Sunteți pe pagina 1din 7

Journal of the American College of Cardiology Vol. 62, No.

2, 2013
 2013 by the American College of Cardiology Foundation ISSN 0735-1097/$36.00
Published by Elsevier Inc. http://dx.doi.org/10.1016/j.jacc.2013.03.070

STATE-OF-THE-ART PAPER

Effects of Nitric Oxide on Cell Proliferation


Novel Insights
Claudio Napoli, MD, PHD,*y Giuseppe Paolisso, MD,z Amelia Casamassimi, BIOLD,*
Mohammed Al-Omran, MD,x Michelangela Barbieri, MD,z Linda Sommese, BIOLD,*
Teresa Infante, BIOLD,y Louis J. Ignarro, PHDk
Naples, Italy; Riyadh, Kingdom of Saudi Arabia; and Los Angeles, California

Nitric oxide (NO) has been suggested to be a pathophysiological modulator of cell proliferation, cell cycle arrest, and
apoptosis. In this context, NO can exert opposite effects under diverse conditions. Indeed, several studies have
indicated that low relative concentrations of NO seem to favor cell proliferation and antiapoptotic responses and
higher levels of NO favor pathways inducing cell cycle arrest, mitochondria respiration, senescence, or apoptosis.
Here we report the effects of NO on both promotion and inhibition of cell proliferation, in particular in regard to
cardiovascular disease, diabetes, and stem cells. Moreover, we focus on molecular mechanisms of action involved
in the control of cell cycle progression, which include both cyclic guanosine monophosphate–dependent
and –independent pathways. This growing field may lead to broad and novel targeted therapies against
cardiovascular diseases, especially concomitant type 2 diabetes, as well as novel bioimaging NO-based diagnostic
tools. (J Am Coll Cardiol 2013;62:89–95) ª 2013 by the American College of Cardiology Foundation

Nitric oxide (NO) is a signaling molecule that regulates involving protein modifications (6). Lower concentrations of
many functions, such as vascular tone, blood pressure, NO have been suggested to exert a direct effect on processes
neurotransmission, immune response, and oxidation-sensitive such as cell proliferation and survival, whereas higher con-
mechanisms (1–4). NO may act as an autocrine or paracrine centrations have an indirect effect through both oxida-
messenger, and its production and degradation are cell type tive and nitrosative stresses (3,5,6). Free radical interactions
dependent. NO synthesis is catalyzed from L-arginine by also influence NO signaling. One of the consequences of
a family of nitric oxide synthases (NOSs), varying from reactive oxygen species (ROS) generation is reduced con-
picomolar/nanomolar range for short periods to micromolar centrations of NO (1). The resulting reactive nitrogen
range for protracted periods (3). NO reacts with molecules species can also have biological effects and increase oxidative
such as oxygen, superoxide or metals, nucleic acids, and and nitrosative stress responses. Overall, cellular responses
proteins. The major reactions involving NO support its rapid are differentially regulated by specific NO concentrations,
oxidation into nitrate and nitrite, which are now considered with lower NO concentrations (picomolar to nanomolar)
not inert end products but actors of a reverse pathway that generally promoting cell survival and proliferation and
represents an alternative source of NO when the endogenous higher concentrations (micromolar) favoring cell cycle arrest,
L-arginine/NOS pathway is dysfunctional (5,6). apoptosis, and senescence (6) (Fig. 1A). However, observ-
The effect of NO production on cellular processes is ed divergent effects can be explained by cellular context,
also dependent on its concentration and on the presence of cell cycle point, and oncogenic state (6). The molecular
other free radicals. Peroxynitrites, generated from reaction mechanisms underlying the proliferative action of NO at
with a superoxide, can interact with several cellular com- low concentrations are not yet clear, but key molecules
ponents and are implicated in NO signaling mechanisms and pathways involved in NO-mediated inhibition have
been better studied. Some basic distinct concentrations of
NO have also been proposed for activity, such as for cyclic
guanosine monophosphate (cGMP)-mediated processes
From the *Department of General Pathology, Excellence Research Centre on (30 nmol/l) and nitrosative stress (1 mmol/l) (6).
Cardiovascular Diseases, U.O.C. Immunohematology, Second University of Naples,
Naples, Italy; zDivision of Geriatrics, 1st School of Medicine, Second University
of Naples, Naples, Italy; yFondazione SDN, IRCCS, Naples, Italy; xCollege of Molecular Mechanisms of NO Involved in
Medicine, King Saud University, Riyadh, Kingdom of Saudi Arabia; and the Progression of the Cell Cycle
kDepartment of Pharmacology, David Geffen School of Medicine, University of
California Los Angeles, Los Angeles, California. This study was supported in part by NO blocks the progression of the cell cycle primarily at the
Regione Campania 2008 Research Funds. The authors have reported that they have no
relationships relevant to the content of this paper to disclose. G1/S transition. Hence, NO-induced G1 arrest has been
Manuscript received February 15, 2013; accepted March 19, 2013. observed in vascular smooth muscle cells (VSMCs) and
90 Napoli et al. JACC Vol. 62, No. 2, 2013
NO and Cell Proliferation July 9, 2013:89–95

