to 61
agonist) urethane) sustained hypoxia remained
unaltered by morphine,
suggesting a reduced HVD
possibly by reducing brain
blood flow response to hypoxia
THE VENTILATORY RESPONSE TO HYPOXIA IN MAMMALS 703

D2 receptor, dopamine 2 receptor; PB-KF, Parabrachial/Kölliker fuse complex; AHR, acute hypoxic ventilatory response; HVD, hypoxic ventilatory decline; HVR, hypoxic ventilatory
Reference symmetry in on- and off-responses observed in halothane-
Nos.

289

289
exposed humans. Manipulating brain blood flow either
92
hemodynamically (for example, by aortic balloon infla-
tion), pharmacologically (for example, by infusion of pa-
No distinction between AHR and

while after microinjection into


paverine to blood perfusing the brain stem), or by expo-

the PB-KF AHR is maintained

suggest a role for PDGF-␤ in


animals compared with their
HVD is made; data suggest a

enhancement of HVD after


sure to central hypoxia produce a similar decrease in V̇I

Control experiments show a


reduced HVD in knockout
systemic MK801 injection,

wild-type littermates; data


that is consistent with washout of central CO2 (60, 293,
reduction in AHR with

while HVD is reduced


542, 823). Finally, we cannot exclude an effect of modu-
Comments

HVD development
lators accumulating during hypoxia in the anesthetized
mammal. Indeed, some enhancement of GABAergic inhi-
bition of V̇I during hypoxia is expected, especially from
anesthetics that have agonistic activity at the GABA re-
ceptor. However, at PO2 values ⬎50 Torr, these effects are
probably small with regard to the reduced tissue PCO2 by
changes in BBF (794).
HVD2 in wild-type
microinjection:

animals only
AHR2 HVD2

IV. GENETIC INFLUENCE ON THE


Effect

VENTILATORY RESPONSE TO HYPOXIA


iv: HVR2

HVR⫽

A. The Genome and HVR

The genotype plays an important role in the magni-


tude of the HVR and consequently explains part of the
Anesthetized (halothane-

large intersubject response variability (734). Measure-


Arousal State

ments of the HVR in humans show that normal healthy


volunteers may vary a factor of 20 in the magnitude of
their response. This range is much larger than expected
urethane)

from just normal physiological variability and technical,


environmental, and/or emotional factors. Furthermore,
Awake

Awake

data from high-altitude residents indicate that genotype


may not only influence the immediate response to hyp-
oxia but possibly also the ventilatory response to sus-
Heterozygous

knockout

tained hypoxia and ventilatory adaptation to high altitude


Animal

PDGF-␤
Rat

Rat

(519).
mice

1. Human studies
PDGF-␤ antagonist microinjected

In humans, evidence for a genetic effect on HVR


response; iv, intravenous; icv, intracerebroventricular.

comes primarily from family and population studies.


PDGF-␤ antagonist (given

First-degree healthy family members of patients with pul-


microinjected in PB-KF
MK801 (NMDA receptor

monary disease associated with hypoventilation and CO2


antagonist) iv and
Agent

intraperitoneally)

retention and nonathletic parents and siblings of endur-


ance athletes show a familial clustering of HVR magni-
in the NTS

tudes at the low end of the spectrum (see Ref. 832 and
references cited therein). Similar observations were made
in families with a history of SIDS and obstructive sleep
2. —Continued

apnea (734). Twin studies show greater similarities in the


magnitude of HVR among homozygotic twins compared
with heterozygotic twins (see Ref. 734 and references
growth factor
NMDA receptor

Platelet-derived

cited therein). Most but not all interpopulation compari-


Target

sons indicate the existence of heritable HVR phenotypes


(PDGF)-␤
receptor

linked to specific populations (96, 97, 152, 783). Strong


TABLE

evidence for a hereditary component comes from studies


in Asians and South Americans. Children from Tibetan

Physiol Rev • VOL 90 • APRIL 2010 • www.prv.org


704 LUC J. TEPPEMA AND ALBERT DAHAN

and Han (Chinese) parents tend to be in the range ob- tactile/auditory stimuli was not different between both
served in the Han population (152). With the use of an- strains (669). The difference in HVR was associated with
cestry-informative genetic markers, an evolutionary ori- structural and functional changes in the carotid bodies
gin was found for the low HVR in Andean high-altitude (858). Compared with the A/J strain, the carotid bodies of
natives of Quechua origin (97). the DBA/2J strain were larger, and acetylcholine in-
creased intracellular Ca2⫹ in more cells. These morpho-
2. Animal studies logical and functional differences are accompanied by
differences in gene expression (30). There were differ-
There is ample proof that the divergence in ventila- ences in expression of genes related to neurotransmitter
tory behavior between different inbred strains of the same metabolism, synaptic vesicles, and neural crest cell mi-
species reflects genetic influences more than effects from gration/development with less expression in A/J mice. An
body mass, age, or sex. This also applies to the ventilatory important difference was the reduced expression of the
response to moderate hypoxia and the survival response ␤-subunit of maxi-K channels in A/J mice. Further studies
to severe hypoxia (874). In rats, most studies show an from Tankersley and co-workers (758, 759, 761) aimed to
effect of strain on the magnitude of HVR (50, 274, 736, 739, uncover the genes involved in the HVR phenotype. Data
834). Some strains are difficult to compare as underlying from an intercross study between C3H/3J and C57BL/6J
disease may affect carotid body function. For example, mice suggest that HVR in terms of respiratory timing is
New Zealand hypertensive rats suffer from sclerotic dam- dependent on only a handful of genes (759). A putative
age to the carotid artery and its branches (50). Strains quantitative trait locus on chromosome 9 could be iden-
may differ in the magnitude of the HVR by up to a factor tified with a significant association with several hypoxic
of seven (834). The differences are not only related to the ventilatory variables (758), while another locus on chro-
carotid body response to hypoxia but also to central mosome 3 was significantly associated with hypoxic in-
neural processing during and immediately after exposure spiratory drive. Both loci explained 26 –30% of the pheno-
to short episodes of hypoxia (274, 739). As stated by typic variation among F2 offspring. The interaction be-
Golder et al. (274), the choice of rat strain can (critically) tween hypoxia and hypercapnia on breathing was linked
influence conclusions concerning the control of breath- to two different loci on chromosomes 1 and 5 (761). The
ing. This is also true for drug studies. For example, in rat, simplicity of this genetic model is appealing and is much
the peripheral dopamine D2 receptor antagonist domperi- less complex than in the rat. It is doubtful whether such a
done and the NOS inhibitor L-NAME differentially affect restricted model also applies to the human HVR phenotype.
ventilation and the HVR depending on the genetic back-
ground (737, 738).
To pinpoint the HVR phenotype to a specific rat B. Mutations and HVR
chromosome, Forster et al. (240) used the technique of
chromosomal substitution in which an entire chromo- Examples of targeted gene deletions in mice to study
some from one inbred rat strain is introgressed into the the process of oxygen sensing at the carotid bodies and/or
genetic background of another to create a consomic rat. to understand the peripheral and central processing of the
Initially, five chromosomes were studied. HVR was not carotid body response to hypoxia are discussed in section
determined by any of the genes on these chromosomes, III. Here we briefly discuss mouse strains developed to
although genes on chromosomes 9 and 18 did contribute study specific diseases and that show altered ventilatory
to HVD and/or a reduced metabolic rate during hypoxia. responses to hypoxia. We further discuss two human
The results of a second study indicate that there are conditions in which one specific mutation is linked to an
complex gene-gender interactions on HVR (199). altered HVR.
Like rats, inbred mice strains vary significantly in the Various mouse strains have been engineered to study
magnitude of their HVR. Tankersley et al. (760) examined the Congenital Central Hypoventilation Syndrome (CCHS),
the HVR in eight inbred strains. The DBA/2J strain had the some of which have recently been reviewed (see Ref.
greatest HVR, while the A/J strain displayed the lowest 259). Particularly genes of the endothelin and c-ret path-
response. Tankersley et al. (760) did not take the brain way have been the focus of interest, for example, endo-
state of the animals into account, and in this this regard, thelin converting enzyme (ECE1), endothelin-1 (EDN1),
it is interesting that A/J mice are more prone to sleep than endothelin-3 (EDN3), endothelin receptor A (Ednra), en-
DBA/2J mice and have a higher arousal threshold in hyp- dothelin receptor B (Ednrb), Mash-1, c-ret, Phox2a, Phox2b,
oxia (669). So possibly the ventilatory response could be brain-derived neurotrophic factor (Bdnf), and orphan ho-
the reflection of a stress response. However, the differ- meobox (Rnx) genes (169, 259, 849). Knockouts at these
ence in frequency of hypoxic events during sleep (larger genes display a variable respiratory phenotype with reduced
in the A/J strain) was inversely related to the hypoxic hypercapnic response and/or HVR in ECE1⫹/⫺, EDN1⫺/⫺,
ventilatory sensitivity, whilst the arousal threshold upon Ednra⫺/⫺ and Mash-1⫹/⫺ newborn mice; however, with the

Physiol Rev • VOL 90 • APRIL 2010 • www.prv.org


THE VENTILATORY RESPONSE TO HYPOXIA IN MAMMALS 705

exception of EDN1⫺/⫺ mice, these genotypes did not display Robbins and co-workers (712) studied the effect of a
abnormalities as adults. This is an important limitation for a mutation in the von Hippel-Lindau tumor suppressor pro-
disease that in humans persists throughout life (259). There tein (VHL) gene. A homozygous missense mutation that
are other mice models associated with CCHS such as the impairs but does not ablate VHL function causes Chuvash
Nurr1 mutant mouse and the microphtalmia-associated tran- polycythemia (47, 213). The disease is endemic to the
scription factor (Mitf) mutant mouse (555, 754). Nurr1 mu- Chuvash population of Russia. In patients with Chuvash
tant mice show complete agenesis of midbrain dopamine polycythemia, binding of VHL to HIF-1␣ is diminished and
cells. Newborn Nurr1 knockout mice have a severely dis- hence normal degradation reduced causing the accumu-
turbed pattern of breathing with a blunted HVR (555). Mitf lation of HIF (in normoxia) and consequent upregulation
plays an essential role in the differentiation of neural crest- of certain target genes such as the erythropoietin gene.
derived melanocytes. Mitf mutant mice display an increased The three patients studied showed several abnormalities
HVR. The authors relate this to a reduction of melanocyte- in respiratory and pulmonary vascular regulation. Most
derived opioids within the CNS (754). This suggests an in- importantly, the isocapnic HVR and pulmonary artery
hibitory role of endogenous opioids on the central ventila- pressure responses to hypoxia were increased compared
tory network and is in agreement with observations in ␮-opi- with healthy and polycythemia control subjects (712). In
oid receptor gene knockout mice (158). other words, cardiorespiratory responses to hypoxia were
A mouse model associated with the Rett syndrome is upregulated in patients with increased HIF levels and up-
represented by a genotype with a ubiquitous deletion of regulated HIF target genes. These data are in agreement
one allele of the X-linked Mecp2 gene, encoding the DNA- with animal studies showing downregulated cardiopulmo-
binding protein methyl CpG (Mecp2) involved in gene nary responses in HIF-1␣⫹/⫺ mice and attenuated adaptive
silencing. Female Mecp2⫹/⫺ mice have a respiratory phe- responses to chronic intermittent hypoxia (see sect. V).
notype resembling that of the Rett syndrome and that is In conclusion, the above examples of the influence of
characterized by episodes of hyperventilation and respi- single gene defects (and their inheritance) on the HVR are
ratory depression (75). The (isocapnic) HVR in heterozy- clear illustrations of a genetic influence. Strain differ-
gous females is larger than in wild types; mice with a ences in animals and differences in HVR between low-
selective deletion of the protein in neurons do also display landers and high-altitude natives illustrate the influence of
an enhanced HVR but do not show the typical episodes of genomic factors, i.e., the interaction between different
respiratory depression (75). It is interesting to note that genes on the one hand and that between genes and the
different respiratory phenotypes also exist between dif- environment on the other hand. Both respiratory and
ferent mouse strains with the gene deletion (references in nonrespiratory stimuli are able to affect the development
Ref. 563). of the HVR (developmental plasticity) causing alterations
In humans, carotid body tumors are rare and often in response that may persist throughout adulthood. In
associated with chronic hypoxia from altitude, cyanotic section II we discussed the influence of changes in oxygen
heart disease, and pulmonary disease. One hereditary tension in the neonatal period. One clear example of a
form of carotid body tumor (as part of the hereditary nonrespiratory stimulus leading to plastic changes is
familiar paraganglioma syndrome) is due to a missense stress from neonatal maternal separation that leads to a
mutation within the gene that encodes succinate dehydro- permanently augmented HVR of male but not female rats
genase D (SDHD; Refs. 48, 154, 213). SDHD is an essential (262). Brain stem areas involved in ventilatory control of the
enzyme of the tricarboxylic acid cycle and part of cyto- neonate have strong plastic capacities depending on specific
chrome b588 of the mitochondrial respiratory chain com- environmental stimuli and genetic background (735).
plex II. All patients are heterozygous: animal studies in-
dicate that homozygotes do not survive and die in utero
V. PATHOPHYSIOLOGICAL INFLUENCES ON
(607). We measured the isocapnic HVR and hypercapnic
THE HYPOXIC VENTILATORY RESPONSE
ventilatory responses in normoxia and hypoxia in 12 tu-
mor-free carriers of the mutation (as tested by MRI; au-
thor’s unpublished observation). There were three main A. Biological Variability
observations: 1) hypercapnic ventilatory responses were
not different from age-matched healthy controls, 2) the Apart from a between-subject variability, one has to
magnitude of the isocapnic HVR was at the low end of consider the appreciable within-subject variation in the
“normal” (mean value 0.3 versus 1.2 l 䡠min⫺1 䡠%⫺1 in con- magnitude of the HVR. The coefficient of variation of
trols), and 3) in contrast to controls, none of the tested repeated HVRs obtained over a 2-h period ranges from 10
carriers of the SDHD gene showed any O2-CO2 interaction to 60% among subjects (mean 20%; Ref. 674). Between-day
on the HCVR. These data may link the SDHD gene to variability is 20 –50% greater than within-day variability.
carotid body oxygen sensing and/or to the well-described The cause of the large between-subject variability may lie
multiplicative action of CO2 on the HVR (see sect. VD). in differences in genetic make-up and race of the different

Physiol Rev • VOL 90 • APRIL 2010 • www.prv.org


706 LUC J. TEPPEMA AND ALBERT DAHAN

subjects tested. However, this explains just part of the morphometric and functional studies in carotid bodies of
variability (see sect. IV). Other causes are related to phys- aged rats (see above). However, it may well be that the
iological factors (such as age, metabolism, body temper- human studies were performed at the older end of normal
ature, the circadian rhythm, hormonal status, and preg- adult function when no functional decline in carotid body
nancy), psychological factors (anxiety, full bladder), function is yet apparent (714). Another possible cause for
pathophysiological factors, the use of pharmacological the difference between morphological appearance of the
agents that may affect HVR, and environmental factors carotid body and its function may be found in the exis-
(previous exposure to hypoxia, altitude). To reduce within- and tence of redundancy in carotid body-mediated processes.
between-subject variability, it is important to perform A reduced number of type I cells may be able to sustain
hypoxic experiments in subjects that feel comfortable with the the functional tasks of the carotid bodies (610). Finally, a
setup, protocol, and laboratory surroundings and have no failing carotid body function in old age may result in
apparent urges that may influence their responses (e.g., a plastic changes in brain stem areas involved in the pro-
full bladder). Also the investigator should do everything cessing of its afferent output, causing the (partial) resto-
not to influence the subject (for example, whispering may ration of the HVR (610).
have a negative influence on the subject).

C. Gender and Pregnancy


B. Ageing
Sex hormones (progesterone and the combination
Breathing is affected by ageing due to a decline in progesterone/estrogen as well as testosterone) have a
function at multiple levels of the neuromechanical link stimulatory effect on breathing and HVR. Progesterone
between chemosensors, brain stem, and ventilatory pump, stimulates breathing via hypothalamic sites through es-
causing a reduced ability to maintain blood gas homeosta- trogen-dependent receptors (46). The role of progester-
sis (360). Morphometric studies of rat carotid bodies in- one in causing an increase in HVR is twofold: it increases
dicate the occurrence of degenerative changes in old age carotid body sensitivity to hypoxia, and in combination
(142, 179, 610). The carotid bodies of aged rats (⬎23 mo) with estrogen, it has a central modulatory effect on its
show an increase in extracellular matrix, a reduction in afferent output (299). Testosterone increases breathing
the number and volume of type I cells, and the presence and the HVR probably through an increase in metabolic
of necrotic cells (142, 179, 610). In the remaining func- rate and central modulatory effects (766, 841).
tional type I cells, mitochondrial volumes and neurose- The majority of human studies could not find a dif-
cretory fill-up of vesicles are reduced (610). Carotid body ference in HVR between men and women (157, 362, 364,
catecholamine content and turnover time is reduced with 418, 677, 680, 681). However, most of the studies involved
age, as is the catecholamine response to hypoxic stimu- did not control for the phase of the menstrual cycle or
lation (142). Conde et al. (142) measured afferent carotid tested women in just one phase (often the follicular
sinus nerve activity during hypoxic and CO2/acidotic stim- phase). The number of animal studies on gender effect on
ulation in young (3 mo) and old (18 mo) rats. While no age HVR is limited, but all, in contrast to the human data,
effect was observed in the nerve response to CO2/ point toward a greater carotid body response to hypoxia
acidosis, in old animals a ⬎50% reduction was observed in in female animals (366, 530, 765). Only part of these sex
the response to hypoxia. Degenerative changes such as differences is related to the acute effects of sex steroids.
fibrosis and loss of glomus tissue have been observed in Castration of the adult animals does lead to a reduced
the human carotid body (311). In 15–30% of humans in HVR in males and females, but the sex differences persist
their eighth and ninth decade, there may be signs of (767). This is not surprising taking into account the long-
chronic carotid glomitis with infiltration of T-cell lympho- term developmental and organizational effects of sex ste-
cytes (311). These data collectively indicate important roids on neuronal systems that occur in prenatal and early
age-dependent morphological and functional changes of postnatal life. In this light, the absence of clear sex dif-
the carotid bodies. ferences in the human population seems remarkable.
A minority of human studies observed a reduced HVR An effect of the menstrual cycle on the HVR is small
in older persons (age range 64 –79 yr; Refs. 412, 604). and ranges from an increased HVR in the luteal phase by
However, the majority of studies observed no differences 10 –20% compared with the follicular phase (157, 840).
in HVR between young (20 –30 yr) and old (60 –79 yr) (6, Pregnancy is associated with hyperventilation due to an
609, 610, 714), while one study observed an increase in increase in metabolic rate and the stimulatory effects of
HVR (124). The overall picture that emerges from the progesterone (636). Also, the HVR is increased during
human literature is that HVR is marginally affected at old pregnancy, an effect that is already apparent at 20 wk and
age and hence suggests that carotid body function re- reaches a maximum at 36 wk (521). More than 60% of the
mains unaltered. This stands in sharp contrast to the increase in HVR is related to sex hormones, the rest to the

Physiol Rev • VOL 90 • APRIL 2010 • www.prv.org


THE VENTILATORY RESPONSE TO HYPOXIA IN MAMMALS 707

increase in metabolic rate (521, 636). A more extensive capnia and hypoxia is not tightly coupled to dopamine
overview of the effect of sex steroids on ventilatory con- release (100, 351, 743). The O2 sensor may behave like a
trol during development and in adult life has been pub- hemoglobin molecule and, analogous to the Bohr shift, O2
lished recently (52). binding may be competitively inhibited by CO2/H⫹, ex-
plaining the finding that the maximal effect of a very high
PCO2 (⬃270 Torr) was the same regardless of the PO2 (180
D. Influence of CO2 on the HVR:
and references therein).
O2-CO2 Interaction

In both humans and animals, there is ample evidence 2. Separate perfusion of peripheral and
for a multiplicative effect of O2 and CO2 on the HVR (12, central chemoreceptors
150, 178, 156, 195, 494, 517, 777). It is uncertain if quanti-
A) DOGS. In a majority of early studies utilizing selec-
tatively this O2-CO2 synergism observed at the ventilation
tive perfusion of the carotid bodies, hypoadditive effects
level is entirely due to a multiplicative interaction in the
on ventilation of peripheral and central stimuli were re-
carotid bodies (423; further references in Ref. 178).
ported (references in Ref. 5). Multiplicative O2-CO2 inter-
However, CO2 does not always act as a multiplier of
action at the level of minute integrated phrenic nerve
the HVR. In conscious cats, O2-CO2 synergism does not
activity was also reported (132). More recently, Smith et
seem to exist, due to a negative interactive effect on tidal
al. (707) showed a clear multiplicative action of periph-
volume (575, 576). When anesthetized, however, this spe-
eral O2 and CO2 on ventilation but also a much larger
cies demonstrates interaction at both carotid body and
inhibiting effect on the HVR of systemic hypocapnia than
ventilation levels (234, 423). Although displaying multipli-
of carotid body hypocapnia alone, thus not supporting a
cative interaction in the carotid bodies (601), the anesthe-
hypoadditive peripheral-central interaction. In the same
tized rat only seems to display a positive synergistic hy-
animal model, it was recently shown that isolated carotid
percapnic-hypoxic action at the level of ventilation at
body perfusion with hyperoxic hypocapnic blood led to a
hypocapnic arterial PCO2 values (145, 613). Earlier studies
substantial decrease in ventilation despite a considerable
showed that in other species O2-CO2 interaction may be
rise in PCO2, not only suggesting an important role of the
hypoadditive or at best additive (references in Ref. 145;
carotid body in euoxic/normocapnic ventilation but also a
more recent studies are discussed below).
hyperadditive peripheral-central interaction (77).
A modulating effect of CO2 on the HVR could origi-
B) GOATS. Awake goats subjected to selective carotid
nate from three sources: the carotid bodies, a central-
body perfusion showed O2-CO2 multiplication at the level
peripheral interaction, and/or central O2-CO2 interaction.
of both carotid bodies and ventilation without any periph-
While a multiplicative interaction in the carotid bodies is
eral-central interaction other than simply additive (163
evident, the brief overview below shows that there is no
164, 165). In awake goats instrumented for ventriculocis-
consensus as to the modality of the peripheral-central
ternal perfusion, carotid body stimulation with NaCN in-
interaction: hyperadditive, simply additive, or hypoaddi-
creased ventilation more at a relatively alkaline perfusate
tive. There is no evidence for central O2-CO2 interaction.
pH, possibly suggesting negative peripheral-central inter-
action. However, in this study an alkaline cerebrospinal
1. Carotid sinus nerve recording: properties of
fluid pH was associated with much higher PaCO2 levels
type I cells
(708), thus almost inevitably leading to more intense cen-
A relatively simple method to study O2-CO2 interac- tral stimulation and potentially to simple additive central
tion in the carotid bodies is with carotid sinus nerve effects consistent with the findings in goats subjected to
recording. Anesthetized cats, rats, lambs, and rabbits selective carotid body perfusion (163, 164, 165).
demonstrate O2-CO2 interaction in the carotid sinus nerve C) CATS. Anesthetized cats subjected to artificial brain
(116, 118, 119, 234, 333, 386, 423, 601, 660; further refer- stem perfusion did not show any central-peripheral inter-
ences in Ref. 233) that most likely can be reduced to the action other than simply additive (314, 793). As in awake
properties of type I cells. The few studies that have been goats and dogs (163, 707), selective manipulation of pe-
performed in type I cells may indeed suggest an interac- ripheral gas tensions showed a multiplicative O2-CO2 ef-
tion between acid/high CO2 and low O2 on intracellular fect on ventilation (314, 793). These findings might not
[Ca2⫹] (167, 668). As outlined by Peers (592), a thorough entirely be compatible with those in the anesthetized cat
quantitative analysis of the relation between neurotrans- showing that, with a constant level of hypoxia and a linear
mitter release and intracellular [Ca2⫹] is required to shed CO2 response of the carotid sinus nerve, CO2 had a
more light on the mechanism of stimulus interaction in smaller effect on ventilation below the eucapnic PCO2 than
type I cells. Other neurotransmitters than dopamine alone above it, suggesting some form of hyperadditive periph-
should be implicated in such analysis because the in- eral-central interaction (422). The disparity between the
crease in afferent carotid sinus nerve activity by hyper- findings in anesthetized and awake cats (awake animals

Physiol Rev • VOL 90 • APRIL 2010 • www.prv.org


708 LUC J. TEPPEMA AND ALBERT DAHAN

showing a negative peripheral-central interaction of tidal relatively slow process may also be responsible for the
volume; Refs. 575, 576) cannot be explained by other than lower CO2 sensitivity and higher resting arterial PCO2 after
unidentified influences of anesthesia and/or arousal ef- carotid body denervation in goats and dogs remains to be
fects of hypoxia during wakefulness. established (579, 663).
D) RATS. Utilizing an in situ dual perfusion system in
the rat to separately perfuse peripheral and central che- 5. Other approaches used in humans
moreceptors, Day and Wilson (172) found a hypoadditive
effect of the central PCO2 on the hypoxic phrenic ampli- In earlier studies, so-called withdrawal tests were
tude response. Perhaps this may explain the large HVR in performed, suddenly removing a central or peripheral
this species at hypocapnic but not hypercapnic PaCO2 stimulus keeping the background peripheral or central
levels (145). gas tensions, respectively, at constant levels (reviewed in
Ref. 150). Underlying assumptions such as lack of activity
3. Dynamic end-tidal forcing and absence of undershoots in carotid sinus nerve dis-
charge in (mild) hyperoxia make these studies difficult to
In both animals and humans, peripheral-central inter-
interpret (e.g., Ref. 325). Other workers compared tran-
action can be studied using end-tidal forcing (DEF), en-
sient and steady stimuli assuming that responses to the
abling one to estimate separate gains of the central (Gc)
former originate at the peripheral chemoreceptors and
and peripheral (Gp) chemoreflex loops, respectively (175,
those to the latter at both peripheral and central sites (see
747; see sect. VI). In the cat, Gc and Gp (assessed with
Ref. 206 for earlier references). The role of HVD could not
both DEF and artificial brain stem perfusion) are indepen-
always be taken into account, as well as the possible
dent, indicating no central-peripheral interaction other
existence of an interaction component with a fast time
than additive (175, 314, 793). In addition, in the cat, O2-
constant (e.g., Ref. 656).
CO2 interaction takes place at the level of the carotid
One way to study peripheral-central interaction is to
sinus nerve (234, 423), so the increase in ventilatory CO2
expose subjects to peripheral stimuli at different central
sensitivity with hypoxia in this species seems entirely due
PCO2 levels. Robbins (656) compared the effects of a step
to a peripheral O2-CO2 interaction, compatible with an
decrease in end-tidal O2 performed just 30 s after impos-
increased Gp but unaltered Gc in overall hypoxia (e.g.,
ing a step decrease in end-tidal CO2 with those performed
Refs. 779, 793). In young piglets, hypoxia increased Gp but
at constant steady-state end-tidal CO2. In two of three
did not alter Gc (845). In humans, an unchanged Gc with
subjects, the response was larger under the former con-
hypoxia was not always observed, with some studies
dition, suggesting a more-than-additive central-peripheral
showing a small but significant rise (226, 227) but others
interaction (alternative explanations were left open; Ref.
reporting no significant change (56, 156, 591). When Gp
656). However, St Croix et al. (724) could not reproduce
was lowered by inhalation of subanesthetic concentra-
this. Clement et al. (136) found no evidence for different
tions of volatile anesthetics, Gc did not change, not sup-
responses to isocapnic changes in arterial pH induced by
porting an other-than-additive central-peripheral interac-
infusion of acid and bicarbonate performed at quite dif-
tion (161, 796, 797).
ferent background PCO2 levels. With the assumption that
with acid-base changes the peripheral chemoreceptors
4. Dynamic end-tidal forcing in carotid body
respond uniquely to changes in arterial [H⫹] (190, 292,
resected subjects
334, 687), these findings provided no evidence for an
Bellville (56) reported an ⬃50% lower central chemo- interaction other than simply additive (136).
sensitivity in carotid body-resected subjects compared Recently, we measured hypoxic sensitivity in hu-
with healthy controls, indicating that a peripheral-central mans, defined as the ratio ⌬V̇I/⌬log PaO2, at three different
interaction other than simply additive cannot be ex- levels of constant end-tidal CO2. The rationale of using the
cluded. One to 26 six years after bilateral removal of the ratio ⌬V̇I/⌬log PaO2 rather than the conventional ratio
carotid bodies due to paraganglioma, subjects displayed a ⌬V̇I/⌬SpO2 is explained in section VIB (see also Ref. 775).
lower central CO2 sensitivity than an age- and sex- As also demonstrated in the cat (650), humans display a
matched control group (227). Three days after carotid steep and sound linear relationship between hypoxic sen-
body resection (due to hereditary paraganglioma), three sitivity and arterial [H⫹], representing a powerful O2-CO2
patients showed a substantially reduced central CO2 sen- interaction (Fig. 7; Ref. 775). But is hypoxic sensitivity a
sitivity that gradually recovered in the following 1–3 years unique function of arterial [H⫹]? This seems unlikely be-
(154). In the anesthetized cat, carotid sinus nerve transec- cause it would imply an almost infinitely high sensitivity
tion did not reduce central CO2 sensitivity in an acute in a metabolic acidosis with a pH of ⬃7.3 ([H⫹] ⬃50 nM)
setting (314). Thus removal of a tonic input from the which, for example, can be induced by the carbonic an-
carotid bodies could lead to a resetting of central CO2 hydrase inhibitor acetazolamide (775, 778). To investigate
sensitivity that needs time to develop. Whether such a this further, bicarbonate was infused isocapnically, and

Physiol Rev • VOL 90 • APRIL 2010 • www.prv.org


THE VENTILATORY RESPONSE TO HYPOXIA IN MAMMALS 709

eucapnia (the stimulus consisted of a rapid desaturation


within 30 s with the end-tidal CO2 briefly adjusted to the
subject’s eucapnic value). These data may suggest that an
augmented carotid body output is translated into an in-
crease in respiratory motor output only at sufficiently
high central chemoreceptor PCO2 levels. Corne et al. (144)
used volume cycle-assisted ventilation to produce steady-
state mild and moderate hypocapnia while maintaining
stable spontaneous breathing. At arterial PCO2 values 5–10
Torr below eucapnia, but not in mild hypocapnia or eu-
capnia, a ventilatory response to hypoxia was totally ab-
sent. Although clearly of clinical significance, a clear pic-
FIG. 7. Relationship between hypoxic ventilatory response and ture of peripheral-central interaction cannot be deduced
arterial H⫹ concentration in humans. Hypoxic sensitivity, defined as from this approach because a separation with O2-CO2
⌬V̇I/⌬log PaO2 as a function of arterial [H⫹]. The right line represents the
condition with normal arterial bicarbonate concentration in the arterial interaction at the carotid bodies cannot be made. In ad-
blood. The acute hypoxic response (i.e., the response 3 min after a step dition, an influence of developing HVD in these subjects
decrease in end-tidal O2) is measured at three constant levels of arterial cannot be excluded even though the hypoxic tests did not
PCO2 (numbers are arterial PCO2 values). There is a sound linear rela-
tionship between hypoxic sensitivity and arterial H⫹ concentration in- outlast 5 min: at the end of the hypoxic test in moderate
dicating a powerful O2/CO2-H⫹ interaction at ventilation level. When hypocapnia (in the absence of an HVR), subjects venti-
subjects are given intravenous bicarbonate (left line), the line shifts to
lower values of arterial [H⫹] in an approximately parallel way. Thus lated less than in the prehypoxic period suggesting HVD
apparently hypoxic sensitivity is not a unique function of the arterial (144).
[H⫹]. Points a and f are virtually isohydric points, and the lower hypoxic Yang et al. (863) introduced a method for estimating
sensitivity in point a is presumably due to the lower PCO2 in the carotid
bodies. See text for further explanation. Data are means ⫾ SE from 8 the dynamic response to simultaneous changes in PCO2
young healthy volunteers. [Adapted from Teppema et al. (775).] and PO2 in humans, and their data could be well described
with a two compartment model, one comprising the slow
this yielded two isocapnic and two virtually isohydric CO2 and the other the fast O2-CO2 interaction component.
situations with different hypoxic sensitivities as shown in The interaction could be entirely modeled by the fast
Figure 7. From these data (775), we can draw several component attributed to the peripheral chemoreceptors,
conclusions. First, O2 sensitivity increases with an isocap- thus not supporting a significant central O2-CO2 interac-
nic rise in arterial [H⫹], and this is presumably due to tion and/or more than simply additive peripheral and
interaction at the carotid bodies (cf. points c and f and b central effects.
and d in Fig. 7). Second, although with acid-base alter- Agents not crossing the blood-brain barrier and not
ations extracellular H⫹ may be the adequate stimulus to influencing central CO2 sensitivity could be used to
the carotid bodies, there is no unique relationship be- investigate possible selective effects on the carotid
tween hypoxic sensitivity and arterial [H⫹] (cf. points f bodies. Acetazolamide, a moderately permeable car-
and a in Fig. 7). Thus, in this regard, our data do not bonic anhydrase inhibitor, will not have penetrated into
support earlier conclusions (e.g., Refs. 250, 349), but nei- the brain in significant amounts 1 h after administration
ther do they provide conclusive evidence for the carotid
of a low dose. About 1 h after infusion of 4 mg/kg, the
bodies as the exclusive site of the O2-CO2/H⫹ interaction.
O2-CO2 interaction in healthy subjects was completely
The lower hypoxic sensitivity in point a compared with f
abolished, and this was reversible with antioxidants
in Figure 7 could be due to the lower carotid body PCO2
(Fig. 8; Ref. 777). Infusion of 500 mg in humans did not
(peripheral O2-CO2 interaction) combined with a dimin-
ished efficacy of afferent carotid body input to increase change hyperoxic CO2 sensitivity, indicating an unal-
ventilation due to the also lower central PCO2. Perhaps a tered central ventilatory CO2 sensitivity (750). With the
more likely explanation is that it is the intracellular envi- assumption of no central effects of acetazolamide,
ronment (pH?) of carotid body cells that, together with these findings suggest that in humans, the O2-CO2 in-
the extracellular pH, determines O2 sensitivity. teraction entirely resides in the carotid bodies. A usual
Roberts et al. (660) subjected humans to mechanical clinical oral dose of acetazolamide does not affect the
ventilation to produce apnea at different end-tidal CO2 interaction, indicating that its effect depends on the
levels varying from 2.5 Torr above to 7.5 Torr below dose and route of administration (750, 775). Animal
eucapnia. The background end-tidal CO2 dose-depen- data suggest that analogous to its effect on hypoxic
dently influenced the magnitude of the EMG response to pulmonary vasoconstriction (749), the inhibitory action
a “standard” peripheral chemoreceptor stimulus that was of acetazolamide on the HVR may occur independently
completely absent at an end-tidal CO2 ⬃7.5 Torr below from carbonic anhydrase inhibition (776).

Physiol Rev • VOL 90 • APRIL 2010 • www.prv.org


710 LUC J. TEPPEMA AND ALBERT DAHAN

FIG. 8. Effect of acetazolamide on O2-CO2 interaction in humans. Left: intravenous low-dose (4 mg/kg) acetazolamide (ACTZ) lowers the acute
hypoxic response in normocapnia (N) but also abolishes the increase in its magnitude in hypercapnia (H). Thus this agent, which at the applied dose
is unlikely to have direct effects on the central nervous system, is able to abolish the O2-CO2 interaction by a peripheral action most likely located
in the carotid bodies. An antioxidant cocktail (AOX; ␣-tocopherol and ascorbic acid) reversed the effects of ACTZ. Right: control study in which
after the ACTZ, subjects received placebo (PLCB) instead of AOX. See text for further explanation. [From Teppema et al. (777).]

6. Conclusion ment of local tumors and/or hyperplasia that, for example,


may arise from genetic defects (see sect. IV) and from
In all species investigated, O2 and CO2 have multipli-
chronic hypoxemia related to cyanotic heart disease, pul-
cative actions on carotid body output both when awake
monary disease, and high altitude (154, 312). Inadvertent
and anesthetized, but the interaction site within type I
loss of carotid body function may arise from surgical
cells has not yet been established. The existence of a
procedures in the neck region, such as carotid endarteri-
multiplicative O2-CO2 interaction on ventilation depends
ectomy and neck dissection as part of the treatment of
on the species, state of consciousness, and the peripheral-
head and neck cancer (524, 812).
central interaction mode. Generally, humans show O2-
Both animal and human studies show immediate hy-
CO2 multiplication, but it appears difficult to characterize
poventilation, respiratory acidosis, and loss of the hyp-
the mode of peripheral-central interaction, although there
oxic drive in response to bilateral carotid body resection
is no conclusive evidence for hypoadditive central and
(Fig. 5; Refs. 154, 473, 474, 492, 570, 664). In a small
peripheral effects. Direct anatomical connections be-
human population tested before and after bilateral re-
tween the first relay station of the peripheral chemore-
moval of the carotid bodies for tumors due to a mutation
ceptors in the NTS and central chemoreceptors in the
in the SDHD gene, end-tidal PCO2 increased by ⬎8 Torr
retrotrapezoid nucleus (RTN) as described for the rat
within the first day after resection (154). Similar observa-
(753), may render a clear separation of central and pe-
tions were made in awake rats over a 75-day period after
ripheral contributions elusive. In addition, a subset of CO2
carotid body excision (570). These data indicate that the
chemoreceptors in the RTN receives an inhibitory input
carotid bodies are essential in maintaining normal venti-
from (mainly slowly adapting) pulmonary stretch recep-
lation during air breathing. Especially in the neonatal
tors (525), providing another complicating factor in the
period, elimination of the carotid bodies can have pro-
interpretation of data from intact as well as vagotomized
found effects on the control of breathing, depending on
animals.
the denervation technique used and the developmental
period in which it is performed (238 and references cited
E. Carotid Body Resection and the HVR therein). Partial restoration of the hypoxic drive is ob-
served in some species (rat, goat, cat, pony, piglet) and is
Resection of the human carotid bodies was per- most likely in neonatal animals (69, 473, 474, 492, 710).
formed for a variety of reasons. Both unilateral and bilat- The rate of recovery is species dependent and ranges
eral resections were carried out in patients with severe from weeks (piglet, rat) to months (goat) to years (pony).
bronchial asthma and chronic obstructive pulmonary dis- The compensatory mechanisms of the partial recovery of
ease. Although the main goal for resection in these pa- the hypoxic drive in these animal studies remain un-
tients was often achieved (that is, the alleviation of known. Potential mechanisms include reinforcement of
breathlessness), the therapy is controversial and cur- aortic bodies, activation of medullary oxygen-sensing
rently not practiced on a wide scale (326, 475, 812). An- neurons, chemosensory input from secondary glomus tis-
other reason for carotid body resection is the develop- sue in the head/neck region (such as the vagal body in the

Physiol Rev • VOL 90 • APRIL 2010 • www.prv.org


THE VENTILATORY RESPONSE TO HYPOXIA IN MAMMALS 711

neck), regeneration of sensory terminals of the sinus during chronic hypoxia, these neurons may receive a
nerve, and/or plastic changes in the central respiratory time-dependent progressively increasing synaptic input
neuronal network. In humans, we were unable to observe that may overcome inhibitory influences of a lower
any recovery in the HVR over a period of 4 years after PCO2 and may thus contribute to VAH.
bilateral removal of the carotid bodies (154). Similar ob-
servations were reported by Wade et al. (812) in patients 2. VAH is associated with an increase of the HVR
up to 8 years postsurgery. Honda et al. (326) did show
Studies in the last two decades have convincingly
some restoration of the HVR in patients two decades after
shown that VAH is associated with a time-dependent aug-
bilateral carotid body resection, although this effect was
mentation of the HVR that at least partly is due to an
dependent on the specific hypoxic test employed.
increased carotid body sensitivity. Initial studies in dogs,
ponies, goats, and rats had already demonstrated that
F. Chronic Hypoxia and the HVR carotid body denervation abolished or at least minimized
VAH (reviewed in Ref. 68). Below we summarize the most
1. Ventilatory acclimatization to chronic hypoxia important studies in goats, cats, rats, and humans show-
ing the crucial role of both the carotid bodies and the CNS
We define chronic hypoxia as exposure to hypoxia or
in mediating an increase in the HVR in chronic hypoxia.
high altitude lasting several hours to days, weeks, or
months. A hallmark of adaptation to high altitude is a
1. Animal studies
gradual rise in ventilation with a time course depending
on the altitude (ventilatory adaptation to high altitude, In goats (mainly awake) with vascularly isolated ca-
VAH). In humans, at very high altitudes (8,000 m and rotid bodies, Bisgard, Forster, and co-workers collected
higher), this may take at least 30 days (839). At less many data on the mechanism of VAH that can be summa-
extreme altitudes, complete VAH requires ⬃10 days (see rized as follows (66, 67, 105, 200, 216, 546, 836; further
Ref. 68 and references cited therein), although at 3,800 m references in Ref. 68). 1) Animals exposed to chronic
ventilation appeared not to have reached a plateau yet hypoxia show complete VAH within 4 – 6 h. 2) Isolated
after 12 days (683). Dogs, goats, and ponies acclimatize CNS hypoxia and/or hypocapnia do not evoke VAH, indi-
much faster and reach complete adaptation after 3, 4 – 6, cating that it must have a peripheral origin. 3) VAH re-
and 30 h, respectively (references in Ref. 68). When ex- quires intact carotid bodies that are perfused by hypox-
posed to a simulated altitude of ⬃4,600 m, cats show a emic blood; isolated carotid body hypercapnia results in a
large decrease in arterial PCO2 after 48 h (809), but accli- rapid sustained rise in ventilation but does not induce the
matization may not yet be complete after this period in time-dependent fall in arterial PCO2 typical for VAH.
this species since at 5,500 m arterial PCO2 continued to fall 4) Both chronic poikilocapnic and isocapnic carotid body
for 10 –12 days (774). Qualitatively, in the rat, VAH is hypoxias increase the isocapnic acute hypoxic response
rather similar to that in humans albeit with a somewhat (AHR), indicating that the enhanced sensitivity develops
faster time course, and this species is considered an regardless of the PCO2 at the carotid bodies. 5) During
adequate animal model for the adaptation in humans isocapnic carotid body hypoxia for 6 h, carotid sinus
(569). nerve afferents of anesthetized goats show a progressive,
Initially it was suggested that VAH could be attrib- time-dependent increase in impulse frequency that quantita-
uted to changes in arterial and later to brain stem acid- tively can account for VAH.
base status, but eventually it became clear that alterations When exposed to a barometric pressure of 460 – 440
in acid-base status during VAH should be considered the Torr for 2–3 days, cats demonstrated a large time-depen-
consequence of ventilatory changes rather than the cause dent drop in end-tidal CO2, increases in both the HVR, and
(178). Here we do not focus on studies aimed at elucidat- afferent carotid sinus nerve activity as well as an aug-
ing the role of the central chemoreceptors in VAH, and the mented isocapnic carotid body sensitivity to hypoxia
reader is referred to excellent reviews on this subject (68, (771, 808). Animals exposed to 10% O2 exhibited a higher
540, 543). Briefly, there is no conclusive evidence for an isocapnic carotid sinus nerve sensitivity to hypoxia after
essential role of the central chemoreceptors, for example, 21 days but not during the initial few hours (34). In
because the time courses of medullary extracellular fluid animals kept at a constant end-tidal O2 of 30 Torr for 7 h,
pH and VAH do not run in parallel (534, 543). Future nerve activity started to increase after 3 h (181). However,
studies are warranted, however, to unmask a possible Lahiri et al. (428) were unable to demonstrate an increase
role of central chemoreceptors in VAH for a reason not in afferent nerve sensitivity to hypoxia after acclimatiza-
anticipated before: during hypoxia in the rat, central tion to 10% O2 for 21–24 days. Finally, Tatsumi et al. (768),
chemoreceptors in the retrotrapezoid nucleus are after keeping cats at 375 Torr for 3– 4 wk, reported a
strongly activated via a glutamatergic pathway from time-dependent decrease in end-tidal CO2 along with a
carotid body projection areas in the NTS (753). Thus, decreased HVR and isocapnic hypoxic carotid sinus nerve