Abbreviations other cell lines (7–9). In other downstream pathways stimulating cell proliferation (33).
and Acronyms contexts and after treatment with In addition, the EGFR trans(auto)phosphorylation inhi-
NO donors of the nonsteroidal bition was found to be independent of cGMP and directly
BMC = bone marrow cell(s)
anti-inflammatory drug family, the inhibited by NO (19). However, an increase in NO-
cGMP = cyclic guanosine
entry into the G2/M phase was mediated cGMP enables the disruption of the downstream
monophosphate
NO sensitive (8,10,11). mitogenic signal through Raf1 phosphorylation (25).
EGFR = epidermal growth
factor receptor
As expected, many cell cycle eNOS S-glutathionylation is a pivotal redox regulator
proteins (p21Cip1/Waf1, p27, cy- of endothelial function and vascular tone. Indeed, in
eNOS = endothelial nitric
oxide synthase
clins, Cdk2, pRb, and the like) endothelial cells, this modification reversibly decreases
EPC = endothelial progenitor
are good candidates for final mo- eNOS activity with increased production of superoxide
cell(s) lecular targets of NO, as revealed anions. Moreover, in hypertensive vessels, eNOS S-gluta-
GC = guanylate cyclase
by exogenous NO, endothelial thionylation is increased with impaired vasodilation (34).
nitric oxide synthase (eNOS) trans- The impact of another mechanism, tyrosine nitration, was
NO = nitric oxide
fection, and whole expression stud- demonstrated in a recent study that identified several nitrated
NOS = nitric oxide synthase
ies (7,9,12–18). NO is also able proteins involved in the different checkpoints toward the cell
ROS = reactive oxygen
to regulate cell proliferation by cycle (35).
species
targeting important mitogenic Another cGMP-independent mechanism involves mito-
T2DM = type 2 diabetes
mellitus
receptors and their downstream chondria that influence several pathways modulating cell
pathways, such as epidermal growth proliferation, cell cycle arrest, and apoptosis through oxi-
VSMC = vascular smooth
muscle cell(s)
factor receptor (EGFR) signaling dative and nitrosative reactions that are mediated by the
pathway (8,19). Moreover, 26S NOS mitochondrial isoform (23,36). Moreover, peroxyni-
proteasome and apoptosis factors represent other important trite can also change calcium homeostasis, allowing the
NO targets (20,21) (Fig. 1B). opening of the mitochondrial permeability transition pore
cGMP-dependent and -independent pathways. A canon- that promotes mitochondrial signaling of cell death (37). In
ical NO pathway involves the selective activation of soluble this context, NO instead exerts a protective action by directly
guanylate cyclase (GC), the generation of cGMP, and the ac- inhibiting the opening of the mitochondrial permeability
tivation of specific cGMP-dependent protein kinases (1,22). transition pore. Furthermore, cytochrome c oxidase is one of
However, further mechanisms have emerged that mainly the most important targets for NO signaling, leading to
include the formation of NO-induced post-translational inhibition of mitochondrial oxidative phosphorylation, the
modifications (8,22,23). Overall, these modifications activate control of apoptosis, and ROS generation (37,38).
pathways that are cGMP independent (Fig. 1B).
The involvement of cGMP in growth inhibition has
Cell Cycle NO-Dependent Effects in the
been described in VSMCs, in which NO activates GC with
Cardiovascular System
a subsequent increase in cGMP leading to the phosphory-
lation of a vasodilator-stimulated phosphoprotein and sub- Cell proliferation, inhibition, and apoptosis. NO is able
sequent inhibition of the epidermal growth factor signaling to inhibit cell growth and proliferation and to induce
pathway (24,25). Recent findings indicate that other nuclear apoptosis in a dose-dependent manner (7,12,20,28,29,39,40)
effects of NO altering the cell cycle can occur. A mechanism (Table 1). Indeed, an adequate concentration of NO is
involved in the regulation of the cell cycle is the direct in- required to induce inhibition of cell proliferation. There is a
teraction of NO-sensitive GC with chromosomes during lack of correlation between increased NOS expression and
mitosis (26). More recently, NO has been shown to mod- inhibition of cell proliferation in some cellular systems,
ulate chromatin folding in human endothelial cells through probably due to the short life of NO that is transformed into
class II histone deacetylases (27). inactive compounds (6,8,28).
The antiproliferative effect of NO does not necessarily NO donors and drugs affecting the NO pathway through
require cGMP as an intermediate effector because it was different mechanisms have been used to inhibit cell pro-
also shown in a fibroblast cell line lacking soluble GC (19) liferation of several cell types. Indeed, it is known that
or with GC inhibitors (16,28,29). Inhibition of the G1/S endogenously produced NO can negatively regulate cell
transition has been shown to be mostly a cGMP-independent proliferation and/or the proliferation of neighboring cells.
process (7,9), although dual mechanisms were found to The increase of endogenous NO depends on the availability
mediate the antimitogenic effects in VSMCs (30). of L-arginine and/or inducible NOS expression mediated by
A mechanism for cGMP-independent proliferative arrest several cytokines (41–43).
is S-nitrosylation, which can reversibly inhibit the catalytic A proliferative arrest induced by endogenous NO pro-
activity of the 26S proteasome and enhance Ras guanine duction has been found in different cell types, including
nucleotide exchange under nitrosative stress (31,32). A re- VSMCs (8). Proliferation of these cells has been accepted
cent study has also demonstrated that NO-mediated Ras as a common event in the pathophysiology of many vascu-
nitrosylation in different subcellular compartments regulates lar diseases. Delivery of L-arginine, pharmacological NO
JACC Vol. 62, No. 2, 2013 Napoli et al. 91
July 9, 2013:89–95 NO and Cell Proliferation

Figure 1 Major Molecular Mechanisms Involved in the Effects of NO on the Cell Cycle

(A) Dual role of different doses of nitric oxide (NO) on the cell cycle and their effects on cardiovascular-related cells. (B) NO synthesis from its precursor L-arginine is catalyzed by
nitric oxide synthases (NOSs). NO is rapidly oxidized into nitrate (NO3) and nitrite (NO2), which can be reduced into bioactive NO in a reverse pathway. NO can also react with
superoxide (O2) to generate peroxynitrite anion (ONOO), which is implicated in NO signaling mechanisms involving post-translational modifications. Molecules involved in both
cyclic guanosine monophosphate–dependent and –independent pathways are shown. NO can influence the epidermal growth factor receptor (EGFR)/Ras/mitogen-activated
protein kinase (MAPK) proliferative pathway by both mechanisms. Examples are EGFR and Ras S-nitrosylation. Mitochondria-related mechanisms, molecules involved in the
apoptosis, insulin/insulin-like growth factor (IGF)-I/Akt, and 26S proteasome pathways are also represented.

donors, NO gas, or overexpression of NOS proteins can apolipoprotein E is able to increase expression of inducible
inhibit proliferation of VSMCs and reduce the injury NOS in VSMCs, thereby inhibiting their proliferation (46).
responses within the blood vessel wall. Preclinical models NO has been implicated in both apoptotic and necrotic
have been essentially used to establish the potential of NO in cell death, depending on several factors including cellular
the inhibition of VSMC proliferation and induction of cell redox state. Additionally, it can be either an antiapoptotic or
cycle arrest in models of neointimal hyperplasia and reste- a proapoptotic regulator, depending on the pathway and
nosis (11,16,44). A recent study also suggested that NO can mechanism involved. For instance, at high concentrations of
inhibit neointimal hyperplasia in a sex- and hormone- NO, programmed cell death was observed after inhibition of
dependent manner (44). cell proliferation in cardiomyocytes (47). Caspase activation
Consistent with these observations, VSMCs transfected and mitochondrial changes are also involved in the apoptosis
with eNOS showed inhibition of cell proliferation and of initiated after treatment with GEA3162 in murine bone
key cell cycle regulatory molecules (45). Moreover, marrow cells (BMCs) (48). NO can also confer protection
92 Napoli et al. JACC Vol. 62, No. 2, 2013
NO and Cell Proliferation July 9, 2013:89–95

Table 1 Effects of Exogenous NO on the Proliferation of Cardiovascular-Related Cells

Cell Type NO Donors NO Donor Action First Author (Ref. #)


Aortic VSMC DETA/NO 100 mmol/l to 1 mmol/l  Tanner et al. (7) and
Kapadia et al. (20)
Umbilical arteries VSMC SNAP 100 mmol/l  Ishida et al. (12,39)
HUVEC/coronary aortic endothelial cells SNAP, S-nitrosoglutathione, SNP 100 mmol/l  Heller et al. (28)
Peripheral blood mononuclear cells GEA3162, GEA3175, SNAP 1–30 mmol/l GEA; 100–500 mmol/l SNAP  Kosonen et al. (29)
HUVEC SNAP, SNP, DETA/NO 100–600 mmol/l  Gooch et al. (40)
Murine bone marrow cells (Jaws II) GEA3162 30–100 mmol/l  Taylor et al. (48)
Embryonic stem cells DETA-NO 2–20 mmol/l þ Tejedo et al. (21)
Murine bone marrow stromal cells (OP9) SNAP 10–50 mmol/l þ Wong et al. (51)