Physiol Rev • VOL 90 • APRIL 2010 • www.prv.org


712 LUC J. TEPPEMA AND ALBERT DAHAN

sensitivity. An altitude-dependent decrease in HVR along 2. Studies in humans


with VAH had already been reported in earlier studies
(e.g., Ref. 774; further references in Ref. 68). Thus, after a In Table 3, we have collected studies mainly per-
few days of hypoxia, cats appear to have an augmented formed over the last 15 years (for a review of earlier
HVR and an increased carotid body sensitivity, but after studies, see Ref. 68). Despite all the distinct exposure
more than 2–3 wk, they show both a blunted HVR and paradigms (stimulus level and duration, hypobaric hyp-
carotid body sensitivity. So, perhaps an increase in ca- oxia, or just lowered inspired O2), techniques and analysis
rotid body sensitivity is sufficient for VAH in the first few methods that make quantitative comparisons very diffi-
days, but other mechanisms manifest after 2–3 wk. cult if not impossible, the outcomes of all these studies
The rat develops an augmented poikilocapnic HVR have two things in common: an increase in resting and, if
and ventilation in the initial few days of chronic hypoxia measured, hyperoxic ventilation and an augmented (acute)
followed by a gradual return of the HVR to control or even hypoxic ventilatory response that develops during the
slightly below at the end of the first week (51, 91, 252, first 4 – 8 h of acclimatization.
811). However, a fast “blunting” of the HVR in the rat has Robbins (659) made considerable efforts to spell out
not been demonstrated very convincingly, for example, the mechanisms of ventilatory adaptation to chronic hyp-
because in all these studies, it was measured under oxia. Apart from a consistent increase in hyperoxic ven-
poikilocapnic conditions. Rather, it may be a manifesta- tilation and an augmented acute response to hypoxia, he
tion of a gradual attenuation of acclimatization as also found the following: 1) in agreement with results obtained
occurs in other species, but without blunting of the HVR from goats, isocapnic and poikilocapnic hypoxias have
(68). Aaron and Powell (1) showed a clear increase in the same effect. 2) Neither eucapnic nor hypocapnic pas-
isocapnic HVR in rats after 7 wk at 380 Torr. Note that sive hyperventilation evokes VAH or an enhanced HVR,
after acclimatization this enhanced HVR was measured indicating that hypocapnia or hyperpnea per se will not
at a constant arterial PCO2 that was lower than in control play a significant role. 3) The effect of hypoxia to induce
by as much as 6 – 8 Torr, whilst both control and acclima- the adaptations is due to the low PO2 rather than to low
tized animals showed a positive O2-CO2 interaction (1). oxygen content. 4) The mechanism underlying the in-
These authors also compared isocapnic HVRs of acclima- crease in the acute response to hypoxia is fast (observ-
tized animals with that of controls that were made slightly able within a few hours as in goats), sensitive (end-tidal
hypercapnic to achieve matching of minute ventilation O2 10 Torr below resting is sufficient to evoke it), inde-
(613). In control animals, the constant arterial PCO2 was pendent of peripheral adrenergic and dopaminergic sys-
kept at a level that was higher than in acclimatized ani- tems, and insensitive to general autonomic blockade (ref-
mals by ⬃9 Torr, but despite this, the HVR in the latter erences in Table 3).
was much higher, suggesting that either the carotid bod- Chronic isocapnic and poikilocapnic hypoxia also
ies had undergone an impressive increase in O2 sensitivity alter peripheral ventilatory CO2 sensitivity. Using dy-
and/or that the translation of afferent carotid body activ- namic end-tidal forcing (see sect. VI) and sophisticated
ity into ventilatory output (CNS gain) was greatly facili- analysis methods, Fatemian and Robbins showed an in-
tated. It would be interesting to compare HVRs at equal crease in CO2 sensitivity that most likely can be attributed
hypercapnic PCO2 levels in control and acclimatized rats, to an increase in sensitivity of the carotid bodies (e.g.,
because it cannot be excluded that a negative O2-CO2 Refs. 228, 229; further references in Table 3). In a study at
interaction (145, 172) may have played a role in the ob- 4,300 m (629), the amount of VAH correlated well with the
servations of Powell et al. (613). magnitude of the HVR at sea level, also supporting an
Dwinell and Powell (201) found a large increase in important role of the peripheral chemoreflex loop.
the CNS gain of a standard electrical input from the In a more recent study, Herigstad et al. (320) exposed
carotid sinus nerve in acclimatized rats (380 Torr, 1 wk). subjects to 8 h of isocapnic hypoxia prior to an incremen-
However, cats exposed to 440 Torr for 48 h developed tal exercise protocol. The exposure resulted in a decrease
both a greater isocapnic HVR and hypoxic carotid sinus in end-tidal PCO2 that was essentially maintained at all
nerve response but did not show an increased CNS gain work loads until exhaustion. According to the alveolar
(808). In goats, the effect of NaCN (assumed to cause ventilation equation, this means that ventilation must
maximal carotid body stimulation) on ventilation was not have increased in proportion to metabolism. The data
different before and after 4 h of isocapnic or poikilocap- from this study also implied that an altered acid-base
nic hypoxia (215), but this does not preclude changes in balance (isocapnia was maintained) or long-lasting in-
CNS gain during prolonged hypoxia in this species (615). creases in ventilation (days) are not required for this
Thus, in the rat, the most frequently used animal model to phenomenon to occur. Thus VAH augments the ventila-
study VAH, an increased central gain has clearly been tory response to exercise and in this sense resembles the
demonstrated, while it cannot be excluded to exist also in effect of hypoxia or carotid body stimulants (references
humans, goats, and ponies (references in Refs. 178, 615). in Ref. 320). Exercise also augments the stimulatory effect

Physiol Rev • VOL 90 • APRIL 2010 • www.prv.org


THE VENTILATORY RESPONSE TO HYPOXIA IN MAMMALS 713

TABLE 3. Carotid body adaptations to chronic hypoxia

Does the Adaptation Increase or


Species Effects and Comments Decrease CB Sensitivity or the HVR? Reference Nos.

Neurochemical adaptations
Dopaminergic system
Rat Upregulation of TH 65, 614 and references
cited therein
Cat TH upregulation 820
Cat CH augments HVR after 2 days at 4,600 m Increase 771
but domperidone fails to further
increase it, suggesting dopaminergic
downregulation
Cat Prolonged hypoxia decreases HVR; Decrease 770
domperidone restores it to control
level, suggesting a role of DA
upregulation in long-term adaptation
Cat Dopaminergic system in efferent Decrease 429
inhibitory fibers in the CSN is
upregulated not supporting disinhibition
of this system in CH
Rat After 5 and 2–3 wk of CH, domperidone Decrease 252, 811
restored “blunted” poikilocapnic HVR,
attributed to increased CB DA content
Rat 1–2 days of CH attenuate dopaminergic Increase and decrease, 51, 342
inhibition of the HVR, but after 8 days respectively
it has increased compared to control
Rat CB D2R decreased by 59% after 2 days of Increase and decrease, 344
CH, but increased by 274 % after ⬃11 respectively
days
Rabbit After 2 days of CH, CB 关DA兴 decreased, ? 343
but 2 days after termination of an 8-day
lasting exposure it was higher than
control
Mice Male D2R⫺/⫺ mice failed to show VAH Increase, but note possible 343
and augmented HVR after 2–8 days of central effects
CH
Rat Type I cells of juvenile rats show ? 355
exaggerated basal DA release
Goat The peripheral DA antagonist Neither 358, 359
domperidone had equal effects in
acclimatized and control animals, and
local CB application of DA did not
influence the time course and
magnitude of VAH
Human Effects of DA and domperidone did not Neither 590
change after 8 h of CH
Norepinephrine
Cat CH for 24–36 h reduced the inhibitory Increase 110
effect of ␣2-adrenergic antagonism,
suggesting downregulation of CB
noradrenergic mechanisms
Various Upregulation of CB NE Decrease References in 65, 278, 818
species
Goat CB sympathectomy did not alter the time Neither 672
course of VAH, the acute HVR and the
inhibitory effects of NE and DA
Human VAH and CH-induced augmentation of the Neither 134, 452, 520
HVR are not affected by ␤-adrenergic
and autonomic ganglion blockade
Acetylcholine
Rat In CB preps from animals exposed to CH Increase 309
for 9–22 days, ACh had a larger
excitatory effect on CSN discharge
compared with control; however, the
nAChR antagonist mecamylamine failed
to cause any reduction in hypoxia-
induced increase in CSN activity

Physiol Rev • VOL 90 • APRIL 2010 • www.prv.org


714 LUC J. TEPPEMA AND ALBERT DAHAN

TABLE 3. —Continued

Does the Adaptation Increase or


Species Effects and Comments Decrease CB Sensitivity or the HVR? Reference Nos.

Rat In type I cells cultured in normoxia Increase 355


mecamylamine did not alter basal DA
release, but in those cultured in hypoxia
for ⬃12 days it completely inhibited it,
possibly explaining its failure to inhibit the
hypoxia-induced rise in the preparation of
He et al. (309)
ATP
Rat In superfused CB preps from rats exposed to Increase 307
CH for 9–16 days, P2X2 purinergic but not
nicotinic antagonists retained the ability to
block hypoxia-induced rise in CSN activity,
suggesting an altered purinerg/cholinergic
balance; afterdischarge in CSN from
acclimatized animals was blocked by
purinergic blockers suggesting prolonged
ATP release as its cause and hypothetically
providing a mechanism for continuous
hyperventilation upon return to normoxia
from high altitude (deacclimatization)
Nitric oxide
Rat Upregulated in hypoxia Decrease 65, 865 for review
Rat In CB-CSN preps from rats exposed to CH Decrease 308
for ⱕ16 days, L-NAME caused a
progressively greater stimulation and an
NO donor a larger inhibition
Substance P
Cat CB Substance P downregulated in animals Decrease 820
exposed to 10% O2 for 2 wk
Endothelin-1
Rat Upregulation of ET1, and ETA receptors in Increase 128
type I cells from animals exposed to CH
for ⱕ16 days. Selective inhibition of ETA
receptors reduced basal CSN activity on
day 0 by 11%, but halved it at day 16
Rat During CH for 16 days, daily administration Increase 129
of the ETA/B antagonist bosentan
diminished the increase in hypoxic CSN
activity and also greatly reduced CB
enlargement and mitotic type I cell activity
Human CH for 4 h increased plasma 关ET-1兴; Increase, assuming a stimulatory 755
continuous infusion of ET-1 during a 4 h effect of ET-1 on the CB
bout of CH exaggerated the increase in
HVR
Angiotensin II
Rat 10% O2 for 4 wk progressively increased CB Increase 431, 432, 443
angiotensinogen mRNA, upregulated AT1
receptors and ACE, and increased CB
sensitivity to ANG II
Proinflammatory
cytokines
Rat Upregulation of IL-1, IL-6, and TNF-␣ and Increase 433
their receptors in type I cells from rat
exposed to 10% O2 for 3, 7 and 28 days;
after CH, the hypoxia-induced rise in
关Ca2⫹兴i was enhanced by the cytokines
Rat Rats exposed to a barometric pressure of 380 Increase 455
Torr for 4 wk showed increased CB 关IL-1␤兴
and 关TNF-␣兴 after 1 day, well before
macrophage invasion; cytokine levels
normalized after 28 days, except IL-6 due
to increased production by the CB;
ibuprofen and dexamethasone inhibited
CH-induced increase in hypoxic CSN
activity

Physiol Rev • VOL 90 • APRIL 2010 • www.prv.org


THE VENTILATORY RESPONSE TO HYPOXIA IN MAMMALS 715

TABLE 3. —Continued

Does the Adaptation Increase or


Species Effects and Comments Decrease CB Sensitivity or the HVR? Reference Nos.

Melatonin
Rat Exposing rats to 10% O2 for 4 wk Increase 785
increased CB melatonin receptor mRNA
Adaptations of ion channels and membrane currents
Maxi-K channels Rat Decrease in K⫹ current density but not Increase 114
that caused by maxi-K channels,
proposed to result in a relative larger
contribution of the latter to the resting
membrane potential and an increased
hypoxia-induced catecholamine release
Sodium current Rat Higher current density in type I cell Increase 726, 727
isolated from 5- to 12-day-old rat and
cultured in hypoxia for 1–2 wk
“Oxygen-sensitive Rabbit In type I cells cultured in hypoxia for Increase 368
and -insensitive” Kv 72 h, potassium channels responsible
channels for an O2-insensitive component of the
K⫹ current were downregulated and
those contributing to the O2 sensitive
component upregulated; thus K⫹
current inhibition by hypoxia was
increased
Sodium channel Rat CB from animals exposed to 10-11% O2 Increase 106
Nav1.1 for 1 wk contained type I cells with
upregulated voltage-gated sodium
(Nav1.1) channels; the sodium channel
activator veratridine caused a
severalfold greater increase in CA
release in these cells, compared with
those from control animals
Gap junctions Rat Exposure to an ambient pressure of 380 Increase? 127
Torr was followed by increased
expression of the gap junctional protein
connexin43 in type I cells and petrosal
ganglion neurons
Morphologic adaptions of the carotid body
Various Organisms living at high altitude have Increased in short-term CH but 19, 209, 419, 818
species enlarged CB decreased in prolonged
hypoxia
Rat CH induces vascular remodeling, Unknown 135, 419, 420, 784, 818
endothelial proliferation, and
vasodilation
Rat Hyperplasia of type I cells during the first Increase 818, 819
week of CH, no further increase
thereafter; newly formed cells can
survive for 1 mo
Mice Exposing mice to CH for 20 days resulted Increase 581
in cell division in CB after 1 day, but
new type I cells appeared after 5 days.
Initially type II cells start to divide and
are replaced by “progenitor,” i.e., stem
cells that differentiate into type I cells
with normal electrophysiological
features. Upon return to normoxia, 50%
of the type I cells are freshly formed
and the total amount gradually returns
to normal

ACh, acetylcholine; ACE, angiotensin converting enzyme; ANG II, angiotensin II; AT1 receptor, angiotensin I receptor; CH, chronic hypoxia;
CA, catecholamine; CSN, carotid sinus nerve; CB, carotid body; DA, dopamine; D2R, dopamine 2 receptor; ET, endothelin; L-NAME, NG-nitro-L-
arginine methyl ester, an inhibitor of NO synthase; HVR, hypoxic ventilatory response; IL, interleukin; NE, norepinephrine; nAchR; nicotinic
acetylcholine receptor; NO, nitric oxide; TH, tyrosine hydroxylase; TNF, tumor necrosis factor; VAH ventilatory adaptation to chronic hypoxia or
high altitude.

Physiol Rev • VOL 90 • APRIL 2010 • www.prv.org


716 LUC J. TEPPEMA AND ALBERT DAHAN

of hypoxia on ventilation, but this interaction is reduced the carotid bodies that act together to increase hypoxic
by a prior exposure to chronic hypoxia, and this phenom- sensitivity. These adaptations occur in parallel, so their
enon will be one of the determinants of the magnitude of separate contribution to the increase in carotid body gain
the HVR at high altitude (848). is difficult to assess. This is especially true for the contri-
A decrease in intercept of the hyperoxic CO2 re- bution of neurotransmitters and/or modulators, some of
sponse curve was not observed by Fatemian and Robbins which have a controversial role in the carotid bodies. For
(228, 229), suggesting that the increase in resting and example, the dopaminergic system seems to be upregu-
hyperoxic ventilation is due to a higher (peripheral) CO2 lated in chronic hypoxia, but although dopamine is gen-
sensitivity. In one study, a leftward shift of the ventilatory erally considered an inhibitory neurotransmitter (mainly
CO2 response curve was found after as long as 75 h of based on observations on the effects of exogenous ap-
poikilocapnic but not isocapnic hypoxia (148). Studying plied agonists and antagonists), the significance of this
the effect of chronic hypoxia with modified rebreathing adaptation is uncertain because endogenous dopamine
(see sect. VI) yields a decrease in intercept of both the may well have excitatory effects on the carotid bodies
hypoxic and hyperoxic CO2 response curves at high alti- (for a discussion on the controversial role of dopamine in
tude, but, possibly related to variation in altitude and the carotid body, see Refs. 278, 283, 869). Both an inhib-
acclimatization period, no consistent increase in periph- itory and excitatory role has also been suggested for
eral CO2 sensitivity (9, 719). norepinephrine (references in Refs. 68, 278). In Table 4,
Two major mechanisms, to be discussed in the fol- we have summarized recent studies on various adapta-
lowing paragraphs, may contribute to the augmented HVR tions of the carotid bodies to chronic hypoxia. We have
in chronic hypoxia: an augmented carotid body sensitivity added a column containing the suggested effects of single
and, as particularly studied in the rat, an increase in CNS adaptations on carotid body sensitivity, but we emphasize
gain. Animal studies do not provide direct evidence for an that with regard to neurotransmitters these must be con-
important role of the sympathetic nervous system (e.g., sidered hypothetical, because what eventually matters is
Ref. 672). In humans, prolonged hypoxia causes a large the total balance between excitatory and inhibitory trans-
increase in sympathetic activity, but how this may influ- mitters. In a recent review, Prabhakar (621) has proposed
ence the HVR is unknown (107, 300). Also, note that the the simultaneous presence of excitatory and inhibitory
chronic hypoxia-induced HVR augmentation in humans transmitters to result in a “push-pull” mechanism between
was reported to be unaltered after autonomic ganglion both that, rather than causing exhaustion in the absence
blockade (452). Neonates and adults adapt differently to of inhibitors, produces sustained activation in their pres-
chronic hypoxia. This is schematically shown in Figure 9. ence. This would be especially effective in the adaptation
chronic hypoxia (621).
3. Chronic hypoxia increases carotid body sensitivity
Chronic hypoxia elicits morphological, neurochemi- 4. Increase of CNS gain during chronic hypoxia
cal, and membrane-electrophysiological adaptations in
An increased CNS gain as observed in the rat could
result from neuroplasticity of the brain stem respiratory
circuitry that processes the afferent input from the carotid
bodies. Below we discuss some mechanisms potentially un-
derlying the increase in CNS gain in chronic hypoxia.
A) CATECHOLAMINERGIC SYSTEM. Rats challenged with 10%
O2 for 1 wk showed a reduced brain dopamine turnover
and an increased carotid body dopamine content (571). In
rats exposed to chronic hypoxia for 2 wk, Schmitt et al.
(685) found a positive correlation between the rise in
ventilation and tyrosine hydroxylase (TH) protein level in
the caudal NTS, i.e., the preferential projection area of
. . chemoafferent fibers. The same area, containing the A2
VI VI
noradrenergic cell group, displayed an upregulation of TH
after 1 wk of chronic hypoxia, but this effect was reversed
after 2 and 3 wk of exposure, respectively (722). When
administered in the NTS, dopamine has a stimulatory
FIG. 9. Comparison of chronic hypoxia in neonates and adults. influence on ventilation (for references, see Ref. 345), and
Effects of chronic hypoxia in neonates, within the critical time window thus an increased tyrosine hydroxylase production may
for developmental respiratory plasticity, compared with those in adults.
Question marks concern unresolved or controversial issues. V̇I, inspired facilitate respiratory output in chronic hypoxia. In male
ventilation. For explanations see text. normoxic mice systemic droperidol, a dopamine antago-

Physiol Rev • VOL 90 • APRIL 2010 • www.prv.org


THE VENTILATORY RESPONSE TO HYPOXIA IN MAMMALS 717

TABLE 4. Chronic hypoxia in humans

Reference
Protocol HVR Measurement Effects on HVR, Comments Nos.

Operation Everest II: subjects Progressive isocapnic rebreathing at 1 iHVR about equal at both pressures. 686
acclimatized to 428 and 305 Torr PETCO2 of ambient air breathing Further elevation upon return to sea
level but note that the test PETCO2
was ⬎10 Torr higher than at the
lowest pressure
Exposure to PB 447 Torr for 7 days Progressive isocapnic rebreathing at 1 iHVR 2–4 days after exposure; 273
a PETCO2 of ambient air breathing postacclimatization resting PETCO2,
however, not provided.
Stay at 4,300 m for 19 days Progressive isocapnic rebreathing at Higher HVR at sea level predicts lower 629
sea level only; serial ventilation PETCO2 during the entire period
and gas measurements from day 1
to 19
pH: 5 days at 3,810 m Stepwise decreases in SpO2 to 77%. Gradual increase of iHVR from 1-5 683
PETCO2 at hyperoxic value with days; test PETCO2 varied between 45
V̇I ⫽140 ml 䡠 min⫺1 䡠 kg⫺1 and 33 Torr
pH: 12 days at 3,810 m Modified rebreathing; PETCO2 at HVR doubled after 12 days; at altitude 682
hyperoxic value with V̇I ⫽140 hyperoxic PETCO2 with V̇I ⫽140
ml 䡠 min⫺1 䡠 kg⫺1 ml 䡠 min⫺1 䡠 kg⫺1 decreased by 8-10
Torr
pH: 1 and 2 days at 5,050 m Modified rebreathing; PETCO2 at 1 iHVR (5- to 7-fold after 1 and 2 348
about resting value at sea level days, respectively)
iH or pH (PETO2 55 Torr) during 8 h HVR not tested Time-dependent rise of V̇I in iH, not pH 338
iH or pH (PETO2 55 Torr) during 8 h DEF, square-wave and saw-tooth 1 iAHR over 8 h; no difference 337
stimuli; analysis of HVR with first- between protocols, suggesting no
order model; PETCO2 1.5 Torr influence of the PCO2; both iH and
above initial resting in all tests pH increased baseline hyperoxic
ventilation and peripheral ventilatory
CO2 sensitivity
iH or pH (PETO2 55 Torr) during 8 h Testing the effects of isocapnic step Time-dependent rise in hyperoxic 762
hyperoxia ventilation in both protocols that
continued for at least 8 h
postexposure
iH or pH (PETO2 60 Torr) during 48 h Idem 1 iAHR over this period in both 763
protocols, gradually reversing over
the subsequent recovery period; no
influence of PCO2
iH or pH (PETO2 55 Torr) during 8 h No testing of HVR, testing overall Increase in CO2 sensitivity after both 228
CO2 sensitivity protocols; no change in x-intercept
of the CO2 response curve
Normobaric iH or pH (PETO2 55 Torr) No HVR testing, testing effect on Overall increase in CO2 sensitivity due 229
during 8 h peripheral and central CO2 to increase in peripheral gain.
sensitivity with DEF and model Contribution, however, of central
analysis chemoreceptors could not be
excluded. No change in x-intercept
of CO2 response curve
8 h of normobaric iH without drug, DEF, square-wave stimuli, model 1 iAHR after 8 h; dopamine and 590
with dopamine and with analysis; PETCO2 1.5 Torr above domperidone elicited equal effects on
domperidone, respectively initial resting in all tests the HVR before and after the iH,
indicating no carotid body
dopaminergic involvement in the
effect of iH
iH (PETO2 50 Torr) for 8 h with and Idem 1 iAHR after 8 h uninfluenced by ␤- 134
without propranolol adrenergic blockade
Decreasing CaO2 for 8 h with lowered Idem 1 iAHR after low PETO2 only, 640
isocapnic PETO2 of 55 Torr, indicating no role of lower oxygen
phlebotomy (500 ml) or 10% COHb content
6 h of hypocapnic (PETCO2 ⫺10 Torr) Idem No change in AHR, suggesting that the 641
or eucapnic hyperventilation increase in AHR during pH and iH
are caused by low PO2 itself;
hypocapnic, but not eucapnic
hyperventilation shifts the hyperoxic
CO2 response curve to the left
pH or poikilocapnic hyperoxia, for 5 Idem 1 iAHR after mild hypoxia and 2 192
days, respectively by maintaining after mild hyperoxia
PETO2 10 Torr below or above
resting

Physiol Rev • VOL 90 • APRIL 2010 • www.prv.org


718 LUC J. TEPPEMA AND ALBERT DAHAN

TABLE 4. —Continued

Reference
Protocol HVR Measurement Effects on HVR, Comments Nos.

iH: PETO2 50 Torr, 8 h, followed by iv Idem 1 iHVR not influenced by autonomic 452
infusion of the ganglion blocker blockade
trimethaphan
iH 3 h in NHC at PETO2 ⫽50 Torr Isoxic hypoxic and hyperoxic CO2 Decrease in threshold, not sensitivity of 483
response curve with modified the peripheral chemoreflex loop at
rebreathing the early stages of CH
pH during 8 h using a facemask Modified rebreathing; PETCO2 at 1 iHVR by 70% but impaired forearm 268
about resting value hypoxic vasodilation
pH: 8 wk at 3,800 m Stepwise decreases in SpO2 to 90 1 iHVR already after 48 h, sustained 346
and 80%; PETCO2 ⬃4 Torr above at 8 wk. Generally test PETCO2 about
resting in ambient conditions 5–6 Torr lower than at sea level
pH for 8 h using a facemask Modified rebreathing PETCO2 at about 1 iHVR by ⬎90% 757
resting value at sea level
5 days at 3,480 m Isoxic hypoxic and hyperoxic CO2 Decreases in the x-intercepts of the 719
response curve with modified hyperoxic and hypoxic CO2 response
rebreathing curves of 12.8 and 9.8 Torr,
respectively, without changes in
slope
pH: 9-day ascent to ⬎5,000 m and Isoxic hypoxic and hyperoxic CO2 Difference in slope between hyperoxic 9
then immediate testing at 3,840 m response curve with modified and hypoxic CO2 rebreathing became
rebreathing larger at high altitude, indicating
increased hypoxic sensitivity
Stay at 1,560 m for 12 days Poikilocapnic response to sustained Peak but not sustained poikilocapnic 8
(20 min) hypoxia (12% O2) HVR response increased

CaO2, arterial oxygen content; iH, isocapnic hypoxia; pH, poikilocapnic hypoxia; iAHR, isocapnic acute hypoxic response; CH, chronic
hypoxia; iHVR, isocapnic hypoxic ventilatory response; DEF, dynamic end-tidal forcing; NHC, normobaric hypoxic chamber; V̇I, inspired minute
ventilation; PB, barometric pressure; PETCO2, end-tidal PCO2; PETO2, end-tidal PO2; SpO2, arterial hemoglobin oxygen saturation.

nist that penetrates into the brain, increased ventilation, to 10% O2 for 7– 8 days contained higher levels of the ␣-
suggesting a predominance of the inhibitory carotid body amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid (AMPA)
dopaminergic system on respiratory output in normoxia receptor subunit glutamate receptor 2 (GluR2), which
(572). However, mice kept in 10% O2 for 10 days re- was thought to contribute to an enhanced glutamatergic
sponded to droperidol with a decrease in ventilation in- synaptic transmission of arterial chemoreceptors inputs
dicating a dominance of the central excitatory dopami- into the NTS (873). NMDA receptors in the medulla of
nergic system in hypoxia (572). This seems consistent mice were upregulated following exposure to 10% O2
with data from mice lacking the dopamine D2 receptor during 2 wk (210; see also sect. VE). In rats exposed to 10%
(D2 R⫺/⫺) that failed to show VAH and an augmented HVR O2 for 9 days, the NMDA antagonist MK-801 had a larger
after 8 days of acclimatization, an effect that was not yet inhibitory effect on hypoxic ventilation than in unaccli-
demonstrable after 2 days (343). These studies suggest matized controls (638).
that a shift from peripheral to central dopaminergic dom- C) PDGF-␤ RECEPTORS. Administration of PDGF-BB in
inance may contribute to VAH. How this can be recon- the NTS decreased the early HVR in rats, and mutant mice
ciled with an increase in carotid body dopamine content heterozygous for the PDGF-␤ receptor showed a reduced
in prolonged hypoxia remains to be seen. Upregulation HVD (see also sect. IIID). This led to the suggestion that
during chronic hypoxia of TH and norepinephrine was that downregulation of PDGF-␤ receptors during chronic
also observed in the in the locus coeruleus of rats (684), hypoxia might contribute to VAH (287, 289). During 14
but its contribution to VAH is difficult to assess because days in 10% O2, rats demonstrated a posttranscriptional
this region is the source of a widespread noradrenergic downregulation of PDGF-␤ receptor protein in the dorso-
network in the brain. caudal brain stem after 7 days, significantly correlated
B) NMDA RECEPTORS. During hypoxia, glutamatergic with VAH, while the attenuation of the HVR by local
NMDA and non-NMDA receptors in the caudal NTS play a PDGF-BB application in the NTS decreased over time and
crucial role in the transmission of afferent impulses from actually was absent from day 7 on (14). In addition to
the carotid body (287, 516, 565, 801 and references PDGF, other growth factors such as brain-derived neuro-
therein). Reeves et al. (634) showed an early upregulation trophic factor (BDNF), located in petrosal ganglion neu-
of the NR1 subunit of the NMDA receptor in the dorsal rons and possibly acting postsynaptically, could also con-
caudal brain stem of rats which had resolved after 30 days tribute to plastic changes within the NTS during chronic
of chronic hypoxia. The caudal NTS of rats exposed hypoxia.

Physiol Rev • VOL 90 • APRIL 2010 • www.prv.org


THE VENTILATORY RESPONSE TO HYPOXIA IN MAMMALS 719

D) NO-RELATED MECHANISMS. As discussed in section since the role of the former isoform in O2 sensing by
IIIC3, NO is an important central mediator of the HVR with central neurons is unknown (501), whereas in unacclima-
a stimulatory effect on the HVR. Thus upregulation of tized condition, the isoform HO-2 seems to be required for
neuronal NOS could also enhance CNS gain. Mice ex- oxygen sensing in neurons of the pre-Bötzinger and C1
posed to 10% O2 for 2 wk had higher medullary neuronal regions (153). These scarce data could suggest a contri-
NOS levels, which seemed related to VAH but not to the bution of central oxygen sensors to the increased CNS
increased HVR: the former was reduced while the latter gain in chronic hypoxia. Isolated central hypoxia, how-
remained unaffected by a specific neuronal NOS inhibitor ever, does not lead to VAH in the goat (836).
(210). Upregulation of neuronal NOS (but not the endo- G) EXPRESSION PROFILE OF ION CHANNELS IN THE NTS. Neu-
thelial isoform) was also shown in peripheral and central rons in slice preparations from rat containing NTS neu-
neurons in rats after 12–24 h in hypobaric hypoxia (618). rons receiving monosynaptic input from the carotid bod-
The hypoxia-induced rise in intracellular NO in the NTS ies respond to hypoxia with an outward potassium cur-
may be one of the secondary consequences of NMDA rent via activation of KATP channels (i.e., ATP-sensitive
receptor activation (references in Ref. 287). Thus, in the potassium channels that close in the presence of high ATP
NTS, a feed-forward mechanism could exist between concentrations; Ref. 872). In slices from rats exposed to
NMDA receptor activation and release of NO to enhance 10% O2 for 1 wk, this current was reduced, and this was
the CNS gain (562). The above-cited work by El-Hasnaoui possibly caused by an alteration of the expression profile
et al. (210) does not directly support this hypothesis. Rats of regulatory subunits of these channels. This then may
exposed to 12% O2 for 2 wk and receiving a continuous have increased both the excitability of these neurons and
intracerebroventricular infusion of the NOS inhibitor CNS gain (872).
L-NAME showed a lower normoxic ventilation than con- H) PLASTICITY-INDEPENDENT MECHANISMS TO INCREASE THE CNS
trols after this period, indicating a reduced VAH. How- GAIN. The brain contains high concentrations of O2-sensi-
ever, their HVR was indistinguishable from that in accli- tive (human tandem P domain) TREK-1 potassium chan-
matized controls not receiving the enzyme inhibitor (688). nels, predominantly in GABAergic inhibitory interneu-
These scarce data raise the impression that upregulated rons, that play a key role in setting the resting membrane
NO in the medulla may contribute to VAH but not to the potential (references in Ref. 511). In human embryonic
increase in HVR. kidney (HEK-293) cells, these background K⫹ channels
E) ERYTHROPOIETIN. Mice overexpressing human eryth- are inhibited by low PO2, in a range relevant for brain
ropoietin (Epo) in the brain without elevation of plasma tissue and also, in an additive way, by intracellular alka-
Epo and hematocrit (Tg21 mice) were reported to show losis (511). Inhibition of these channels in GABAergic
larger increases in ventilation and HVR in chronic hyp- inhibitory interneurons by mild alkalosis might then be of
oxia, even after carotid sinus nerve section (716). Female physiological significance especially at the background of
(but not male) mice with increased peripheral and central hypoxia-induced alkalosis (511). In the rat, an additional
Epo expression (Tg6 mice) were reported to have an way of increasing CNS gain could exist in a negative
augmented HVR (717). However, another study in Tg6 peripheral-central interaction (145, 172, 613; see sect. VD).
mice (sex not mentioned) reported no difference in HVR The time-dependent fall in PCO2 in the early acclimatiza-
compared with wild types; after acclimatization, VAH and tion phase could augment the central response to carotid
an augmented HVR were clearly present in wild types but body hypoxia, thus increasing ventilation and the HVR.
totally absent in the Tg6 genotype, and this was suggested Isocapnia should then be expected to decrease the mag-
to be related to upregulated carotid body dopaminergic nitude of these adaptations, but this has not been system-
mechanisms (804). Finally, infusion into the brain of the atically investigated in the rat.
soluble Epo receptor, that normally is downregulated in Many of the above modulations and adaptations in
chronic hypoxia thus increasing Epo bioavailability, re- both the carotid bodies and CNS are undoubtedly the
versed VAH in normal mice (715). consequence of altered gene expression induced by
F) CENTRAL OXYGEN-SENSING NEURONS. In the initial phase HIF-1. Rats exposed to chronic hypoxia (0.5 atm, 3 wk)
of VAH (10% O2 for 4 –5 days), O2-sensitive cells in the displayed accumulation of HIF-1␣ in neurons, astrocytes,
RVLM displayed an increased O2 sensitivity in vitro that in ependymal cells, as well as endothelial cells and also
later stages (9 –10 days) of acclimatization was offset, showed upregulation of genes with hypoxia-responsive
possibly due to an altered neurotransmitter balance or elements (126). HIF-1␣⫹/⫺ mice lack an increase in ca-
other processes decreasing membrane excitability (554). rotid sinus nerve activity in response to hypoxia (not to
In both normoxia and hypoxia, the C1 region and the NaCN) but increase their ventilation, which is initiated by
pre-Bötzinger complex show a constitutive expression of the aortic bodies. Ventilatory adaptation to a 3-day bout of
HO-2 (501). Placing rats in 10% O2 for 10 days induced hypoxia does not occur in these animals (388). Chronic
HO-1 but not HO-2 in the ventral medulla. At this stage, hypoxia induces carotid body expression of both HIF-1␣,
however, the significance of this observation is unclear HIF-2␣, and HIF-3␣, which all contribute to morphologi-

Physiol Rev • VOL 90 • APRIL 2010 • www.prv.org


720 LUC J. TEPPEMA AND ALBERT DAHAN

cal and neurochemical adaptations (434, 584, 614, 784 for the spinal cord of rats is sufficient to evoke LTF. In
further references). Humans with an impaired degrada- addition, when BDNF translation and new protein synthe-
tion rate of HIF-1␣ in normoxia (Chuvash polycythemia; sis is prevented, episodic hypoxia does not induce LTF
see sect. IV) have an exaggerated acute HVR and low (28). BDNF is thought to act through the tyrosine kinase
resting PCO2 much similar to VAH (712). The contribution B receptor which then can lead to the facilitation of
of HIF-1 in both the carotid bodies and the CNS to VAH is glutamatergic transmission via several pathways (28). Ki-
recently reviewed by Powell and Fu (614). nases and phosphatases relevant for LTF are sensitive to
Other transcription factors also are involved. For ROS, explaining the inhibitory effects of antioxidants on
example, a role of activator protein-1 (AP-1) is indicated LTF. An important source of ROS in various cell types
by the finding that mice lacking the fosB gene (a member (e.g., motoneurons and carotid body cells) is NADPH
of the fos family of immediate early genes), although oxidase. Depending on the cell type, this enzyme can be
having a normal HVR when kept in normoxia, do not activated in hypoxia, upon reoxygenation as well as via
exhibit VAH and an increase in HVR when exposed to membrane-bound receptors such as 5-HT2A receptors.
chronic hypoxia (486). Phrenic LTF can be inhibited by serotonin receptor block-
Chronic hypoxia may also recruit additional oxygen ers, antioxidants, and inhibitors of NADPH oxidase, re-
sensing mechanisms that are silent in a normoxic condi- spectively (480, 481). Finally, repetitive spinal injections
tion. of 5-HT induce phrenic LTF, even without hypoxia. So the
working model of episodic hypoxia-induced phrenic LTF
is that it is mediated by a release of serotonin, the acti-
G. Chronic Intermittent Hypoxia and the HVR
vation of NADPH oxidase, and increased production of
BDNF and ROS with facilitation of glutamatergic synaptic
1. Episodic hypoxia induces phrenic and
transmission in the phrenic motor nucleus as the eventual
ventilatory LTF
result (27, 28, 230, 447, 480, 481 and references cited
In awake rats, dogs, and goats, the termination of therein). In both artificially ventilated and spontaneously
episodic hypoxia, for example, a challenge with repeated breathing rats, LTF is substantially reduced by NMDA
hypoxia of short duration (mostly 1–5 min) interrupted by antagonists injected in the phrenic motor nucleus (447).
short periods of reoxygenation, is followed by a pro-
longed increase in ventilatory motor output, a phenome- 2. Chronic intermittent hypoxia augments LTF,
non that is known as LTF (112, 506, 568, 792). Sustained sensitizes the carotid bodies in animals, and
hypoxia of short duration does not evoke the phenome- increases the HVR
non (27). At the level of phrenic and hypoglossal motor
outputs, LTF can be evoked by intermittent electrical Following three 5-min episodes of isocapnic hypoxia,
stimulation of the carotid sinus nerve and/or continuous the phrenic neurogram of rats showed a somewhat in-
stimulation of medullary raphe obscurus neurons in anes- creased baseline amplitude that further increased over
thetized, (mostly) vagotomized animals (244, 305, 495, time to become significantly greater than the prestimulus
512, 513) or by repetitive hypoxia (23, 245, 387, 504). LTF amplitude after 30 and 60 min, respectively, indicating
requires an intact serotonergic system with neurons in LTF (448). Exposing rats to chronic continuous alterna-
medullary raphe nuclei playing a crucial role (23, 506, tions of 5 min of hypoxia (11–12% O2) and 5 min of air for
514). 12 h/night for 7 days, i.e., chronic intermittent hypoxia,
We briefly summarize the main underlying mecha- augmented the acute phrenic response to episodic mild
nisms of phrenic LTF as far as they may also be relevant hypoxia and, via a serotonergic pathway, increased LTF
for (plastic changes of) the HVR in chronic intermittent (448). This suggests that the chronic intermittent hypoxia
hypoxia (references in Refs. 230, 447, 480, 481). During had resulted in an augmented carotid body sensitivity to
episodic hypoxia, raphe serotonergic neurons in the me- hypoxia and/or increased CNS gain. Electrical carotid
dulla are activated resulting in serotonin release in the sinus nerve stimulation now resulted in an enhanced ac-
vicinity of brain stem respiratory neurons and spinal mo- tivation of the phrenic nerve, indicating an increased CNS
toneurons. Each hypoxic episode triggers a release of gain (448). In conscious rats, however, not all these find-
5-HT. One of the downstream cascades then activates ings could be confirmed at the level of minute ventilation
several protein kinases that may phosphorylate glutamate because a similar chronic intermittent hypoxia protocol
receptor proteins, thus increasing synaptic strength by induced ventilatory LTF but no increase in the HVR (505,
enhancing NMDA receptor currents. Some phosphates, 506). Similar to episodic hypoxia-induced LTF, the effect
on the other hand, may lay a constraint on LTF (refer- of chronic intermittent hypoxia to augment its magnitude
ences in Ref. 481). A key role in LTF is played by BDNF. is a NMDA receptor-related mechanism; however, the
During episodic hypoxia, the synthesis of this neurotro- serotonin subtype involved may be a different one: a
phin in the spinal cord increases; application of BDNF in 5-HT2 subtype in episodic hypoxia, and a 5-HT6 and/or

Physiol Rev • VOL 90 • APRIL 2010 • www.prv.org


THE VENTILATORY RESPONSE TO HYPOXIA IN MAMMALS 721

5-HT7 subtype in the chronic intermittent hypoxia-in- chronic intermittent hypoxia in these animals are un-
duced augmentation (447). known.
These chronic protocols with relatively long dura- An increase in the sustained, but not acute, HVR was
tions of mild hypoxia in rats were not directly intended to reported in rats after alternations of 10 and 21% O2 every
mimic the rapid and more severe hypoxic swings that are 90 s, continuously applied during 30 days (634). Challeng-
experienced by sleep apnea patients but were mainly ing cats with alternations of 90 s hypoxia with ⬃5 min
aimed at studying the plasticity of the neuronal network normoxia, 8 h/day for 2– 4 days enhanced the in vivo basal
shaping the HVR. Peng and Prabhakar (597) have ex- discharge and hypoxic sensitivity of the carotid sinus
tended the above observations on the effects of episodic nerve, upregulated carotid body ET-1 by a factor of ⬃10
hypoxia by imposing hypoxic episodes lasting seconds and shifted the hypoxic response curve of their in vitro
rather than minutes, a stimulus pattern more relevant to vascularly perfused carotid bodies to the right (643). Sub-
sleep apnea. They showed that conditioning rats with sequently, a chronic intermittent hypoxia-induced upregu-
chronic intermittent hypoxia consisting of 15 s hypoxia lation of carotid body ETB but not ETA receptors was
alternated with 5 min of normoxia, 9 cycles/h for 8 h/day shown, and it was proposed that upregulation of the
during 10 consecutive days, augmented the episodic hyp- endothelin system would increase carotid body sensitivity
oxia-induced phrenic LTF, which was absent in animals by modulating blood flow (642). Recently, it was sug-
exposed to sustained hypoxia with a comparable cumu- gested that, similar to its facilitatory effect on synaptic
lative duration (597). Combining chronic intermittent hyp- transmission in the NTS, ET-1 may also stimulate the
oxia conditioning with daily administration of a superox- carotid body by augmenting the excitatory effect of glu-
ide dismutase (SOD) mimetic prevented this LTF augmen- tamate (456). Rats exposed to chronic intermittent hyp-
tation (597). Subsequently, they showed in the rat that oxia for 3 wk showed upregulation of carotid body NR1,
chronic intermittent hypoxia: 1) evoked episodic hypoxia- and NR2, NMDA receptor subunits, and in contrast to
induced LTF in the carotid sinus nerve (“sensory LTF”) controls, carotid body baseline activity in these animals
was enhanced by infusion of NMDA without altering the
that was prevented with a SOD mimetic; 2) reduced both
sensitivity to hypoxia. The NMDA antagonist MK-801 not
aconitase activity in the carotid bodies and the activity of
only reversed this increase in baseline activity but also
mitochondrial complex I; and 3) increased hypoxic but
inhibited the stimulatory effect of ET-1 (456). So, although
not hypercapnic sensitivity of the in vivo and in vitro
probably not directly involved in O2 sensing, NMDA re-
carotid body, which was prevented by scavenging super-
ceptors may play a functional role in the adaptation of the
oxide radicals and could be reversed by subsequent ad-
carotid bodies to chronic intermittent hypoxia.
aptation to normoxia (585, 595–597). This functional ca-
Chronic intermittent hypoxia will not only initiate
rotid body plasticity occurred without macroscopic mor-
plastic changes in the carotid bodies. In NTS neurons
phological changes (585, 596). receiving afferent input from the carotid bodies of rats
Additional studies in mice with a partial deficiency exposed to chronic intermittent hypoxia, dose-dependent
for HIF-1␣ (HIF-1␣⫹/⫺) revealed that, in contrast to wild currents induced by AMPA were increased whilst NMDA-
types, these animals did not respond to chronic intermit- activated currents were reduced (174). Reeves et al. (634)
tent hypoxia with increases in HVR and carotid body showed a chronic hypoxia-induced increase in the expres-
sensitivity. Also, the heterozygous animals did not accu- sion of the NR1, NR2A, and NR2B NMDA receptor subtypes
mulate ROS and showed no upregulation of HIF 1-␣ (600). in the dorsocaudal brain stem. Another factor contribut-
Prabhakar and colleagues (536) proposed a working ing to a centrally mediated HVR plasticity may be platelet-
model of chronic intermittent hypoxia in which oxidative activator factor (PAF). In an attempt to gain insight into
stress leads to ROS generation (presumably by increased the role of this phospholipid, Reeves and Gozal (630)
NADPH oxidase activity and/or inhibition of mitochon- compared the effects of a chronic (30 day) intermittent
drial complex I), upregulation of HIF-1 and induction of hypoxic paradigm in mice deficient for the PAF receptor
many genes by this transcription factor that together help (PAFR) with those in their wild-type littermates. The
to sensitize the carotid bodies. Other transcription factors results were somewhat contradictory. On the one hand,
and posttranscriptional phosphorylation of proteins also unlike wild-type mice, PAFR⫺/⫺ mice failed to show an
play an important role (414, 536). increase in normoxic ventilation after the challenge, indi-
NO could also be involved in chronic intermittent cating a role of PAF in chronic intermittent hypoxia-
hypoxia-induced LTF and carotid body sensitization. Mice induced ventilatory LTF. On the other hand, over the
deficient in neuronal NOS have an augmented HVR but period of 30 days, both genotypes showed an equally
show an attenuated episodic hypoxia-induced ventilatory robust time-dependent increase in the response to sus-
LTF; the effect of chronic intermittent hypoxia in these tained (20 min) hypoxia (630). Thus the precise role of
mice was not studied (387). Mice deficient in endothelial PAF in the chronic intermittent hypoxia-induced augmen-
NOS have a blunted O2 response (390); the effect of tation of the HVR remains to be determined.