DETA/NO ¼ N-{4-[1-(3-aminopropyl)-2-hydroxy-2 nitrosohydrazino]butyl}propane-1,3-diamine; GEA3162 ¼ 1,2,3,4-oxatriazolium,5-amino-3-(3,4-dichlorophenyl)-chloride; GEA3175 ¼ 1,2,3,4-oxatriazolium,


3-(3-chloro-2-methylphenyl)-5-[[(4-methylphenyl) sulfonyl]amino], hydroxide inner salt; HUVEC ¼ human umbilical vein endothelial cells; NO, nitric oxide; SNAP ¼ S-nitroso-N-acetylpenicillamine; SNP ¼ sodium
nitroprusside; VSMC ¼ vascular smooth muscle cells.

from apoptosis and promote survival of embryonic stem cells, in cardiac progenitors and stem cells remains poorly
thus delaying their differentiation (21). NO donors were understood.
found to cause a dramatic and concentration-dependent NO scavenger treatment can increase the proliferation of
induction of apoptosis in a caspase-dependent manner in bone marrow–derived endothelial progenitor cells (EPCs),
human neutrophils and murine BMCs (8,48). a subpopulation of adult stem cells that are recruited from
Effects on cell proliferation. NO can also promote cell bone marrow to the injured vessel to promote endothelial
proliferation under certain conditions, although the mech- regeneration and neovascularization (53). Data from several
anisms responsible for this effect have remained poorly studies indicate that the NO pathway may improve the
understood (2,6). Interestingly, a recent in vivo study (49) paracrine and angiogenesis efficiency of BMCs in experi-
showed that eNOS deficiency may cause collateral vessel mental hind limb ischemia (56–58) and in patients with
rarefaction, thus impairing the activation of a cell cycle gene chronic critical limb ischemia (59,60).
network during arteriogenesis; this finding has suggested a
novel role for eNOS in maintaining native collateral density The NO Pathway and Cell Proliferation
during growth to adulthood and in collateral remodeling in in Type 2 Diabetes
obstructive disease through regulation of cell proliferation
(49). Similarly, NO derived from eNOS has been shown to Hyperglycemia, associated with endothelial dysfunction and
play an important role in cardiomyocyte proliferation and reduced new blood vessel growth, is a primary cause of
maturation during early neonatal heart development (50). vascular complications in diabetes. Moreover, the ability of
Still, NO donor treatment determines cell proliferation in patients with type 2 diabetes mellitus (T2DM) to develop
several contexts (Table 1) (8,21,51). Interestingly, an NO coronary collateral vessels is diminished due to a diabetes-
donor at low concentrations (10 to 50 mmol/l) protected associated impairment of EPC count and mobilization
murine bone marrow stromal cells against spontaneous (61). The EPC count of patients with T2DM and periph-
apoptosis (51). Similarly, exposure of embryonic stem cells eral arterial disease is substantially lower than that of healthy
to low concentrations of NO (2 to 20 mmol/l) can regulate subjects, nondiabetic patients with vascular disease, and
their differentiation by arresting the loss of self-renewal patients with T2DM who do not have vascular disease
markers and promoting cell survival through inhibition of (53,60,62). Moreover, EPCs isolated from patients with
apoptosis (21). Overall, these studies support the concept T2DM displayed impaired proliferation and function (63).
that the final effect of NO on the cell cycle is dependent on Additionally, proliferation of EPCs was inversely correlated
both concentration and the surrounding context (Fig. 1A). with plasma glycated hemoglobin levels of these patients,
The existence of multipotent resident cardiac stem cells suggesting a relationship between glycemic control and EPC
and adult BMCs that participate in homeostasis of the heart number and proliferation (63).
and regeneration of the injured tissue make these cells a EPCs require NO-cGMP signaling for proper function,
valuable resource for the treatment of cardiovascular disease including migration, and decreased bioavailability of NO has
(52,53). The involvement of NO signaling in stem cell been proposed as one of the determinants of vascular
cardiovascular biology was demonstrated in the differentia- damage in diabetes (61). Patients with T2DM have a lower
tion of embryonic stem cells into myocardial cells (54). overall systemic fraction of L-arginine that is converted to
Furthermore, a recent study in a porcine model showed NO compared with healthy individuals (64). Interestingly,
that activation of the NO pathway directs BMCs to eNOS uncoupling in diabetic EPCs resulted in excessive
a preferential cardiomyogenic phenotype and also stimulates superoxide anion production and reduced bioavailability of
cell proliferation (55). However, the precise role of NO NO, implying an intimate relationship between oxidative
JACC Vol. 62, No. 2, 2013 Napoli et al. 93
July 9, 2013:89–95 NO and Cell Proliferation

stress and EPC damage (65). Prolonged exposure of early diabetes, where EPCs are impaired. Moreover, other drugs
and late EPCs to high glucose concentrations reduces their such as angiotensin-converting enzyme inhibitors also pre-
number and proliferative ability and the extent of phosphor- vent endothelial dysfunction by increasing eNOS protein
ylation of eNOS and some members of the PI3-kinase/Akt expression and activity (71–73).
signaling pathway (66). This study also showed that The biology of NO has stimulated the development of
impaired NO-related rather than oxidative stress–related pharmacological agents with different actions on cell pro-
mechanisms are involved in the EPC dysfunction induced liferation; for example, some agents are able to release NO,
by high levels of glucose (66). In light of such evidence, such as NO-donating nonsteroidal anti-inflammatory drugs
pharmacological or genetic interventions affecting the mole- and NO-aspirin, and may also be used against vascular
cular pathway involving eNOS activity have been hypothe- damage. Indeed, NCX-4016, an aspirin-like NO donor, has
sized to counteract diabetic EPC dysfunction. Thus, the been found to reduce experimental restenosis (11) and the
stimulation of NO production or its signaling cascades may development of atherosclerosis (74), which are linked to
increase the number and function of EPCs and attenuate proliferation of VSMCs. Similarly, nebivolol, a beta-blocker
endothelium damage, independently of the vasodilatory that releases NO, was able to reduce experimental athero-
effects of NO. In this regard, a recent study showed that sclerosis (75). Consistently, there are numerous NO-based
treatment of glucose-stressed EPCs with superoxide dis- therapies for pulmonary hypertension (76), another disease
mutase attenuated generation of O2, restored production of linked to proliferation of VSMCs. Preclinical studies
NO, and partially restored their ability to form colonies, revealed a potent effect of inhaled nitrite in the inhibition of
whereas treatment with insulin increased production of NO experimental pulmonary arterial hypertrophy and prolifera-
but did not change generation of O2 and their ability to form tion of VSMCs, although further studies are required to
colonies. However, this ability was fully restored after better establish doses, potential toxic effects, and mecha-
combined treatment with superoxide dismutase and insulin nisms of their therapeutic action (77). The chemical versa-
(61). Additionally, Chen et al. reported that in vitro treat- tility of NO has led to the synthesis of a wide range of NO
ment of EPCs from patients with T2DM with an NO donor donors, each with a different mode and rate of release and
drug could also reverse impairment of EPCs induced by high action, suitable for different disease targets. However, long-
levels of glucose acting on cell proliferation (66). term use of current NO donors is limited by development
of tolerance and toxicity issues, suggesting that novel alter-
New Concepts in the Role of NO natives should be identified. Nitrosyl-cobinamide has been
in the Cell Cycle and Clinical Insights found to be an effective NO-releasing compound in several
models, suggesting that it could be a useful drug for treat-
The NO pathway is involved in many cardiovascular ing hypertension and cardiovascular disease (78). Further-
conditions, and delivery of specific concentrations of exog- more, a recent study showed the positive effect of infusion
enous NO is an attractive therapeutic option for these of NO-releasing hydrogel/glass hybrid nanoparticles that
disorders, including those that are cell proliferation based. A induce a reduction of blood pressure and an increase in
well-known example of cell proliferation–based pathology is vascular relaxation without any tolerance and immunologic
neointimal hyperplasia, which has been found to decrease response (79).
with NO-based therapies in animal model studies. A recent Additionally, in vitro and in vivo studies have shown that
study suggested that particular attention should be paid to some mitochondrial antioxidants are able to decrease ROS
the patient’s sex and hormone status when developing these production, leading to reduced apoptosis and improved
therapies (44). A significant protective role of NO is also cardiac function. However, the use of these molecules in
implicated in therapeutic neoangiogenesis associated with humans require the development of specific and sensitive
peripheral ischemia (56–60). tools to monitor mitochondrial oxidative stress and the
Interestingly, vasorelaxant prostanoids used in clinical development of orally available compounds (80).
practice, such as prostacyclin or its derivatives, have been One of the major weaknesses in studying ROS and reac-
shown to exert protective effects on endothelial cells by tive nitrogen species is the lack of proper tools to monitor
mechanisms that partly involve cyclic adenosine mono- their production in vivo. Thus, sensitive and specific detec-
phosphate–mediated formation of NO (67). Indeed, pros- tion methods can be useful for elucidation of the effects of
tacyclin analogues such as Beraprost or Iloprost can increase NO on cell proliferation in pathophysiological conditions
the number and migration of EPCs in humans and in both in vitro and in vivo. To date, fluorescent probes based
ischemic tissues of experimental animal models (68,69). on small organic molecules provide dynamic information
Prostacyclin has been found to exert a direct effect on the concerning the localization and quantity of biological mole-
function of EPCs in an autocrine or a paracrine manner cules of interest (81). This approach could be worthwhile to
through an NO-dependent mechanism (70). In this regard, evaluate their functions in the living body by using less
NO-dependent vasoprotective agents such as prostacyclin or invasive techniques, without the need for isolating tissues or
statins could have a significant therapeutic role in cardio- cellular constituents. Today, several design strategies for
vascular diseases under pathological conditions, such as specific reactive nitrogen species fluorescent probes to use in
94 Napoli et al. JACC Vol. 62, No. 2, 2013
NO and Cell Proliferation July 9, 2013:89–95