Physiol Rev • VOL 90 • APRIL 2010 • www.prv.org


722 LUC J. TEPPEMA AND ALBERT DAHAN

After exposure to chronic intermittent hypoxia,


anesthetized rats showed a rise in blood pressure and
an augmented preganglionic sympathetic nerve re-
sponse to acute hypoxia (294). Conscious rats re-
sponded with an increase in the hypoxic response of
the renal sympathetic nerve (339). Chronic intermittent
hypoxia has a profound influence on the oxidative sta-
tus of the brain cortex and leads to protein and nucleic
acid oxidation, lipid peroxidation, and apoptosis, a pic-
ture that is mitigated in mice overexpressing Cu,Zn- .
VI
SOD (857). ROS induced specific enzymatic changes in .
VI
the brain stem of rats exposed to chronic intermittent
hypoxia that eventually led to enhanced ␣-amidation of
neuropeptides such as substance P and neuropeptide Y
that thereby become biologically active (699). Many
neuropeptides are operating in central neuronal net-
works, so similar mechanisms may also contribute to
chronic intermittent hypoxia-induced changes in the
HVR and sympathetic chemoreflex sensitivity.
In summary, in animals and across species, chronic
intermittent hypoxia leads to an augmented episodic hyp- FIG. 10. Comparison of chronic intermittent hypoxia in neonates
and adults. Effects of chronic intermittent hypoxia in neonates (within
oxia-induced LTF at the level of carotid bodies, phrenic the critical time window for developmental respiratory plasticity) and in
nerve, and ventilation. Carotid body sensitivity is in- adults. Question marks concern unresolved or controversial issues. V̇I,
creased and the HVR is enhanced. These adaptive inspired ventilation; LTF, long-term facilitation. In the adult, ventilatory
LTF can be the result of sensory LTF and LTF in major respiratory motor
changes are mediated by ROS-related HIF-1␣ stabilization nuclei such as the phrenic and/or hypoglossal motor nuclei and inter-
and gene expression resulting in adaptations in the sig- costal motor neurons. In the adult, sensory LTF in the carotid bodies
naling-transduction cascade in the carotid bodies and may tend to augment carotid body sensitivity and thus destabilize
breathing, but LTF in motor nuclei may have a stabilizing effect. For
plastic changes within the central respiratory neural net- example, if the phenomenon would exist in the hypoglossal (and
work. As was also the case for chronic hypoxia, chronic phrenic) nucleus in humans with obstructive apnea, the chronic inter-
intermittent hypoxia has different effects in neonates and mittent hypoxia in these patients may tend to limit the occurrence of
upper airway collapse. For further explanation, see text.
adults. This is schematically shown in Figure 10.

3. Chronic intermittent hypoxia increases the HVR If it is accepted that the peripheral chemoreceptors
in humans mediate the initial hypoxia-induced rise in ventilation,
enhancement of the acute HVR (8, 10, 11, 253, 484) is most
Protocols used in humans to produce chronic inter- probably due to an increase in carotid body sensitivity. A
mittent hypoxia vary widely in total duration (days to higher ventilation during hypoxia combined with an also
weeks), severity, and length of individual hypoxic expo- higher arterial oxygen saturation is compatible with an
sures. In a minority of cases, isocapnia was maintained. increased slope of the ventilation-SaO2 relationship (e.g.,
Data we have collected from a number of recent studies Ref. 53). It is not easy, however, to assess the precise
are shown in Table 5. The paradigms employed do not quantitative contribution of the carotid bodies to the aug-
exactly mimic the swings in PO2 and PCO2 that are encoun- mented HVR: isocapnic progressive hypoxic tests lasting
tered in sleep apnea syndrome wherein hypoxic episodes 5–9 min are associated with increases in CBF and central
are shorter, 20 s on average, and accompanied by hyper- hypocapnia and may also be accompanied by a rapidly
capnia (242). Rather, they may represent time courses of developing HVD (see sect. III). The fact that chronic inter-
prolonged hypoxic episodes that can occur in patients mittent hypoxia induces augmented cerebrovascular O2
with chronic obstructive pulmonary disease (COPD) and CO2 sensitivities should also be taken into account (8,
and/or severe heart failure. With only a few exceptions, 395).
paradigms of chronic intermittent hypoxia in humans re- The mechanisms of a chronic intermittent hypoxia-
sult in an increase in the HVR, which can be accompanied induced increase in carotid body sensitivity in humans
by a rise in resting normoxic ventilation and drop in remain elusive, but plastic changes as described for ani-
end-tidal CO2. In cases where isocapnic HVRs were mea- mals could well operate in humans also. Like in animals,
sured at resting end-tidal CO2 levels (Table 5), the effects the increase in HVR may depend on stimulus intensity, the
may even have been underestimated (postexposure end- number of deoxygenation/reoxygenation periods, and to-
tidal CO2 levels mostly being lower). tal exposure length. Exposing humans to a chronic inter-

Physiol Rev • VOL 90 • APRIL 2010 • www.prv.org


THE VENTILATORY RESPONSE TO HYPOXIA IN MAMMALS 723

TABLE 5. Chronic intermittent hypoxia in humans

Protocol HVR Measurement Effects on HVR, Comments Reference Nos.

5 min isocapnic hypoxia (12% Progressive isocapnic hypoxia to 1 iHVR by 37% on day 12, 241
O2) alternated by 5 min SaO2 ⬃75 %; PETCO2 at resting recovery on day 17; no
normoxia for 1 h, 12 days values difference between both
(protocol 1) versus 30 min protocols; no changes in
isocapnic 12% O for 12 days resting V̇I and PETCO2
(protocol 2); interruption of
protocols on days 6 and 7
FIO2 0.13 for 2 h/day for 12 days Step decrease in FIO2 from 0.3 to Max 1 of iAHR, on day 5 and 253
0.13; PETCO2 at level causing a then gradually decreasing to
hyperoxic V̇I of 15 control on day12; isocapnic
ml/min⫺1 䡠 kg⫺1 PETCO2 after hypoxia ⬃1.5
Torr lower
Daily exposure to a barometric Progressive isocapnic hypoxia at HVR, defined as the constant of 376
pressure of 432 Torr for 1 h, eucapnic PETCO2 the hyperbolic V̇I-PaO2
7 days relationship, increases by 73
%; no change in resting PETCO2
Daily three 6-min bouts of Progressive isocapnic rebreathing 1 HVR by ⬃50%, no change in 695 (in which many studies in the
isocapnic hypoxia with PETO2 to FIO2 8-7% at constant resting PETCO2 former Sovjet Union are
50-35 Torr interspersed with eucapnic PETCO2 reviewed).
normoxia within 30 min, for
14 days
Decompression to 15.5 or 12.3 Progressive isocapnic hypoxia to 1 iHVR only after 12.3 % O2 by 374 (More data from the same
% O2 for 1 h/day for 1 wk SaO2 ⬃75 %; PETCO2 at resting ⬃25%, indicating an influence authors in ref, 373, 375–377)
values of the depth of hypoxia; no
changes in resting PETCO2 and
resting V̇I
12% O2 for 5 min, poikilocapnic, Isocapnic progressive hypoxia to No change in iHVR, resting 623
followed by 5 min normoxia, SaO2 ⬃75%; PETCO2 at resting PETCO2 somewhat lower after
for 1 h, 10 days values. CIH
FIO2 0.12, isocapnic for 5 min, Isocapnic progressive hypoxia to No change in iHVR, neither in 700
followed by 5 min 0.21, 4 SaO2 ⬃85%; PETCO2 at resting fit, nor in sedentary men; no
times/day for 10 days, men values change in resting PETCO2
60–74 yr old
12 times 5 min poikilocapnic Isocapnic progressive hypoxia 1 iHVR by ⬃50% after both 393
12% O2, 5 min normoxia daily until SaO2 ⬃80 %; measured on SDIH and LDIH, max effect
for 7 days SDIH versus 60 7 consecutive days; PETCO2 reached after 3 days; CO2
min poikilocapnic 12% O2 kept at resting values in all threshold (from modified
daily, 7 LDIH in the same conditions rebreathing) decreased after
subjects both protocols
5 min 12% O2, 5 min 21% O2 for Poikilocapnic response to acute Acute, but not sustained pHVR 8
90 min during 10–12 and sustained (20 min) hypoxia response increased; no
consecutive days (12% O2) change in resting PETCO2
60 min of isocapnic hypoxia Progressive isocapnic hypoxia to Sustained (20 min) iHVR 478
(SaO2 ⬃80%) for 10 SaO2 ⬃75% and then doubles, no change in resting
consecutive days maintained for another 20 min; V̇I and resting PETCO2
PETCO2 at resting values.
Sleeping for 5 consecutive iAHR determined from six Mean iAHR more than doubles, 10, 11
nights with FIO2 ⬃0.14 descending PETO2 steps with baseline ventilation increased,
DEF technique; PETCO2 kept at resting PETCO2 fell by ⬃3.5
⬃1.5 Torr above resting Torr
14 nights (9 h) of sustained Isocapnic progressive hypoxia to iHVR slope increases ⬃2.5-fold, 269
SaO2 ⬃84% SaO2 ⬃80% increase in baseline
ventilation; resting [paco2]
lower by 3.3 Torr
20 min of isocapnic (eucapnic) Isocapnic steps in hypoxia daily; AHR but not sustained HVR 484
hypoxia (FIO2 ⫽ 0.10) daily modified rebreathing to increased; augmented AHR
for 14 consecutive days determine thresholds and ascribed to decrease in
sensitivities both before and peripheral CO2 threshold
after isocapnic hypoxic studies because the threshold of the
hypoxic, not the hyperoxic
CO2 rebreathing curve
decreased. Sustained HVR not
increased because of HVD,
ascribed to an increase in
peripheral CO2 threshold

Physiol Rev • VOL 90 • APRIL 2010 • www.prv.org


724 LUC J. TEPPEMA AND ALBERT DAHAN

TABLE 5. —Continued

Protocol HVR Measurement Effects on HVR, Comments Reference Nos.

20 consecutive nights with FIO2 Isocapnic progressive hypoxia to Time-dependent increase in 787
0.163 (Pl) versus 4 blocks of SaO2 ⬃75% HVR in both P1 and P2; effect
5 nights with each block of P1⬎ P2. HVR expressed
interspersed with 2 nights of both as linear V̇I-SaO2 and
normoxia (P2). “Living high, hyperbolic V̇I-PaO2
training low” in athletes relationships. Resting PETCO2
lower after both P1 and P2
4 consecutive days with cycling Acute isocapnic HVR with DEF; Time-dependent increase in the 605
between 2 min PETO2 ⫽45 hypoxic sensitivity defined as acute hypoxic response that
Torr and 2 min at 88 Torr the ratio delta ventilation over after 4 days was ⬃80%. The
during 6 h in awake humans delta SaO2 increase was correlated with
an increased production of
reactive oxygen species.
Subjects slept during 8-9 h/day Progressive isocapnic hypoxic Doubling of the HVR after 2 wk; 756
in 13% O2 during 2 and 4 wk, test; HVR defined as the ratio in fact, this protocol consisted
respectively, and were delta ventilation over delta of two CIH paradigms
exposed overnight to 30 SaO2 superimposed on each other.
cycles/h of reoxygenation Sleeping in a hypoxic
environment alone also
increases the HVR (e.g., see
Refs. 10 and 269) and a
control group not subjected
to the reoxygenation cycles
was not included. An
interesting observation was
that the hypopneas associated
with reoxygenation were
followed by arousals.

iH, isocapnic hypoxia; pH, poikilocapnic hypoxia; iAHR, isocapnic acute hypoxic response (3-min hypoxic ventilatory response); iHVR,
isocapnic hypoxic ventilatory response; pHVR, poikilocapnic hypoxic ventilatory response; HVD, hypoxic ventilatory decline; DEF, dynamic
end-tidal forcing; V̇I, inspired minute ventilation; FIO2, oxygen fraction of inspired air; PETCO2, end-tidal PCO2; PETO2, end-tidal PO2; SaO2, arterial
hemoglobin oxygen saturation; CH, chronic hypoxia; CIH, chronic intermittent hypoxia; SDID, short-duration intermittent hypoxia; LDIH, long-
duration intermittent hypoxia.

mittent hypoxia protocol with hypoxic episodes of 12% O2 sensitivity elicits higher sympathetic activity and even-
led to an increase in oxidative plasma biomarkers and a tually results in hypertension in many of these patients
decrease in antioxidant capacity that were not observed (372, 537, 548).
with 15% O2 (817). Katayama et al. (374) reported an
increase in HVR by a protocol incorporating 12.3% O2 but 5. Different effects of chronic hypoxia and chronic
not 15.5%. Patients with obstructive sleep apnea show an intermittent hypoxia
increase in plasma biomarkers of oxidative stress and
ROS production in leukocytes (202, 365). A recent study The effects of chronic hypoxia and chronic inter-
in healthy young subjects showed that after 4 days of mittent hypoxia may be difficult to compare. For exam-
intermittent hypoxia, the acute ventilatory response to ple, differences in the duration and severity of individ-
hypoxia increased, which was correlated with an aug- ual hypoxic episodes, total exposure time to hypoxia,
mented production of ROS (605). and the large variation in cycle number of intermittent
Long-term chronic intermittent hypoxia in humans hypoxic paradigms render it hard to define one single
has pathophysiological consequences that are recently effect chronic intermittent hypoxia. Some protocols
reviewed elsewhere (242). Briefly, it leads to an in- may comprise both chronic hypoxia and chronic inter-
crease in (muscle nerve) sympathetic activity, impaired mittent hypoxia or repetitive cycles with different
baroreflex, increased production of ET-1 by enhanced lengths. Protocols with long uninterrupted exposures,
shear stress, decreased NO bioavailability, impaired e.g., during long nights spent in hypoxia (e.g., Refs. 11,
vasodilation, and hypertension. Increased production 269) lead to increased carotid body sensitivity and
of ROS causes upregulation of genes encoding factors periodic breathing and an ensuing additional, chronic
that elicit inflammation, platelet aggregation, and ath- intermittent (hypercapnic) hypoxia superimposed on
erosclerosis (242). Chronic intermittent hypoxia as ex- the experimental one. This may be a confounding factor
perienced by patients with a sleep apnea syndrome will in studying the effects of “chronic hypoxia” during long
tend to increase their HVR. An increase in chemoreflex stays at high altitude.

Physiol Rev • VOL 90 • APRIL 2010 • www.prv.org


THE VENTILATORY RESPONSE TO HYPOXIA IN MAMMALS 725

The effect of chronic intermittent hypoxia clearly response. The network activated by chronic hypoxia
depends on the pattern of the hypoxic exposures. For included the hub nodes with vascular endothelial
example, rats exposed to chronic intermittent hypoxia growth factor, nuclear factor- ␬ B and transforming
with long uninterrupted hypoxic episodes did not man- growth factor-␤, all of which are known to play a role in
ifest an augmented carotid body sensitivity, in contrast pulmonary hypertension (850). This interesting ap-
to those challenged with hypoxia of equal total duration proach awaits application in the carotid bodies.
but with many oxygenation-reoxygenation cycles; the
former animals may have experienced a milder oxida-
tive stress compared with the latter (598). The produc- VI. MEASURING, EXPRESSING, AND MODELING
tion of ROS will widely vary not only with the length THE HYPOXIC VENTILATORY RESPONSE
and severity of the hypoxic episodes but also on the
number of reoxygenation periods. A single bout of A. Measuring the HVR
hypoxia and reoxygenation produce ROS, and this ef-
fect is amplified by repetitive hypoxia/reoxygenation Techniques used to measure the human HVR can be
(857, 867). divided into five groups: 1) steady-state techniques involv-
Some of the reported different consequences of ing fixed inspired O2 and CO2 concentrations, 2) single or
chronic hypoxia and chronic intermittent hypoxia can double breath tests involving transient hypoxia or hyper-
be listed as follows. 1) In contrast to chronic hypoxia, oxia for no more than a few breaths, 3) progressive
a specific chronic intermittent paradigm did not lead to hypoxia tests involving the rebreathing of a gas mixture
morphological changes in the carotid bodies (585, 596). from a rebreathing bag or circuit with CO2 absorption
2) In PC12 cells, the time course and magnitude of allowing control of end-tidal CO2, 4) step hypoxic tests in
HIF-1␣ expression differed between the two paradigms: which rapid decreases in end-tidal O2 are performed while
chronic intermittent hypoxia was more potent in in- maintaining end-tidal CO2 constant, and 5) via measure-
creasing HIF-1␣ protein, and the elevated levels were ment of isoxic hypoxic and hyperoxic ventilatory CO2
sustained for longer periods of time (references in Ref. responses.
536). 3) Another study reported that in contrast to The HVR obtained with steady-state hypoxic tests
chronic hypoxia, chronic intermittent hypoxia did not (test 1) is a mixture of the acute effects of hypoxia and
enhance HIF-1␣ mRNA in the rat carotid body, while its central depressant effects. Hence, the response is
both paradigms augmented the expression of HIF-2␣ “contaminated” by HVD. These tests were applied in a
and HIF-3␣ (434). 4) In the dorsocaudal brain stem of time and age in which there was little knowledge on
the rat, chronic intermittent hypoxia was associated HVD. Consequently, steady-state tests are considered
with an increase in the expression of the NR2A and outdated (871). Dejours et al. (177) developed the sin-
NR2B subunits of the NMDA receptor that did not occur gle breath test (test 2) in which after several control
in chronic hypoxia (634). The authors of this study breaths the subject inhales one vital capacity of a test
related this to the less pronounced presence of HVD gas (100% O2 or 100 N2 with or without the addition of
after the chronic intermittent compared with the CO2). The change in ventilation during the next few
chronic protocol (634). 5) As discussed in section VG1, breaths is a measure of hypoxic sensitivity. While this
in the rat, chronic intermittent but not chronic hypoxia test is not contaminated by HVD, it does not allow the
augmented the magnitude of LTF (597). development of the full HVR, and isocapnia is not main-
Using a systems biology approach, Wu et al. (850) tained during the test. Furthermore, to avoid random
studied the expression of genes, sets of genes, and genetic effects from the background variation in ventilation,
networks and found different temporal expression pat- the test has to be repeated frequently (411). Finally,
terns in chronic- and chronic intermittent hypoxia in the quantification of the small and short-lived change in
lungs of male rats. The gene encoding the estrogen ventilation is difficult (628), and therefore, the test
receptor 1 (Esr-1) was unmasked as an important reg- should be just considered a qualitative measure of pe-
ulator of the response to chronic intermittent but not to ripheral chemoreceptor activity. At present, most pop-
chronic hypoxia. Increased expression of Esr-1 also ular tests are those that use progressive (test 3) and
resulted in upregulation of ⬎200 target genes of the step hypoxia (test 4) and those that measure the HVR
ESR-1 protein, for example, the P2X2 purinergic recep- via the V̇I-CO2 responses at hyperoxia and hypoxia
tor. In addition, ESR-1 closely interacted with gene (test 5).
networks that already were known to play key roles in
hypoxia, for example, a hub node around HIF-1␤ and 1. Progressive hypoxic tests
vascular endothelial growth factor (VEGF). Chronic
hypoxia acutely induced a different set of genes that Over the years, various progressive hypoxic tests
were overrepresented by those involved in the immune have been designed (143, 272, 411, 458, 485, 626 – 628,

Physiol Rev • VOL 90 • APRIL 2010 • www.prv.org


726 LUC J. TEPPEMA AND ALBERT DAHAN

833). Techniques differ in depth of hypoxia, steepness of DEF is good, with standard deviations of the end-tidal
the hypoxic ramp (and linked to this the duration of the concentrations over time ⬍0.6 Torr (161). The main ad-
test), and the technique to keep end-tidal CO2 constant. vantage of DEF is its ability to obtain an open-loop esti-
Weil et al. (833) published their method in 1970: subjects mation of ventilatory responses within a short period of
breathed sequentially three gas mixtures for 3–5 min time, something that is impossible under closed-loop con-
(room air, 10 and 8.5% O2 in N2) causing a slow (15 min) ditions.
progressive isocapnic decline in alveolar PO2 to ⬃40 Torr. The technique has the disadvantage that relatively
Isocapnia was maintained by adding CO2 to the inspira- complex apparatus is required. As a result, less sophisti-
tory limb of the breathing circuit. Godfrey et al. (272) cated techniques are utilized more frequently using con-
adapted the CO2 rebreathing technique developed by tainers with preset hypoxic mixtures and/or adaptations
Read (624) in such a way that hypoxia developed progres- of the inspired oxygen concentrations by hand to obtain
sively over 5– 8 min. The subjects rebreathed from a cir- the desired end-tidal values (204, 370, 683, 705). Often
cuit that included a CO2 absorber allowing the mainte- isocapnia is maintained by using a rebreathing circuit
nance of isocapnia. The initial gas mixture contains 25–30% O2 (683), otherwise by adding CO2 to the inspiratory limb of
in N2, and as the subjects consumes oxygen, the oxygen the apparatus (204).
concentration of the bag decreases. During the course of One variant of the step hypoxic test is the application
rebreathing, arterial PO2 drops from 130 to ⬃30 Torr. of randomly modulated sequences in hypoxia and hyper-
Rebuck et al. (627) and later Rebuck and Campbell (626) capnia (863). This computer-driven technique requires
described a rebreathing technique similar to that of God- mathematical modeling to obtain an indication of the
frey et al. (272). The “Rebuck” approach involves re- magnitude of the HVR and its interaction with CO2. While
breathing from a 6-liter bag containing a gas mixture of 7% of great value in the characterization of the ventilatory
CO2 and 22–24% O2 in N2. The obvious problem with this control system (see also sect. VIC), it is of lesser practical
technique is that the ensuing HVR is due not only to the applicability.
progressive decline in PO2 but also to the step increase in
PCO2 (658). In a modified approach, the high PCO2 level is 3. Measuring HVR via the V̇I-CO2 response
kept constant for 6 min before beginning to rebreathe
Various authors derive the HVR from isoxic hypoxic
from the bag (485). As pointed out by Robbins and Zhang
and hyperoxic V̇I-CO2 response curves (195, 349, 411, 547,
(658), the later description of the modified “Rebuck re-
628, 720). Most frequent studied PO2 values for hyperoxia
breathing technique” by Rebuck and Slutsky (628) does
are 150 –250 Torr and for hypoxia are 40 –50 Torr. Sever-
not make it clear that the original technique is seriously
inghaus defines the hypoxic response as the increase in
flawed. See Figure 11A for a description of the apparatus
ventilation at an interpolated end-tidal CO2 going from
used by Rebuck and colleagues. It is important to be
hyperoxia to hypoxia (⌬V̇40 when a 40 Torr hypoxic level
aware that rebreathing methods have the potential for
is chosen, Ref. 683). Others use the change in slope going
systematic errors due to contamination by HVD when
from hyperoxia to hypoxia (⌬S) or the ratio of the slopes
tests last longer than 5 min (411, 612).
(S40/S200) as well as the change in the position of the
response curves (195, 628). When the isoxic V̇I-CO2 re-
2. Step hypoxic tests sponse curve is assessed using steady-state methods at a
hypoxic background, an appreciable effect of HVD on the
A sophisticated method to obtain ventilatory re-
response occurs. To overcome this problem, some au-
sponses to steps into and out of hypoxia is by using the
thors use the (modified) CO2 rebreathing procedure
computer-controlled DEF technique, developed by Swan-
according to Read (624). Note, however, that the Read-
son and Bellville in the late 1960s and early 1970s in
rebreathing technique, although relatively simple in per-
Stanford, California (56, 747, 748). Originally DEF was
formance, contains inherent difficulties that are not al-
used to measure the V̇I-CO2 response and quantify the
ways appreciated (155). For example, abolishing the
contribution of the central and peripheral chemoreflex
mixed venous-to-arterial PCO2 gradient, one of the key
loops to total ventilation (56). Not much later, it was
features of the Read-rebreathing technique (624), is not
adopted by several research teams to study step and
enough to reduce the brain tissue-arterial PCO2 gradient
sinusoidal hypoxic responses (176, 655, 822). DEF (see
sufficiently to zero to cause equilibration between arterial
Fig. 11B) allows manipulation of the inspired PO2 and PCO2
and brain tissue PCO2 (155).
to induce specific waveforms in end-tidal gas concentra-
tions independent of the ventilatory response and venous
4. The choice of method of measuring HVR
return. Feedback/forward loop algorithms are applied to
anticipate and compensate for the respiratory response to A) THE CHOICE OF TEST END-TIDAL PCO2. There is no
a desired end-tidal gas concentration on a breath-to-breath consensus on defining the test end-tidal or arterial PCO2
basis. The control of the end-tidal gas concentrations with (696). Some investigators raise the end-tidal value ar-

Physiol Rev • VOL 90 • APRIL 2010 • www.prv.org


THE VENTILATORY RESPONSE TO HYPOXIA IN MAMMALS 727

FIG. 11. Experimental set-up to measure the HVR. A: setup used by Rebuck and Slutsky to measure the ventilatory response to hypoxia. The
DC pump draws gas from the bag through the soda lime and returns the gas to the end of the bag (arrows). [Adapted from Rebuck and Slutsky (628).]
B: experimental setup used in Leiden to apply dynamic end-tidal forcing of oxygen and carbon dioxide. There are two computer subunits, one to
apply control signals to the mass flow controllers, which sets the desired inspiration oxygen and carbon dioxide fractions, and one to collect the
end-tidal data.

Physiol Rev • VOL 90 • APRIL 2010 • www.prv.org


728 LUC J. TEPPEMA AND ALBERT DAHAN

bitrarily (e.g., to 45 Torr), and others use a fixed in- excitatory substances in the brain (e.g., NO, glutamate,
crease relative to baseline end-tidal CO2 (e.g., ⫹5 Torr ROS). Although these components seem of lesser im-
above resting) or a value set to achieve equal back- portance during mild hyperoxia (140 –250 Torr), we
ground central drive throughout the hypoxic experi- cannot exclude some excitatory effect on breathing
ment (for example, by using a end-tidal CO2 value at from even brief increases in O2 (see Fig. 13; Ref. 613).
which ventilation in hyperoxia equals 4 l/m2 per min or With all of the above taken into account, our first
140 ml 䡠 min⫺1 䡠 kg⫺1), presumably to prevent a possible choice would be to assess the HVR using a normoxic
confounding influence of central-peripheral interaction baseline level. The choice of a hyperoxic baseline level
(195, 696, 720). The method of choice of the end-tidal has little or no advantage, and in addition, from a
CO2 is of importance when studying the effect of spe- physiological view point, hyperoxia does not represent
cific interventions, such as high altitude, acid-base al- a realistic ambient state.
terations, or pharmacological agents, on the HVR. An C) A PROPOSAL FOR MEASUREMENT OF HVR. Over the years,
insightful and acceptable alternative to the measure- Severinghaus and others made several attempts to
ment of HVR at a fixed end-tidal PCO2 would be the reach consensus on the HVR methods (131, 195, 696,
assessment of HVR at two to three PCO2 values (Fig. 12). 697). Consensus is required to overcome “lack of com-
Alternatives could be ⌬S or S40/S200. parability between results obtained by various groups
B) THE CHOICE OF BASELINE END-TIDAL PO2. When study- due to their variety in methods, definitions, choice of
ing HVR, the baseline oxygen level chosen is sometimes isocapnic PCO2 and duration of hypoxia” (696). In the
hyperoxic (end-tidal PO2 150 –250 Torr; Refs. 195, 696). so-called “proposed Lake Louise HVR Methods,” Sever-
This is done to “silence the peripheral chemoreflex inghaus (696) proposes three tests (Table 6): 1) an
response to CO2” (195). Whether this is achieved at isocapnic HVR (iHVR) to determine the hypoxic sensi-
mild hyperoxia is doubtful. Data from Pedersen et al. tivity of the carotid bodies lasting no longer than 5 min
(591) indicate that at 200 Torr, the fast peripheral com- and 2) a poikilocapnic HVR (pHVR) lasting 20 min to
ponent of the V̇I-CO2 response remains present in hu- predict the response to high altitude. In both tests,
mans (mean peripheral component was 27% of total baseline (prehypoxia) conditions are hyperoxic (150 ⬍
CO2 sensitivity). Even at much “deeper” levels of hy- end-tidal PO2 ⬍ 250 Torr). 3) The iHVR may be pro-
peroxia (end-tidal PO2 ⬎500 Torr), the human carotid longed to get an indication of the isocapnic HVD. Duffin
bodies remain active with an average 13% contribution (195) proposes three tests (Table 6): 1) the assessment
of the peripheral component to total CO2 sensitivity of peripheral chemoreflex responsiveness by measure-
(156). Furthermore, high oxygen levels affect central ment of the ventilatory response to CO2 at hyperoxia
respiratory drive via the Haldane effect and a reduction (150 Torr) and hypoxia (40 Torr) preferentially per-
of brain blood flow (this effect is independent of any formed by using the modified Read-rebreathing test (or
CO2 inhalation), causing a rise in brain tissue PCO2 alternatively performing 5-min isocapnic steps from
(156). High oxygen may also cause the formation of hyperoxia to hypoxia); 2) to get an indication of HVD

FIG. 12. Protocol to measure the ventilatory response to hypoxia. A: measuring the isocapnic ventilatory response to hypoxia (iHVR) at
three arterial PCO2 levels using the “Leiden” approach (test 1 in Table 1) in an awake healthy 30-year-old male volunteer. At each CO2 level,
the arterial PO2 is lowered from 100 mmHg (closed data points) to 45 mmHg and kept constant for 5 min (open data points). The ventilatory
increase is depicted by the arrows. The lines through the data are obtained from linear regression and are the post hoc steady-state normoxic
and acute hypoxic V̇I-CO2 response curves. B: the hypoxic response, expressed as ⌬V̇I/⌬SpO2, is plotted for each arterial CO2 concentration
(open squares and broken line). The O2/CO2 interaction (i.e., the carotid body-induced increase in hypoxic sensitivity at increasing levels of
arterial CO2) is abolished when the experiment is repeated during the inhalation of 0.15 end-tidal percent halothane.

Physiol Rev • VOL 90 • APRIL 2010 • www.prv.org


THE VENTILATORY RESPONSE TO HYPOXIA IN MAMMALS 729

FIG. 13. Measuring the hypoxic ventilatory response at three constant levels of end-tidal PCO2. A: isocapnic ventilatory responses to hypoxia
and hyperoxia at three arterial PCO2 levels obtained in a healthy 28-year-old female volunteer. Normoxic, hyperoxic (arterial PO2 ⫽ 140 mmHg), and
hypoxic (arterial PO2 ⫽ 45 mmHg) data points are given. The data points form post hoc steady-state V̇I-CO2 response curves in normoxia, hyperoxia,
and acute hypoxia (lines through the data points derived from linear regression). The left shift of the hyperoxic V̇I-CO2 response curve relative to
the normoxic curve is due to the central stimulatory effects from even mild hyperoxia. Consequently, using the hyperoxic data as a measure of the
isocapnic HVR (see Table 1) makes data interpretation difficult if not impossible. B: the data in these graphs are obtained by using a small adaptation
to the test 1 of the “Leiden proposal” of Table 6. The top graph shows the imposed end-tidal fractions of O2 (continuous line) and CO2 (dotted line).
In between the normoxic and hypoxic exposures, a 7-min isocapnic hyperoxic exposure (PO2 ⫽ 140 mmHg, measured in arterial blood) was added.
The middle graph shows the measured arterial O2-Hb saturation using pulse-oximetry. The bottom graph shows the measured inspired ventilation.
Each data point is one breath.

the first procedure is repeated before and after 20-min normoxic rather than hyperoxic baseline states to prevent
of hypoxia (or alternatively performing a 20-min iso- any excitatory effect of even mild hyperoxia. A transition
capnic hypoxic response); and 3) a 20-min poikilocap- from hyperoxia to hypoxia will change central ventilatory
nic step hypoxic response is performed to mimic the drive more than the transition from normoxia to hypoxia
response to environmental hypoxia. (see sect. VIA4B). This will affect the position of the V̇I-CO2
Our proposal is a modification of the methods out- response causing a leftward shift (Fig. 13). Of equal im-
lined by Severinghaus and Duffin (Table 6: the “Leiden” portance is the fact that hyperoxia does not silence the
proposal). Similar to the other proposals, we suggest peripheral chemoreceptors. Furthermore, we refrain from
performing three distinct tests. In test 1, 5-min step re- obtaining V̇I-CO2 responses curves at different PO2 levels
sponses into hypoxia (arterial PO2 45–50 Torr) are per- as proposed by Duffin (195) as ion channel and neuro-
formed at three different arterial PCO2 levels (e.g., ⫹2, ⫹4, transmitter responses to low oxygen and high PCO2/low
and ⫹8 Torr above resting). However, in contrast to the pH stimulation at the carotid bodies may differ and, as is
previous proposals, baseline oxygen levels are normoxic also important, arterial PO2 is a separate stimulus at the
(arterial PO2 ⫽ 100 Torr). One is now able to calculate the peripheral chemoreceptors (277). Furthermore, by per-
acute hypoxic response (i.e., hypoxic sensitivity), and forming step hypoxic tests, one may decide to use peak
since the responses are obtained at various PCO2 levels, hypoxic ventilation rather than 5 min of hypoxia as hy-
one gets a full characterization of O2-CO2 interaction, a poxic measurement point. This is especially important
phenomenon most likely originating at the peripheral che- when the temporal profile of the response indicates that
moreceptors (see also sect. VD; Fig. 12). Tests 2 and 3 are HVD is apparent at times ⬍5 min of hypoxia (see also Fig.
similar to the ones proposed by Severinghaus but with 13 for an example).

Physiol Rev • VOL 90 • APRIL 2010 • www.prv.org


730 LUC J. TEPPEMA AND ALBERT DAHAN

TABLE 6. Proposed methods to measure the hypoxic ventilatory response

Proposed Methods to Measure Hypoxic Ventilatory Response

“Lake Louise HVR Method proposal” (696)


Test 1: isocapnic HVR (iHVR) to assess the hypoxic
sensitivity of the carotid bodies
Baseline oxygen level 150 ⬍ end-tidal PO2 ⬍250 Torr for 10 min
Hypoxic test Reduce the inspired O2 level such that the SpO2 is rapidly lowered and maintained
at 80% (range 75–85%) for 5 min
PCO2 Keep the end-tidal PCO2 constant at a level causing hyperoxic ventilation to rise to
a standard, e.g., 140 ml 䡠 min⫺1⫺kg⫺1
Expression of response iHVR ⫽ ⌬V̇I/⌬SpO2 where ⌬V̇I ⫽ the increase in V̇I from baseline to the 5th min of
hypoxia
Test 2: measuring the isocapnic hypoxic ventilatory
decline (iHVD)
Hypoxic test Expand test 1 to 20 min of isocapnic hypoxia
Expression of iHVD % fall in ⌬V̇I from the 5th to the 20th min
Test 3: poikilocapnic HVR (pHVR) to predict the HVR at
altitude
Baseline oxygen level 150 ⬍ end-tidal PO2 ⬍250 Torr for 10 min
Hypoxic test Inspiration of a O2/N2 gas mixture (e.g., 12 % O2) to
Reduce SpO2 to 80% for 20 min pHVR ⫽ ⌬V̇I/⌬SpO2
Expression of response pHPR ⫽ ⌬PETCO2/⌬SpO2
The “Ontario proposal” (195)
Test 1: measuring peripheral chemoreflex sensitivity
Hypoxic test* Ventilatory response to CO2 at an isoxic high (150 Torr) and an isoxic low (40
Torr) oxygen tension using the modified Read-rebreathing test
Expression of response Differences in hyperoxic and hypoxic V̇I-CO2 sensitivities and position of the curves
Test 2: measuring the hypoxic ventilatory decline (HVD)
Hypoxic testⴱⴱ Ventilatory response to CO2 at an isoxic high (150 Torr) and an isoxic low (40
Torr) oxygen tension using the modified Read-rebreathing test (lasting no longer
than 5 min) performed before and after 20 min of hypoxia
Expression of response As in test 1
Test 3: measuring the poikilocapnic HVR (pHVR)
Hypoxic test Step reduction in end-tidal PO2 from hyperoxia (150 Torr) to hypoxia lasting 20 min
Expression of response ⌬V̇I from baseline to 20th min of hypoxia at a specified O2 level
⌬CO2 from baseline to 20th min of hypoxia
The “Leiden proposal” (2010)
Test 1: isocapnic HVR (iHVR) to assess the hypoxic
sensitivity and O2/CO2 interaction of the carotid
bodies
Baseline oxygen level Arterial PO2 ⫽ 100 Torr for 10 min
Hypoxic test Step reduction in arterial PO2 to 45–50 Torr, which is kept constant for 5 min
PCO2 The test is performed at three arterial PCO2 levels, e.g., ⫹2, ⫹5 and ⫹12 Torr
above resting. In between tests there is a 15- to 30-min rest period
Expression of response HVR ⫽ ⌬V̇I/⌬SpO2; an alternative could be HVR ⫽ ⌬V̇I /⌬log PO2 (units L/min) if in
humans ventilation would appear to be linearly related to log PO2 below 100
Torr (but ⬎10 Torr)
where ⌬V̇I ⫽ the increase in V̇I from baseline to the 5th min of hypoxia. PO2 is
arterial (preferred) or end-tidal PO2
Test 2: isocapnic hypoxic ventilatory decline (iHVD)
Baseline oxygen level Arterial PO2 ⫽ 100 Torr for 5–10 min
Hypoxic test Step reduction in arterial PO2 to 45–50 Torr, which is kept constant for 20 min
PCO2 The test is performed at one constant arterial PCO2 levels, e.g., ⫹2 or ⫹4 Torr
above resting
Expression of response ⌬V̇I from baseline to 20th min of hypoxia at a specified O2 level
Test 3: poikilocapnic HVR (pHVR)
Baseline oxygen level Inspired oxygen concentration ⫽20.8% for 5–10 min
Hypoxic test Step reduction in inspired O2 concentration (e.g. to 10%), which is kept constant
for 20 min
PCO2 CO2 concentrations remains not controlled throughout the test
Expression of response ⌬V̇I from baseline to 20th min of hypoxia at a specified O2 level
⌬CO2 from baseline to min 20 of hypoxia

*An alternative is the measurement of a three-point steady-state V̇I-CO2 responses performed at hyperoxia and hypoxia. **An alternative test
is an isocapnic 20-min step hypoxic test as suggested by Severinghaus (696). iHVR, isocapnic hypoxic ventilatory response; pHVR, poikilocapnic
hypoxic ventilatory response; pHPR, response of the end-tidal PCO2 to poikilocapnic hypoxia; SpO2, arterial Hb oxygen saturation as determined by
pulse oximetry; PETCO2, end-tidal PCO2; V̇I, inspired minute ventilation.