bioimaging technologies, including photo-induced electron 19. Garg UC, Hassid A. Nitric oxide-generating vasodilators inhibit
mitogenesis and proliferation of BALB/C 3T3 fibroblasts by a cyclic
transfer, are well established and have been applied to many GMP-independent mechanism. Biochem Biophys Res Commun 1990;
probes. The use of this approach should be suitable for 171:474–9.
real-time analysis of NO-dependent mechanisms (82–84). 20. Kapadia MR, Eng JW, Jiang Q, Stoyanovsky DA, Kibbe MR. Nitric
oxide regulates the 26S proteasome in vascular smooth muscle cells.
Nitric Oxide 2009;20:279–88.
21. Tejedo JR, Tapia-Limonchi R, Mora-Castilla S, et al. Low concen-
Reprint requests and correspondence: Prof. Claudio Napoli, trations of nitric oxide delay the differentiation of embryonic stem cells
Department of General Pathology, Excellence Research Centre on and promote their survival. Cell Death Dis 2010;1:e80.
Cardiovascular Diseases, U.O.C. Immunohematology, 1st School 22. Bian K, Ke Y, Kamisaki Y, Murad F. Proteomic modification by nitric
of Medicine, Second University of Naples, 80138 Naples, Italy. oxide. J Pharmacol Sci 2006;101:271–9.
23. Martínez-Ruiz A, Cadenas S, Lamas S. Nitric oxide signaling: clas-
E-mail: claunap@tin.it or claudio.napoli@unina2.it.
sical, less classical, and nonclassical mechanisms. Free Radic Biol Med
2011;51:17–29.
24. Chen L, Daum G, Chitaley K, et al. Vasolidator-stimulated phos-
REFERENCES phoprotein regulates proliferation and growth inhibition by nitric oxide
in vascular smooth muscle cells. Arterioscler Thromb Vasc Bio 2004;
1. Napoli C, Ignarro LJ. Nitric oxide and atherosclerosis. Nitric Oxide 24:1403–8.
2001;5:88–97. 25. Yu SM, Hung LM, Lin CC. cGMP-elevating agents suppress
2. Bian K, Murad F. Nitric oxide (NO)dbiogeneration, regulation, and proliferation of vascular smooth muscle cells by inhibiting the activation
relevance to human diseases. Front Biosci 2003;8:d264–78. of epidermal growth factor signaling pathway. Circulation 1997;95:
3. Alderton WK, Cooper CE, Knowles RG. Nitric oxide synthases: 1269–77.
structure, function and inhibition. Biochem J 2001;357:593–615. 26. Pifarré P, Baltrons MA, Földi I, García A. NO-sensitive guanylyl
4. Furchgott RF. Introduction to EDRF research. J Cardiovasc Phar- cyclase beta1 subunit is peripherally associated to chromosomes during
macol 1993;22 Suppl 7:S1–2. mitosis. Novel role in chromatin condensation and cell cycle progres-
5. Lundberg JO, Weitzberg E. NO generation from inorganic nitrate and sion. Int J Biochem Cell Biol 2009;41:1719–30.
nitrite: Role in physiology, nutrition and therapeutics. Arch Pharm Res 27. Illi B, Dello Russo C, Colussi C, et al. Nitric oxide modulates chro-
2009;32:1119–26. matin folding in human endothelial cells via protein phosphatase 2A
6. Thomas DD, Ridnour LA, Isenberg JS, et al. The chemical biology of activation and class II histone deacetylases nuclear shuttling. Circ Res
nitric oxide: implications in cellular signaling. Free Radic Biol Med 2008;102:51–8.
2008;45:18–31. 28. Heller R, Polack T, Gräbner R, Till U. Nitric oxide inhibits prolifer-
7. Tanner FC, Meier P, Greutert H, Champion C, Nabel EG, ation of human endothelial cells via a mechanism independent of
Lüscher TF. Nitric oxide modulates expression of cell cycle regulatory cGMP. Atherosclerosis 1999;144:49–57.
proteins: a cytostatic strategy for inhibition of human vascular smooth 29. Kosonen O, Kankaanranta H, Vuorinen P, Moilanen E. Inhibition of
muscle cell proliferation. Circulation 2000;101:1982–9. human lymphocyte proliferation by nitric oxide-releasing oxatriazole
8. Villalobo A. Nitric oxide and cell proliferation. FEBS J 2006;273: derivatives. Eur J Pharmacol 1997;337:55–61.
2329–44. 30. Sarkar R, Gordon D, Stanley JC, Webb RC. Dual cell cycle-specific
9. Cui X, Zhang J, Ma P, et al. cGMP-independent nitric oxide signaling mechanisms mediate the antimitogenic effects of nitric oxide in
and regulation of the cell cycle. BMC Genomics 2005;6:151. vascular smooth muscle cells. J Hypertens 1997;15:275–83.