Physiol Rev • VOL 90 • APRIL 2010 • www.prv.org


THE VENTILATORY RESPONSE TO HYPOXIA IN MAMMALS 731

B. Expressing the Steady-State HVR artifact with an incorrect calculation of HVR (613). For
example, a metabolic acidosis increases P50 and changes
Mathematical expressions of the HVR have to take the value of ⌬V̇I/⌬SpO2 without any change in the rela-
into account the fact that the steady-state relationships tionship between V̇I and PO2 and hence of the HVR. And
between ventilation and arterial and end-tidal PO2 are from a technical point of view, it is our experience that at
nonlinear. The HVR has been quantified using hyperbolic low saturation levels (⬍ 80%), measurements derived
and exponential expressions (411, 697), and there is no from pulse oximetry are a less reliable measure of the
physiological reason to select one over the other. actual arterial HbO2 saturation.
The hyperbolic expression (457, 697, 833) In the cat, it has been demonstrated that in the
hypoxic range, ventilation is a linear function of log PaO2,
V̇ I ⫽ V̇ 0 ⫹ A/(PO2 ⫺ C) (1) while hypoxic sensitivity, defined as the ratio ⌬V̇I/⌬log
PaO2, is linearly related to the arterial H⫹ concentration
where V̇0 is the horizontal asymptote in V̇I for infinite high (650). Recently, we found that in humans relative changes
PO2 values, A is the hypoxic sensitivity, and C is the in hypoxic sensitivity induced by alterations in arterial
vertical asymptote in PO2 for infinite high V̇I values. In PCO2 are the same regardless of how the sensitivity was
most studies, the asymptote C is fixed to 32 Torr, which defined, either as ⌬V̇I/⌬log PaO2 or as ⌬V̇I/⌬SpO2 (775). As
causes a bias in the estimation of the parameter A (214). in cats, humans also showed a linear relationship between
Bayesian estimation schemes using soft constraints on C are hypoxic sensitivity (defined as ⌬V̇I/⌬log PaO2) and arterial
advocated to improve the estimation of parameter A (214). H⫹ concentration (see Fig. 7; Ref. 775). Future studies in
The hyperbolic expression indicates that the reciprocal of humans are warranted to see if, as in the cat, ventilation
ventilation versus PO2 is a linear relationship, an observation is a linear function of log PaO2 in the hypoxic range. If this
that has been established experimentally (272). The expo- would appear to be the case, then we would suggest the
nential expression (58, 176, 411, 697) use of the expression ⌬V̇I/⌬log PO2 to estimate hypoxic
sensitivity, with end-tidal O2 but preferentially arterial PO2
as input. It is a simple approach, alike to the use of oxygen
V̇I ⫽ V̇0 ⫹ G 䡠 exp共⫺D 䡠 PO2兲 (2)
saturation but without the confounders that are inher-
ently present in the use of SpO2. When performing multi-
where V̇0 is V̇I for infinite high PO2 values, G is the hypoxic ple steps into hypoxia (e.g., at three different background
sensitivity, and D is a shape parameter. The shape param- PCO2 levels), it further enables the description of O2-CO2
eter is often fixed to a specific value (e.g., to 0.29%⫺1; Ref. interaction (see Figs. 7 and 12) and the effects that drugs
824). This is done when the O2 input is not rich enough to may have on this important feature of the peripheral
provide an estimation of D (e.g., when employing single chemoreceptors (such as abolishment of O2-CO2 interac-
steps into hypoxia). This value of D has been estimated by tion by inhalational anesthetics).
fitting an exponential through the function 100 ⫺ Z, where
Z is the saturation calculated from the Hill model of the
oxygen dissociation curve (824). The use of an exponen- C. Modeling the HVR
tial model is equivalent to the empirically linear relation
of log dV̇I to PO2 as used by Kronenberg et al. (411). Several models allow the estimation of physiologi-
Since there is an empirical linear relationship be- cally relevant parameters (hypoxic sensitivities/gains and
tween V̇I and blood oxygen desaturation (626, 628), a time constants) from single experimental data sets. Initial
more simple approach would be to express ventilation as mathematical models of the ventilatory response to iso-
a linear function of oxygen saturation capnic hypoxia consisted of two additive components: an
initial stimulating component arising from the carotid
V̇I ⫽ V̇0 ⫹ S/关SpO2共0兲 ⫺ SpO2兴 (3) bodies and a second, slower component that causes a
decline in ventilation (176, 824). These models were un-
where V̇0 and SpO2(0) are prehypoxic ventilation and able to fit human responses into and out of hypoxia
saturation, respectively, and S is the hypoxic sensitivity simultaneously due to the asymmetry in the magnitude of
(units, L 䡠min⫺1 䡠%⫺1). There are several reasons, how- the fast on- and off-transients (824). The model proposed
ever, why saturation may not be the most appropriate tool by DeGoede et al. (176) is an exponential expression, in
to express the HVR. Oxygen saturation is not the stimulus which the two components differ with respect to their
to the oxygen sensors at the carotid bodies (e.g., Ref. dynamics (expressed by a time constant or ␶), gain values,
313). The V̇I-SpO2 relationship is not under all circum- and input latencies. One of the first human studies (824)
stances and protocols linear (159, 336, 384). And, most yielded the following parameter values: fast component:
importantly, a change in the position of the oxygen dis- ␶ ⫽ 6 s, gain of response into hypoxia 50 l/min, gain of
sociation curve (reflected in a change in P50) creates an response out of hypoxia 30 l/min; slow component: ␶ ⫽ 4

Physiol Rev • VOL 90 • APRIL 2010 • www.prv.org


732 LUC J. TEPPEMA AND ALBERT DAHAN

min, gain of response into hypoxia ⫺40 l/min (i.e., HVD), account (e.g., by subtracting the variations in ventilation
gain of response out of hypoxia ⫺20 l/min (relief of HVD). by performing additional isohypercapnic experiments or
The asymmetry in the gain values suggests modulation performing hypoxic experiments just above resting end-
of the peripheral component during sustained hypoxia as tidal PCO2 levels), we suggest adding a trend term in the
the cause of HVD (cf. Ref. 162). analysis. Such an approach has been applied successfully
Khamnei and Robbins (382) examined how HVD is in studies assessing central and peripheral CO2 chemo-
best incorporated in models of the HVR. They concluded sensitivity yielding significant values for a trend in the
that modulation of the peripheral chemoreflex gain during order of 70 ml/min2 (156).
sustained hypoxia consistently describes the human data
in contrast to models in which hypoxic depression is
modeled as an additive component or as a modulator of VII. COMMENTS AND FUTURE CHALLENGES
central or total CO2 sensitivity (382). In a quantitative
modeling study (by assuming that the peripheral gain At sea level, hypoxia is a relatively rare event in
term is a linear function of arterial oxygen saturation), healthy individuals, except in utero and perhaps during a
this was confirmed and moreover allowed (the asymme- neonatal period in which irregular breathing is not un-
try of) the ventilatory response into and out of hypoxia to common, or during sleep when wakefulness drive is ab-
be analyzed simultaneously (577). However, in some sub- sent and one may sometimes discontinue breathing. When
jects, the description of the response out of hypoxia (the healthy individuals are exposed to inspired oxygen ten-
off-transient) remained inadequate. Therefore, to opti- sions lower or higher than normal, for example, at high
mize the model, a further model expansion was tested in altitude or in an experimental setting, the respiratory
which HVD was modeled by modulation of the peripheral system responds in a way that critically depends on the
chemoreflex gain (577) combined with a component that stage of maturation (neonates show different responses
is independent of the peripheral chemoreflex loop (445). than adults), the physiological background condition
The extended model yielded significantly better fits in just (e.g., CO2 and exercise increase the HVR), the intensity of
two of six subjects. Both had a chemoreflex-independent the stimulus, and, last but not least, the time pattern and
HVD component ⬎50%. This contrasts with a value of duration of the exposure. Acute, chronic, and episodic
⬃10% in the four other subjects examined. Liang et al. hypoxia with or without a chronic character all have
(445) concluded that in some subjects, but not others, different effects on ventilatory control. Both in neonates
there might be a component of HVD that is independent of and adults, the respiratory system displays remarkable
the peripheral chemoreflex sensitivity. plasticity when exposed to intermittent and/or chronic
Some studies, human and animal, indicate that the hypoxic paradigms that lead to changes in HVR pheno-
fast hyperventilatory response to isocapnic hypoxia is types. Animal studies show that when hypoxic stimulus
best described by a fast and a slower component (58, 198, paradigms (or environmental factors such as nicotine
384, 550) with time constants for the fast and slow com- exposure or maternal separation) are imposed during a
ponents of 2–3 s and 70 –100 s, respectively (58, 550). neonatal critical time window, plastic changes persist into
Possibly, the slow peripheral component is a manifesta- adulthood and are difficult to reverse, if at all. An impor-
tion of central neuronal dynamics, equivalent to the slow tant challenge for future research is to examine whether
component observed when the carotid sinus nerve is the underlying mechanisms that are responsible for these
electrically stimulated (211). This then suggests that the changes also apply in humans.
HVR must be modeled by at least three components, two An important circumstance that mediates plasticity-
of peripheral origin, one of which (or both?) is modulated induced respiratory changes in rodents is neonatal oxida-
by central hypoxia, and in some subjects a peripheral tive stress. Rats exposed to chronic intermittent hypoxia
chemoreflex loop independent HVD component. develop an augmented carotid body sensitivity and an
Finally, studies that strictly control end-tidal PCO2 increased HVR, changes that persist into adulthood (585;
levels during HVR measurement may be influenced by a note, however, that as explained in section II, the increase
slow increase in ventilation that occurs upon the induc- in HVR is not without controversy). If translatable to
tion of isohypercapnia. Tansley et al. (764) show a pro- human newborns, this raises a number of clinically rele-
gressive increase in ventilation lasting 2– 8 h during expo- vant issues. Newborn babies, especially when premature,
sure to isohypercapnia (end-tidal PCO2 ⫽ 6.5 Torr above display irregular breathing that can be accompanied by
resting levels). The increase in ventilation is slow and frequent apneic events. Does this disturb a normal matu-
most prominent during the first 2 h of exposure (⬃66 ration of the HVR in premature babies, and might this lead
ml/min for the first 2 h). This varying effect of baseline to sensitization of the carotid bodies thereby destabilizing
ventilation may hamper proper assessment of the HVR breathing just like in animals? Further destabilizing
irrespective of the method chosen to measure the HVR. breathing might lead to a higher frequency of apneic
Although there are various ways to take this effect into events, an increase in the number of deoxygenation/

Physiol Rev • VOL 90 • APRIL 2010 • www.prv.org


THE VENTILATORY RESPONSE TO HYPOXIA IN MAMMALS 733

reoxygenation cycles, and finally to an increased produc- with an enhanced sympathetic activity, systemic hyper-
tion of ROS that would further aggravate the oxidative tension, and an augmented HVR. Similar to animals ex-
stress (in neonatal rats the augmented carotid body sen- posed to chronic intermittent hypoxia, OSA patients dis-
sitivity is a ROS-related phenomenon; Ref. 586). And can play increased production of ROS (202, 365). The in-
possible changes in the HVR be reversed at adult or creases in both systemic blood pressure and HVR can be
possibly at earlier age? An important difference between reversed with nasal continuous positive airway pressure
newborn and adult rats is the much higher sensitivity to treatment (790, 791). To mimic the complex events that
chronic intermittent hypoxia of the former with respect to occur in OSA in a laboratory setting may be elusive in the
the effects on carotid body sensitization, morphological sense that (awake) healthy subjects do not display the
changes (e.g., hyperplasia), and reversibility (see Fig. 10; increase in resting tone of upper airway dilator muscles
Ref. 585). that in OSA patients serves to compensate for a decreased
In neonatal animals, within a critical time window of patency of the upper airways. This may well be one of the
plasticity in the period after birth, chronic hypoxia im- reasons why it is difficult if not impossible to evoke LTF
pairs carotid body maturation and O2 sensitivity, causes of upper airway dilator muscles in healthy sleeping sub-
degenerative changes in glomus cells and sinus nerve jects but less so in snorers with upper airway flow limi-
afferents, and reduces both the CNS gain and the HVR tations (21). Apart from the absence of obstructive ap-
(Fig. 9). The fact that at the level of ventilation the dimin- neas, an additional limitation of studies in awake healthy
ished response to hypoxia persists into adulthood indi- subjects is the lack of arousals that in sleeping OSA
cates that at least some of these adaptations are perma- patients may play a crucial (albeit not clearly defined) role
nent and perhaps difficult to reverse. If similar mecha- and the absence of the influences of sleep itself on the
nisms are operating in humans, the question arises as to control of breathing. And finally, in OSA patients, apneic
whether chronic hypoxia in newborn babies, when un- events followed by hyperventilation are associated with
treated, may have similar deleterious effects that ulti- alternating episodes of low and high PCO2, which makes
mately may lead to a decreased ability to respond to an the stimulus paradigm fundamentally different from that
asphyxic stimulus, long-lasting exposures to hypoxia, and in most laboratory studies in which the hypoxic episodes
failure to autoresuscitate. However, at least in the west- are hypocapnic or at best isocapnic (Table 5). On the
ern world, the clinical relevance of this issue may be other hand, studies in animals and healthy humans are of
limited because when newborn babies are hypoxemic, for considerable interest because they can uncover mecha-
example, due to hypoventilation, ventilation perfusion nisms by which chronic intermittent hypoxia by itself can
mismatch, intrapulmonary or cardiac shunts, anemia, etc., lead to maladapations such as inflammation, endothelial
they will be rapidly treated with oxygen. On the other dysfunction, atherosclerosis, and other cardiovascular pa-
hand however, apart from the well-known detrimental thology.
side effects of oxygen (for example, retinopathy; see Ref. From studies over the last decade, the picture
816 for further references), a permanently reduced HVR emerges that oxygen sensing by the carotid bodies occurs
(from chronic oxygen therapy) in these children that per- via several parallel pathways. The key role of potassium
sists into adulthood cannot be excluded. In rats, neonatal channel species is well established, but identification of
chronic hyperoxia can cause a reduced HVR at adult age individual members involved is complicated by species-
depending on the level of hyperoxia and the duration of and even strain-related differences in potassium channel
the neonatal exposure (41, 71, 72). regulation by hypoxia. AMP-activated protein kinase
An important point to consider when putting data (AMPK), an enzyme considered to play a key role in
from neonatal animals into the context of newborn babies cellular metabolism, has been identified as a link between
is relative maturity at birth. If 1- to 5-day-old rats resemble hypoxia-induced changes in mitochondrial metabolism
preterm babies in their development of the HVR (73), the and potassium channel activity. Mice deficient in the ␣2
findings from this species may be less relevant for term subunit of AMPK show an attenuated frequency response
babies. This, however, does not obviate the need to focus to hypoxia in vivo (222). A possible role of AMPK in
attention on possible adverse effects of neonatal oxida- glucose sensing by the carotid bodies, in the adaptation to
tive stress (both hypoxia and hyperoxia) on the HVR in high altitude, as well as in exercise remain interesting
both preterm and term babies. subjects for further study. To uncover the role of the
In adult healthy humans, chronic intermittent hyp- numerous individual type I cell neurotransmitters in the
oxia results in an augmented HVR, presumably related to integrated response of the carotid body to hypoxia is
oxidative stress (increased production of ROS; Refs. 374, another challenging issue.
605, 817). Studies in humans on the effects of chronic A fascinating example of respiratory plasticity at
intermittent hypoxia are often aimed at mimicking adult age becomes manifest during adaptation to chronic
hypoxic episodes that occur in patients with obstructive hypoxia. In fact, all elements in the carotid bodies and
sleep apnea (OSA). This chronic disease is associated CNS known to participate in the hypoxia signaling trans-

Physiol Rev • VOL 90 • APRIL 2010 • www.prv.org


734 LUC J. TEPPEMA AND ALBERT DAHAN

duction cascade undergo plastic changes during chronic Finally, the method of measuring the HVR, especially
hypoxia. How does the organism manage to orchestrate in humans, remains a topic of intense discussion. We
so many changes (recalibrations) at the same time with- propose a standardized method (the Leiden protocol) that
out losing control over system stability? One of the master allows measurement of the HVR with as few problems as
regulators now identified is HIF-1, a transcription factor possible in its interpretation. Adoption of this protocol
that regulates the expression of numerous genes encod- will enable simple comparisons of physiological and phar-
ing elements involved in improving tissue oxygenation macological studies on HVR between laboratories.
(HVR, erythropoiesis, angiogenesis, etc.) and in metabolic
ACKNOWLEDGMENTS
adaptations (increased ATP supply from glycolysis, opti-
mization, and reduction of oxidative phosphorylation). We thank Dr. Egbert Lakke for making Figure 4.
Systems biology approaches are currently used to map Address for reprint requests and other correspondence:
individual genes, untangle genetic networks, and identify L. J. Teppema, Dept. of Anesthesiology, Leiden University Med-
ical Center, PO Box 9600, 2300 RC Leiden, The Netherlands
their hubs (441, 850). Systems biology also offers an im-
(e-mail: l.j.s.m.teppema@lumc.nl).
portant avenue to discover cross-talk between hypoxia
and other key signaling mechanisms. For example, it has
been shown that angiogenesis in hypoxia is induced by REFERENCES
cross-talk with notch signaling (441); notch receptors are
1. Aaron EA, Powell FL. Effect of chronic hypoxia on hypoxic
highly conserved transmembrane proteins thought to be ventilatory response in awake rats. J Appl Physiol 74: 1635–1640,
involved in cell-to-cell communication and cell differenti- 1993.
ation (references in Ref. 441). Similar mechanisms may 2. Acker H. Mechanisms and meaning of cellular oxygen sensing in
the organism. Respir Physiol 95: 1–10, 1994.
operate in the carotid bodies, but efforts to examine this 3. Acker H. The oxygen sensing signal cascade under the influence of
have not yet been undertaken. reactive oxygen species. Philos Trans R Soc Lond B Biol Sci 360:
How hypoxia influences the stability and activity of 2201–2210, 2005.
4. Adachi T, Ishikawa K, Hida W, Matsumoto H, Masuda T, Date
transcription factors is also focus of current interest. The F, Ogawa K, Takeda K, Furuyama K, Zhang Y, Kitamuro T,
fact that transcription factors may also undergo posttran- Ogawa H, Maruyama Y, Shibahara S. Hypoxemia and blunted
scriptional modification independent of hypoxia is an in- hypoxic ventilatory responses in mice lacking heme oxygenase-2.
Biochem Biophys Res Commun 320: 514 –522, 2004.
teresting but complicating factor in the interpretation of 5. Adams JM, Severns ML. Interaction of chemoreceptor effects
these studies because hypoxia-independent pathways and its dependence on the intensity of stimuli. J Appl Physiol 52:
may then autonomously influence the expression of genes 602– 606, 1982.
6. Ahmed M, Giesbrecht GG, Serrette C, Georgopoulos D, An-
involved in the adaptation of the HVR to chronic condi- thonisen NR. Ventilatory response to hypoxia in elderly humans.
tions. For example, HIF-1␣ can be stabilized by NO-me- Respir Physiol 83: 343–351, 1991.
diated S-nitrosylation and by heat shock protein 90, and 7. Aicher SA, Kurucz OS, Reis DJ, Milner TA. Nucleus tractus
solitarius efferent terminals synapse on neurons in the caudal
both processes are hypoxia independent (692). With re- ventrolateral medulla that project to the rostral ventrolateral me-
gard to HIF-1␣, targeting its stability independent of the dulla. Brain Res 693: 51– 63, 1995.
PO2 may appear to open new treatment options of HIF-1- 8. Ainslie PN, Barach A, Cummings KJ, Murrell C, Hamlin M,
Hellemans J. Cardiorespiratory and cerebrovascular responses to
related pathology. For example, an interesting recent ob- acute poikilocapnic hypoxia following intermittent and continuous
servation in humans is the reversal of hypoxia-induced exposure to hypoxia in humans. J Appl Physiol 102: 1953–1961,
pulmonary hypertension by iron supplementation (713; 2007.
9. Ainslie PN, Burgess KR. Cardiorespiratory and cerebrovascular
further references in Refs. 441, 692). responses to hyperoxic and hypoxic rebreathing: effects of accli-
Animal studies indicate an important effect of geno- matization to high altitude. Respir Physiol Neurobiol 161: 201–209,
type on the HVR phenotype (i.e., the HVR magnitude). 2008.
10. Ainslie PN, Kolb JC, Ide K, Poulin MJ. Effects of five nights of
With the relatively large effect of environment and pre- normobaric hypoxia on the ventilatory responses to acute hypoxia
frontal cortex on the magnitude and variability of HVR and hypercapnia. Respir Physiol Neurobiol 138: 193–204, 2003.
taken into account, a genetic influence seems hard to 11. Ainslie PN, Kolb JC, Ide K, Poulin MJ. Effect of five nights of
normobaric hypoxia on cardiovascular responses to acute isocap-
discover from human studies. However, in certain popu- nic hypoxia in humans: relationship to ventilatory chemosensitiv-
lations and patient groups, the genotype may be the more ity. Ergonomics 48: 1523–1534, 2005.
dominant factor. An important question that so far has 12. Ainslie PN, Poulin MJ. Ventilatory, cerebrovascular, and cardio-
vascular interactions in acute hypoxia: regulation by carbon diox-
received little attention is whether linking of the genotype ide. J Appl Physiol 97: 149 –159, 2004.
to magnitude of HVR is of clinical importance. Does a 13. Aizad T, Bodani J, Cates D, Horvath L, Rigatto H. Effect of a
HVR risk phenotype exist which leads to an increased risk single breath of 100% oxygen on respiration in neonates during
sleep. J Appl Physiol 57: 1531–1535, 1984.
for adverse respiratory events? And is this phenotype 14. Alea OA, Czapla MA, Lasky JA, Simakajornboon N, Gozal E,
linked to an increase in morbidity and mortality? This is, Gozal D. PDGF-beta receptor expression and ventilatory acclima-
for example, of importance for patients with central or tization to hypoxia in the rat. Am J Physiol Regul Integr Comp
Physiol 279: R1625–R1633, 2000.
obstructive sleep apnea or patients receiving respiratory 15. Alvaro R, Alvarez J, Kwiatkowski K, Cates D, Rigatto H. Small
depressants in the perioperative setting. preterm infants less than or equal to 1500 g have only a sustained

Physiol Rev • VOL 90 • APRIL 2010 • www.prv.org


THE VENTILATORY RESPONSE TO HYPOXIA IN MAMMALS 735

decrease in ventilation in response to hypoxia. Pediatr Res 32: 37. Bascom DA, Clement ID, Cunningham DA, Painter R, Robbins
403– 406, 1992. PA. Changes in peripheral chemoreflex sensitivity during sus-
16. Alvaro RE, Robertson M, Al Saedi S, Lemke RP, Cates DB, tained, isocapnic hypoxia. Respir Physiol 82: 161–176, 1990.
Rigatto H. Preliminary characterization of a placental factor in- 38. Bascom DA, Clement ID, Dorrington KL, Robbins PA. Effects
hibiting breathing in fetal sheep. Reprod Fertil Dev 9: 641– 649, of dopamine and domperidone on ventilation during isocapnic
1997. hypoxia in humans. Respir Physiol 85: 319 –328, 1991.
17. Andronikou S, Shirahata M, Mokashi A, Lahiri S. Carotid body 39. Bavis RW. Developmental plasticity of the hypoxic ventilatory
chemoreceptor and ventilatory responses to sustained hypoxia and response after perinatal hyperoxia and hypoxia. Respir Physiol
hypercapnia in the cat. Respir Physiol 72: 361–374, 1988. Neurobiol 149: 287–299, 2005.
18. Ang RC, Hoop B, Kazemi H. Role of glutamate as the central 40. Bavis RW, Olson EB Jr, Mitchell GS. Critical developmental
neurotransmitter in the hypoxic ventilatory response. J Appl period for hyperoxia-induced blunting of hypoxic phrenic re-
Physiol 72: 1480 –1487, 1992. sponses in rats. J Appl Physiol 92: 1013–1018, 2002.
19. Arias-Stella J, Valcarcel J. Chief cell hyperplasia in the human 41. Bavis RW, Olson EB Jr, Vidruk EH, Bisgard GE, Mitchell GS.
carotid body at high altitudes: physiologic and pathologic signifi- Level and duration of developmental hyperoxia influence impair-
cance. Hum Pathol 7: 361–373, 1976. ment of hypoxic phrenic responses in rats. J Appl Physiol 95:
20. Astrom K, Cohen JE, Willett-Brozick JE, Aston CE, Baysal 1550 –1559, 2003.
BE. Altitude is a phenotypic modifier in hereditary paraganglioma 42. Bavis RW, Olson EB Jr, Vidruk EH, Fuller DD, Mitchell GS.
type-1: evidence for an oxygen-sensing defect. Hum Genet 113: Developmental plasticity of the hypoxic ventilatory response in
228 –237, 2003. rats induced by neonatal hypoxia. J Physiol 557: 645– 660, 2004.
21. Babcock M, Shkoukani M, Aboubakr SE, Badr MS. Determi- 43. Bavis RW, Russell KE, Simons JC, Otis JP. Hypoxic ventilatory
nants of long-term facilitation in humans during NREM sleep. responses in rats after hypercapnic hyperoxia and intermittent
J Appl Physiol 94: 53–59, 2003. hyperoxia. Respir Physiol Neurobiol 155: 193–202, 2007.
22. Baby S, Roy A, Lahiri S. Role of mitochondria in the regulation of 44. Bavis RW, Wenninger JM, Miller BM, Dmitrieff EF, Olson EB
hypoxia-inducible factor-1␣ in the rat carotid body glomus cells. Jr, Mitchell GS, Bisgard GE. Respiratory plasticity after perina-
Histochemy Cell Biol 124: 69 –76, 2005. tal hyperoxia is not prevented by antioxidant supplementation.
23. Bach KB, Mitchell GS. Hypoxia-induced long-term facilitation of Respir Physiol Neurobiol 160: 301–312, 2008.
respiratory activity is serotonin dependent. Respir Physiol 104: 45. Bavis RW, Mitchell GS. Long-term effects of the perinatal envi-
251–260, 1996. ronment on respiratory control. J Appl Physiol 104: 1220 –1229,
24. Baier RJ, Fajardo C, Alvarez J, Cates DB, Nowaczyk B, 2008.
Rigatto H. The effects of gestational age and labour on the breath- 46. Bayliss DA, Millhorn DE. Central neural mechanisms of proges-
ing and behaviour response to oxygen and umbilical cord occlusion terone action: application to the respiratory system. J Appl Physiol
in the fetal sheep. J Dev Physiol 18: 93–98, 1992. 73: 393– 404, 1992.
25. Bailey PL, Lu JK, Pace NL, Orr JA, White JL, Hamber EA, 47. Baysal BE. A phenotypic perspective on mammalian oxygen sen-
Slawson MH, Crouch DJ, Rollins DE. Effects of intrathecal sor candidates. Ann NY Acad Sci 1073: 221–233, 2006.
morphine on the ventilatory response to hypoxia. N Engl J Med 48. Baysal BE, Ferrell RE, Willett-Brozick JE, Lawrence EC,
343: 1228 –1234, 2000. Myssiorek D, Bosch A, van der MA, Taschner PE, Rubinstein
26. Bairam A, Carroll JL. Neurotransmitters in carotid body devel- WS, Myers EN, Richard CW III, Cornelisse CJ, Devilee P,
opment. Respir Physiol Neurobiol 149: 217–232, 2005. Devlin B. Mutations in SDHD, a mitochondrial complex II gene, in
27. Baker TL, Fuller DD, Zabka AG, Mitchell GS. Respiratory hereditary paraganglioma. Science 287: 848 – 851, 2000.
plasticity: differential actions of continuous and episodic hypoxia 49. Becker RF, King JE, Marsh RH, Wyrick AD. Intraauterine res-
and hypercapnia. Respir Physiol 129: 25–35, 2001. piration in the rat fetus. 1. Direct observations–Comparison with
28. Baker-Herman TL, Fuller DD, Bavis RW, Zabka AG, Golder the guinea pig. Am J Obstet Gynecol 90: 236 –246, 1964.
FJ, Doperalski NJ, Johnson RA, Watters JJ, Mitchell GS. 50. Bee D, Barer G, Wach R, Pallot D, Emery C, Jones S. Structure
BDNF is necessary and sufficient for spinal respiratory plasticity and function of the carotid body in New Zealand genetically hyper-
following intermittent hypoxia. Nat Neurosci 7: 48 –55, 2004. tensive rats. Q J Exp Physiol 74: 691–701, 1989.
29. Balbir A, Lande B, Fitzgerald RS, Polotsky V, Mitzner W, 51. Bee D, Pallot DJ. Acute hypoxic ventilation, carotid body cell
Shirahata M. Behavioral and respiratory characteristics during division, and dopamine content during early hypoxia in rats. J Appl
sleep in neonatal DBA/2J and A/J mice. Brain Res 1241: 84 –91, Physiol 79: 1504 –1511, 1995.
2008. 52. Behan M, Wenninger JM. Sex steroidal hormones and respiratory
30. Balbir A, Lee H, Okumura M, Biswal S, Fitzgerald RS, Shira- control. Respir Physiol Neurobiol 164: 213–221, 2008.
hata M. A search for genes that may confer divergent morphology 53. Beidleman BA, Muza SR, Fulco CS, Cymerman A, Ditzler D,
and function in the carotid body between two strains of mice. Am J Stulz D, Staab JE, Skrinar GS, Lewis SF, Sawka MN. Intermit-
Physiol Lung Cell Mol Physiol 292: L704 –L715, 2007. tent altitude exposures reduce acute mountain sickness at 4300 m.
31. Bamford O, Hawkins RL. Central effects of an alpha 2-adrenergic Clin Sci Lond 106: 321–328, 2004.
antagonist on fetal lambs: a possible mechanism for hypoxic apnea. 54. Bekedam DJ, Mulder EJ, Snijders RJ, Visser GH. The effects of
J Dev Physiol 13: 353–358, 1990. maternal hyperoxia on fetal breathing movements, body move-
32. Bamford OS, Dawes GS, Denny R, Ward RA. Effects of the alpha ments and heart rate variation in growth retarded fetuses. Early
2-adrenergic agonist clonidine and its antagonist idazoxan on the Hum Dev 27: 223–232, 1991.
fetal lamb. J Physiol 381: 29 –37, 1986. 55. Bell EL, Chandel NS. Genetics of mitochondrial electron trans-
33. Bamford OS, Sterni LM, Wasicko MJ, Montrose MH, Carroll port chain in regulating oxygen sensing. In: Methods in Enzymol-
JL. Postnatal maturation of carotid body and type I cell chemore- ogy. Oxygen Biology and Hypoxia, edited by H. Sies and B. Brune.
ception in the rat. Am J Physiol Lung Cell Mol Physiol 276: Orlando, FL: Academic, 2007, p. 447– 461.
L875–L884, 1999. 56. Bellville JW, Whipp BJ, Kaufman RD, Swanson GD, Aqleh KA,
34. Barnard P, Andronikou S, Pokorski M, Smatresk N, Mokashi Wiberg DM. Central and peripheral chemoreflex loop gain in
A, Lahiri S. Time-dependent effect of hypoxia on carotid body normal and carotid body-resected subjects. J Appl Physiol 46:
chemosensory function. J Appl Physiol 63: 685– 691, 1987. 843– 853, 1979.
35. Barros RC, Branco LG. Central dopamine modulates anapyrexia 57. Berkenbosch A, Dahan A, DeGoede J, Olievier IC. The venti-
but not hyperventilation induced by hypoxia. J Appl Physiol 92: latory response to CO2 of the peripheral and central chemoreflex
975–981, 2002. loop before and after sustained hypoxia in man. J Physiol 456:
36. Barros RC, Branco LG, Carnio EC. Evidence for thermoregula- 71– 83, 1992.
tion by dopamine D1 and D2 receptors in the anteroventral preop- 58. Berkenbosch A, DeGoede J, Ward DS, Olievier CN, Van-
tic region during normoxia and hypoxia. Brain Res 1030: 165–171, Hartevelt J. Dynamic response of the peripheral chemoreflex loop
2004. to changes in end-tidal O2. J Appl Physiol 71: 1123–1128, 1991.

Physiol Rev • VOL 90 • APRIL 2010 • www.prv.org


736 LUC J. TEPPEMA AND ALBERT DAHAN

59. Berkenbosch A, Heeringa J, Olievier CN, Kruyt EW. Artificial 81. Blanco CE, Hanson MA, Johnson P, Rigatto H. Breathing pat-
perfusion of the ponto-medullary region of cats. A method for tern of kittens during hypoxia. J Appl Physiol 56: 12–17, 1984.
separation of central and peripheral effects of chemical stimulation 82. Blanco CE, Hanson MA, McCooke HB. Effects on carotid che-
of ventilation. Respir Physiol 37: 347–364, 1979. moreceptor resetting of pulmonary ventilation in the fetal lamb in
60. Berkenbosch A, Olievier CN, DeGoede J, Kruyt EW. Effect on utero. J Dev Physiol 10: 167–174, 1988.
ventilation of papaverine administered to the brain stem of the 83. Blood AB, Hunter CJ, Power GG. The role of adenosine in
anaesthetized cat. J Physiol 443: 457– 468, 1991. regulation of cerebral blood flow during hypoxia in the near-term
61. Berkenbosch A, Teppema LJ, Olievier CN, Dahan A. Influences fetal sheep. J Physiol 543: 1015–1023, 2002.
of morphine on the ventilatory response to isocapnic hypoxia. 84. Blood AB, Hunter CJ, Power GG. Adenosine mediates decreased
Anesthesiology 86: 1342–1349, 1997. cerebral metabolic rate and increased cerebral blood flow during
62. Berkenbosch A, van Beek JH, Olievier CN, De Goede J, Quan- acute moderate hypoxia in the near-term fetal sheep. J Physiol 553:
jer PH. Central respiratory CO2 sensitivity at extreme hypocapnia. 935–945, 2003.
Respir Physiol 55: 95–102, 1984. 85. Bocking AD. Assessment of fetal heart rate and fetal movements
63. Berra E, Benizri E, Ginouves A, Volmat V, Roux D, Pouysse- in detecting oxygen deprivation in-utero. Eur J Obstet Gynecol
gur J. HIF prolyl-hydroxylase 2 is the key oxygen sensor setting Reprod Biol 110 Suppl 1: S108 –S112, 2003.
low steady-state levels of HIF-1alpha in normoxia. EMBO J 22: 86. Bocking AD, Gagnon R, Milne KM, White SE. Behavioral activ-
4082– 4090, 2003. ity during prolonged hypoxemia in fetal sheep. J Appl Physiol 65:
64. Bin-Jaliah I, Maskell PD, Kumar P. Indirect sensing of insulin- 2420 –2426, 1988.
induced hypoglycaemia by the carotid body in the rat. J Physiol 87. Bocking AD, Harding R. Effects of reduced uterine blood flow on
556: 255–266, 2004. electrocortical activity, breathing, and skeletal muscle activity in
65. Bisgard GE. Carotid body mechanisms in acclimatization to hyp- fetal sheep. Am J Obstet Gynecol 154: 655– 662, 1986.
oxia. Respir Physiol 121: 237–246, 2000. 88. Boddy K, Dawes GS, Fisher R, Pinter S, Robinson JS. Foetal
66. Bisgard GE, Busch MA, Daristotle L, Berssenbrugge AD, For- respiratory movements, electrocortical and cardiovascular re-
ster HV. Carotid body hypercapnia does not elicit ventilatory sponses to hypoxaemia and hypercapnia in sheep. J Physiol 243:
acclimatization in goats. Respir Physiol 65: 113–125, 1986. 599 – 618, 1974.
67. Bisgard GE, Busch MA, Forster HV. Ventilatory acclimatization 89. Bonora M, Gautier H. Maturational changes in body temperature
to hypoxia is not dependent on cerebral hypocapnic alkalosis. and ventilation during hypoxia in kittens. Respir Physiol 68: 359 –
J Appl Physiol 60: 1011–1015, 1986. 370, 1987.
68. Bisgard GE, Forster HV. Ventilatory responses to acute and 90. Bonora M, Marlot D, Gautier H, Duron B. Effects of hypoxia on
chronic hypoxia. In: Handbook of Physiology. Environmental ventilation during postnatal development in conscious kittens.
Physiology, edited by Frely MJ and Blatteis CM. New York: Oxford J Appl Physiol 56: 1464 –1471, 1984.
Univ. Press, 1996, p. 1207–1239. 91. Bonora M, Vizek M. Ventilation, EELV and diaphragmatic activity
in rats during the early phase of normobaric hypoxia. Respir
69. Bisgard GE, Forster HV, Klein JP. Recovery of peripheral che-
Physiol 128: 131–145, 2001.
moreceptor function after denervation in ponies. J Appl Physiol 49:
92. Boon JA, Milsom WK. NMDA receptor-mediated processes in the
964 –970, 1980.
Parabrachial/Kölliker fuse complex influence respiratory re-
70. Bisgard GE, Forster HV, Orr JA, Buss DD, Rawlings CA,
sponses directly and indirectly via changes in cortical activation
Rasmussen B. Hypoventilation in ponies after carotid body dener-
state. Respir Physiol Neurobiol 162: 63–72, 2008.
vation. J Appl Physiol 40: 184 –190, 1976.
93. Brady JP, Ceruti E. Chemoreceptor reflexes in the new-born
71. Bisgard GE, Olson EB Jr, Wang ZY, Bavis RW, Fuller DD,
infant: effects of varying degrees of hypoxia on heart rate and
Mitchell GS. Adult carotid chemoafferent responses to hypoxia
ventilation in a warm environment. J Physiol 184: 631– 645, 1966.
after 1, 2, and 4 wk of postnatal hyperoxia. J Appl Physiol 95: 94. Brady R, Zaidi SI, Mayer C, Katz DM. BDNF is a target-derived
946 –952, 2003. survival factor for arterial baroreceptor and chemoafferent primary
72. Bisgard GE, OlsonEB Jr, Bavis RW, Wenninger J, Nordheim sensory neurons. J Neurosci 19: 2131–2142, 1999.
EV, Mitchell GS. Carotid chemoafferent plasticity in adult rats 95. Breen S, Rees S, Walker D. Identification of brainstem neurons
following developmental hyperoxia. Respir Physiol Neurobiol 145: responding to hypoxia in fetal and newborn sheep. Brain Res 748:
3–11, 2005. 107–121, 1997.
73. Bissonnette JM. Mechanisms regulating hypoxic respiratory de- 96. Brutsaert TD. Population genetic aspects and phenotypic plastic-
pression during fetal and postnatal life. Am J Physiol Regul Integr ity of ventilatory responses in high altitude natives. Respir Physiol
Comp Physiol 278: R1391–R1400, 2000. Neurobiol 158: 151–160, 2007.
74. Bissonnette JM, Hohimer AR. Acute anemic hypoxemia pro- 97. Brutsaert TD, Parra EJ, Shriver MD, Gamboa A, Rivera-Ch M,
duces a transient depression in fetal respiratory activity. J Appl Leon-Velarde F. Ancestry explains the blunted ventilatory re-
Physiol 63: 1942–1946, 1987. sponse to sustained hypoxia and lower exercise ventilation of
75. Bissonnette JM, Knopp SJ. Separate respiratory phenotypes in Quechua altitude natives. Am J Physiol Regul Integr Comp Physiol
methyl-CpG-binding protein 2 Mecp2 deficient mice. Pediatr Res 289: R225–R234, 2005.
59: 513–518, 2006. 98. Buckler KJ. TASK-like potassium channels and oxygen sensing in
76. Bissonnette JM, Reddington M. Autoradiographic localization of the carotid body. Respir Physiol Neurobiol 157: 55– 64, 2007.
adenosine A1 receptors in brainstem of fetal sheep. Brain Res 61: 99. Buckler KJ, Vaughan-Jones RD. Effects of mitochondrial uncou-
111–115, 1991. plers on intracellular calcium, pH and membrane potential in rat
77. Blain GM, Smith CA, Henderson KS, Dempsey JA. Contribu- carotid body type I cells. J Physiol 513: 819 – 833, 1998.
tion of the carotid body chemoreceptors to eupneic ventilation in 100. Buerk DG, Osanai S, Mokashi A, Lahiri S. Dopamine, sensory
the intact, unanesthetized dog. J Appl Physiol 106: 1564 –1573, discharge, and stimulus interaction with CO2 and O2 in cat carotid
2009. body. J Appl Physiol 85: 1719 –1726, 1998.
78. Blanco CE, Chen V, Maertzdorf W, Bamford OS, Hanson M. 101. Bunn HF, Poyton RO. Oxygen sensing and molecular adaptation
Effect of hyperoxia PaO2 50 –90 mmHg on fetal breathing move- to hypoxia. Physiol Rev 76: 839 – 885, 1996.
ments in the unanaesthetized fetal sheep. J Dev Physiol 14: 235– 102. Bureau MA, Begin R. Postnatal maturation of the respiratory
241, 1990. response to O2 in awake newborn lambs. J Appl Physiol 52: 428 –
79. Blanco CE, Dawes GS, Hanson MA, McCooke HB. The re- 433, 1982.
sponse to hypoxia of arterial chemoreceptors in fetal sheep and 103. Bureau MA, Lamarche J, Foulon P, Dalle D. The ventilatory
new-born lambs. J Physiol 351: 25–37, 1984. response to hypoxia in the newborn lamb after carotid body de-
80. Blanco CE, Dawes GS, Walker DW. Effects of hypoxia on nervation. Respir Physiol 60: 109 –119, 1985.
polysynaptic hind-limb reflexes in new-born lambs before and after 104. Burton MD, Kazemi H. Neurotransmitters in central respiratory
carotid denervation. J Physiol 339: 467– 474, 1983. control. Respir Physiol 122: 111–121, 2000.

Physiol Rev • VOL 90 • APRIL 2010 • www.prv.org


THE VENTILATORY RESPONSE TO HYPOXIA IN MAMMALS 737

105. Busch MA, Bisgard GE, Forster HV. Ventilatory acclimatization body to chronic hypoxia. Am J Physiol Lung Cell Mol Physiol 282:
to hypoxia is not dependent on arterial hypoxemia. J Appl Physiol L1314 –L1323, 2002.
58: 1874 –1880, 1985. 129. Chen J, He L, Liu X, Dinger B, Stensaas L, Fidone S. Effect of
106. Caceres AI, Obeso A, Gonzalez C, Rocher A. Molecular identi- the endothelin receptor antagonist bosentan on chronic hypoxia-
fication and functional role of voltage-gated sodium channels in rat induced morphological and physiological changes in rat carotid
carotid body chemoreceptor cells. Regulation of expression by body. Am J Physiol Lung Cell Mol Physiol 292: L1257–L1262, 2007.
chronic hypoxia in vivo. J Neurochem 102: 231–245, 2007. 130. Cheng Z, Guo SZ, Lipton AJ, Gozal D. Domoic acid lesions in
107. Calbet JA. Chronic hypoxia increases blood pressure and nor- nucleus of the solitary tract: time-dependent recovery of hypoxic
adrenaline spillover in healthy humans. J Physiol 551: 379 –386, ventilatory response and peripheral afferent axonal plasticity.
2003. J Neurosci 22: 3215–3226, 2002.
108. Calder NA, Williams BA, Kumar P, Hanson MA. The respiratory 131. Cherniack NS, Dempsey J, Fencl V, Fitzgerald RS, Lourenco
response of healthy term infants to breath-by-breath alternations in RV, Rebuck AS, Rigg J, Severinghaus JW, Weil JW, Whitelaw
inspired oxygen at two postnatal ages. Pediatr Res 35: 321–324, WA, Zwilich CW. Workshop on assessment of respiratory control
1994. in humans. I. Methods of measurement of ventilatory responses to
109. Campanucci VA, Zhang M, Vollmer C, Nurse CA. Expression of hypoxia and hypercapnia. Am Rev Respir Dis 115: 177–181, 1977.
multiple P2X receptors by glossopharyngeal neurons projecting to 132. Cherniack NS, Edelman NH, Lahiri S. Hypoxia and hypercapnia
rat carotid body O2-chemoreceptors: role in nitric oxide-mediated as respiratory stimulants and depressants. Respir Physiol 11: 113–
efferent inhibition. J Neurosci 26: 9482–9493, 2006. 126, 1970.
110. Cao H, Kuo YR, Prabhakar NR. Absence of chemoreceptor inhi- 133. Chowdhury R, Hardy A, Schofield CJ. The human oxygen sens-
bition by alpha-2 adrenergic receptor agonist in cats exposed to ing machinery and its manipulation. Chem Soc Rev 37: 1308 –1319,
low PO2 (Abstract). FASEB J 5: A1118, 1991. 2008.
111. Cao KY, Sullivan CE, Berthon-Jones M, Zwillich CW. Breath- 134. Clar C, Dorrington KL, Robbins PA. Ventilatory effects of 8 h of
ing route and resistive loading: influence on ventilatory response to isocapnic hypoxia with and without beta-blockade in humans.
hypoxia in conscious dogs. J Appl Physiol 75: 1247–1255, 1993. J Appl Physiol 86: 1897–1904, 1999.
112. Cao KY, Zwillich CW, Berthon-Jones M, Sullivan CE. In- 135. Clarke JA, Daly MB, Marshall JM, Ead HW, Hennessy EM.
creased normoxic ventilation induced by repetitive hypoxia in Quantitative studies of the vasculature of the carotid body in the
conscious dogs. J Appl Physiol 73: 2083–2088, 1992. chronically hypoxic rat. Braz J Med Biol Res 33: 331–340, 2000.
113. Cao KY, Zwillich CW, Berthon-Jones M, Sullivan CE. Ventila- 136. Clement ID, Pandit JJ, Bascom DA, Dorrington KL, O’Connor
tory response to sustained eucapnic hypoxia in the adult conscious DF, Robbins PA. An assessment of central-peripheral ventilatory
dog. Respir Physiol 89: 65–73, 1992. chemoreflex interaction using acid and bicarbonate infusions in
114. Carpenter E, Bee D, Peers C. Ionic currents in carotid body type humans. J Physiol 485: 561–570, 1995.
I cells isolated from normoxic and chronically hypoxic adult rats. 137. Clewlow F, Dawes GS, Johnston BM, Walker DW. Changes in
Brain Res 811: 79 – 87, 1998. breathing, electrocortical and muscle activity in unanaesthetized
115. Carroll JL. Developmental plasticity in respiratory control. J Appl fetal lambs with age. J Physiol 341: 463– 476, 1983.
Physiol 94: 375–389, 2003. 138. Cohen G, Katz-Salamon M. Development of chemoreceptor re-
116. Carroll JL, Bamford OS, Fitzgerald RS. Postnatal maturation of sponses in infants. Respir Physiol Neurobiol 149: 233–242, 2005.
carotid chemoreceptor responses to O2 and CO2 in the cat. J Appl 139. Cohen G, Malcolm G, Henderson-Smart D. Ventilatory response
Physiol 75: 2383–2391, 1993. of the newborn infant to mild hypoxia. Pediatr Pulmonol 24:
117. Carroll JL, Bureau MA. Decline in peripheral chemoreceptor 163–172, 1997.
excitatory stimulation during acute hypoxia in the lamb. J Appl 140. Coles SK, Dick TE. Neurones in the ventrolateral pons are re-
Physiol 63: 795– 802, 1987. quired for post-hypoxic frequency decline in rats. J Physiol 497:
118. Carroll JL, Bureau MA. Peripheral chemoreceptor CO2 response 79 –94, 1996.
during hyperoxia in the 14-day-old awake lamb. Respir Physiol 73: 141. Conde SV, Obeso A, Gonzalez C. Low glucose effects on rat
339 –349, 1988. carotid body chemoreceptor cells’ secretory responses and action
119. Carroll JL, Canet E, Bureau MA. Dynamic ventilatory responses potential frequency in the carotid sinus nerve. J Physiol 585: 721–
to CO2 in the awake lamb: role of the carotid chemoreceptors. 730, 2007.
J Appl Physiol 71: 2198 –2205, 1991. 142. Conde SV, Obeso A, Rigual R, Monteiro EC, Gonzalez C.
120. Cartwright CR, Henson LC, Ward DS. Effects of alfentanil on Function of the rat carotid body chemoreceptors in ageing. J Neu-
the ventilatory response to sustained hypoxia. Anesthesiology 89: rochem 99: 711–723, 2006.
612– 619, 1998. 143. Cormack RS, Cunningham DJ, Gee JB. The effect of carbon
121. Cattarossi L, Haxhiu-Poskurica B, Haxhiu MA, Litmanovitz I, dioxide on the respiratory response to want of oxygen in man. Q J
Martin RJ, Carlo WA. Carotid bodies and ventilatory response to Exp Physiol Cogn Med Sci 42: 303–319, 1957.
hypoxia in aminophylline-treated piglets. Pediatr Pulmonol 20: 144. Corne S, Webster K, Younes M. Hypoxic respiratory response
94 –100, 1995. during acute stable hypocapnia. Am J Respir Crit Care Med 167:
122. Chandel NS, Budinger GR. The cellular basis for diverse re- 1193–1199, 2003.
sponses to oxygen. Free Radic Biol Med 42: 165–174, 2007. 145. Cragg PA, Drysdale DB. Interaction of hypoxia and hypercapnia
123. Chandel NS, Schumacker PT. Cellular oxygen sensing by mito- on ventilation, tidal volume and respiratory frequency in the an-
chondria: old questions, new insight. J Appl Physiol 88: 1880 –1889, aesthetized rat. J Physiol 341: 477– 493, 1983.
2000. 146. Cross AR, Henderson L, Jones OT, Delpiano MA, Hentschel J,
124. Chapman KR, Cherniack NS. Aging effects on the interaction of Acker H. Involvement of an NADPH oxidase as a PO2 sensor
hypercapnia and hypoxia as ventilatory stimuli. J Gerontol 42: protein in the rat carotid body. Biochem J 272: 743–747, 1990.
202–209, 1987. 147. Cross KW, Oppe TE. The effect of inhalation of high and low
125. Chau A, Koos BJ. Metabolic and cardiorespiratory responses to concentrations of oxygen on the respiration of the premature in-
hypoxia in fetal sheep: adenosine receptor blockade. Am J Physiol fant. J Physiol 117: 38 –55, 1952.
Regul Integr Comp Physiol 276: R1805–R1811, 1999. 148. Cruz JC, Reeves JT, Grover RF, Maher JT, McCullough RE,
126. Chavez JC, Agani F, Pichiule P, LaManna JC. Expression of Cymerman A, Denniston JC. Ventilatory acclimatization to high
hypoxia-inducible factor-1alpha in the brain of rats during chronic altitude is prevented by CO2 breathing. Respiration 39: 121–130,
hypoxia. J Appl Physiol 89: 1937–1942, 2000. 1980.
127. Chen J, He L, Dinger B, Stensaas L, Fidone S. Chronic hypoxia 149. Cummins EP, Taylor CT. Hypoxia-responsive transcription fac-
upregulates connexin43 expression in rat carotid body and petrosal tors. Pflügers Arch 450: 363–371, 2005.
ganglion. J Appl Physiol 92: 1480 –1486, 2002. 150. Cunningham DJC, Robbins PA, Wolff CB. Integration of respi-
128. Chen J, He L, Dinger B, Stensaas L, Fidone S. Role of endo- ratory response to changes in alveolar partial pressures in CO2 and
thelin and endothelin A-type receptor in adaptation of the carotid O2 and in arterial pH. In: Handbook of Physiology. The Respiratory

Physiol Rev • VOL 90 • APRIL 2010 • www.prv.org


738 LUC J. TEPPEMA AND ALBERT DAHAN

System. Control of Breathing. Bethesda, MD: Am. Physiol. Soc., 172. Day TA, Wilson RJ. Brainstem PCO2 modulates phrenic responses
1986, sect. 3, vol. II, pt. 2, chapt. 15, p. 475–528. to specific carotid body hypoxia in an in situ dual perfused rat
151. Curran AK, Rodman JR, Eastwood PR, Henderson KS, Demp- preparation. J Physiol 578: 843– 857, 2007.
sey JA, Smith CA. Ventilatory responses to specific CNS hypoxia 173. De Paula PM, Branco LG. Nitric oxide in the rostral ventrolateral
in sleeping dogs. J Appl Physiol 88: 1840 –1852, 2000. medulla modulates hyperpnea but not anapyrexia induced by hyp-
152. Curran LS, Zhuang J, Sun SF, Moore LG. Ventilation and oxia. Brain Res 977: 231–238, 2003.
hypoxic ventilatory responsiveness in Chinese-Tibetan residents at 174. De Paula PM, Tolstykh G, Mifflin S. Chronic intermittent hyp-
3,658 m. J Appl Physiol 83: 2098 –2104, 1997. oxia alters NMDA and AMPA-evoked currents in NTS neurons
153. D’Agostino D, Mazza E Jr, Neubauer JA. Heme oxygenase is receiving carotid body chemoreceptor inputs. Am J Physiol Regul
necessary for the excitatory response of cultured neonatal rat Integr Comp Physiol 292: R2259 –R2265, 2007.
rostral ventrolateral medulla neurons to hypoxia. Am J Physiol 175. DeGoede J, Berkenbosch A, Ward DS, Bellville JW, Olievier
Regul Integr Comp Physiol 296: R102–R118, 2009. CN. Comparison of chemoreflex gains obtained with two different
154. Dahan A, Nieuwenhuijs D, Teppema L. Plasticity of central methods in cats. J Appl Physiol 59: 170 –179, 1985.
chemoreceptors: effect of bilateral carotid body resection on cen- 176. DeGoede J, VanderHoeven N, Berkenbosch A, Olievier CN,
tral CO2 sensitivity. PLoS Medicine 4: e239, 2007. and van Beek JHGM. Ventilatory responses to sudden isocapnic
155. Dahan A, Berkenbosch A, DeGoede J, Olievier IC, Bovill JG. changes in end-tidal O2 in cats. In: Modelling and Control of
On a pseudo-rebreathing technique to assess the ventilatory sensi- Breathing, edited by Whipp BJ and Wiberg DM. New York: Elsevier
tivity to carbon dioxide in man. J Physiol 423: 615– 629, 1990. Biomedical, 1983, p. 37– 45.
156. Dahan A, DeGoede J, Berkenbosch A, Olievier IC. The influ- 177. Dejours P, Labrousse Y, Raynoud J, Teillac A. Oxygen che-
ence of oxygen on the ventilatory response to carbon dioxide in moreflex stimulus in ventilation at low altitude in man. I. At rest.
man. J Physiol 428: 485– 499, 1990. J Physiol 49: 115–120, 1957.
157. Dahan A, Sarton E, Teppema L, Olievier C. Sex-related differ- 178. Dempsey JA, Forster HV. Mediation of ventilatory adaptations.
ences in the influence of morphine on ventilatory control in hu- Physiol Rev 62: 262–346, 1982.
mans. Anesthesiology 88: 903–913, 1998. 179. DiGiulio C, Cacchio M, Bianchi G, Rapino C, Di IlioC. Selected
158. Dahan A, Sarton E, Teppema L, Olievier C, Nieuwenhuijs D, contribution: carotid body as a model for aging studies: is there a
Matthes HW, Kieffer BL. Anesthetic potency and influence of link between oxygen and aging? J Appl Physiol 95: 1755–1758,
morphine and sevoflurane on respiration in mu-opioid receptor 2003.
knockout mice. Anesthesiology 94: 824 – 832, 2001. 180. DiGiulio C, Huang W, Mokashi A, Lahiri S. Further character-
159. Dahan A, Sarton E, van den Elsen M, van Kleef J, Teppema L, ization of stimulus interaction of cat carotid chemoreceptors. J
Berkenbosch A. Ventilatory response to hypoxia in humans. In- Auton Nerv Syst 71: 196 –200, 1998.
fluences of subanesthetic desflurane. Anesthesiology 85: 60 – 68, 181. DiGiulio C, Huang WX, Mokashi A, Roy A, Cacchio M, Macri
1996. MA, Lahiri S. Sustained hypoxia promotes hyperactive response
160. Dahan A, van den Elsen MJ, Berkenbosch A, DeGoede J, of carotid body in the cat. Respir Physiol Neurobiol 134: 69 –74,
Olievier IC, van Kleef JW, Bovill JG. Effects of subanesthetic 2003.
halothane on the ventilatory responses to hypercapnia and acute 182. Dillon GH, Waldrop TG. In vitro responses of caudal hypotha-
hypoxia in healthy volunteers. Anesthesiology 80: 727–738, 1994. lamic neurons to hypoxia and hypercapnia. Neuroscience 51: 941–
161. Dahan A, van den Elsen MJLJ, de Goede J, Olievier ICW, 950, 1992.
Burm AGL, van Kleef JW. Influence of subanesthetic concentra- 183. Dillon GH, Waldrop TG. Responses of feline caudal hypothalamic
tion of halothane on the ventilatory response to step changes into cardiorespiratory neurons to hypoxia and hypercapnia. Exp Brain
and out of sustained isocapnic hypoxia in healthy-volunteers. An- Res 96: 260 –272, 1993.
esthesiology 81: 850 – 859, 1994. 184. Dinger B, He L, Chen J, Liu X, Gonzalez C, Obeso A, Sanders
162. Dahan A, Ward D, van den Elsen M, Temp J, Berkenbosch A. K, Hoidal J, Stensaas L, Fidone S. The role of NADPH oxidase
Influence of reduced carotid body drive during sustained hypoxia in carotid body arterial chemoreceptors. Respir Physiol Neurobiol
on hypoxic depression of ventilation in humans. J Appl Physiol 81: 157: 45–54, 2007.
565–572, 1996. 185. Donnelly DF. Chemoreceptor nerve excitation may not be pro-
163. Daristotle L, Berssenbrugge AD, Bisgard GE. Hypoxic-hyper- portional to catecholamine secretion. J Appl Physiol 81: 657– 664,
capnic ventilatory interaction at the carotid body of awake goats. 1996.
Respir Physiol 70: 63–72, 1987. 186. Donnelly DF. Development of carotid body/petrosal ganglion re-
164. Daristotle L, Bisgard GE. Central-peripheral chemoreceptor ven- sponse to hypoxia. Respir Physiol Neurobiol 149: 191–199, 2005.
tilatory interaction in awake goats. Respir Physiol 76: 383–391, 187. Donnelly DF, Bavis RW, Kim I, Dbouk HA, Carroll JL. Time
1989. course of alterations in pre- and post-synaptic chemoreceptor func-
165. Daristotle L, Engwall MJ, Niu WZ, Bisgard GE. Ventilatory tion during developmental hyperoxia. Respir Physiol Neurobiol
effects and interactions with change in PaO2 in awake goats. J Appl 168: 189 –197, 2009.
Physiol 71: 1254 –1260, 1991. 188. Donnelly DF, Doyle TP. Developmental changes in hypoxia-in-
166. Darnall RA, Green G, Pinto L, Hart N. Effect of acute hypoxia duced catecholamine release from rat carotid body, in vitro.
on respiration and brain stem blood flow in the piglet. J Appl J Physiol 475: 267–275, 1994.
Physiol 70: 251–259, 1991. 189. Donnelly DF, Kim I, Carle C, Carroll JL. Perinatal hyperoxia for
167. Dasso LL, Buckler KJ, Vaughan-Jones RD. Interactions be- 14 days increases nerve conduction time and the acute unitary
tween hypoxia and hypercapnic acidosis on calcium signaling in response to hypoxia of rat carotid body chemoreceptors. J Appl
carotid body type I cells. Am J Physiol Lung Cell Mol Physiol 279: Physiol 99: 114 –119, 2005.
L36 –L42, 2000. 190. Donnelly DF, Smith E, Dutton RE. Carbon dioxide versus H ion
168. Daudu PA, Roy A, Rozanov C, Mokashi A, Lahiri S. Extra- and as a chemoreceptor stimulus. Brain Res 245: 136 –138, 1982.
intracellular free iron and the carotid body responses. Respir 191. Donnelly DF. Developmental aspects of oxygen sensing by the
Physiol Neurobiol 130: 21–31, 2002. carotid body. J Appl Physiol 88: 2296 –2301, 2000.
169. Dauger S, Pattyn A, Lofaso F, Gaultier C, Goridis C, Gallego 192. Donoghue S, Fatemian M, Balanos GM, Crosby A, Liu C,
J, Brunet JF. Phox2b controls the development of peripheral O’Connor D, Talbot NP, Robbins PA. Ventilatory acclimatization
chemoreceptors and afferent visceral pathways. Development 130: in response to very small changes in PO2 in humans. J Appl Physiol
6635– 6642, 2003. 98: 1587–1591, 2005.
170. Dawes GS. The central control of fetal breathing and skeletal 193. Drorbaugh JE, Fenn WO. A barometric method for measuring
muscle movements. J Physiol 346: 1–18, 1984. ventilation in newborn infants. Pediatrics 16: 81– 87, 1955.
171. Dawes GS, Gardner WN, Johnston BM, Walker DW. Breathing 194. Duchen MR, Biscoe TJ. Relative mitochondrial membrane poten-
in fetal lambs: the effect of brain stem section. J Physiol 335: tial and [Ca2⫹]i in type I cells isolated from the rabbit carotid body.
535–553, 1983. J Physiol 450: 33– 61, 1992.

Physiol Rev • VOL 90 • APRIL 2010 • www.prv.org


THE VENTILATORY RESPONSE TO HYPOXIA IN MAMMALS 739

195. Duffin J. Measuring the ventilatory response to hypoxia. J Physiol 218. Enhorning G, van Schaik S, Lundgren C, Vargas I. Whole-body
584: 285–293, 2007. plethysmography, does it measure tidal volume of small animals?
196. Duprat F, Lauritzen I, Patel A, Honore E. The TASK back- Can J Physiol Pharmacol 76: 945–951, 1998.
ground K2P channels: chemo- and nutrient sensors. Trends Neu- 219. Epstein RA, Epstein MA, Haddad GG, Mellins RB. Practical
rosci 30: 573–580, 2007. implementation of the barometric method for measurement of tidal
197. Durand E, Lofaso F, Dauger S, Vardon G, Gaultier C, Gallego volume. J Appl Physiol 49: 1107–1115, 1980.
J. Intermittent hypoxia induces transient arousal delay in newborn 220. Erickson JT, Mayer C, Jawa A, Ling L, Olson EB Jr, Vidruk
mice. J Appl Physiol 96: 1216 –1222, 2004. EH, Mitchell GS, Katz DM. Chemoafferent degeneration and
198. Dutton RE, Smith EJ, Ghatak PK, Davies DG. Dynamics of the carotid body hypoplasia following chronic hyperoxia in newborn
respiratory controller during carotid body hypoxia. J Appl Physiol rats. J Physiol 509: 519 –526, 1998.
35: 844 – 850, 1973. 221. Erickson JT, Millhorn DE. Hypoxia and electrical stimulation of
199. Dwinell MR, Forster HV, Petersen J, Rider A, Kunert MP, the carotid sinus nerve induce Fos-like immunoreactivity within
Cowley AW Jr, Jacob HJ. Genetic determinants on rat chromo- catecholaminergic and serotoninergic neurons of the rat brainstem.
some 6 modulate variation in the hypercapnic ventilatory response J Comp Neurol 348: 161–182, 1994.
using consomic strains. J Appl Physiol 98: 1630 –1638, 2005. 222. Evans AM, Hardie DG, Peers C, Wyatt CN, Viollet B, Kumar P,
200. Dwinell MR, Janssen PL, Pizarro J, Bisgard GE. Effects of Dallas ML, Ross F, Ikematsu N, Jordan HL, Barr BL, Rafferty
JN, Ogunbayo O. Ion channel regulation by AMPK: the route of
carotid body hypocapnia during ventilatory acclimatization to hyp-
hypoxia-response coupling in thecarotid body and pulmonary ar-
oxia. J Appl Physiol 82: 118 –124, 1997.
tery. Ann NY Acad Sci 1177: 89 –100, 2009.
201. Dwinell MR, Powell FL. Chronic hypoxia enhances the phrenic
223. Evans AM, Mustard KJ, Wyatt CN, Peers C, Dipp M, Kumar P,
nerve response to arterial chemoreceptor stimulation in anesthe-
Kinnear NP, Hardie DG. Does AMP-activated protein kinase cou-
tized rats. J Appl Physiol 87: 817– 823, 1999. ple inhibition of mitochondrial oxidative phosphorylation by hyp-
202. Dyugovskaya L, Lavie P, Lavie L. Increased adhesion molecules oxia to calcium signaling in O2-sensing cells? J Biol Chem 280:
expression and production of reactive oxygen species in leuko- 41504 – 41511, 2005.
cytes of sleep apnea patients. Am J Respir Crit Care Med 165: 224. Fabris G, Anselmo-Franci JA, Branco LG. Role of nitric oxide in
934 –939, 2002. hypoxia-induced hyperventilation and hypothermia: participation
203. Easton PA, Anthonisen NR. Ventilatory response to sustained of the locus coeruleus. Braz J Med Biol Res 32: 1389 –1398, 1999.
hypoxia after pretreatment with aminophylline. J Appl Physiol 64: 225. Fabris G, Steiner AA, Anselmo-Franci JA, Branco LG. Role of
1445–1450, 1988. nitric oxide in rat locus coeruleus in hypoxia-induced hyperventi-
204. Easton PA, Slykerman LJ, Anthonisen NR. Ventilatory re- lation and hypothermia. Neuroreport 11: 2991–2995, 2000.
sponse to sustained hypoxia in normal adults. J Appl Physiol 61: 226. Fatemian M, Gamboa A, Leon-Velarde F, Rivera-Ch M, Pala-
906 –911, 1986. cios JA, Robbins PA. Selected contribution: ventilatory response
205. Easton PA, Slykerman LJ, Anthonisen NR. Recovery of the to CO2 in high-altitude natives and patients with chronic mountain
ventilatory response to hypoxia in normal adults. J Appl Physiol sickness. J Appl Physiol 94: 1279 –1287, 2003.
64: 521–528, 1988. 227. Fatemian M, Nieuwenhuijs DJ, Teppema LJ, Meinesz S, van
206. Edelman NH, Epstein PE, Lahiri S, Cherniack NS. Ventilatory der Mey AG, Dahan A, Robbins PA. The respiratory response to
responses to transient hypoxia and hypercapnia in man. Respir carbon dioxide in humans with unilateral and bilateral resections
Physiol 17: 302–314, 1973. of the carotid bodies. J Physiol 549: 965–973, 2003.
207. Eden GJ, Hanson MA. Effects of chronic hypoxia from birth on 228. Fatemian M, Robbins PA. Human ventilatory response to CO2
the ventilatory response to acute hypoxia in the newborn rat. after 8 h of isocapnic or poikilocapnic hypoxia. J Appl Physiol 85:
J Physiol 392: 11–19, 1987. 1922–1928, 1998.
208. Eden GJ, Hanson MA. Maturation of the respiratory response to 229. Fatemian M, Robbins PA. Selected contribution: chemoreflex
acute hypoxia in the newborn rat. J Physiol 392: 1–9, 1987. responses to CO2 before and after an 8-h exposure to hypoxia in
209. Edwards C, Heath D, Harris P, Castillo Y, Kruger H, Arias- humans. J Appl Physiol 90: 1607–1614, 2001.
Stella J. The carotid body in animals at high altitude. J Pathol 104: 230. Feldman JL, Mitchell GS, Nattie EE. Breathing: rhythmicity
231–238, 1971. plasticity chemosensitivity. Annu Rev Neurosci 26: 239 –266, 2003.
210. El Hasnaoui-Saadani R, Alayza RC, Launay T, Pichon A, 231. Filuk RB, Berezanski DJ, Anthonisen NR. Depression of
Quidu P, Beaudry M, Leon-Velarde F, Richalet JP, Duvallet A, hypoxic ventilatory response in humans by somatostatin. J Appl
Favret F. Brain stem NO modulates ventilatory acclimatization to Physiol 65: 1050 –1054, 1988.
hypoxia in mice. J Appl Physiol 103: 1506 –1512, 2007. 232. Finley JC, Katz DM. The central organization of carotid body
211. Eldridge FL, Millhorn DE. Oscillation, gating and memory in the afferent projections to the brainstem of the rat. Brain Res 572:
respiratory control system. In: Handbook of Physiology. The Re- 108 –116, 1992.
233. Fitzgerald RS, Lahiri S. Reflex responses to chemoreceptors
spiratory System. Control of Breathing. Bethesda, MD: Am.
stimulation. In: Handbook of Physiology. The Respiratory System.
Physiol. Soc., 1986, sect. 3, vol. II, pt. 1, chapt. 3, p. 93–114.
Control of Breathing. Bethesda, MD: Am. Physiol. Soc., 1986, sect.
212. Elnazir B, Marshall JM, Kumar P. Postnatal development of the
3, vol. II, pt. 1, chapt. 10, p. 313–362.
pattern of respiratory and cardiovascular response to systemic
234. Fitzgerald RS, Parks DC. Effect of hypoxia on carotid chemore-
hypoxia in the piglet: the roles of adenosine. J Physiol 492: 573– ceptor response to carbon dioxide in cats. Respir Physiol 12:
585, 1996. 218 –229, 1971.
213. Eng C, Kiuru M, Fernandez MJ, Aaltonen LA. A role for mito- 235. Foo IT, Martin SE, Lee RJ, Drummond GB, Warren PM. Inter-
chondrial enzymes in inherited neoplasia and beyond. Nat Rev action of the effects of domperidone and sub-anaesthetic concen-
Cancer 3: 193–202, 2003. trations of isoflurane on the immediate and sustained hypoxic
214. Engeman RM, Sherrill DL, Swanson GD. On the estimation of ventilatory response in humans. Br J Anaesth 74: 134 –140, 1995.
hypoxic ventilatory response. Comput Biomed Res 18: 553–562, 236. Foo IT, Warren PM, Drummond GB. Influence of oral clonidine
1985. on the ventilatory response to acute and sustained isocapnic hyp-
215. Engwall MJ, Bisgard GE. Ventilatory responses to chemorecep- oxia in human males. Br J Anaesth 76: 214 –220, 1996.
tor stimulation after hypoxic acclimatization in awake goats. J Appl 237. Fordyce WE. Hypoxic ventilatory control in the awake cat five
Physiol 69: 1236 –1243, 1990. years after carotid body resection. Respir Physiol 69: 209 –225,
216. Engwall MJ, Vidruk EH, Nielsen AM, Bisgard GE. Response of 1987.
the goat carotid body to acute and prolonged hypercapnia. Respir 238. Forster HV. Plasticity in the control of breathing following sen-
Physiol 74: 335–344, 1988. sory denervation. J Appl Physiol 94: 784 –794, 2003.
217. Enhorning G. Inhibition of fetal breathing: a pilot study. Pediatr 239. Forster HV, Bisgard GE, Rasmussen B, Orr JA, Buss DD,
Res 34: 834 – 836, 1993. Manohar M. Ventilatory control in peripheral chemoreceptor-den-

Physiol Rev • VOL 90 • APRIL 2010 • www.prv.org


740 LUC J. TEPPEMA AND ALBERT DAHAN

ervated ponies during chronic hypoxemia. J Appl Physiol 41: 878 – hypoxic ventilatory response in awake rat. J Physiol 554: 543–557,
885, 1976. 2004.
240. Forster HV, Dwinell MR, Hodges MR, Brozoski D, Hogan GE. 263. Georgopoulos D, Berezanski D, Anthonisen NR. Effects of CO2
Do genes on rat chromosomes 9, 13, 16, 18, and 20 contribute to breathing on ventilatory response to sustained hypoxia in normal
regulation of breathing? Respir Physiol Neurobiol 135: 247–261, adults. J Appl Physiol 66: 1071–1078, 1989.
2003. 264. Georgopoulos D, Berezanski D, Anthonisen NR. Effect of di-
241. Foster GE, McKenzie DC, Milsom WK, Sheel AW. Effects of chloroacetate on ventilatory response to sustained hypoxia in nor-
two protocols of intermittent hypoxia on human ventilatory, car- mal adults. Respir Physiol 82: 115–122, 1990.
diovascular and cerebral responses to hypoxia. J Physiol 567: 265. Georgopoulos D, Holtby SG, Berezanski D, Anthonisen NR.
689 – 699, 2005. Aminophylline effects on ventilatory response to hypoxia and hy-
242. Foster GE, Poulin MJ, Hanly PJ. Intermittent hypoxia and vas- peroxia in normal adults. J Appl Physiol 67: 1150 –1156, 1989.
cular function: implications for obstructive sleep apnoea. Exp 266. Georgopoulos D, Walker S, Anthonisen NR. Increased chemo-
Physiol 92: 51– 65, 2007. receptor output and ventilatory response to sustained hypoxia.
243. Frappell P, Lanthier C, Baudinette RV, Mortola JP. Metabo- J Appl Physiol 67: 1157–1163, 1989.
lism and ventilation in acute hypoxia: a comparative analysis in 267. Georgopoulos D, Walker S, Anthonisen NR. Effect of sustained
small mammalian species. Am J Physiol Regul Integr Comp hypoxia on ventilatory response to CO2 in normal adults. J Appl
Physiol 262: R1040 –R1046, 1992. Physiol 68: 891– 896, 1990.
244. Fregosi RF, Mitchell GS. Long-term facilitation of inspiratory 268. Gilmartin G, Tamisier R, Anand A, Cunnington D, Weiss JW.
intercostal nerve activity following carotid sinus nerve stimulation Evidence of impaired hypoxic vasodilation after intermediate-du-
in cats. J Physiol 477: 469 – 479, 1994. ration hypoxic exposure in humans. Am J Physiol Heart Circ
245. Fuller DD, Baker TL, Behan M, Mitchell GS. Expression of Physiol 291: H2173–H2180, 2006.
hypoglossal long-term facilitation differs between substrains of 269. Gilmartin GS, Tamisier R, Curley M, Weiss JW. Ventilatory,
Sprague-Dawley rat. Physiol Genomics 4: 175–181, 2001. hemodynamic, sympathetic nervous system, and vascular reactiv-
246. Fuller DD, Bavis RW, Vidruk EH, Wang ZY, Olson EB Jr, ity changes after recurrent nocturnal sustained hypoxia in humans.
Bisgard GE, Mitchell GS. Life-long impairment of hypoxic Am J Physiol Heart Circ Physiol 295: H778 –H785, 2008.
phrenic responses in rats following 1 month of developmental 270. Giussani DA, Moore PJ, Bennet L, Spencer JA, Hanson MA.
hyperoxia. J Physiol 538: 947–955, 2002. Alpha 1- and alpha 2-adrenoreceptor actions of phentolamine and
247. Fuller DD, Wang ZY, Ling L, Olson EB, Bisgard GE, Mitchell prazosin on breathing movements in fetal sheep in utero. J Physiol
GS. Induced recovery of hypoxic phrenic responses in adult rats 486: 249 –255, 1995.
exposed to hyperoxia for the first month of life. J Physiol 536: 271. Gluckman PD, Johnston BM. Lesions in the upper lateral pons
917–926, 2001. abolish the hypoxic depression of breathing in unanaesthetized
248. Fung ML, Dong X. Absence of MK801-induced inspiratory prolon- fetal lambs in utero. J Physiol 382: 373–383, 1987.
272. Godfrey S, Edwards RHT, Copland GM, Gross PL. Chemosen-
gation in chronically hypoxic rats. Life Sci 69: 2319 –2326, 2001.
sitivity in normal subjects, athletes, and patients with chronic
249. Fung ML, Wang W, Darnall RA, St John WM. Characterization
airways obstruction. J Appl Physiol 30: 193–199, 1971.
of ventilatory responses to hypoxia in neonatal rats. Respir Physiol
273. Goldberg SV, Schoene RB, Haynor D, Trimble B, Swenson ER,
103: 57– 66, 1996.
Morrison JB, Banister EJ. Brain tissue pH and ventilatory accli-
250. Gabel RA, Weiskopf RB. Ventilatory interaction between hypoxia
matization to high altitude. J Appl Physiol 72: 58 – 63, 1992.
and [H⫹] at chemoreceptors of man. J Appl Physiol 39: 292–296,
274. Golder FJ, Zabka AG, Bavis RW, Baker-Herman T, Fuller DD,
1975.
Mitchell GS. Differences in time-dependent hypoxic phrenic re-
251. Gagnon R, Hunse C, Vijan S. The effect of maternal hyperoxia on
sponses among inbred rat strains. J Appl Physiol 98: 838 – 844,
behavioral activity in growth-retarded human fetuses. Am J Obstet
2005.
Gynecol 163: 1894 –1899, 1990. 275. Gomez-Nino A, Agapito MT, Obeso A, Gonzalez C. Effects of
252. Gamboa J, Macarlupu JL, Rivera-Chira M, Monge C, Leon- mitochondrial poisons on glutathione redox potential and carotid
Velarde F. Effect of domperidone on ventilation and polycythemia body chemoreceptor activity. Respir Physiol Neurobiol 165: 104 –
after 5 weeks of chronic hypoxia in rats. Respir Physiol Neurobiol 111, 2009.
135: 1– 8, 2003. 276. Gonzalez C, Agapito MT, Rocher A, Gonzalez-Martin MC,
253. Garcia N, Hopkins SR, Powell FL. Effects of intermittent hyp- Vega-Agapito V, Gomez-Nino A, Rigual R, Castaneda J, Obeso
oxia on the isocapnic hypoxic ventilatory response and erythropoi- A. Chemoreception in the context of the general biology of ROS.
esis in humans. Respir Physiol 123: 39 – 49, 2000. Respir Physiol Neurobiol 157: 30 – 44, 2007.
254. Garcia-Fernandez M, Ortega-Saenz P, Castellano A, Lopez- 277. Gonzalez C, Almaraz L, Obeso A, Rigual R. Oxygen and acid
Barneo J. Mechanisms of low-glucose sensitivity in carotid body chemoreception in the carotid body chemoreceptors. Trends Neu-
glomus cells. Diabetes 56: 2893–2900, 2007. rosci 15: 146 –153, 1992.
255. Gargaglioni LH, Bicego KC, Branco LG. Brain monoaminergic 278. Gonzalez C, Almaraz L, Obeso A, Rigual R. Carotid body che-
neurons and ventilatory control in vertebrates. Respir Physiol moreceptors: from natural stimuli to sensory discharges. Physiol
Neurobiol 164: 112–122, 2008. Rev 74: 829 – 898, 1994.
256. Gargaglioni LH, Coimbra NC, Branco LG. The nucleus raphe 279. Gonzalez C, Lopez-Lopez JR, Obeso A, Perez-Garcia MT,
magnus modulates hypoxia-induced hyperventilation but not ana- Rocher A. Cellular mechanisms of oxygen chemoreception in the
pyrexia in rats. Neurosci Lett 347: 121–125, 2003. carotid body. Respir Physiol 102: 137–147, 1995.
257. Garthwaite J. Glutamate, nitric oxide and cell-cell signalling in 280. Gonzalez C, Rocher A, Zapata P. Arterial chemoreceptors: cel-
the nervous system. Trends Neurosci 14: 60 – 67, 1991. lular and molecular mechanisms in the adaptative and homeostatic
258. Gauda EB, Lawson EE. Developmental influences on carotid function of the carotid body. Rev Neurol 36: 239 –254, 2003.
body responses to hypoxia. Respir Physiol 121: 199 –208, 2000. 281. Gonzalez C, Sanz-Alyayate G, Agapito MT, Obeso A. Effects of
259. Gaultier C, Dauger S, Simonneau M, Gallego J. Genes modu- reducing agents on glutathione metabolism and the function of
lating chemical breathing control: lessons from mutant animals. carotid body chemoreceptor cells. Biol Chem 385: 265–274, 2004.
Respir Physiol Neurobiol 136: 105–114, 2003. 282. Gonzalez C, Sanz-Alfayate G, Agapito MT, Gomez-Nino A,
260. Gautier H. Interactions among metabolic rate, hypoxia, and con- Rocher A, Obeso A. Significance of ROS in oxygen sensing in cell
trol of breathing. J Appl Physiol 81: 521–527, 1996. systems with sensitivity to physiological hypoxia. Respir Physiol
261. Gautier H, Bonora M. Possible alterations in brain monoamine Neurobiol 132: 17– 41, 2002.
metabolism during hypoxia-induced tachypnea in cats. J Appl 283. Gonzalez-Guerrero PR, Rigual R, Gonzalez C. Effects of
Physiol 49: 769 –777, 1980. chronic hypoxia on opioid peptide and catecholamine levels and on
262. Genest SE, Gulemetova R, Laforest S, Drolet G, Kinkead R. the release of dopamine in the rabbit carotid body. J Neurochem
Neonatal maternal separation and sex-specific plasticity of the 60: 1769 –1776, 1993.