10. Su X, Takahashi A, Kondo N, et al. Nitric oxide radical-induced 31. Lima B, Forrester MT, Hess DT, Stamler JS. S-nitrosylation in
radioadaptation and radiosensitization are G2/M phase-dependent. cardiovascular signaling. Circ Res 2010;106:633–46.
J Radiat Res 2011;52:609–15. 32. Schulman IH, Hare JM. Regulation of cardiovascular cellular processes
11. Napoli C, Cirino G, Del Soldato P, et al. Effects of nitric oxide- by S-nitrosylation. Biochim Biophys Acta 2012;1820:752–62.
releasing aspirin versus aspirin on restenosis in hypercholesterolemic 33. Batista WL, Ogata FT, Curcio MF, et al. S-nitrosoglutathione and
mice. Proc Natl Acad Sci U S A 2001;98:2860–4. endothelial nitric oxide synthase-derived nitric oxide regulate com-
12. Ishida A, Sasaguri T, Miwa Y, Kosaka C, Taba Y, Abumiya T. Tumor partmentalized Ras S-nitrosylation and stimulate cell proliferation.
suppressor p53 but not cGMP mediates NO-induced expression of Antioxid Redox Signal 2013;18:221–38.
p21Waf1/Cip1/Sdi1 in vascular smooth muscle cells. Mol Pharmacol 1999; 34. Chen CA, Wang TY, Varadharaj S, et al. S-glutathionylation uncou-
56:938–46. ples eNOS and regulates its cellular and vascular function. Nature 2010;
13. Bauer PM, Buga GM, Fukuto JM, Pegg AE, Ignarro LJ. Nitric oxide 468:1115–8.
inhibits ornithine decarboxylase via S-nitrosylation of cystein 360 in the 35. Jia M, Mateoiu C, Souchelnytskyi S. Protein tyrosine nitration in the
active site of the enzyme. J Biol Chem 2001;276:34458–64. cell cycle. Biochem Biophys Res Commun 2011;413:270–6.
14. Gu M, Lynch J, Brecher P. Nitric oxide increases p21Waf1/Cip1 36. Carreras MC, Poderoso JJ. Mitochondrial nitric oxide in the signaling
expression by a cGMP-dependent pathway that includes activation of of cell integrated responses. Am J Physiol Cell Physiol 2007;292:
extracellular signal-regulated kinase and p70S6k. J Biol Chem 2000; C1569–80.
275:11389–96. 37. Borutaite V, Morkuniene R, Arandarcikaite O, Jekabsone A,
15. Condorelli G, Aycock JK, Frati G, Napoli C. Mutated p21/WAF/CIP Barauskaite J, Brown GC. Nitric oxide protects the heart from
transgene overexpression reduces smooth muscle cell proliferation, ischemia-induced apoptosis and mitochondrial damage via protein
macrophage deposition, oxidation-sensitive mechanisms, and restenosis kinase G mediated blockage of permeability transition and cytochrome
in hypercholesterolemic apolipoprotein E knockout mice. FASEB J c release. J Biomed Sci 2009;16:70.
2001;15:2162–70. 38. Erusalimsky JD, Moncada S. Nitric oxide and mitochondrial signaling:
16. Kibbe MR, Li J, Nie S, et al. Inducible nitric oxide synthase (iNOS) from physiology to pathophysiology. Arterioscler Throm Vasc Biol
expression upregulates p21 and inhibits vascular smooth muscle cell 2007;27:2524–31.
proliferation through p42/44 mitogen-activated protein kinase activa- 39. Ishida A, Sasaguri T, Kosaka C, Nojima H, Ogata J. Induction of the
tion and independent of p53 and cyclic guanosine monophosphate. cyclin-dependent kinase inhibitor p21Sdi1/Cip1/Waf1 by nitric oxide-
J Vasc Surg 2000;31:1214–28. generating vasodilator in vascular smooth muscle cells. J Biol Chem
17. Maeda Y, Ikeda U, Oya KI, et al. Endogenously generated nitric oxide 1997;272:10050–7.
by nitric-oxide synthase gene transfer inhibits cellular proliferation. 40. Gooch KJ, Dangler CA, Frangos JA. Exogenous, basal, and flow-
J Pharmacol Exp Ther 2000;292:387–93. induced nitric oxide production and endothelial cell proliferation.
18. Vadiveloo PK, Keramidaris E, Morrison WA, Stewart AG. J Cell Physiol 1997;171:252–8.
Lipopolysaccharide-induced cell cycle arrest in macrophages occurs 41. Ignarro LJ, Napoli C, Loscalzo J. Nitric oxide donors and cardiovas-
independently of nitric oxide synthase II induction. Biochim Biophys cular agents modulating the bioactivity of nitric oxide: an overview. Circ
Acta 2001;1539:140–6. Res 2002;90:21–8.
JACC Vol. 62, No. 2, 2013 Napoli et al. 95
July 9, 2013:89–95 NO and Cell Proliferation