Physiol Rev • VOL 90 • APRIL 2010 • www.prv.org


THE VENTILATORY RESPONSE TO HYPOXIA IN MAMMALS 741

284. Gourine AV, Llaudet E, Dale N, Spyer KM. Release of ATP in 307. He L, Chen J, Dinger B, Stensaas L, Fidone S. Effect of chronic
the ventral medulla during hypoxia in rats: role in hypoxic venti- hypoxia on purinergic synaptic transmission in rat carotid body.
latory response. J Neurosci 25: 1211–1218, 2005. J Appl Physiol 100: 157–162, 2006.
285. Gozal D, Gaultier C. Evolving concepts of the maturation of 308. He L, Chen J, Liu X, Dinger B, Fidone S. Enhanced nitric
central pathways underlying the hypoxic ventilatory response. oxide-mediated chemoreceptor inhibition and altered cyclic GMP
Am J Respir Crit Care Med 164: 325–329, 2001. signaling in rat carotid body following chronic hypoxia. Am J
286. Gozal D, Gozal E. Episodic hypoxia enhances late hypoxic ven- Physiol Lung Cell Mol Physiol 293: L1463–L1468, 2007.
tilation in developing rat: putative role of neuronal NO synthase. 309. He L, Dinger B, Fidone S. Effect of chronic hypoxia on cholin-
Am J Physiol Regul Integr Comp Physiol 276: R17–R22, 1999. ergic chemotransmission in rat carotid body. J Appl Physiol 98:
287. Gozal D, Gozal E, Simakajornboon N. Signaling pathways of the 614 – 619, 2005.
acute hypoxic ventilatory response in the nucleus tractus soli- 310. He L, Dinger B, Sanders K, Hoidal J, Obeso A, Stensaas L,
tarius. Respir Physiol 121: 209 –221, 2000. Fidone S, Gonzalez C. Effect of p47phox gene deletion on ROS
288. Gozal D, Gozal E, Torres JE, Gozal YM, Nuckton TJ, Hornby production and oxygen sensing in mouse carotid body chemore-
PJ. Nitric oxide modulates ventilatory responses to hypoxia in the ceptor cells. Am J Physiol Lung Cell Mol Physiol 289: L916 –L924,
developing rat. Am J Respir Crit Care Med 155: 1755–1762, 1997. 2005.
289. Gozal D, Simakajornboon N, Czapla MA, Xue YD, Gozal E, 311. Heath D. The human carotid body in health and disease. J Pathol
Vlasic V, Lasky JA, Liu JY. Brainstem activation of platelet-
164: 1– 8, 1991.
derived growth factor-beta receptor modulates the late phase of the
312. Heath D, Smith P, Fitch R, Harris P. Comparative pathology of
hypoxic ventilatory response. J Neurochem 74: 310 –319, 2000.
the enlarged carotid body. J Comp Pathol 95: 259 –271, 1985.
290. Gozal D, Torres JE, Gozal YM, Littwin SM. Effect of nitric
313. Hebbel RP, Kronenberg RS, Eaton JW. Hypoxic ventilatory
oxide synthase inhibition on cardiorespiratory responses in the
conscious rat. J Appl Physiol 81: 2068 –2077, 1996. response in subjects with normal and high oxygen affinity hemo-
291. Granjeiro EM, Machado BH. NO in the caudal NTS modulates globins. J Clin Invest 60: 1211–1215, 1977.
the increase in respiratory frequency in response to chemoreflex 314. Heeringa J, Berkenbosch A, De Goede J, Olievier CN. Relative
activation in awake rats. Respir Physiol Neurobiol 166: 32– 40, contribution of central and peripheral chemoreceptors to the ven-
2009. tilatory response to CO2 during hyperoxia. Respir Physiol 37:
292. Gray BA. Response of the perfused carotid body to changes in pH 365–379, 1979.
and PCO2. Respir Physiol 4: 229 –245, 1968. 315. Hempleman SC. Sodium and potassium current in neonatal rat
293. Green GA, Darnall RA, Bierd TB, Adams JM. Effect of aortic carotid body cells following chronic in vivo hypoxia. Brain Res
balloon inflation on ventilation and brain stem blood flow in piglets. 699: 42–50, 1995.
J Appl Physiol 66: 2174 –2180, 1989. 316. Hempleman SC. Increased calcium current in carotid body glo-
294. Greenberg HE, Sica A, Batson D, Scharf SM. Chronic intermit- mus cells following in vivo acclimatization to chronic hypoxia.
tent hypoxia increases sympathetic responsiveness to hypoxia and J Neurophysiol 76: 1880 –1886, 1996.
hypercapnia. J Appl Physiol 86: 298 –305, 1999. 317. Henderson-Smart DJ, Cohen GL. Chemical control of breathing
295. Guzy RD, Schumacker PT. Oxygen sensing by mitochondria at in early life. Ann NY Acad Sci 533: 276 –288, 1988.
complex III: the paradox of increased reactive oxygen species 318. Henderson-Smart DJ, Pettigrew AG, Campbell DJ. Clinical
during hypoxia. Exp Physiol 91: 807– 819, 2006. apnea and brain-stem neural function in preterm infants. N Engl
296. Guzy RD, Mack MM, Schumacker PT. Mitochondrial complex III J Med 308: 353–357, 1983.
is required for hypoxia-induced ROS production and gene tran- 319. Henderson-Smart DJ, Read DJ. Ventilatory responses to
scription in yeast. Antioxid Redox Signal 9: 1317–1328, 2007. hypoxaemia during sleep in the newborn. J Dev Physiol 1: 195–208,
297. Haddad GG, Donnelly DF. O2 deprivation induces a major depo- 1979.
larization in brain stem neurons in the adult but not in the neonatal 320. Herigstad M, Fatemian M, Robbins PA. Respiratory control
rat. J Physiol 429: 411– 428, 1990. during air-breathing exercise in humans following an 8 h exposure
298. Hall SM, Gorenflo M, Reader J, Lawson D, Haworth SG. Neo- to hypoxia. Respir Physiol Neurobiol 162: 169 –175, 2008.
natal pulmonary hypertension prevents reorganisation of the pul- 321. Hildebrandt W, Alexander S, Bartsch P, Droge W. Effect of
monary arterial smooth muscle cytoskeleton after birth. J Anat N-acetyl-cysteine on the hypoxic ventilatory response and erythro-
196: 391– 403, 2000. poietin production: linkage between plasma thiol redox state and
299. Hannhart B, Pickett CK, Weil JV, Moore LG. Influence of O2 chemosensitivity. Blood 99: 1552–1555, 2002.
pregnancy on ventilatory and carotid body neural output respon- 322. Hodges MR, Richerson GB. Contributions of 5-HT neurons to
siveness to hypoxia in cats. J Appl Physiol 67: 797– 803, 1989. respiratory control: neuromodulatory and trophic effects. Respir
300. Hansen J, Sander M. Sympathetic neural overactivity in healthy Physiol Neurobiol 164: 222–232, 2008.
humans after prolonged exposure to hypobaric hypoxia. J Physiol 323. Hodges MR, Richerson GB. Interaction between defects in ven-
546: 921–929, 2003.
tilatory and thermoregulatory control in mice lacking 5-HT neu-
301. Hanson MA, Kumar P, Williams BA. The effect of chronic hyp-
rons. Respir Physiol Neurobiol 164: 350 –357, 2008.
oxia upon the development of respiratory chemoreflexes in the
324. Hodges MR, Tattersall GJ, Harris MB, McEvoy SD, Richerson
newborn kitten. J Physiol 411: 563–574, 1989.
DN, Deneris ES, Johnson RL, Chen ZF, Richerson GB. Defects
302. Harding R. Fetal pulmonary development: the role of respiratory
movements. Equine Vet J Suppl 32–39, 1997. in breathing and thermoregulation in mice with near-complete
303. Hatton CJ, Carpenter E, Pepper DR, Kumar P, Peers C. De- absence of central serotonin neurons. J Neurosci 28: 2495–2505,
velopmental changes in isolated rat type I carotid body cell K⫹ 2008.
currents and their modulation by hypoxia. J Physiol 501: 49 –58, 325. Honda Y, Hata N, Sakakibara Y, Nishino T, Satomura Y. Cen-
1997. tral hypoxic-hypercapnic interaction in mild hypoxia in man.
304. Hatton CJ, Peers C. Effects of cytochrome P-450 inhibitors on Pflügers Arch 391: 289 –295, 1981.
ionic currents in isolated rat type I carotid body cells. Am J Physiol 326. Honda Y, Watanabe S, Hashizume I, Satomura Y, Hata N,
Cell Physiol 271: C85–C92, 1996. Sakakibara Y, Severinghaus JW. Hypoxic chemosensitivity in
305. Hayashi F, Coles SK, Bach KB, Mitchell GS, McCrimmon DR. asthmatic patients two decades after carotid body resection. J Appl
Time-dependent phrenic nerve responses to carotid afferent acti- Physiol 46: 632– 638, 1979.
vation: intact vs. decerebellate rats. Am J Physiol Regul Integr 327. Hoop B, Beagle JL, Maher TJ, Kazemi H. Brainstem amino acid
Comp Physiol 265: R811–R819, 1993. neurotransmitters and hypoxic ventilatory response. Respir
306. He L, Chen J, Dinger B, Sanders K, Sundar K, Hoidal J, Physiol 118: 117–129, 1999.
Fidone S. Characteristics of carotid body chemosensitivity in 328. Hooper SB, Coulter CL, Deayton JM, Harding R, Thorburn
NADPH oxidase-deficient mice. Am J Physiol Cell Physiol 282: GD. Fetal endocrine responses to prolonged hypoxemia in sheep.
C27–C33, 2002. Am J Physiol Regul Integr Comp Physiol 259: R703–R708, 1990.

Physiol Rev • VOL 90 • APRIL 2010 • www.prv.org


742 LUC J. TEPPEMA AND ALBERT DAHAN

329. Horn EM, Dillon GH, Fan YP, Waldrop TG. Developmental 352. Ivan M, Kondo K, Yang H, Kim W, Valiando J, Ohh M, Salic A,
aspects and mechanisms of rat caudal hypothalamic neuronal re- Asara JM, Lane WS, Kaelin WG Jr. HIF␣ targeted for VHL-
sponses to hypoxia. J Neurophysiol 81: 1949 –1959, 1999. mediated destruction by proline hydroxylation: implications for O2
330. Horn EM, Waldrop TG. Oxygen-sensing neurons in the caudal sensing. Science 292: 464 – 468, 2001.
hypothalamus and their role in cardiorespiratory control. Respir 353. Jaakkola P, Mole DR, Tian YM, Wilson MI, Gielbert J, Gaskell
Physiol 110: 219 –228, 1997. SJ, Kriegsheim Av Hebestreit HF, Mukherji M, Schofield CJ,
331. Horn EM, Waldrop TG. Suprapontine control of respiration. Re- Maxwell PH, Pugh CW, Ratcliffe PJ. Targeting of HIF-alpha to
spir Physiol 114: 201–211, 1998. the von Hippel-Lindau ubiquitylation complex by O2-regulated
332. Hornbein TF. The puzzle of periodicity. Pediatrics 50: 183–185, prolyl hydroxylation. Science 292: 468 – 472, 2001.
1972. 354. Jackson A, Nurse C. Dopaminergic properties of cultured rat
333. Hornbein TF, Griffo ZJ, Roos A. Quantitation of chemoreceptor carotid body chemoreceptors grown in normoxic and hypoxic
activity: interrelation of hypoxia and hypercapnia. J Neurophysiol environments. J Neurochem 69: 645– 654, 1997.
24: 561–568, 1961. 355. Jackson A, Nurse CA. Role of acetylcholine receptors and dopa-
334. Hornbein TF, Roos A. Specificity of H ion concentration as a mine transporter in regulation of extracellular dopamine in rat
carotid chemoreceptor stimulus. J Appl Physiol 18: 580 –584, 1963. carotid body cultures grown in chronic hypoxia or nicotine. J Neu-
335. Housley GD, Martin-Body RL, Dawson NJ, Sinclair JD. Brain rochem 70: 653– 662, 1998.
stem projections of the glossopharyngeal nerve and its carotid 356. Jacky JP. Barometric measurement of tidal volume: effects of
sinus branch in the rat. Neuroscience 22: 237–250, 1987. pattern and nasal temperature. J Appl Physiol 49: 319 –325, 1980.
336. Howard LS, Robbins PA. Problems with determining the hypoxic 357. Jansen AH, Chernick V. Development of respiratory control.
response in humans using stepwise changes in end-tidal PO2. Re- Physiol Rev 63: 437– 483, 1983.
spir Physiol 98: 241–249, 1994. 358. Janssen PL, Dwinell MR, Pizarro J, Bisgard GE. Intracarotid
337. Howard LS, Robbins PA. Alterations in respiratory control during dopamine infusion does not prevent acclimatization to hypoxia.
8 h of isocapnic and poikilocapnic hypoxia in humans. J Appl Respir Physiol 111: 33– 43, 1998.
Physiol 78: 1098 –1107, 1995. 359. Janssen PL, O’Halloran KD, Pizarro J, Dwinell MR, Bisgard
338. Howard LS, Robbins PA. Ventilatory response to 8 h of isocapnic GE. Carotid body dopaminergic mechanisms are functional after
and poikilocapnic hypoxia in humans. J Appl Physiol 78: 1092– acclimatization to hypoxia in goats. Respir Physiol 111: 25–32,
1097, 1995. 1998.
339. Huang J, Lusina S, Xie T, Ji E, Xiang S, Liu Y, Weiss JW. 360. Janssens JP, Pache JC, Nicod LP. Physiological changes in
Sympathetic response to chemostimulation in conscious rats ex- respiratory function associated with ageing. Eur Respir J 13: 197–
posed to chronic intermittent hypoxia. Respir Physiol Neurobiol 205, 1999.
166: 102–106, 2009.
361. Javaheri S, Teppema LJ, Evers JA. Effects of aminophylline on
340. Huang J, Suguihara C, Hehre D, Lin J, Bancalari E. Effects of
hypoxemia-induced ventilatory depression in the cat. J Appl
GABA receptor blockage on the respiratory response to hypoxia in
Physiol 64: 1837–1843, 1988.
sedated newborn piglets. J Appl Physiol 77: 1006 –1010, 1994.
362. Jensen D, Wolfe LA, O’Donnell DE, Davies GA. Chemoreflex
341. Huang LE, Arany Z, Livingston DM, Bunn HF. Activation of
control of breathing during wakefulness in healthy men and
hypoxia-inducible transcription factor depends primarily upon re-
women. J Appl Physiol 98: 822– 828, 2005.
dox-sensitive stabilization of its alpha subunit. J Biol Chem 271:
363. Johnston BM, Gluckman PD. Peripheral chemoreceptors re-
32253–32259, 1996.
spond to hypoxia in pontine-lesioned fetal lambs in utero. J Appl
342. Huey KA, Brown IP, Jordan MC, Powell FL. Changes in dopa-
Physiol 75: 1027–1034, 1993.
mine D2-receptor modulation of the hypoxic ventilatory response
with chronic hypoxia. Respir Physiol 123: 177–187, 2000. 364. Jordan AS, Catcheside PG, Orr RS, O’Donoghue FJ, Saunders
343. Huey KA, Low MJ, Kelly MA, Juarez R, Szewczak JM, Powell NA, McEvoy RD. Ventilatory decline after hypoxia and hypercap-
FL. Ventilatory responses to acute and chronic hypoxia in mice: nia is not different between healthy young men and women. J Appl
effects of dopamine D2 receptors. J Appl Physiol 89: 1142–1150, Physiol 88: 3–9, 2000.
2000. 365. Jordan W, Cohrs S, Degner D, Meier A, Rodenbeck A, Mayer
344. Huey KA, Powell FL. Time-dependent changes in dopamine D2- G, Pilz J, Ruther E, Kornhuber J, Bleich S. Evaluation of
receptor mRNA in the arterial chemoreflex pathway with chronic oxidative stress measurements in obstructive sleep apnea syn-
hypoxia. Brain Res 75: 264 –270, 2000. drome. J Neural Transm 113: 239 –254, 2006.
345. Huey KA, Szewczak JM, Powell FL. Dopaminergic mechanisms 366. Joseph V, Soliz J, Pequignot J, Sempore B, Cottet-Emard JM,
of neural plasticity in respiratory control: transgenic approaches. Dalmaz Y, Favier R, Spielvogel H, Pequignot JM. Gender dif-
Respir Physiol Neurobiol 135: 133–144, 2003. ferentiation of the chemoreflex during growth at high altitude:
346. Hupperets MD, Hopkins SR, Pronk MG, Tiemessen IJ, Garcia functional and neurochemical studies. Am J Physiol Regul Integr
N, Wagner PD, Powell FL. Increased hypoxic ventilatory re- Comp Physiol 278: R806 –R816, 2000.
sponse during 8 weeks at 3800 m altitude. Respir Physiol Neuro- 367. Julien C, Bairam A, Joseph V. Chronic intermittent hypoxia
biol 142: 145–152, 2004. reduces ventilatory long-term facilitation and enhances apnea fre-
347. Ide K, Worthley M, Anderson T, Poulin MJ. Effect of systemic quency in newborn rats. Am J Physiol Regul Integr Comp Physiol
administration of the nitric oxide synthase inhibitor L-NMMA on 294: R1356 –R1366, 2008.
the human ventilatory response to hypoxia. Adv Exp Med Biol 605: 368. Kaab S, Miguel-Velado E, Lopez-Lopez JR, Perez-Garcia MT.
41– 45, 2008. Downregulation of Kv3.4 channels by chronic hypoxia increases
348. Insalaco G, Romano S, Salvaggio A, Braghiroli A, Lanfranchi acute oxygen sensitivity in rabbit carotid body. J Physiol 566:
P, Patruno V, Donner CF, Bonsignore G. Cardiovascular and 395– 408, 2005.
ventilatory response to isocapnic hypoxia at sea level and at 5,050 369. Kaelin J, Ratcliffe PJ. Oxygen sensing by metazoans: the central
m. J Appl Physiol 80: 1724 –1730, 1996. role of the HIF hydroxylase pathway. Mol Cell 30: 393– 402, 2008.
349. Irsigler GB, Stafford MJ, Severinghaus JW. Relationship of 370. Kagawa S, Stafford MJ, Waggener TB, Severinghaus JW. No
CSF pH, O2, and CO2 responses in metabolic acidosis and alkalosis effect of naloxone on hypoxia-induced ventilatory depression in
in humans. J Appl Physiol 48: 355–361, 1980. adults. J Appl Physiol 52: 1030 –1034, 1982.
350. Iturriaga R, Alcayaga J. Neurotransmission in the carotid body: 371. Kaluz S, Kaluzova M, Stanbridge EJ. Regulation of gene expres-
transmitters and modulators between glomus cells and petrosal sion by hypoxia: integration of the HIF-transduced hypoxic signal
ganglion nerve terminals. Brain Res 47: 46 –53, 2004. at the hypoxia-responsive element. Clin Chim Acta 395: 6 –13,
351. Iturriaga R, Alcayaga J, Zapata P. Dissociation of hypoxia- 2008.
induced chemosensory responses and catecholamine efflux in cat 372. Kara T, Narkiewicz K, Somers VK. Chemoreflexes: physiology
carotid body superfused in vitro. J Physiol 497: 551–564, 1996. and clinical implications. Acta Physiol Scand 177: 377–384, 2003.

Physiol Rev • VOL 90 • APRIL 2010 • www.prv.org


THE VENTILATORY RESPONSE TO HYPOXIA IN MAMMALS 743

373. Katayama K, Fujita H, Sato K, Ishida K, Iwasaki K, Miyamura 395. Kolb JC, Ainslie PN, Ide K, Poulin MJ. Effects of five consec-
M. Effect of a repeated series of intermittent hypoxic exposures on utive nocturnal hypoxic exposures on the cerebrovascular re-
ventilatory response in humans. High Alt Med Biol 6: 50 –59, 2005. sponses to acute hypoxia and hypercapnia in humans. J Appl
374. Katayama K, Sato K, Hotta N, Ishida K, Iwasaki K, Miyamura Physiol 96: 1745–1754, 2004.
M. Intermittent hypoxia does not increase exercise ventilation at 396. Koos BJ, Chao A, Doany W. Adenosine stimulates breathing in
simulated moderate altitude. Int J Sports Med 28: 480 – 487, 2007. fetal sheep with brain stem section. J Appl Physiol 72: 94 –99, 1992.
375. Katayama K, Sato Y, Ishida K, Mori S, Miyamura M. The effects 397. Koos BJ, Chau A, Matsuura M, Punla O, Kruger L. Thalamic
of intermittent exposure to hypoxia during endurance exercise locus mediates hypoxic inhibition of breathing in fetal sheep.
training on the ventilatory responses to hypoxia and hypercapnia in J Neurophysiol 79: 2383–2393, 1998.
humans. Eur J Appl Physiol Occup Physiol 78: 189 –194, 1998. 398. Koos BJ, Chau A, Matsuura M, Punla O, Kruger L. Thalamic
376. Katayama K, Sato Y, Morotome Y, Shima N, Ishida K, Mori S, lesions dissociate breathing inhibition by hypoxia and adenosine in
Miyamura M. Intermittent hypoxia increases ventilation and SaO2 fetal sheep. Am J Physiol Regul Integr Comp Physiol 278: R831–
during hypoxic exercise and hypoxic chemosensitivity. J Appl R837, 2000.
Physiol 90: 1431–1440, 2001. 399. Koos BJ, Kawasaki Y, Hari A, Bohorquez F, Jan C, Roostaeian
377. Katayama K, Sato Y, Shima N, Qiu JC, Ishida K, Mori S, J, Wilson CL, Kruger L. Electrical stimulation of the posterome-
Miyamura M. Enhanced chemosensitivity after intermittent dial thalamus modulates breathing in unanesthetized fetal sheep.
hypoxic exposure does not affect exercise ventilation at sea level. J Appl Physiol 96: 115–123, 2004.
Eur J Appl Physiol 87: 187–191, 2002. 400. Koos BJ, Kawasaki Y, Kim YH, Bohorquez F. Adenosine A2A-
378. Kawai Y, Qi J, Comer AM, Gibbons H, Win J, Lipski J. Effects receptor blockade abolishes the roll-off respiratory response to
of cyanide and hypoxia on membrane currents in neurones acutely hypoxia in awake lambs. Am J Physiol Regul Integr Comp Physiol
dissociated from the rostral ventrolateral medulla of the rat. Brain 288: R1185–R1194, 2005.
Res 830: 246 –257, 1999. 401. Koos BJ, Kitanaka T, Matsuda K, Gilbert RD, Longo LD. Fetal
379. Kazemi H, Hoop B. Glutamic acid and gamma-aminobutyric acid breathing adaptation to prolonged hypoxaemia in sheep. J Dev
neurotransmitters in central control of breathing. J Appl Physiol Physiol 10: 161–166, 1988.
70: 1–7, 1991. 402. Koos BJ, Kruger L, Murray TF. Source of extracellular brain
380. Kemp PJ. Detecting acute changes in oxygen: will the real sensor adenosine during hypoxia in fetal sheep. Brain Res 778: 439 – 442,
please stand up? Exp Physiol 91: 829 – 834, 2006. 1997.
381. Kenneth NS, Rocha S. Regulation of gene expression by hypoxia. 403. Koos BJ, Maeda T, Jan C, Lopez G. Adenosine A2A receptors
Biochem J 414: 19 –29, 2008. mediate hypoxic inhibition of fetal breathing in sheep. Am J Obstet
382. Khamnei S, Robbins PA. Hypoxic depression of ventilation in Gynecol 186: 663– 668, 2002.
humans: alternative models for the chemoreflexes. Respir Physiol 404. Koos BJ, Mason BA, Punla O, Adinolfi AM. Hypoxic inhibition
81: 117–134, 1990. of breathing in fetal sheep: relationship to brain adenosine concen-
383. Kimura H, Tanaka M, Nagao K, Niijima M, Masuyama S,
trations. J Appl Physiol 77: 2734 –2739, 1994.
Mizoo A, Uruma T, Tatsumi K, Kuriyama T, Masuda A, Koba-
405. Koos BJ, Matsuda K. Fetal breathing, sleep state, and cardiovas-
yashi T, Honda Y. A new aspect of the carotid body function
cular responses to adenosine in sheep. J Appl Physiol 68: 489 – 495,
controlling hypoxic ventilatory decline in humans. Appl Human
1990.
Sci 17: 131–137, 1998.
406. Koos BJ, Sameshima H. Effects of hypoxaemia and hypercapnia
384. Kirby TP, Wraith PK, De Cort SC, Airlie MA, Hill JE, Carson
on breathing movements and sleep state in sinoaortic-denervated
ER, Flenley DC, Warren PM. Modelling the dynamic ventilatory
fetal sheep. J Dev Physiol 10: 131–144, 1988.
response to hypoxia in normal subjects. J Theor Biol 166: 135–147,
407. Koos BJ, Sameshima H, Power GG. Fetal breathing movement,
1994.
385. Kita I, Sato-Suzuki I, Oguri M, Arita H. Yawning responses sleep state and cardiovascular responses to an inhibitor of mito-
induced by local hypoxia in the paraventricular nucleus of the rat. chondrial ATPase in sheep. J Dev Physiol 8: 67–75, 1986.
Behav Brain Res 117: 119 –126, 2000. 408. Koos BJ, Sameshima H, Power GG. Fetal breathing, sleep state,
386. Kiwull-Schone H, Kiwull P, Muckenhoff K, Both W. The role of and cardiovascular responses to graded anemia in sheep. J Appl
carotid chemoreceptors in the regulation of arterial oxygen trans- Physiol 63: 1463–1468, 1987.
port under hypoxia with and without hypercapnia. Adv Exp Med 409. Kourie JI. Interaction of reactive oxygen species with ion trans-
Biol 75: 469 – 476, 1976. port mechanisms. Am J Physiol Cell Physiol 275: C1–C24, 1998.
387. Kline DD, Overholt JL, Prabhakar NR. Mutant mice deficient in 410. Kramer JM, Nolan PC, Waldrop TG. In vitro responses of neu-
NOS-1 exhibit attenuated long-term facilitation and short-term po- rons in the periaqueductal gray to hypoxia and hypercapnia. Brain
tentiation in breathing. J Physiol 539: 309 –315, 2002. Res 835: 197–203, 1999.
388. Kline DD, Peng YJ, Manalo DJ, Semenza GL, Prabhakar NR. 411. Kronenberg R, Hamilton FN, Gabel R, Hickey R, Read DJ,
Defective carotid body function and impaired ventilatory re- Severinghaus J. Comparison of three methods for quantitating
sponses to chronic hypoxia in mice partially deficient for hypoxia- respiratory response to hypoxia in man. Respir Physiol 16: 109 –
inducible factor 1 alpha. Proc Natl Acad Sci USA 99: 821– 826, 2002. 125, 1972.
389. Kline DD, Yang T, Huang PL, Prabhakar NR. Altered respiratory 412. Kronenberg RS, Drage CW. Attenuation of the ventilatory and
responses to hypoxia in mutant mice deficient in neuronal nitric heart rate responses to hypoxia and hypercapnia with aging in
oxide synthase. J Physiol 511: 273–287, 1998. normal men. J Clin Invest 52: 1812–1819, 1973.
390. Kline DD, Yang T, Premkumar DR, Thomas AJ, Prabhakar NR. 413. Kuipers IM, Maertzdorf WJ, Keunen H, De Jong DS, Hanson
Blunted respiratory responses to hypoxia in mutant mice deficient MA, Blanco CE. Effect of maternal hypoxemia on behavior in
in nitric oxide synthase-3. J Appl Physiol 88: 1496 –1508, 2000. unanesthetized normoxic or mildly hyperoxic fetal lambs. J Appl
391. Kobayashi K, Lemke RP, Greer JJ. Ultrasound measurements of Physiol 76: 2535–2540, 1994.
fetal breathing movements in the rat. J Appl Physiol 91: 316 –320, 414. Kumar GK, Prabhakar NR. Post-translational modification of
2001. proteins during intermittent hypoxia. Respir Physiol Neurobiol
392. Kobertz WR, Williams C, Miller C. Hanging gondola structure of 164: 272–276, 2008.
the T1 domain in a voltage-gated K⫹ channel. Biochemistry 39: 415. Kumar P. Sensing hypoxia in the carotid body: from stimulus to
10347–10352, 2000. response. Essays Biochem 43: 43– 60, 2007.
393. Koehle MS, Sheel AW, Milsom WK, McKenzie DC. Two pat- 416. Kumar P, Bin-Jaliah I. Adequate stimuli of the carotid body: more
terns of daily hypoxic exposure and their effects on measures of than an oxygen sensor? Respir Physiol Neurobiol 157: 12–21, 2007.
chemosensitivity in humans. J Appl Physiol 103: 1973–1978, 2007. 417. Kumar P, Peers C. AMP-activated protein kinase: function and
394. Koerner P, Hesslinger C, Schaefermeyer A, Prinz C, Gratzl M. dysfunction in health and disease. J Physiol 574: 3– 6, 2006.
Evidence for histamine as a transmitter in rat carotid body sensor 418. Kunitomo F, Kimura H, Tatsumi K, Kuriyama T, Watanabe S,
cells. J Neurochem 91: 493–500, 2004. Honda Y. Sex differences in awake ventilatory drive and abnormal

Physiol Rev • VOL 90 • APRIL 2010 • www.prv.org


744 LUC J. TEPPEMA AND ALBERT DAHAN

breathing during sleep in eucapnic obesity. Chest 93: 968 –976, carotid body receptor stimulation. Auton Neurosci 141: 73– 82,
1988. 2008.
419. Kusakabe T, Hirakawa H, Oikawa S, Matsuda H, Kawakami T, 441. Lendahl U, Lee KL, Yang H, Poellinger L. Generating specificity
Takenaka T, Hayashida Y. Morphological changes in the rat and diversity in the transcriptional response to hypoxia. Nat Rev
carotid body 1, 2, 4, and 8 weeks after the termination of chroni- Genet 10: 821– 832, 2009.
cally hypocapnic hypoxia. Histol Histopathol 19: 1133–1140, 2004. 442. Leonard TO, Lydic R. Pontine nitric oxide modulates acetylcho-
420. Kusakabe T, Matsuda H, Hayashida Y. Hypoxic adaptation of line release, rapid eye movement sleep generation, and respiratory
the rat carotid body. Histol Histopathol 20: 987–997, 2005. rate. J Neurosci 17: 774 –785, 1997.
421. Kuwaki T. Orexinergic modulation of breathing across vigilance 443. Leung PS, Fung ML, Tam MS. Renin-angiotensin system in the
states. Respir Physiol Neurobiol 164: 204 –212, 2008. carotid body. Int J Biochem Cell Biol 35: 847– 854, 2003.
422. Lahiri S, DeLaney RG. Relationship between carotid chemore- 444. Li A, Nattie E. Serotonin transporter knockout mice have a re-
ceptor activity and ventilation in the cat. Respir Physiol 24: 267– duced ventilatory response to hypercapnia predominantly in males
286, 1975. but not to hypoxia. J Physiol 586: 2321–2329, 2008.
423. Lahiri S, DeLaney RG. Stimulus interaction in the responses of 445. Liang PJ, Bascom DA, Robbins PA. Extended models of the
carotid body chemoreceptor single afferent fibers. Respir Physiol ventilatory response to sustained isocapnic hypoxia in humans.
24: 249 –266, 1975. J Appl Physiol 82: 667– 677, 1997.
424. Lahiri S, Ehleben W, Acker H. Chemoreceptor discharges and 446. Lin LH, Emson PC, Talman WT. Apposition of neuronal elements
cytochrome redox changes of the rat carotid body: role of heme containing nitric oxide synthase and glutamate in the nucleus
ligands. Proc Natl Acad Sci USA 96: 9427–9432, 1999. tractus solitarii of rat: a confocal microscopic analysis. Neuro-
425. Lahiri S, Mitchell CH, Reigada D, Roy A, Cherniack NS. Pu- science 96: 341–350, 2000.
rines, the carotid body and respiration. Respir Physiol Neurobiol 447. Ling L. Serotonin and NMDA receptors in respiratory long-term
157: 123–129, 2007. facilitation. Respir Physiol Neurobiol 164: 233–241, 2008.
426. Lahiri S, Roy A, Baby SM, Hoshi T, Semenza GL, Prabhakar 448. Ling L, Fuller DD, Bach KB, Kinkead R, Olson EB Jr, Mitchell
NR. Oxygen sensing in the body. Prog Biophys Mol Biol 91: 249 – GS. Chronic intermittent hypoxia elicits serotonin-dependent plas-
286, 2006. ticity in the central neural control of breathing. J Neurosci 21:
427. Lahiri S, Rumsey WL, Wilson DF, Iturriaga R. Contribution of 5381–5388, 2001.
in vivo microvascular PO2 in the cat carotid body chemotransduc- 449. Ling L, Olson EB Jr, Vidruk EH, Mitchell GS. Attenuation of the
tion. J Appl Physiol 75: 1035–1043, 1993. hypoxic ventilatory response in adult rats following one month of
428. Lahiri S, Smatresk N, Pokorski M, Barnard P, Mokashi A. perinatal hyperoxia. J Physiol 495: 561–571, 1996.
Efferent inhibition of carotid body chemoreception in chronically 450. Ling L, Olson EB Jr, Vidruk EH, Mitchell GS. Integrated
hypoxic cats. Am J Physiol Regul Integr Comp Physiol 245: R678 – phrenic responses to carotid afferent stimulation in adult rats
R683, 1983. following perinatal hyperoxia. J Physiol 500: 787–796, 1997.
429. Lahiri S, Smatresk N, Pokorski M, Barnard P, Mokashi A, 451. Lipton AJ, Johnson MA, Macdonald T, Lieberman MW, Gozal
McGregor KH. Dopaminergic efferent inhibition of carotid body D, Gaston B. S-nitrosothiols signal the ventilatory response to
chemoreceptors in chronically hypoxic cats. Am J Physiol Regul hypoxia. Nature 413: 171–174, 2001.
Integr Comp Physiol 247: R24 –R28, 1984. 452. Liu C, Smith TG, Balanos GM, Brooks J, Crosby A, Herigstad
430. Lahiri S. Historical perspectives of cellular oxygen sensing and M, Dorrington KL, Robbins PA. Lack of involvement of the
responses to hypoxia. J Appl Physiol 88: 1467–1473, 2000. autonomic nervous system in early ventilatory and pulmonary vas-
431. Lam SY, Fung ML, Leung PS. Regulation of the angiotensin- cular acclimatization to hypoxia in humans. J Physiol 579: 215–225,
converting enzyme activity by a time-course hypoxia in the carotid 2007.
body. J Appl Physiol 96: 809 – 813, 2004. 453. Liu J, Boujedaini N, Cazin L, Mallet E, Clabaut M. Develop-
432. Lam SY, Leung PS. Chronic hypoxia activates a local angiotensin- mental changes in cardio-respiratory responses to hypoxia and
generating system in rat carotid body. Mol Cell Endocrinol 203: hypercapnia in anesthetized low-birth-weight rats. Respir Physiol
147–153, 2003. 123: 189 –199, 2000.
433. Lam SY, Tipoe GL, Liong EC, Fung ML. Chronic hypoxia up- 454. Liu Q, Lowry TF, Wong-Riley MT. Postnatal changes in ventila-
regulates the expression and function of proinflammatory cyto- tion during normoxia and acute hypoxia in the rat: implication for
kines in the rat carotid body. Histochem Cell Biol 130: 549 –559, a sensitive period. J Physiol 577: 957–970, 2006.
2008. 455. Liu X, He L, Stensaas L, Dinger B, Fidone S. Adaptation to
434. Lam SY, Tipoe GL, Liong EC, Fung ML. Differential expressions chronic hypoxia involves immune cell invasion and increased ex-
and roles of hypoxia-inducible factor-1alpha, -2alpha and -3alpha in pression of inflammatory cytokines in rat carotid body. Am J
the rat carotid body during chronic and intermittent hypoxia. His- Physiol Lung Cell Mol Physiol 296: L158 –L166, 2009.
tol Histopathol 23: 271–280, 2008. 456. Liu Y, Ji ES, Xiang S, Tamisier R, Tong J, Huang J, Weiss JW.
435. LaManna JC, Haxhiu MA, Kutina-Nelson KL, Pundik S, Exposure to cyclic intermittent hypoxia increases expression of
Erokwu B, Yeh ER, Lust WD, Cherniack NS. Decreased energy functional NMDA receptors in the rat carotid body. J Appl Physiol
metabolism in brain stem during central respiratory depression in 106: 259 –267, 2009.
response to hypoxia. J Appl Physiol 81: 1772–1777, 1996. 457. Lloyd BB, Cunningham DJC. A quantitative approach to the
436. LaManna JC, Kutina-Nelson KL, Hritz MA, Huang Z, Wong- regulation of human respiration. In: The Regulation of Human
Riley MT. Decreased rat brain cytochrome oxidase activity after Respiration, edited by Cunningham DJC and Lloyd BB. Oxford UK:
prolonged hypoxia. Brain Res 720: 1– 6, 1996. Blackwell Science, 1963, p. 331–349.
437. Landauer RC, Pepper DR, Kumar P. Effect of chronic hypoxae- 458. Loeschcke HH, Gertz KH. Effect of oxygen pressure in inspired
mia from birth upon chemosensitivity in the adult rat carotid body air on respiratory activity of the human, tested under the constant
in vitro. J Physiol 485: 543–550, 1995. behavior of alveolar carbon dioxide pressure. Pflügers Arch 267:
438. Lazarov N, Rozloznik M, Reindl S, Rey-Ares V, Dutschmann 460 – 477, 1958.
M, Gratzl M. Expression of histamine receptors and effect of 459. Long GR, Filuk R, Balakumar M, Easton PA, Anthonisen NR.
histamine in the rat carotid body chemoafferent pathway. Eur Ventilatory response to sustained hypoxia: effect of methysergide
J Neurosci 24: 3431–3444, 2006. and verapamil. Respir Physiol 75: 173–182, 1989.
439. Lee YM, Kim BJ, Chun YS, So I, Choi H, Kim MS, Park JW. 460. Long W, Anthonisen NR. Dopaminergic influence on the ventila-
NOX4 as an oxygen sensor to regulate TASK-1 activity. Cell Signal tory response to sustained hypoxia in the cat. J Appl Physiol 78:
18: 499 –507, 2006. 1250 –1255, 1995.
440. Lemus M, Montero S, Cadenas JL, Lara JJ, Tejeda-Chavez 461. Long W, Lobchuk D, Anthonisen NR. Ventilatory responses to
HR, Alvarez-Buylla R, de Alvarez-Buylla ER. GABA-B receptors CO2 and hypoxia after sustained hypoxia in awake cats. J Appl
activation in the NTS blocks the glycemic responses induced by Physiol 76: 2262–2266, 1994.

Physiol Rev • VOL 90 • APRIL 2010 • www.prv.org


THE VENTILATORY RESPONSE TO HYPOXIA IN MAMMALS 745

462. Long WA, Lawson EE. Neurotransmitters and biphasic respira- immediate early gene fosB. Respir Physiol Neurobiol 145: 23–31,
tory response to hypoxia. J Appl Physiol 57: 213–222, 1984. 2005.
463. Long WQ, Anthonisen NR. Aminophylline partially blocks venti- 487. Marchal F, Bairam A, Haouzi P, Crance JP, Di GC, Vert P,
latory depression with hypoxia in the awake cat. Can J Physiol Lahiri S. Carotid chemoreceptor response to natural stimuli in the
Pharmacol 72: 673– 678, 1994. newborn kitten. Respir Physiol 87: 183–193, 1992.
464. Long WQ, Giesbrecht GG, Anthonisen NR. Ventilatory response 488. Martin CB Jr, Murata Y, Petrie RH, Parer JT. Respiratory
to moderate hypoxia in awake chemodenervated cats. J Appl movements in fetal rhesus monkeys. Am J Obstet Gynecol 119:
Physiol 74: 805– 810, 1993. 939 –948, 1974.
465. Lopez-Barneo J, del Toro R, Levitsky KL, Chiara MD, Ortega- 489. Martin RJ, Difiore JM, Jana L, Davis RL, Miller MJ, Coles SK,
Saenz P. Regulation of oxygen sensing by ion channels. J Appl Dick TE. Persistence of the biphasic ventilatory response to hyp-
Physiol 96: 1187–1195, 2004. oxia in preterm infants. J Pediatr 132: 960 –964, 1998.
466. Lopez-Barneo J, Lopez-Lopez JR, Urena J, Gonzalez C. Che- 490. Martin RJ, Van LE, Haxhiu MA, Carlo WA. Upper airway mus-
motransduction in the carotid body: K⫹ current modulated by PO2 cle and diaphragm responses to hypoxia in the piglet. J Appl
in type I chemoreceptor cells. Science 241: 580 –582, 1988. Physiol 68: 672– 677, 1990.
467. Lopez-Barneo J, Ortega-Saenz P, Pardal R, Pascual A, Piruat 491. Martin-Body RL, Johnston BM. Central origin of the hypoxic
JI. Carotid body oxygen sensing. Eur Respir J 32: 1386 –1398, 2008. depression of breathing in the newborn. Respir Physiol 71: 25–32,
468. Lopez-Barneo J, Pardal R, Montoro RJ, Smani T, Garcia- 1988.
Hirschfeld J, Urena J. K⫹ and Ca2⫹ channel activity and cytosolic 492. Martin-Body RL, Robson GJ, Sinclair JD. Restoration of
[Ca2⫹] in oxygen-sensing tissues. Respir Physiol 115: 215–227, hypoxic respiratory responses in the awake rat after carotid body
1999. denervation by sinus nerve section. J Physiol 380: 61–73, 1986.
469. Lopez-Barneo J, Pardal R, Ortega-Saenz P. Cellular mechanism 493. Massaro GD, Olivier J, Dzikowski C, Massaro D. Postnatal
of oxygen sensing. Annu Rev Physiol 63: 259 –287, 2001. development of lung alveoli: suppression by 13% O2 and a critical
470. Lopez-Lopez JR, Perez-Garcia MT. Oxygen sensitive Kv chan- period. Am J Physiol Lung Cell Mol Physiol 258: L321–L327, 1990.
nels in the carotid body. Respir Physiol Neurobiol 157: 65–74, 2007. 494. Masuda A, Ohyabu Y, Kobayashi T, Yoshino C, Sakakibara Y,
471. Lorier AR, Huxtable AG, Robinson DM, Lipski J, Housley GD, Komatsu T, Honda Y. Lack of positive interaction between CO2
Funk GD. P2Y1 receptor modulation of the pre-Botzinger complex and hypoxic stimulation for PCO2-VAS response slope in humans.
inspiratory rhythm generating network in vitro. J Neurosci 27: Respir Physiol 126: 173–181, 2001.
993–1005, 2007. 495. Mateika JH, Fregosi RF. Long-term facilitation of upper airway
472. Lovering AT, Dunin-Barkowski WL, Vidruk EH, Orem JM. muscle activities in vagotomized and vagally intact cats. J Appl
Ventilatory response of the cat to hypoxia in sleep and wakeful- Physiol 82: 419 – 425, 1997.
ness. J Appl Physiol 95: 545–554, 2003. 496. Matsuda Y, Patrick J, Carmichael L, Fraher L, Richardson B.
473. Lowry TF, Forster HV, Pan LG, Korducki MA, Probst J, Fran- Recovery of the ovine fetus from sustained hypoxia: effects on
endocrine, cardiovascular, and biophysical activity. Am J Obstet
ciosi RA, Forster M. Effect of carotid body denervation on
Gynecol 170: 1433–1441, 1994.
breathing in neonatal goats. J Appl Physiol 87: 1026 –1034, 1999.
497. Matsuoka T, Yoda T, Ushikubo S, Matsuzawa S, Sasano T,
474. Lowry TF, Forster HV, Pan LG, Serra A, Wenninger J, Nash R,
Komiyama A. Repeated acute hypoxia temporarily attenuates the
Sheridan D, Franciosi RA. Effects on breathing of carotid body
ventilatory respiratory response to hypoxia in conscious newborn
denervation in neonatal piglets. J Appl Physiol 87: 2128 –2135, 1999.
rats. Pediatr Res 46: 120 –125, 1999.
475. Lugliani R, Whipp BJ, Seard C, Wasserman K. Effect of bilateral
498. Maxova H, Vizek M. Ventilatory response to sustained hypoxia in
carotid-body resection on ventilatory control at rest and during
carotid body denervated rats. Physiol Res 50: 327–331, 2001.
exercise in man. N Engl J Med 285: 1105–1111, 1971.
499. Maxwell PH, Wiesener MS, Chang GW, Clifford SC, Vaux EC,
476. Lumbroso D, Joseph V. Impaired acclimatization to chronic hyp-
Cockman ME, Wykoff CC, Pugh CW, Maher ER, Ratcliffe PJ.
oxia in adult male and female rats following neonatal hypoxia. The tumour suppressor protein VHL targets hypoxia-inducible fac-
Am J Physiol Regul Integr Comp Physiol 297: R421–R427, 2009. tors for oxygen-dependent proteolysis. Nature 399: 271–275, 1999.
477. Lundblad LK, Irvin CG, Adler A, Bates JH. A reevaluation of the 500. Mazza E Jr, Edelman NH, Neubauer JA. Hypoxic excitation in
validity of unrestrained plethysmography in mice. J Appl Physiol neurons cultured from the rostral ventrolateral medulla of the
93: 1198 –1207, 2002. neonatal rat. J Appl Physiol 88: 2319 –2329, 2000.
478. Lusina SJ, Kennedy PM, Inglis JT, McKenzie DC, Ayas NT, 501. Mazza E, Thakkar-Varia S, Tozzi CA, Neubauer JA. Expres-
Sheel AW. Long-term intermittent hypoxia increases sympathetic sion of heme oxygenase in the oxygen-sensing regions of the rostral
activity and chemosensitivity during acute hypoxia in humans. ventrolateral medulla. J Appl Physiol 91: 379 –385, 2001.
J Physiol 575: 961–970, 2006. 502. McCrimmon DR, Dekin MS, Mitchell GS. Glutamate, GABA and
479. Lutz PL. Mechanisms for anoxic survival in the vertebrate brain. serotonin in ventilatory control. In: Regulation of Breathing, edited
Annu Rev Physiol 54: 601– 618, 1992. by Dempsey JA and Pack AI. New York: Dekker, 1995, p. 151–218.
480. Macfarlane PM, Satriotomo I, Windelborn JA, Mitchell GS. 503. McCrimmon DR, Dempsey JA, Olson EB Jr. Effect of catechol-
NADPH oxidase activity is necessary for acute intermittent amine depletion on ventilatory control in unanesthetized normoxic
hypoxia-induced phrenic long-term facilitation. J Physiol 587: and hypoxic rats. J Appl Physiol 55: 522–528, 1983.
1931–1942, 2009. 504. McGuire M, MacDonald SM, Song G, Poon CS. Phrenic long-
481. Macfarlane PM, Wilkerson JE, Lovett-Barr MR, Mitchell GS. term facilitation is robust to hypercapnia and hypocapnia but not
Reactive oxygen species and respiratory plasticity following inter- hyperventilatory hypotension under PEEP. Respir Physiol Neuro-
mittent hypoxia. Respir Physiol Neurobiol 164: 263–271, 2008. biol 158: 107–111, 2007.
482. Madden CJ, Morrison SF. Hypoxic activation of arterial chemo- 505. McGuire M, Zhang Y, White DP, Ling L. Chronic intermittent
receptors inhibits sympathetic outflow to brown adipose tissue in hypoxia enhances ventilatory long-term facilitation in awake rats.
rats. J Physiol 566: 559 –573, 2005. J Appl Physiol 95: 1499 –1508, 2003.
483. Mahamed S, Cunningham DA, Duffin J. Changes in respiratory 506. McGuire M, Zhang Y, White DP, Ling L. Serotonin receptor
control after three hours of isocapnic hypoxia in humans. J Physiol subtypes required for ventilatory long-term facilitation and its en-
547: 271–281, 2003. hancement after chronic intermittent hypoxia in awake rats. Am J
484. Mahamed S, Duffin J. Repeated hypoxic exposures change respi- Physiol Regul Integr Comp Physiol 286: R334 –R341, 2004.
ratory chemoreflex control in humans. J Physiol 534: 595– 603, 507. Melton JE, Chae LO, Neubauer JA, Edelman NH. Extracellular
2001. potassium homeostasis in the cat medulla during progressive brain
485. Mahutte CK, Rebuck AS. Influence of rate of induction of hyp- hypoxia. J Appl Physiol 70: 1477–1482, 1991.
oxia on the ventilatory response. J Physiol 284: 219 –227, 1978. 508. Melton JE, Neubauer JA, Edelman NH. GABA antagonism re-
486. Malik MT, Peng YJ, Kline DD, Adhikary G, Prabhakar NR. verses hypoxic respiratory depression in the cat. J Appl Physiol 69:
Impaired ventilatory acclimatization to hypoxia in mice lacking the 1296 –1301, 1990.

Physiol Rev • VOL 90 • APRIL 2010 • www.prv.org


746 LUC J. TEPPEMA AND ALBERT DAHAN

509. Miller MJ, Haxhiu MA, Haxhiu-Poskurica B, Dreshaj IA, Di- 532. Mortola JP. Influence of temperature on metabolism and breath-
fiore JM, Martin RJ. Recurrent hypoxic exposure and reflex ing during mammalian ontogenesis. Respir Physiol Neurobiol 149:
responses during development in the piglet. Respir Physiol 123: 155–164, 2005.
51– 61, 2000. 533. Mulligan EM. Discharge properties of carrotid bodies: develop-
510. Miller MJ, Tenney SM. Hypoxia-induced tachypnea in carotid- mental aspects. In: Developmental Neurobiology of Breathing, ed-
deafferented cats. Respir Physiol 23: 31–39, 1975. ited by Haddad GG and Farber JP. New York: Dekker, 1990, p.
511. Miller P, Peers C, Kemp PJ. Polymodal regulation of hTREK1 by 321–340.
pH, arachidonic acid, and hypoxia: physiological impact in acidosis 534. Musch TI, Dempsey JA, Smith CA, Mitchell GS, Bateman NT.
and alkalosis. Am J Physiol Cell Physiol 286: C272–C282, 2004. Metabolic acids and [H⫹] regulation in brain tissue during acclima-
512. Millhorn DE. Stimulation of raphe obscurus nucleus causes long- tization to chronic hypoxia. J Appl Physiol 55: 1486 –1495, 1983.
term potentiation of phrenic nerve activity in cat. J Physiol 381: 535. Nagyova B, Dorrington KL, Robbins PA. Effect of low-dose
169 –179, 1986. enflurane on the ventilatory response to hypoxia in humans. Br J
513. Millhorn DE, Eldridge FL, Waldrop TG. Prolonged stimulation Anaesth 72: 509 –514, 1994.
of respiration by a new central neural mechanism. Respir Physiol 536. Nanduri J, Yuan G, Kumar GK, Semenza GL, Prabhakar NR.
41: 87–103, 1980. Transcriptional responses to intermittent hypoxia. Respir Physiol
514. Millhorn DE, Eldridge FL, Waldrop TG. Prolonged stimulation Neurobiol 164: 277–281, 2008.
of respiration by endogenous central serotonin. Respir Physiol 42: 537. Narkiewicz K, van de Borne PJ, Pesek CA, Dyken ME, Mon-
171–188, 1980. tano N, Somers VK. Selective potentiation of peripheral chemore-
515. Mitchell GS, Baker TL, Nanda SA, Fuller DD, Zabka AG, flex sensitivity in obstructive sleep apnea. Circulation 99: 1183–
Hodgeman BA, Bavis RW, Mack KJ, Olson EB Jr. Physiological 1189, 1999.
and genomic consequences of intermittent hypoxia: invited review: 538. Natsui T, Kuwana S. Central hypoxic depression of respiration
intermittent hypoxia and respiratory plasticity. J Appl Physiol 90: determined by perfusing a recipient cat’s carotid bodies with blood
2466 –2475, 2001. from a donor cat. Jpn J Physiol 44: 561–574, 1994.
516. Mizusawa A, Ogawa H, Kikuchi Y, Hida W, Kurosawa H, Ok- 539. Nattie E, Li A. Muscimol dialysis into the caudal aspect of the
abe S, Takishima T, Shirato K. In vivo release of glutamate in nucleus tractus solitarii of conscious rats inhibits chemoreception.
nucleus tractus solitarii of the rat during hypoxia. J Physiol 478: Respir Physiol Neurobiol 164: 394 – 400, 2008.
55– 66, 1994. 540. Neubauer JA, Melton JE, Edelman NH. Modulation of respira-
517. Mohan R, Duffin J. The effect of hypoxia on the ventilatory tion during brain hypoxia. J Appl Physiol 68: 441– 451, 1990.
response to carbon dioxide in man. Respir Physiol 108: 101–115, 541. Neubauer JA, Posner MA, Santiago TV, Edelman NH. Nalox-
1997. one reduces ventilatory depression of brain hypoxia. J Appl
518. Moon RY, Horne RS, Hauck FR. Sudden infant death syndrome. Physiol 63: 699 –706, 1987.
Lancet 370: 1578 –1587, 2007. 542. Neubauer JA, Santiago TV, Posner MA, Edelman NH. Ventral
519. Moore LG. Comparative human ventilatory adaptation to high medullary pH and ventilatory responses to hyperperfusion and
altitude. Respir Physiol 121: 257–276, 2000. hypoxia. J Appl Physiol 58: 1659 –1668, 1985.
520. Moore LG, Cymerman A, Huang SY, McCullough RE, 543. Neubauer JA, Simone A, Edelman NH. Role of brain lactic
McCullough RG, Rock PB, Young A, Young P, Weil JV, Reeves acidosis in hypoxic depression of respiration. J Appl Physiol 65:
JT. Propranolol blocks metabolic rate increase but not ventilatory 1324 –1331, 1988.
acclimatization to 4300 m. Respir Physiol 70: 195–204, 1987. 544. Neubauer JA, Sunderram J. Oxygen-sensing neurons in the cen-
521. Moore LG, McCullough RE, Weil JV. Increased HVR in preg- tral nervous system. J Appl Physiol 96: 367–374, 2004.
nancy: relationship to hormonal and metabolic changes. J Appl 545. Nieber K, Sevcik J, Illes P. Hypoxic changes in rat locus coer-
Physiol 62: 158 –163, 1987. uleus neurons in vitro. J Physiol 486: 33– 46, 1995.
522. Moore PJ, Ackland GL, Hanson MA. Unilateral cooling in the 546. Nielsen AM, Bisgard GE, Vidruk EH. Carotid chemoreceptor
region of locus coeruleus blocks the fall in respiratory output activity during acute and sustained hypoxia in goats. J Appl Physiol
during hypoxia in anaesthetized neonatal sheep. Exp Physiol 81: 65: 1796 –1802, 1988.
983–994, 1996. 547. Nielsen M, Smith H. Studies on the regulation of respiration in
523. Moore PJ, Parkes MJ, Nijhuis JG, Hanson MA. The incidence acute hypoxia: with an appendix on respiratory control during
of breathing movements of fetal sheep in normoxia and hypoxia prolonged hypoxia. Acta Physiol Scand 24: 293–313, 1952.
after peripheral chemodenervation and brain-stem transection. J 548. Nieto FJ, Young TB, Lind BK, Shahar E, Samet JM, Redline S,
Dev Physiol 11: 147–151, 1989. D’Agostino RB, Newman AB, Lebowitz MD, Pickering TG.
524. Moorthy SS, Sullivan TY, Fallon JH, Dierdorf SF, Radpour S, Association of sleep-disordered breathing, sleep apnea, and hyper-
DeAtley RE. Loss of hypoxic ventilatory response following bilat- tension in a large community-based study. Sleep Heart Health
eral neck dissection. Anesth Analg 76: 791–794, 1993. Study. JAMA 283: 1829 –1836, 2000.
525. Moreira TS, Takakura AC, Colombari E, West GH, Guyenet 549. Nieuwenhuijs D, Sarton E, Teppema L, Dahan A. Propofol for
PG. Inhibitory input from slowly adapting lung stretch receptors to monitored anesthesia care: implications on hypoxic control of
retrotrapezoid nucleus chemoreceptors. J Physiol 580: 285–300, cardiorespiratory responses. Anesthesiology 92: 46 –54, 2000.
2007. 550. Nieuwenhuijs D, Sarton E, Teppema LJ, Kruyt E, Olievier I,
526. Mortola JP. How newborn mammals cope with hypoxia. Respir van Kleef J, Dahan A. Respiratory sites of action of propofol:
Physiol 116: 95–103, 1999. absence of depression of peripheral chemoreflex loop by low-dose
527. Mortola JP, Morgan CA, Virgona V. Respiratory adaptation to propofol. Anesthesiology 95: 889 – 895, 2001.
chronic hypoxia in newborn rats. J Appl Physiol 61: 1329 –1336, 551. Nitsos I, Walker DW. Characterization of pontine neurons which
1986. respond to hypoxia in fetal sheep. Neurosci Lett 266: 33–36, 1999.
528. Mortola JP, Rezzonico R. Metabolic and ventilatory rates in 552. Nock ML, Difiore JM, Arko MK, Martin RJ. Relationship of the
newborn kittens during acute hypoxia. Respir Physiol 73: 55– 67, ventilatory response to hypoxia with neonatal apnea in preterm
1988. infants. J Pediatr 144: 291–295, 2004.
529. Mortola JP, Rezzonico R, Lanthier C. Ventilation and oxygen 553. Nolan PC, Waldrop TG. In vivo and in vitro responses of neurons
consumption during acute hypoxia in newborn mammals: a com- in the ventrolateral medulla to hypoxia. Brain Res 630: 101–114,
parative analysis. Respir Physiol 78: 31– 43, 1989. 1993.
530. Mortola JP, Saiki C. Ventilatory response to hypoxia in rats: 554. Nolan PC, Waldrop TG. In vitro responses of VLM neurons to
gender differences. Respir Physiol 106: 21–34, 1996. hypoxia after normobaric hypoxic acclimatization. Respir Physiol
531. Mortola JP. Implications of hypoxic hypometabolism during 105: 23–33, 1996.
mammalian ontogenesis. Respir Physiol Neurobiol 141: 345–356, 555. Nsegbe E, Wallen-Mackenzie A, Dauger S, Roux JC, Shvarev
2004. Y, Lagercrantz H, Perlmann T, Herlenius E. Congenital hy-

Physiol Rev • VOL 90 • APRIL 2010 • www.prv.org


THE VENTILATORY RESPONSE TO HYPOXIA IN MAMMALS 747

poventilation and impaired hypoxic response in Nurr1 mutant afferents in control of breathing. J Appl Physiol 85: 1299 –1306,
mice. J Physiol 556: 43–59, 2004. 1998.
556. Nucci TB, Branco LG, Gargaglioni LH. Nitric oxide pathway in 580. Pardal R, Lopez-Barneo J. Low glucose-sensing cells in the ca-
the nucleus raphe magnus modulates hypoxic ventilatory response rotid body. Nat Neurosci 5: 197–198, 2002.
but not anapyrexia in rats. Brain Res 1017: 39 – 45, 2004. 581. Pardal R, Ortega-Saenz P, Duran R, Lopez-Barneo J. Glia-like
557. Nucci TB, Branco LG, Gargaglioni LH. 5-HT1A, but not 5-HT2 and stem cells sustain physiologic neurogenesis in the adult mamma-
5-HT7, receptors in the nucleus raphe magnus modulate hypoxia- lian carotid body. Cell 131: 364 –377, 2007.
induced hyperpnoea. Acta Physiol Oxf 193: 403– 414, 2008. 582. Park SJ, Chun YS, Park KS, Kim SJ, Choi SO, Kim HL, Park
558. Nurse CA. Neurotransmission and neuromodulation in the chemo- JW. Identification of subdomains in NADPH oxidase-4 NOX-4 crit-
sensory carotid body. Auton Neurosci 120: 1–9, 2005. ical for the oxygen-dependent regulation of TASK-1 K⫹ channels.
559. Nurse CA, Zhang M. Synaptic mechanisms during re-innervation Am J Physiol Cell Physiol 2009.
of rat arterial chemoreceptors in co-culture. Comp Biochem 583. Pascual O, Morin-Surun MP, Barna B, avit-Saubie M,
Physiol A Mol Integr Physiol 130: 241–251, 2001. Pequignot JM, Champagnat J. Progesterone reverses the neuro-
560. Obeso A, Almaraz L, Gonzalez C. Effects of 2-deoxy-D-glucose nal responses to hypoxia in rat nucleus tractus solitarius in vitro.
on in vitro cat carotid body. Brain Res 371: 25–36, 1986. J Physiol 544: 511–520, 2002.
561. Obeso A, Gomez-Nino A, Gonzalez C. NADPH oxidase inhibition 584. Paulding WR, Schnell PO, Bauer AL, Striet JB, Nash JA,
does not interfere with low PO2 transduction in rat and rabbit CB Kuznetsova AV, Czyzyk-Krzeska MF. Regulation of gene ex-
chemoreceptor cells. Am J Physiol Cell Physiol 276: C593–C601, pression for neurotransmitters during adaptation to hypoxia in
1999. oxygen-sensitive neuroendocrine cells. Microsc Res Tech 59: 178 –
562. Ogawa H, Mizusawa A, Kikuchi Y, Hida W, Miki H, Shirato K. 187, 2002.
Nitric oxide as a retrograde messenger in the nucleus tractus 585. Pawar A, Peng YJ, Jacono FJ, Prabhakar NR. Comparative
solitarii of rats during hypoxia. J Physiol 486: 495–504, 1995. analysis of neonatal and adult rat carotid body responses to
563. Ogier M, Katz DM. Breathing dysfunction in Rett syndrome: chronic intermittent hypoxia. J Appl Physiol 104: 1287–1294, 2008.
understanding epigenetic regulation of the respiratory network. 586. Pawar A, Nanduri J, Yuan G, Khan SA, Wang N, Kumar GK,
Respir Physiol Neurobiol 164: 55– 63, 2008. Prabhakar NR. Reactive oxygen species-dependent endothelin
564. Ohtake PJ, Simakajornboon N, Fehniger MD, Xue YD, Gozal signaling is required for augmented hypoxic sensory response of
D. N-methyl-D-aspartate receptor expression in the nucleus tractus the neonatal carotid body by intermittent hypoxia. Am J Physiol
solitarii and maturation of hypoxic ventilatory response in the rat. Regul Integr Comp Physiol 296: R735–R742, 2009.
Am J Respir Crit Care Med 162: 1140 –1147, 2000. 587. Paxinos G, Watson C. The Rat Brain in Stereotaxic Coordinates.
565. Ohtake PJ, Torres JE, Gozal YM, Graff GR, Gozal D. NMDA San Diego, CA: Academic, 1997.
receptors mediate peripheral chemoreceptor afferent input in the 588. Pearce W. Hypoxic regulation of the fetal cerebral circulation.
conscious rat. J Appl Physiol 84: 853– 861, 1998. J Appl Physiol 100: 731–738, 2006.
566. Okubo S, Mortola JP. Long-term respiratory effects of neonatal 589. Pedersen ME, Dorrington KL, Robbins PA. Effects of haloper-
hypoxia in the rat. J Appl Physiol 64: 952–958, 1988. idol on ventilation during isocapnic hypoxia in humans. J Appl
567. Okubo S, Mortola JP. Control of ventilation in adult rats hypoxic Physiol 83: 1110 –1115, 1997.
in the neonatal period. Am J Physiol Regul Integr Comp Physiol 590. Pedersen ME, Dorrington KL, Robbins PA. Effects of dopamine
259: R836 –R841, 1990. and domperidone on ventilatory sensitivity to hypoxia after 8 h of
568. Olson EB Jr, Bohne CJ, Dwinell MR, Podolsky A, Vidruk EH, isocapnic hypoxia. J Appl Physiol 86: 222–229, 1999.
Fuller DD, Powell FL, Mitchel GS. Ventilatory long-term facili- 591. Pedersen ME, Fatemian M, Robbins PA. Identification of fast
tation in unanesthetized rats. J Appl Physiol 91: 709 –716, 2001. and slow ventilatory responses to carbon dioxide under hypoxic
569. Olson EB Jr, Dempsey JA. Rat as a model for humanlike venti- and hyperoxic conditions in humans. J Physiol 521: 273–287, 1999.
latory adaptation to chronic hypoxia. J Appl Physiol 44: 763–769, 592. Peers C. Interactions of chemostimuli at the single cell level:
1978. studies in a model system. Exp Physiol 89: 60 – 65, 2004.
570. Olson EB Jr, Vidruk EH, Dempsey JA. Carotid body excision 593. Peers C, Buckler KJ. Transduction of chemostimuli by the type I
significantly changes ventilatory control in awake rats. J Appl carotid body cell. J Membr Biol 144: 1–9, 1995.
Physiol 64: 666 – 671, 1988. 594. Peers C, Wyatt CN. The role of maxiK channels in carotid body
571. Olson EB Jr Vidruk EH, McCrimmon DR, Dempsey JA. Mono- chemotransduction. Respir Physiol Neurobiol 157: 75– 82, 2007.
amine neurotransmitter metabolism during acclimatization to hyp- 595. Peng Y, Yuan G, Overholt JL, Kumar GK, Prabhakar NR.
oxia in rats. Respir Physiol 54: 79 –96, 1983. Systemic and cellular responses to intermittent hypoxia: evidence
572. Olson LG, Saunders NA. Effect of a dopamine antagonist on for oxidative stress and mitochondrial dysfunction. Adv Exp Med
ventilation during sustained hypoxia in mice. J Appl Physiol 62: Biol 536: 559 –564, 2003.
1222–1226, 1987. 596. Peng YJ, Overholt JL, Kline D, Kumar GK, Prabhakar NR.
573. Ortega-Saenz P, Pardal R, Garcia-Fernandez M, Lopez-Bar- Induction of sensory long-term facilitation in the carotid body by
neo J. Rotenone selectively occludes sensitivity to hypoxia in rat intermittent hypoxia: implications for recurrent apneas. Proc Natl
carotid body glomus cells. J Physiol 548: 789 – 800, 2003. Acad Sci USA 100: 10073–10078, 2003.
574. Ortega-Saenz P, Pascual A, Gomez-Diaz R, Lopez-Barneo J. 597. Peng YJ, Prabhakar NR. Reactive oxygen species in the plasticity
Acute oxygen sensing in heme oxygenase-2 null mice. J Gen of respiratory behavior elicited by chronic intermittent hypoxia.
Physiol 128: 405– 411, 2006. J Appl Physiol 94: 2342–2349, 2003.
575. Ou LC, Tenney SM. Hypoxia and carbon dioxide as separate and 598. Peng YJ, Prabhakar NR. Effect of two paradigms of chronic
interactive depressants of ventilation. Respir Physiol 28: 347–358, intermittent hypoxia on carotid body sensory activity. J Appl
1976. Physiol 96: 1236 –1242, 2004.
576. Ou LC, Tenney SM. The role of brief hypocapnia in the ventilatory 599. Peng YJ, Rennison J, Prabhakar NR. Intermittent hypoxia aug-
response to CO2 with hypoxia. Respir Physiol 28: 333–346, 1976. ments carotid body and ventilatory response to hypoxia in neonatal
577. Painter R, Khamnei S, Robbins P. A mathematical model of the rat pups. J Appl Physiol 97: 2020 –2025, 2004.
human ventilatory response to isocapnic hypoxia. J Appl Physiol 600. Peng YJ, Yuan G, Ramakrishnan D, Sharma SD, Bosch-Marce
74: 2007–2015, 1993. M, Kumar GK, Semenza GL, Prabhakar NR. Heterozygous HIF-
578. Palmer LA, Doctor A, Chhabra P, Sheram ML, Laubach VE, 1alpha deficiency impairs carotid body-mediated systemic re-
Karlinsey MZ, Forbes MS, Macdonald T, Gaston B. S-nitroso- sponses and reactive oxygen species generation in mice exposed to
thiols signal hypoxia-mimetic vascular pathology. J Clin Invest intermittent hypoxia. J Physiol 577: 705–716, 2006.
117: 2592–2601, 2007. 601. Pepper DR, Landauer RC, Kumar P. Postnatal development of
579. Pan LG, Forster HV, Martino P, Strecker PJ, Beales J, Serra CO2-O2 interaction in the rat carotid body in vitro. J Physiol 485:
A, Lowry TF, Forster MM, Forster AL. Important role of carotid 531–541, 1995.

Physiol Rev • VOL 90 • APRIL 2010 • www.prv.org


748 LUC J. TEPPEMA AND ALBERT DAHAN

602. Perez H, Ruiz S. Medullary responses to chemoreceptor activa- 625. Read DJ, Henderson-Smart DJ. Regulation of breathing in the
tion are inhibited by locus coeruleus and nucleus raphe magnus. newborn during different behavioral states. Annu Rev Physiol 46:
Neuroreport 6: 1373–1376, 1995. 675– 685, 1984.
603. Perez-Garcia MT, Lopez-Lopez JR, Gonzalez C. Kvbeta1.2 sub- 626. Rebuck AS, Campbell EJ. A clinical method for assessing the
unit coexpression in HEK293 cells confers O2 sensitivity to Kv4.2 ventilatory response to hypoxia. Am Rev Respir Dis 109: 345–350,
but not to Shaker channels. J Gen Physiol 113: 897–907, 1999. 1974.
604. Peterson DD, Pack AI, Silage DA, Fishman AP. Effects of aging 627. Rebuck AS, Kangalee M, Pengelly LD, Campbell EJ. Correla-
on ventilatory and occlusion pressure responses to hypoxia and tion of ventilatory responses to hypoxia and hypercapnia. J Appl
hypercapnia. Am Rev Respir Dis 124: 387–391, 1981. Physiol 35: 173–177, 1973.
605. Pialoux V, Hanly PJ, Foster GE, Brugniaux JV, Beaudin AE, 628. Rebuck AS, Slutsky AS. Measurement of the ventilatory response
Hartmann SE, Pun M, Duggan CT, Poulin MJ. Effects of expo- to hypercapnia and hypoxia. In: Reulation of Breathing, Part II,
sure to intermittent hypoxia on oxidative stress and acute hypoxic edited by Hornbein TF. New York: Dekker, 1981, p. 745–772.
ventilatory response in humans. Am J Respir Crit Care Med 180: 629. Reeves JT, McCullough RE, Moore LG, Cymerman A, Weil JV.
1002–1009, 2009. Sea-level PCO2 relates to ventilatory acclimatization at 4,300 m.
606. Piruat JI, Lopez-Barneo J. Oxygen tension regulates mitochon- J Appl Physiol 75: 1117–1122, 1993.
drial DNA-encoded complex I gene expression. J Biol Chem 280: 630. Reeves SR, Gozal D. Platelet-activating factor receptor modulates
42676 – 42684, 2005. respiratory adaptation to long-term intermittent hypoxia in mice.
607. Piruat JI, Pintado CO, Ortega-Saenz P, Roche M, Lopez-Bar- Am J Physiol Regul Integr Comp Physiol 287: R369 –R374, 2004.
neo J. The mitochondrial SDHD gene is required for early embry- 631. Reeves SR, Gozal D. Respiratory and metabolic responses to
ogenesis, and its partial deficiency results in persistent carotid early postnatal chronic intermittent hypoxia and sustained hypoxia
body glomus cell activation with full responsiveness to hypoxia. in the developing rat. Pediatr Res 60: 680 – 686, 2006.
Mol Cell Biol 24: 10933–10940, 2004. 632. Reeves SR, Gozal D. Developmental plasticity of respiratory con-
608. Pokorski M, Marczak M. Ascorbic acid enhances hypoxic venti- trol following intermittent hypoxia. Respir Physiol Neurobiol 149:
latory reactivity in elderly subjects. J Int Med Res 31: 448 – 457, 301–311, 2005.
2003. 633. Reeves SR, Gozal D. Changes in ventilatory adaptations associ-
609. Pokorski M, Marczak M. Ventilatory response to hypoxia in ated with long-term intermittent hypoxia across the age spectrum
elderly women. Ann Hum Biol 30: 53– 64, 2003. in the rat. Respir Physiol Neurobiol 150: 135–143, 2006.
610. Pokorski M, Walski M, Dymecka A, Marczak M. The aging 634. Reeves SR, Gozal E, Guo SZ, Sachleben LR Jr, Brittian KR,
carotid body. J Physiol Pharmacol 55 Suppl 3: 107–113, 2004. Lipton AJ, Gozal D. Effect of long-term intermittent and sus-
611. Porzionato A, Macchi V, Parenti A, Matturri L, De Caro R. tained hypoxia on hypoxic ventilatory and metabolic responses in
Peripheral chemoreceptors: postnatal development and cytochem- the adult rat. J Appl Physiol 95: 1767–1774, 2003.
ical findings in Sudden Infant Death Syndrome. Histol Histopathol 635. Reeves SR, Mitchell GS, Gozal D. Early postnatal chronic inter-
mittent hypoxia modifies hypoxic respiratory responses and long-
23: 351–365, 2008.
term phrenic facilitation in adult rats. Am J Physiol Regul Integr
612. Powell FL. Lake Louise consensus methods for measuring the
Comp Physiol 290: R1664 –R1671, 2006.
hypoxic ventilatory response. Adv Exp Med Biol 588: 271–276,
636. Regensteiner JG, Woodard WD, Hagerman DD, Weil JV, Pick-
2006.
ett CK, Bender PR, Moore LG. Combined effects of female
613. Powell FL, Dwinell MR, Aaron EA. Measuring ventilatory accli-
hormones and metabolic rate on ventilatory drives in women.
matization to hypoxia: comparative aspects. Respir Physiol 122:
J Appl Physiol 66: 808 – 813, 1989.
271–284, 2000.
637. Rehan V, Haider AZ, Alvaro RE, Nowaczyk B, Cates DB, Kwi-
614. Powell FL, Fu Z. HIF-1 and ventilatory acclimatization to chronic
atkowski K, Rigatto H. The biphasic ventilatory response to
hypoxia. Respir Physiol Neurobiol 164: 282–287, 2008. hypoxia in preterm infants is not due to a decrease in metabolism.
615. Powell FL, Huey KA, Dwinell MR. Central nervous system mech- Pediatr Pulmonol 22: 287–294, 1996.
anisms of ventilatory acclimatization to hypoxia. Respir Physiol 638. Reid SG, Powell FL. Effects of chronic hypoxia on MK-801-
121: 223–236, 2000. induced changes in the acute hypoxic ventilatory response. J Appl
616. Prabhakar NR, Dinerman JL, Agani FH, Snyder SH. Carbon Physiol 99: 2108 –2114, 2005.
monoxide: a role in carotid body chemoreception. Proc Natl Acad 639. Ren X, Dorrington KL, Maxwell PH, Robbins PA. Effects of
Sci USA 92: 1994 –1997, 1995. desferrioxamine on serum erythropoietin and ventilatory sensitiv-
617. Prabhakar NR, Peng YJ, Kumar GK, Pawar A. Altered carotid ity to hypoxia in humans. J Appl Physiol 89: 680 – 686, 2000.
body function by intermittent hypoxia in neonates and adults: 640. Ren X, Dorrington KL, Robbins PA. Respiratory control in hu-
relevance to recurrent apneas. Respir Physiol Neurobiol 157: 148 – mans after 8 h of lowered arterial PO2, hemodilution, or carboxy-
153, 2007. hemoglobinemia. J Appl Physiol 90: 1189 –1195, 2001.
618. Prabhakar NR, Pieramici SF, Premkumar DR, Kumar GK, 641. Ren X, Robbins PA. Ventilatory responses to hypercapnia and
Kalaria RN. Activation of nitric oxide synthase gene expression by hypoxia after 6 h passive hyperventilation in humans. J Physiol
hypoxia in central and peripheral neurons. Brain Res 43: 341–346, 514: 885– 894, 1999.
1996. 642. Rey S, Corthorn J, Chacon C, Iturriaga R. Expression and
619. Prabhakar NR. NO and CO as second messengers in oxygen immunolocalization of endothelin peptides and its receptors, ETA
sensing in the carotid body. Respir Physiol 115: 161–168, 1999. and ETB, in the carotid body exposed to chronic intermittent
620. Prabhakar NR. Oxygen sensing by the carotid body chemorecep- hypoxia. J Histochem Cytochem 55: 167–174, 2007.
tors. J Appl Physiol 88: 2287–2295, 2000. 643. Rey S, Del Rio R, Iturriaga R. Contribution of endothelin-1 to the
621. Prabhakar NR. O2 sensing at the mammalian carotid body: why enhanced carotid body chemosensory responses induced by
multiple O2 sensors and multiple transmitters? Exp Physiol 91: chronic intermittent hypoxia. Brain Res 1086: 152–159, 2006.
17–23, 2006. 644. Reyes EP, Fernandez R, Larrain C, Zapata P. Carotid body
622. Prieto-Lloret J, Caceres AI, Rigual R, Obeso A, Rocher A, chemosensory activity and ventilatory chemoreflexes in cats per-
Bustamante R, Castaneda J, Lopez-Lopez JR, Perez-Garcia T, sist after combined cholinergic-purinergic block. Respir Physiol
Agapito T, Gonzalez C. Effects of perinatal hyperoxia on carotid Neurobiol 156: 23–32, 2007.
body chemoreceptor activity in vitro. Adv Exp Med Biol 536: 517– 645. Reyes EP, Fernandez R, Larrain C, Zapata P. Effects of com-
524, 2003. bined cholinergic-purinergic block upon cat carotid body chemo-
623. Querido JS, Godwin JB, Sheel AW. Intermittent hypoxia re- receptors in vitro. Respir Physiol Neurobiol 156: 17–22, 2007.
duces cerebrovascular sensitivity to isocapnic hypoxia in humans. 646. Rezzonico R, Mortola JP. Respiratory adaptation to chronic hy-
Respir Physiol Neurobiol 161: 1–9, 2008. percapnia in newborn rats. J Appl Physiol 67: 311–315, 1989.
624. Read DJ. A clinical method for assessing the ventilatory response 647. Richardson BS, Carmichael L, Homan J, Patrick JE. Electro-
to carbon dioxide. Australas Ann Med 16: 20 –32, 1967. cortical activity, electroocular activity, and breathing movements

Physiol Rev • VOL 90 • APRIL 2010 • www.prv.org


THE VENTILATORY RESPONSE TO HYPOXIA IN MAMMALS 749

in fetal sheep with prolonged and graded hypoxemia. Am J Obstet and DBA/2J mouse strains. Am J Respir Crit Care Med 168: 1520 –
Gynecol 167: 553–558, 1992. 1527, 2003.
648. Richardson HL, Parslow PM, Walker AM, Harding R, Horne 670. Rurak DW, Gruber NC. Increased oxygen consumption associ-
RS. Maturation of the initial ventilatory response to hypoxia in ated with breathing activity in fetal lambs. J Appl Physiol 54:
sleeping infants. J Sleep Res 16: 117–127, 2007. 701–707, 1983.
649. Richter DW, Schmidt-Garcon P, Pierrefiche O, Bischoff AM, 671. Ryan JW, Waldrop TG. Hypoxia sensitive neurons in the caudal
Lalley PM. Neurotransmitters and neuromodulators controlling hypothalamus project to the periaqueductal gray. Respir Physiol
the hypoxic respiratory response in anaesthetized cats. J Physiol 100: 185–194, 1995.
514: 567–578, 1999. 672. Ryan ML, Hedrick MS, Pizarro J, Bisgard GE. Effects of carotid
650. Riedstra JW. Influence of central and peripheral PCO2 pH on the body sympathetic denervation on ventilatory acclimatization to
ventilatory response to hypoxic chemoreceptor stimulation. Acta hypoxia in the goat. Respir Physiol 99: 215–224, 1995.
Physiol Pharmacol Neerl 12: 407– 452, 1963. 673. Saha S, Batten TF, McWilliam PN. Glutamate, gamma-aminobu-
651. Rigatto H. Control of ventilation in the newborn. Annu Rev tyric acid and tachykinin-immunoreactive synapses in the cat nu-
Physiol 46: 661– 674, 1984. cleus tractus solitarii. J Neurocytol 24: 55–74, 1995.
652. Rigatto H, Brady JP. Periodic breathing and apnea in preterm 674. Sahn SA, Zwillich CW, Dick N, McCullough RE, Lakshmi-
infants. II. Hypoxia as a primary event. Pediatrics 50: 219 –228, narayan S, Weil JV. Variability of ventilatory responses to hyp-
1972. oxia and hypercapnia. J Appl Physiol 43: 1019 –1025, 1977.
653. Rigatto H, Wiebe C, Rigatto C, Lee DS, Cates D. Ventilatory 675. Saiki C, Matsuoka T, Mortola JP. Metabolic-ventilatory interac-
response to hypoxia in unanesthetized newborn kittens. J Appl tion in conscious rats: effect of hypoxia and ambient temperature.
Physiol 64: 2544 –2551, 1988. J Appl Physiol 76: 1594 –1599, 1994.
654. Ritchie K. The fetal response to changes in the composition of 676. Saiki C, Mortola JP. Ventilatory control in infant rats after daily
maternal inspired air in human pregnancy. Semin Perinatol 4: episodes of anoxia. Pediatr Res 35: 490 – 493, 1994.
295–299, 1980. 677. Sajkov D, Neill A, Saunders NA, McEvoy RD. Comparison of
655. Robbins PA. The ventilatory response of the human respiratory effects of sustained isocapnic hypoxia on ventilation in men and
system to sine waves of alveolar carbon dioxide and hypoxia. women. J Appl Physiol 83: 599 – 607, 1997.
J Physiol 350: 461– 474, 1984. 678. Sanders KA, Sundar KM, He L, Dinger B, Fidone S, Hoidal JR.
656. Robbins PA. Evidence for interaction between the contributions Role of components of the phagocytic NADPH oxidase in oxygen
to ventilation from the central and peripheral chemoreceptors in sensing. J Appl Physiol 93: 1357–1364, 2002.
man. J Physiol 401: 503–518, 1988. 679. Sanz-Alfayate G, Obeso A, Agapito MT, Gonzalez C. Reduced
657. Robbins PA. Hypoxic ventilatory decline: site of action. J Appl to oxidized glutathione ratios and oxygen sensing in calf and rabbit
Physiol 79: 373–374, 1995. carotid body chemoreceptor cells. J Physiol 537: 209 –220, 2001.
658. Robbins PA, Zhang S. Ventilatory response to hypoxia in humans. 680. Sarton E, Teppema L, Dahan A. Sex differences in morphine-
Respir Physiol 115: 333–335, 1999. induced ventilatory depression reside within the peripheral che-
659. Robbins PA. Role of the peripheral chemoreflex in the early stages moreflex loop. Anesthesiology 90: 1329 –1338, 1999.
of ventilatory acclimatization to altitude. Respir Physiol Neurobiol 681. Sarton E, van der Wal M, Nieuwenhuijs D, Teppema L, Ro-
158: 237–242, 2007. botham JL, Dahan A. Sevoflurane-induced reduction of hypoxic
660. Roberts CA, Corfield DR, Murphy K, Calder NA, Hanson MA, drive is sex-independent. Anesthesiology 90: 1288 –1293, 1999.
Adams L, Guz A. Modulation by “central” PCO2 of the response to 682. Sato M, Severinghaus JW, Bickler P. Time course of augmen-
carotid body stimulation in man. Respir Physiol 102: 149 –161, tation and depression of hypoxic ventilatory responses at altitude.
1995. J Appl Physiol 77: 313–316, 1994.
661. Rocha S. Gene regulation under low oxygen: holding your breath 683. Sato M, Severinghaus JW, Powell FL, Xu FD, Spellman MJ Jr.
for transcription. Trends Biochem Sci 32: 389 –397, 2007. Augmented hypoxic ventilatory response in men at altitude. J Appl
662. Rocher A, Geijo-Barrientos E, Caceres AI, Rigual R, Gonzalez Physiol 73: 101–107, 1992.
C, Almaraz L. Role of voltage-dependent calcium channels in 684. Schmitt P, Pequignot J, Garcia C, Pujol JF, Pequignot JM.
stimulus-secretion coupling in rabbit carotid body chemoreceptor Regional specificity of the long-term regulation of tyrosine hydrox-
cells. J Physiol 562: 407– 420, 2005. ylase in some catecholaminergic rat brainstem areas. I. Influence of
663. Rodman JR, Curran AK, Henderson KS, Dempsey JA, Smith long-term hypoxia. Brain Res 611: 53– 60, 1993.
CA. Carotid body denervation in dogs: eupnea and the ventilatory 685. Schmitt P, Soulier V, Pequignot JM, Pujol JF, Avit-Saubie M.
response to hyperoxic hypercapnia. J Appl Physiol 91: 328 –335, Ventilatory acclimatization to chronic hypoxia: relationship to nor-
2001. adrenaline metabolism in the rat solitary complex. J Physiol 477:
664. Roux JC, Pequignot JM, Dumas S, Pascual O, Ghilini G, 331–337, 1994.
Pequignot J, Mallet J, Denavit-Saubi M. O2-sensing after ca- 686. Schoene RB, Roach RC, Hackett PH, Sutton JR, Cymerman A,
rotid chemodenervation: hypoxic ventilatory responsiveness and Houston CS. Operation Everest II: ventilatory adaptation during
upregulation of tyrosine hydroxylase mRNA in brainstem cat- gradual decompression to extreme altitude. Med Sci Sports Exerc
echolaminergic cells. Eur J Neurosci 12: 3181–3190, 2000. 22: 804 – 810, 1990.
665. Roux JC, Peyronnet J, Pascual O, Dalmaz Y, Pequignot JM. 687. Schuitmaker JJ, Berkenbosch A, De Goede J, Olievier CN.
Ventilatory and central neurochemical reorganisation of O2 che- Effects of CO2 and H⫹ on the ventilatory response to peripheral
moreflex after carotid sinus nerve transection in rat. J Physiol 522: chemoreceptor stimulation. Respir Physiol 64: 69 –79, 1986.
493–501, 2000. 688. Schwenke DO, Pearson JT, Kangawa K, Shirai M. Does central
666. Roy A, Mokashi A, Rozanov C, Daudu PA, Lahiri S. Reduced nitric oxide chronically modulate the acute hypoxic ventilatory
glutathione, dithiothreitol and cytochrome P-450 inhibitors do not response in conscious rats? Acta Physiol Oxf 186: 309 –318, 2006.
influence hypoxic chemosensory responses in the rat carotid body. 689. Semenza GL. Regulation of mammalian O2 homeostasis by
Brain Res 889: 131–137, 2001. hypoxia-inducible factor 1. Annu Rev Cell Dev Biol 15: 551–578,
667. Roy A, Rozanov C, Mokashi A, Daudu P, Al mehdi AB, Shams 1999.
H, Lahiri S. Mice lacking in gp91 phox subunit of NADPH oxidase 690. Semenza GL. HIF-1: mediator of physiological and pathophysio-
showed glomus cell [Ca2⫹]i and respiratory responses to hypoxia. logical responses to hypoxia. J Appl Physiol 88: 1474 –1480, 2000.
Brain Res 872: 188 –193, 2000. 691. Semenza GL. O2-regulated gene expression: transcriptional con-
668. Roy A, Rozanov C, Mokashi A, Lahiri S. PO2-PCO2 stimulus trol of cardiorespiratory physiology by HIF-1. J Appl Physiol 96:
interaction in [Ca2⫹]i and CSN activity in the adult rat carotid body. 1173–1177, 2004.
Respir Physiol 122: 15–26, 2000. 692. Semenza GL. Regulation of oxygen homeostasis by hypoxia-in-
669. Rubin AE, Polotsky VY, Balbir A, Krishnan JA, Schwartz AR, ducible factor 1. Physiology 24: 97–106, 2009.
Smith PL, Fitzgerald RS, Tankersley CG, Shirahata M, 693. Semenza GL, Shimoda LA, Prabhakar NR. Regulation of gene
O’Donnell CP. Differences in sleep-induced hypoxia between A/J expression by HIF-1. Novartis Found Symp 272: 2– 8, 2006.

Physiol Rev • VOL 90 • APRIL 2010 • www.prv.org


750 LUC J. TEPPEMA AND ALBERT DAHAN

694. Semenza GL. Life with Oxygen. Science 318: 62– 64, 2007. 715. Soliz J, Gassmann M, Joseph V. Soluble erythropoietin receptor
695. Serebrovskaya TV. Intermittent hypoxia research in the former is present in the mouse brain and is required for the ventilatory
Soviet Union and the Commonwealth of Independent States: his- acclimatization to hypoxia. J Physiol 583: 329 –336, 2007.
tory and review of the concept and selected applications. High Alt 716. Soliz J, Joseph V, Soulage C, Becskei C, Vogel J, Pequignot
Med Biol 3: 205–221, 2002. JM, Ogunshola O, Gassmann M. Erythropoietin regulates
696. Severinghaus J. Draft: 2006 Lake Louise HVR methods. http:// hypoxic ventilation in mice by interacting with brainstem and
www.hypoxia.net/forum/HVR.pdf. 2006. carotid bodies. J Physiol 568: 559 –571, 2005.
697. Severinghaus JW. Proposed standard determination of ventila- 717. Soliz J, Thomsen JJ, Soulage C, Lundby C, Gassmann M.
tory responses to hypoxia and hypercapnia in man. Chest 70: Sex-dependent regulation of hypoxic ventilation in mice and hu-
129 –131, 1976. mans is mediated by erythropoietin. Am J Physiol Regul Integr
698. Severinghaus JW. Fire-air and dephlogistication. Revisionisms of Comp Physiol 296: R1837–R1846, 2009.
oxygen’s discovery. Adv Exp Med Biol 543: 7–19, 2003. 718. Solomon IC, Edelman NH, Neubauer JA. Pre-Botzinger com-
699. Sharma SD, Raghuraman G, Lee MS, Prabhakar NR, Kumar plex functions as a central hypoxia chemosensor for respiration in
GK. Intermittent hypoxia activates peptidylglycine ␣-amidating vivo. J Neurophysiol 83: 2854 –2868, 2000.
monooxygenase in rat brain stem via reactive oxygen species- 719. Somogyi RB, Preiss D, Vesely A, Fisher JA, Duffin J. Changes
mediated proteolytic processing. J Appl Physiol 106: 12–19, 2009. in respiratory control after 5 days at altitude. Respir Physiol Neu-
700. Shatilo VB, Korkushko OV, Ischuk VA, Downey HF, Sere- robiol 145: 41–52, 2005.
brovskaya TV. Effects of intermittent hypoxia training on exercise 720. Sorensen SC, Severinghaus JW. Respiratory sensitivity to acute
performance, hemodynamics, and ventilation in healthy senior hypoxia in man born at sea level living at high altitude. J Appl
men. High Alt Med Biol 9: 43–52, 2008. Physiol 25: 211–216, 1968.
701. Shirahata M, Balbir A, Otsubo T, Fitzgerald RS. Role of ace- 721. Soulier V, Dalmaz Y, Cottet-Emard JM, Lagercrantz H,
tylcholine in neurotransmission of the carotid body. Respir Physiol Pequignot JM. Long-term influence of neonatal hypoxia on cate-
Neurobiol 157: 93–105, 2007. cholamine activity in carotid bodies and brainstem cell groups of
702. Simakajornboon N, Kuptanon T. Maturational changes in neu- the rat. J Physiol 498: 523–530, 1997.
romodulation of central pathways underlying hypoxic ventilatory 722. Soulier V, Gestreau C, Borghini N, Dalmaz Y, Cottet-Emard
response. Respir Physiol Neurobiol 149: 273–286, 2005. JM, Pequignot JM. Peripheral chemosensitivity and central inte-
703. Sladek M, Parker RA, Grogaard JB, Sundell HW. Long-lasting gration: neuroplasticity of catecholaminergic cells under hypoxia.
effect of prolonged hypoxemia after birth on the immediate venti- Comp Biochem Physiol A Physiol 118: 1–7, 1997.
latory response to changes in arterial partial pressure of oxygen in 723. Sovik S, Lossius K. Development of ventilatory response to tran-
young lambs. Pediatr Res 34: 821– 828, 1993. sient hypercapnia and hypercapnic hypoxia in term infants. Pediatr
704. Slegel P, Kitagawa H, Maguire MH. Determination of adenosine Res 55: 302–309, 2004.
in fetal perfusates of human placental cotyledons using fluores- 724. St Croix CM, Cunningham DA, Paterson DH. Nature of the
cence derivatization and reversed-phase high-performance liquid interaction between central and peripheral chemoreceptor drives
chromatography. Anal Biochem 171: 124 –134, 1988. in human subjects. Can J Physiol Pharmacol 74: 640 – 646, 1996.
705. Slessarev M, Han J, Mardimae A, Prisman E, Preiss D, 725. Stark RI, Daniel SS, Garland M, Jaille-Marti JC, Kim YI,
Volgyesi G, Ansel C, Duffin J, Fisher JA. Prospective targeting Leung K, Myers MM, Tropper PJ. Fetal hiccups in the baboon.
and control of end-tidal CO2 and O2 concentrations. J Physiol 581: Am J Physiol Regul Integr Comp Physiol 267: R1479 –R1487, 1994.
1207–1219, 2007. 726. Stea A, Jackson A, Macintyre L, Nurse CA. Long-term modu-
706. Smith CA, Bisgard GE, Nielsen AM, Daristotle L, Kressin NA, lation of inward currents in O2 chemoreceptors by chronic hypoxia
Forster HV, Dempsey JA. Carotid bodies are required for venti- and cyclic AMP in vitro. J Neurosci 15: 2192–2202, 1995.
latory acclimatization to chronic hypoxia. J Appl Physiol 60: 1003– 727. Stea A, Jackson A, Nurse CA. Hypoxia and N6,O2⬘-dibutyrylad-
1010, 1986. enosine 3⬘,5⬘-cyclic monophosphate, but not nerve growth factor,
707. Smith CA, Harms CA, Henderson KS, Dempsey JA. Ventilatory induce Na⫹ channels and hypertrophy in chromaffin-like arterial
effects of specific carotid body hypocapnia and hypoxia in awake chemoreceptors. Proc Natl Acad Sci USA 89: 9469 –9473, 1992.
dogs. J Appl Physiol 82: 791–798, 1997. 728. Steinback CD, Poulin MJ. Ventilatory responses to isocapnic and
708. Smith CA, Jameson LC, Mitchell GS, Musch TI, Dempsey JA. poikilocapnic hypoxia in humans. Respir Physiol Neurobiol 155:
Central-peripheral chemoreceptor interaction in awake cerebrospi- 104 –113, 2007.
nal fluid-perfused goats. J Appl Physiol 56: 1541–1549, 1984. 729. Stephenson R, Liao KS, Hamrahi H, Horner RL. Circadian
709. Smith DW, Buller KM, Day TA. Role of ventrolateral medulla rhythms and sleep have additive effects on respiration in the rat.
catecholamine cells in hypothalamic neuroendocrine cell re- J Physiol 536: 225–235, 2001.
sponses to systemic hypoxia. J Neurosci 15: 7979 –7988, 1995. 730. Sterni LM, Bamford OS, Tomares SM, Montrose MH, Carroll
710. Smith PG, Mills E. Restoration of reflex ventilatory response to JL. Developmental changes in intracellular Ca2⫹ response of ca-
hypoxia after removal of carotid bodies in the cat. Neuroscience 5: rotid chemoreceptor cells to hypoxia. Am J Physiol Lung Cell Mol
573–580, 1980. Physiol 268: L801–L808, 1995.
711. Smith TG, Balanos GM, Croft QP, Talbot NP, Dorrington KL, 731. Sterni LM, Bamford OS, Wasicko MJ, Carroll JL. Chronic
Ratcliffe PJ, Robbins PA. The increase in pulmonary arterial hypoxia abolished the postnatal increase in carotid body type I cell
pressure caused by hypoxia depends on iron status. J Physiol 586: sensitivity to hypoxia. Am J Physiol Lung Cell Mol Physiol 277:
5999 – 6005, 2008. L645–L652, 1999.
712. Smith TG, Brooks JT, Balanos GM, Lappin TR, Layton DM, 732. Stornetta RL. Identification of neurotransmitters and co-localiza-
Leedham DL, Liu C, Maxwell PH, McMullin MF, McNamara tion of transmitters in brainstem respiratory neurons. Respir
CJ, Percy MJ, Pugh CW, Ratcliffe PJ, Talbot NP, Treacy M, Physiol Neurobiol 164: 18 –27, 2008.
Robbins PA. Mutation of von Hippel-Lindau tumour suppressor 733. Streller T, Huckstorf C, Pfeiffer CAH. Unusual cytochrome
and human cardiopulmonary physiology. PLoS Med 3: e290, 2006. a592 with low PO2 affinity correlates as putative oxygen sensor with
713. Smith TG, Talbot NP, Privat C, Rivera-Ch M, Nickol AH, rat carotid body chemoreceptor discharge. FASEB J 16: 1277–1279,
Ratcliffe PJ, Dorrington KL, Leon-Velarde F, Robbins PA. 2002.
Effects of iron supplementation and depletion on hypoxic pulmo- 734. Strohl KP. Inheritance and ventilatory behavior in animal models.
nary hypertension: two randomized controlled trials. JAMA 302: In: Pharmacology and Pathophysiology of the Control of Breath-
1444 –1450, 2009. ing, edited by Ward DS, Dahan A, and Teppema LJ. New York:
714. Smith WD, Poulin MJ, Paterson DH, Cunningham DA. Dy- Taylor & Francis, 2005, p. 261–291.
namic ventilatory response to acute isocapnic hypoxia in septua- 735. Strohl KP, Thomas AJ. Neonatal conditioning for adult respira-
genarians. Exp Physiol 86: 117–126, 2001. tory behavior. Respir Physiol 110: 269 –275, 1997.

Physiol Rev • VOL 90 • APRIL 2010 • www.prv.org


THE VENTILATORY RESPONSE TO HYPOXIA IN MAMMALS 751

736. Strohl KP, Thomas AJ, St Jean P, Schlenker EH, Koletsky RJ, 759. Tankersley CG, Elston RC, Schnell AH. Genetic determinants
Schork NJ. Ventilation and metabolism among rat strains. J Appl of acute hypoxic ventilation: patterns of inheritance in mice. J Appl
Physiol 82: 317–323, 1997. Physiol 88: 2310 –2318, 2000.
737. Subramanian S, Dostal J, Erokwu B, Han F, Dick TE, Strohl 760. Tankersley CG, Fitzgerald RS, Kleeberger SR. Differential con-
KP. Domperidone and ventilatory behavior: Sprague-Dawley ver- trol of ventilation among inbred strains of mice. Am J Physiol
sus Brown Norway rats. Respir Physiol Neurobiol 155: 22–28, 2007. Regul Integr Comp Physiol 267: R1371–R1377, 1994.
738. Subramanian S, Erokwu B, Han F, Dick TE, Strohl KP. 761. Tankersley CG, Broman KW. Interactions in hypoxic and hyper-
L-NAME differentially alters ventilatory behavior in Sprague-Daw- capnic breathing are genetically linked to mouse chromosomes 1
ley and Brown Norway rats. J Appl Physiol 93: 984 –989, 2002. and 5. J Appl Physiol 97: 77– 84, 2004.
739. Subramanian S, Han F, Erokwu BO, Dick TE, Strohl KP. Do 762. Tansley JG, Clar C, Pedersen ME, Robbins PA. Human venti-
genetic factors influence the Dejours phenomenon? Adv Exp Med latory response to acute hyperoxia during and after 8 h of both
Biol 499: 209 –214, 2001. isocapnic and poikilocapnic hypoxia. J Appl Physiol 82: 513–519,
740. Suguihara C, Hehre D, Bancalari E. Effect of dopamine on 1997.
hypoxic ventilatory response of sedated piglets with intact and 763. Tansley JG, Fatemian M, Howard LS, Poulin MJ, Robbins PA.
denervated carotid bodies. J Appl Physiol 77: 285–289, 1994. Changes in respiratory control during and after 48 h of isocapnic
741. Sun MK, Jeske IT, Reis DJ. Cyanide excites medullary sympa- and poikilocapnic hypoxia in humans. J Appl Physiol 85: 2125–
thoexcitatory neurons in rats. Am J Physiol Regul Integr Comp 2134, 1998.
Physiol 262: R182–R189, 1992. 764. Tansley JG, Pedersen ME, Clar C, Robbins PA. Human venti-
742. Sun MK, Reis DJ. Central neural mechanisms mediating excita- latory response to 8 h of euoxic hypercapnia. J Appl Physiol 84:
tion of sympathetic neurons by hypoxia. Prog Neurobiol 44: 197– 431– 434, 1998.
219, 1994. 765. Tatsumi K, Hannhart B, Pickett CK, Weil JV, Moore LG.
743. Sun MK, Reis DJ. Dopamine or transmitter release from rat Influences of gender and sex hormones on hypoxic ventilatory
carotid body may not be essential to hypoxic chemoreception. response in cats. J Appl Physiol 71: 1746 –1751, 1991.
Am J Physiol Regul Integr Comp Physiol 267: R1632–R1639, 1994. 766. Tatsumi K, Hannhart B, Pickett CK, Weil JV, Moore LG.
744. Sun MK, Reis DJ. Hypoxia selectively excites vasomotor neurons Effects of testosterone on hypoxic ventilatory and carotid body
of rostral ventrolateral medulla in rats. Am J Physiol Regul Integr neural responsiveness. Am J Respir Crit Care Med 149: 1248 –1253,
Comp Physiol 266: R245–R256, 1994. 1994.
745. Sun MK, Reis DJ. Hypoxia-activated Ca2⫹ currents in pacemaker 767. Tatsumi K, Pickett CK, Jacoby CR, Weil JV, Moore LG. Role of
neurones of rat rostral ventrolateral medulla in vitro. J Physiol 476: endogenous female hormones in hypoxic chemosensitivity. J Appl
101–116, 1994. Physiol 83: 1706 –1710, 1997.
746. Suzuki A, Nishimura M, Yamamoto H, Miyamoto K, Kishi F, 768. Tatsumi K, Pickett CK, Weil JV. Attenuated carotid body
Kawakami Y. No effect of brain blood flow on ventilatory depres- hypoxic sensitivity after prolonged hypoxic exposure. J Appl
sion during sustained hypoxia. J Appl Physiol 66: 1674 –1678, 1989.
Physiol 70: 748 –755, 1991.
747. Swanson GD, Bellville JW. Step changes in end-tidal CO2: meth-
769. Tatsumi K, Pickett CK, Weil JV. Effects of haloperidol and
ods and implications. J Appl Physiol 39: 377–385, 1975.
domperidone on ventilatory roll off during sustained hypoxia in
748. Swanson GD, Carpenter TM Jr, Snider DE, Bellville JW. An
cats. J Appl Physiol 72: 1945–1952, 1992.
on-line hybrid computing system for dynamic respiratory response
770. Tatsumi K, Pickett CK, Weil JV. Decreased carotid body
studies. Comput Biomed Res 4: 205–215, 1971.
hypoxic sensitivity in chronic hypoxia: role of dopamine. Respir
749. Swenson ER. Carbonic anhydrase inhibitors and hypoxic pulmo-
Physiol 101: 47–57, 1995.
nary vasoconstriction. Respir Physiol Neurobiol 151: 209 –216,
771. Tatsumi K, Pickett CK, Weil JV. Possible role of dopamine in
2006.
ventilatory acclimatization to high altitude. Respir Physiol 99: 63–
750. Swenson ER, Hughes JM. Effects of acute and chronic acetazol-
amide on resting ventilation and ventilatory responses in men. 73, 1995.
J Appl Physiol 74: 230 –237, 1993. 772. Taylor NC, Li A, Nattie EE. Ventilatory effects of muscimol
751. Szewczak JM, Powell FL. Open-flow plethysmography with pres- microdialysis into the rostral medullary raphe region of conscious
sure-decay compensation. Respir Physiol Neurobiol 134: 57– 67, rats. Respir Physiol Neurobiol 153: 203–216, 2006.
2003. 773. Temp JA, Henson LC, Ward DS. Does a subanesthetic concen-
752. Tabata M, Kurosawa H, Kikuchi Y, Hida W, Ogawa H, Okabe S, tration of isoflurane blunt the ventilatory response to hypoxia?
Tun Y, Hattori T, Shirato K. Role of GABA within the nucleus Anesthesiology 77: 1116 –1124, 1992.
tractus solitarii in the hypoxic ventilatory decline of awake rats. 774. Tenney SM, Ou LC. Hypoxic ventilatory response of cats at high
Am J Physiol Regul Integr Comp Physiol 281: R1411–R1419, 2001. altitude: an interpretation of “blunting.” Respir Physiol 30: 185–
753. Takakura AC, Moreira TS, Colombari E, West GH, Stornetta 199, 1977.
RL, Guyenet PG. Peripheral chemoreceptor inputs to retrotrap- 775. Teppema LJ, van Dorp ELA, Dahan A. Arterial [H⫹] and the
ezoid nucleus RTN CO2-sensitive neurons in rats. J Physiol 572: ventilatory response to hypoxia in humans: influence of acetazol-
503–523, 2006. amide-induced metabolic acidosis. Am J Physiol Lung Cell Mol
754. Takeda K, Adachi T, Han F, Yokoyama S, Yamamoto H, Hida Physiol 298: L89 –L95, 2010.
W, Shibahara S. Augmented chemosensitivity in black-eyed white 776. Teppema LJ, Bijl H, Mousavi GB, Dahan A. The carbonic an-
Mitfmi-bw mice, lacking melanocytes. J Biochem 141: 327–333, hydrase inhibitors methazolamide and acetazolamide have differ-
2007. ent effects on the hypoxic ventilatory response in the anaesthetized
755. Talbot NP, Balanos GM, Robbins PA, Dorrington KL. Intrave- cat. J Physiol 574: 565–572, 2006.
nous endothelin-1 and ventilatory sensitivity to hypoxia in humans. 777. Teppema LJ, Bijl H, Romberg RR, Dahan A. Antioxidants re-
Adv Exp Med Biol 605: 57– 62, 2008. verse depression of the hypoxic ventilatory response by acetazol-
756. Tamisier R, Gilmartin GS, Launois SH, Pepin JL, Nespoulet amide in man. J Physiol 572: 849 – 856, 2006.
H, Thomas R, Levy P, Weiss JW. A new model of chronic inter- 778. Teppema LJ, Dahan A. Acetazolamide and breathing. Does a
mittent hypoxia in humans: effect on ventilation, sleep, and blood clinical dose alter peripheral and central CO2 sensitivity? Am J
pressure. J Appl Physiol 107: 17–24, 2009. Respir Crit Care Med 160: 1592–1597, 1999.
757. Tamisier R, Hunt BE, Gilmartin GS, Curley M, Anand A, 779. Teppema LJ, Dahan A, Olievier CN. Low-dose acetazolamide
Weiss JW. Hemodynamics and muscle sympathetic nerve activity reduces CO2-O2 stimulus interaction within the peripheral chemo-
after 8 h of sustained hypoxia in healthy humans. Am J Physiol receptors in the anaesthetised cat. J Physiol 537: 221–229, 2001.
Heart Circ Physiol 293: H3027–H3035, 2007. 780. Teppema LJ, Nieuwenhuijs D, Sarton E, Romberg R, Olievier
758. Tankersley CG. Selected contribution: variation in acute hypoxic CN, Ward DS, Dahan A. Antioxidants prevent depression of the
ventilatory response is linked to mouse chromosome 9. J Appl acute hypoxic ventilatory response by subanaesthetic halothane in
Physiol 90: 1615–1622, 2001. men. J Physiol 544: 931–938, 2002.

Physiol Rev • VOL 90 • APRIL 2010 • www.prv.org


752 LUC J. TEPPEMA AND ALBERT DAHAN

781. Teppema LJ, Romberg RR, Dahan A. Antioxidants reverse re- 802. Vicario I, Obeso A, Rocher A, Lopez-Lopez JR, Gonzalez C.
duction of the human hypoxic ventilatory response by subanes- Intracellular Ca2⫹ stores in chemoreceptor cells of the rabbit ca-
thetic isoflurane. Anesthesiology 102: 747–753, 2005. rotid body: significance for chemoreception. Am J Physiol Cell
782. Teppema LJ, Veening JG, Kranenburg A, Dahan A, Berken- Physiol 279: C51–C61, 2000.
bosch A, Olievier C. Expression of c-fos in the rat brainstem after 803. Viemari JC. Noradrenergic modulation of the respiratory neural
exposure to hypoxia and to normoxic and hyperoxic hypercapnia. network. Respir Physiol Neurobiol 164: 123–130, 2008.
J Comp Neurol 388: 169 –190, 1997. 804. Villafuerte FC, Cardenas-Alayza R, Macarlupu JL, Monge C,
783. Terblanche JS, Tolley KA, Fahlman A, Myburgh KH, Jackson Leon-Velarde F. Ventilatory response to acute hypoxia in trans-
S. The acute hypoxic ventilatory response: testing the adaptive genic mice over-expressing erythropoietin: effect of acclimation to
significance in human populations. Comp Biochem Physiol A Mol 3-week hypobaric hypoxia. Respir Physiol Neurobiol 158: 243–250,
Integr Physiol 140: 349 –362, 2005. 2007.
784. Tipoe GL, Fung ML. Expression of HIF-1alpha, VEGF and VEGF 805. Vincent SG, Waddell AE, Caron MG, Walker JK, Fisher JT. A
receptors in the carotid body of chronically hypoxic rat. Respir murine model of hyperdopaminergic state displays altered respira-
Physiol Neurobiol 138: 143–154, 2003. tory control. FASEB J 21: 1463–1471, 2007.
785. Tjong YW, Chen Y, Liong EC, Tipoe GL, Fung ML. Chronic 806. Vizek M, Bonora M. Diaphragmatic activity during biphasic ven-
hypoxia modulates the function and expression of melatonin re- tilatory response to hypoxia in rats. Respir Physiol 111: 153–162,
ceptors in the rat carotid body. J Pineal Res 40: 125–134, 2006. 1998.
786. Towler MC, Hardie DG. AMP-activated protein kinase in meta- 807. Vizek M, Pickett CK, Weil JV. Biphasic ventilatory response of
bolic control and insulin signaling. Circ Res 100: 328 –341, 2007. adult cats to sustained hypoxia has central origin. J Appl Physiol
787. Townsend NE, Gore CJ, Hahn AG, McKenna MJ, Aughey RJ, 63: 1658 –1664, 1987.
Clark SA, Kinsman T, Hawley JA, Chow CM. Living high-train- 808. Vizek M, Pickett CK, Weil JV. Increased carotid body hypoxic
ing low increases hypoxic ventilatory response of well-trained sensitivity during acclimatization to hypobaric hypoxia. J Appl
endurance athletes. J Appl Physiol 93: 1498 –1505, 2002. Physiol 63: 2403–2410, 1987.
788. Trapp S, Aller MI, Wisden W, Gourine AV. A role for TASK-1 809. Vizek M, Pickett CK, Weil JV. Interindividual variation in
KCNK3 channels in the chemosensory control of breathing. J Neu- hypoxic ventilatory response: potential role of carotid body. J Appl
rosci 28: 8844 – 8850, 2008. Physiol 63: 1884 –1889, 1987.
789. Trippenbach T. Ventilatory and metabolic effects of repeated 810. Vlasic V, Simakajornboon N, Gozal E, Gozal D. PDGF-beta
hypoxia in conscious newborn rabbits. Am J Physiol Regul Integr receptor expression in the dorsocaudal brainstem parallels
Comp Physiol 266: R1584 –R1590, 1994. hypoxic ventilatory depression in the developing rat. Pediatr Res
790. Tun Y, Hida W, Okabe S, Kikuchi Y, Kurosawa H, Tabata M, 50: 236 –241, 2001.
Shirato K. Effects of nasal continuous positive airway pressure on 811. Wach RA, Bee D, Barer GR. Dopamine and ventilatory effects of
awake ventilatory responses to hypoxia and hypercapnia in pa- hypoxia and almitrine in chronically hypoxic rats. J Appl Physiol
tients with obstructive sleep apnea. Tohoku J Exp Med 190: 157–
67: 186 –192, 1989.
168, 2000.
812. Wade JG, Larson CP Jr, Hickey RF, Ehrenfeld WK, Severing-
791. Tun Y, Hida W, Okabe S, Ogawa H, Kikuchi Y, Oikawa M,
haus JW. Effect of carotid endarterectomy on carotid chemore-
Kitamuro T, Kunio S. Can nasal continuous positive airway pres-
ceptor and baroreceptor function in man. N Engl J Med 282:
sure decrease clinic blood pressure in patients with obstructive
823– 829, 1970.
sleep apnea? Tohoku J Exp Med 201: 181–190, 2003.
813. Waldrop TG, Bauer RM, Iwamoto GA. Microinjection of GABA
792. Turner DL, Mitchell GS. Long-term facilitation of ventilation
antagonists into the posterior hypothalamus elicits locomotor ac-
following repeated hypoxic episodes in awake goats. J Physiol 499:
tivity and a cardiorespiratory activation. Brain Res 444: 84 –94,
543–550, 1997.
793. Van Beek JH, Berkenbosch A, De Goede J, Olievier CN. Influ- 1988.
ence of peripheral O2 tension on the ventilatory response to CO2 in 814. Walker DW. Hypoxic inhibition of breathing and motor activity in
cats. Respir Physiol 51: 379 –390, 1983. the foetus and newborn. Clin Exp Pharmacol Physiol 22: 533–536,
794. Van Beek JH, Berkenbosch A, De Goede J, Olievier CN. Ef- 1995.
fects of brain stem hypoxaemia on the regulation of breathing. 815. Walker DW, Lee B, Nitsos I. Effect of hypoxia on respiratory
Respir Physiol 57: 171–188, 1984. activity in the foetus. Clin Exp Pharmacol Physiol 27: 110 –113,
795. Van Bockstaele EJ, Peoples J, Telegan P. Efferent projections 2000.
of the nucleus of the solitary tract to peri-locus coeruleus dendrites 816. Walsh BK, Brooks TM, Grenier BM. Oxygen therapy in the
in rat brain: evidence for a monosynaptic pathway. J Comp Neurol neonatal care environment. Respir Care 54: 1193–1202, 2009.
412: 410 – 428, 1999. 817. Wang JS, Chen LY, Fu LL, Chen ML, Wong MK. Effects of
796. Van den Elsen M, Dahan A, DeGoede J, Berkenbosch A, van moderate and severe intermittent hypoxia on vascular endothelial
Kleef J. Influences of subanesthetic isoflurane on ventilatory con- function and haemodynamic control in sedentary men. Eur J Appl
trol in humans. Anesthesiology 83: 478 – 490, 1995. Physiol 100: 127–135, 2007.
797. Van den Elsen M, Sarton E, Teppema L, Berkenbosch A, 818. Wang ZY, Bisgard GE. Chronic hypoxia-induced morphological
Dahan A. Influence of 0.1 minimum alveolar concentration of and neurochemical changes in the carotid body. Microsc Res Tech
sevoflurane, desflurane and isoflurane on dynamic ventilatory re- 59: 168 –177, 2002.
sponse to hypercapnia in humans. Br J Anaesth 80: 174 –182, 1998. 819. Wang ZY, Olson EB Jr, Bjorling DE, Mitchell GS, Bisgard GE.
798. Van den Elsen MJ, Dahan A, Berkenbosch A, DeGoede J, van Sustained hypoxia-induced proliferation of carotid body type I cells
Kleef JW, Olievier IC. Does subanesthetic isoflurane affect the in rats. J Appl Physiol 104: 803– 808, 2008.
ventilatory response to acute isocapnic hypoxia in healthy volun- 820. Wang ZZ, Dinger B, Fidone SJ, Stensaas LJ. Changes in ty-
teers? Anesthesiology 81: 860 – 867, 1994. rosine hydroxylase and substance P immunoreactivity in the cat
799. Varas R, Alcayaga J, Iturriaga R. ACh and ATP mediate excita- carotid body following chronic hypoxia and denervation. Neuro-
tory transmission in cat carotid identified chemoreceptor units in science 83: 1273–1281, 1998.
vitro. Brain Res 988: 154 –163, 2003. 821. Ward DS. The effects of dopamine on the ventilatory response to
800. Varas R, Wyatt CN, Buckler KJ. Modulation of TASK-like back- sustained hypoxia in humans. In: Control of Breathing and Its
ground potassium channels in rat arterial chemoreceptor cells by Modeling Perspective, edited by Honda Y, Miyamoto Y, Konno K
intracellular ATP and other nucleotides. J Physiol 583: 521–536, and Widdicombe JG. New York: Plenum, 1992, p. 291–298.
2007. 822. Ward DS. Stimulation of hypoxic ventilatory drive by droperidol.
801. Vardhan A, Kachroo A, Sapru HN. Excitatory amino acid recep- Anesth Analg 63: 106 –110, 1984.
tors in commissural nucleus of the NTS mediate carotid chemore- 823. Ward DS, Berkenbosch A, DeGoede J, Olievier CN. Dynamics
ceptor responses. Am J Physiol Regul Integr Comp Physiol 264: of the ventilatory response to central hypoxia in cats. J Appl
R41–R50, 1993. Physiol 68: 1107–1113, 1990.

Physiol Rev • VOL 90 • APRIL 2010 • www.prv.org


THE VENTILATORY RESPONSE TO HYPOXIA IN MAMMALS 753

824. Ward DS, Dahan A, Mann CB. Modelling the dynamic ventilatory 847. Wong-Riley MT, Liu Q. Neurochemical and physiological corre-
response to hypoxia in humans. Ann Biomed Eng 20: 181–194, lates of a critical period of respiratory development in the rat.
1992. Respir Physiol Neurobiol 164: 28 –37, 2008.
825. Ward DS, Voter WA, Karan S. The effects of hypo- and hyper- 848. Wood HE, Fatemian M, Robbins PA. Prior sustained hypoxia
glycaemia on the hypoxic ventilatory response in humans. attenuates interaction between hypoxia and exercise as ventilatory
J Physiol 582: 859 – 869, 2007. stimuli in humans. Exp Physiol 92: 273–286, 2007.
826. Wasicko MJ, Sterni LM, Bamford OS, Montrose MH, Carroll 849. Wrobel LJ, Ogier M, Chatonnet F, Autran S, Mezieres V,
JL. Resetting and postnatal maturation of oxygen chemosensitivity Thoby-Brisson M, McLean H, Taeron C, Champagnat J. Ab-
in rat carotid chemoreceptor cells. J Physiol 514: 493–503, 1999. normal inspiratory depth in Phox2a haploinsufficient mice. Neuro-
827. Waters KA, Gozal D. Responses to hypoxia during early develop- science 145: 384 –392, 2007.
ment. Respir Physiol Neurobiol 136: 115–129, 2003. 850. Wu W, Dave NB, Yu G, Strollo PJ, Kovkarova-Naumovski E,
828. Waters KA, Laferriere A, Paquette J, Goodyer C, Moss IR. Ryter SW, Reeves SR, Dayyat E, Wang Y, Choi AM, Gozal D,
Curtailed respiration by repeated vs. isolated hypoxia in maturing Kaminski N. Network analysis of temporal effects of intermittent
piglets is unrelated to NTS ME or SP levels. J Appl Physiol 83: and sustained hypoxia on rat lungs. Physiol Genomics 36: 24 –34,
522–529, 1997. 2008.
829. Waters KA, Tinworth KD. Depression of ventilatory responses 851. Wyatt CN, Buckler KJ. The effect of mitochondrial inhibitors on
after daily, cyclic hypercapnic hypoxia in piglets. J Appl Physiol 90: membrane currents in isolated neonatal rat carotid body type I
1065–1073, 2001. cells. J Physiol 556: 175–191, 2004.
830. Watson CS, Schaefer R, White SE, Homan JH, Fraher L, 852. Wyatt CN, Evans AM. AMP-activated protein kinase and chemo-
Harding R, Bocking AD. Effect of intermittent umbilical cord transduction in the carotid body. Respir Physiol Neurobiol 157:
occlusion on fetal respiratory activity and brain adenosine in late- 22–29, 2007.
gestation sheep. Reprod Fertil Dev 14: 35– 42, 2002. 853. Wyatt CN, Peers C. Ca2⫹-activated K⫹ channels in isolated type I
831. Watson CS, White SE, Homan J, Abdollah S, Brien JF, Challis cells of the neonatal rat carotid body. J Physiol 483: 559 –565, 1995.
JR, Bocking AD. Fetal breathing is not inhibited by ethanol expo- 854. Wyatt CN, Wright C, Bee D, Peers C. O2-sensitive K⫹ currents in
sure during prolonged reduced uterine blood flow. Can J Physiol carotid body chemoreceptor cells from normoxic and chronically
Pharmacol 74: 1016 –1024, 1996. hypoxic rats and their roles in hypoxic chemotransduction. Proc
832. Weil JV. Variation in human ventilatory control-genetic influence Natl Acad Sci USA 92: 295–299, 1995.
on the hypoxic ventilatory response. Respir Physiol Neurobiol 135: 855. Wyatt CN, Mustard KJ, Pearson SA, Dallas ML, Atkinson L,
239 –246, 2003. Kumar P, Peers C, Hardie DG, Evans AM. AMP-activated pro-
833. Weil JV, Byrne-Quinn E, Sodal IE, Friesen WO, Underhill B, tein kinase mediates carotid body excitation by hypoxia. J Biol
Filley GF, Grover RF. Hypoxic ventilatory drive in normal man. Chem 282: 8092– 8098, 2007.
J Clin Invest 49: 1061–1072, 1970. 856. Xu F, Xu J, Tse FW, Tse A. Adenosine stimulates depolarization
834. Weil JV, Stevens T, Pickett CK, Tatsumi K, Dickinson MG,
and rise in cytoplasmic [Ca2⫹] in type I cells of rat carotid bodies.
Jacoby CR, Rodman DM. Strain-associated differences in hypoxic
Am J Physiol Cell Physiol 290: C1592–C1598, 2006.
chemosensitivity of the carotid body in rats. Am J Physiol Lung
857. Xu W, Chi L, Row BW, Xu R, Ke Y, Xu B, Luo C, Kheirandish
Cell Mol Physiol 274: L767–L774, 1998.
L, Gozal D, Liu R. Increased oxidative stress is associated with
835. Weir EK, Lopez-Barneo J, Buckler KJ, Archer SL. Acute oxy-
chronic intermittent hypoxia-mediated brain cortical neuronal cell
gen-sensing mechanisms. N Engl J Med 353: 2042–2055, 2005.
apoptosis in a mouse model of sleep apnea. Neuroscience 126:
836. Weizhen N, Engwall MJ, Daristotle L, Pizarro J, Bisgard GE.
313–323, 2004.
Ventilatory effects of prolonged systemic CNS hypoxia in awake
858. Yamaguchi S, Balbir A, Schofield B, Coram J, Tankersley CG,
goats. Respir Physiol 87: 37– 48, 1992.
837. Wenger RH. Mitochondria: oxygen sinks rather than sensors? Med Fitzgerald RS, O’Donnell CP, Shirahata M. Structural and func-
Hypotheses 66: 380 –383, 2006. tional differences of the carotid body between DBA/2J and A/J
838. Wenninger JM, Olson EB, Wang Z, Keith IM, Mitchell GS, strains of mice. J Appl Physiol 94: 1536 –1542, 2003.
Bisgard GE. Carotid sinus nerve responses and ventilatory accli- 859. Yamamoto M, Nishimura M, Kobayashi S, Akiyama Y, Miy-
matization to hypoxia in adult rats following 2 weeks of postnatal amoto K, Kawakami Y. Role of endogenous adenosine in hypoxic
hyperoxia. Respir Physiol Neurobiol 150: 155–164, 2006. ventilatory response in humans: a study with dipyridamole. J Appl
839. West JB. Rate of ventilatory acclimatization to extreme altitude. Physiol 76: 196 –203, 1994.
Respir Physiol 74: 323–333, 1988. 860. Yamamoto Y, Konig P, Henrich M, Dedio J, Kummer W. Hyp-
840. White DP, Douglas NJ, Pickett CK, Weil JV, Zwillich CW. oxia induces production of nitric oxide and reactive oxygen spe-
Sexual influence on the control of breathing. J Appl Physiol 54: cies in glomus cells of rat carotid body. Cell Tissue Res 325: 3–11,
874 – 879, 1983. 2006.
841. White DP, Schneider BK, Santen RJ, McDermott M, Pickett 861. Yamamoto Y, Kummer W, Atoji Y, Suzuki Y. TASK-1, TASK-2,
CK, Zwillich CW, Weil JV. Influence of testosterone on ventila- TASK-3 and TRAAK immunoreactivities in the rat carotid body.
tion and chemosensitivity in male subjects. J Appl Physiol 59: Brain Res 950: 304 –307, 2002.
1452–1457, 1985. 862. Yan X, Koos BJ, Kruger L, Linden J, Murray TF. Characteriza-
842. Williams BA, Buckler KJ. Biophysical properties and metabolic tion of [125I]ZM 241385 binding to adenosine A2A receptors in the
regulation of a TASK-like potassium channel in rat carotid body pineal of sheep brain. Brain Res 1096: 30 –39, 2006.
type 1 cells. Am J Physiol Lung Cell Mol Physiol 286: L221–L230, 863. Yang F, Khoo MC. Ventilatory response to randomly modulated
2004. hypercapnia and hypoxia in humans. J Appl Physiol 76: 2216 –2223,
843. Williams BA, Smyth J, Boon AW, Hanson MA, Kumar P, 1994.
Blanco CE. Development of respiratory chemoreflexes in re- 864. Yang JJ, Chou YC, Lin MT, Chiu TH. Hypoxia-induced differen-
sponse to alternations of fractional inspired oxygen in the newborn tial electrophysiological changes in rat locus coeruleus neurons.
infant. J Physiol 442: 81–90, 1991. Life Sci 61: 1763–1773, 1997.
844. Williams SE, Wootton P, Mason HS, Bould J, Iles DE, Riccardi 865. Ye JS, Tipoe GL, Fung PC, Fung ML. Augmentation of
D, Peers C, Kemp PJ. Hemoxygenase-2 is an oxygen sensor for a hypoxia-induced nitric oxide generation in the rat carotid body
calcium-sensitive potassium channel. Science 306: 2093–2097, 2004. adapted to chronic hypoxia: an involvement of constitutive and induc-
845. Wolsink JG, Berkenbosch A, DeGoede J, Olievier CN. Venti- ible nitric oxide synthases. Pflügers Arch 444: 178 –185, 2002.
latory interaction between hypoxia and hypercapnia in piglets 866. Yelmen NK, Turgut G, Sahin G, Oruc T. Effects of intravenous
shortly after birth. Respir Physiol 96: 25–35, 1994. and intracerebroventricular theophylline on hypoxic ventilatory
846. Wong-Riley MT, Liu Q. Neurochemical development of brain depression in anesthetized cats. Med Princ Pract 13: 277–281, 2004.
stem nuclei involved in the control of respiration. Respir Physiol 867. Yuan G, Adhikary G, McCormick AA, Holcroft JJ, Kumar GK,
Neurobiol 149: 83–98, 2005. Prabhakar NR. Role of oxidative stress in intermittent hypoxia-

Physiol Rev • VOL 90 • APRIL 2010 • www.prv.org


754 LUC J. TEPPEMA AND ALBERT DAHAN

induced immediate early gene activation in rat PC12 cells. 872. Zhang W, Carreno FR, Cunningham JT, Mifflin SW. Chronic
J Physiol 557: 773–783, 2004. sustained and intermittent hypoxia reduce the function of ATP-
868. Zapata P. Is ATP a suitable co-transmitter in carotid body arterial sensitive potassium channels in the nucleus of the solitary tract.
chemoreceptors? Respir Physiol Neurobiol 157: 106 –115, 2007. Am J Physiol Regul Integr Comp Physiol 295: R1555–R1562,
869. Zapata P, Torrealba F. Blockade of dopamine-induced chemosen- 2008.
sory inhibition by domperidone. Neurosci Lett 51: 359 –364, 1984. 873. Zhang W, Carreno FR, Cunningham JT, Mifflin SW. Chronic
870. Zhang M, Buttigieg J, Nurse CA. Neurotransmitter mechanisms sustained hypoxia enhances both evoked EPSCs and norepineph-
mediating low-glucose signalling in cocultures and fresh tissue rine inhibition of glutamatergic afferent inputs in the nucleus of the
slices of rat carotid body. J Physiol 578: 735–750, 2007. solitary tract. J Neurosci 29: 3093–3102, 2009.
871. Zhang S, Robbins PA. Methodological and physiological variabil- 874. Zwemer CF, Song MY, Carello KA, D’Alecy LG. Strain differ-
ity within the ventilatory response to hypoxia in humans. J Appl ences in response to acute hypoxia: CD-1 versus C57BL/6J mice.
Physiol 88: 1924 –1932, 2000. J Appl Physiol 102: 286 –293, 2007.

Physiol Rev • VOL 90 • APRIL 2010 • www.prv.org


Luc J. Teppema and Albert Dahan
Physiol Rev 90:675-754, 2010. doi:10.1152/physrev.00012.2009

You might find this additional information useful...

This article cites 858 articles, 463 of which you can access free at:
http://physrev.physiology.org/cgi/content/full/90/2/675#BIBL

Medline items on this article's topics can be found at http://highwire.stanford.edu/lists/artbytopic.dtl


on the following topics:
Oncology .. Transcription Factors
Oncology .. Hypoxia
Physiology .. Carotid Chemoreceptors
Medicine .. Environment and Public Health
Medicine .. Respiration
Medicine .. Anoxia
Updated information and services including high-resolution figures, can be found at:
http://physrev.physiology.org/cgi/content/full/90/2/675

Additional material and information about Physiological Reviews can be found at:
http://www.the-aps.org/publications/prv

This information is current as of September 2, 2010 .

Physiological Reviews provides state of the art coverage of timely issues in the physiological and biomedical sciences. It is
published quarterly in January, April, July, and October by the American Physiological Society, 9650 Rockville Pike, Bethesda MD
20814-3991. Copyright © 2010 by the American Physiological Society. ISSN: 0031-9333, ESSN: 1522-1210. Visit our website at
http://www.the-aps.org/.

S-ar putea să vă placă și