42. Napoli C, Ignarro LJ. Nitric oxide-releasing drugs. Annu Rev Phar- 63. Tepper OM, Galiano RD, Capla JM, et al. Human endothelial
macol Toxicol 2003;47:97–123. progenitor cells from type II diabetics exhibit impaired proliferation,
43. Pautz A, Art J, Hahn S, Nowag S, Voss C, Kleinert H. Regulation of adhesion, and incorporation into vascular structures. Circulation 2002;
the expression of inducible nitric oxide synthase. Nitric Oxide 2010;23: 106:2781–6.
75–93. 64. Avogaro A, Toffolo G, Kiwanuka E, et al. L-Arginine-nitric oxide
44. Hogg ME, Varu VN, Vavra AK, et al. Effect of nitric oxide on neo- kinetics in normal and type 2 diabetic subjects: a stable-labelled 15N
intimal hyperplasia based on sex and hormone status. Free Radic Biol arginine approach. Diabetes 2003;52:795–802.
Med 2011;50:1065–74. 65. Thum T, Fraccarollo D, Schultheiss M, et al. Endothelial nitric oxide
45. Sharma RV, Tan E, Fang S, Gurjar MV, Bhalla RC. NOS gene synthase uncoupling impairs endothelial progenitor cell mobilization
transfer inhibits expression of cell cycle regulatory molecules in vascular and function in diabetes. Diabetes 2007;56:666–74.
smooth muscle cells. Am J Physiol 1999;276:H1450–9. 66. Chen YH, Lin SJ, Lin FY, et al. High glucose impairs early and late
46. Ishigami M, Swertfeger DK, Hui MS, Granholm NA, Hui DY. endothelial progenitor cells by modifying nitric oxide-related but not
Apolipoprotein E inhibition of vascular smooth muscle cell prolifera- oxidative stress-mediated mechanisms. Diabetes 2007;56:1559–68.
tion but not the inhibition of migration is mediated through activation 67. Niwano K, Arai M, Tomaru K, Uchiyama T, Ohyama Y,
of inducible nitric oxide synthase. Arterioscler Thromb Vasc Biol 2000; Kurabayashi M. Transcriptional stimulation of the eNOS gene by the
20:1020–6. stable prostacyclin analogue beraprost is mediated through cAMP-
47. Pignatti C, Tantini B, Stefanelli C, et al. Nitric oxide mediates either responsive element in vascular endothelial cells: close link between
proliferation or cell death in cardiomyocytes: involvement of poly- PGI2 signal and NO pathways. Circ Res 2003;93:523–30.
amines. Amino Acids 1999;16:181–90. 68. Miyahara Y, Ohnishi S, Obata H, et al. Beraprost sodium enhances
48. Taylor EL, Li JT, Tupper JC, Rossi AG, Winn RK, Harlan JM. GEA neovascularization in ischemic myocardium by mobilizing bone marrow
3162, a peroxynitrite donor, induces Bcl-2-sensitive, p53-independent cells in rats. Biochem Biophys Res Commun 2006;349:1242–9.
apoptosis in murine bone marrow cells. Biochem Pharmacol 2007;74: 69. Di Stefano R, Barsotti MC, Melillo E, et al. The prostacyclin analogue
1039–49. iloprost increases circulating endothelial progenitor cells in patients
49. Dai X, Faber JE. Endothelial nitric oxide synthase deficiency causes with critical limb ischemia. Thromb Haemost 2008;100:871–7.
collateral vessel rarefaction and impairs activation of a cell cycle gene 70. Kawabe J, Yuhki K, Okada M, et al. Prostaglandin I2 promotes
network during arteriogenesis. Circ Res 2010;106:1870–81. recruitment of endothelial progenitor cells and limits vascular remod-
50. Lepic E, Burger D, Lu X, Song W, Feng Q. Lack of endothe- eling. Arterioscler Thromb Vasc Biol 2010;30:464–70.
lial nitric oxide synthase decreases cardiomyocyte proliferation and 71. Comini L, Bachetti T, Cargnoni A, et al. Therapeutic modulation of
delays cardiac maturation. Am J Physiol Cell Physiol 2006;291: the nitric oxide: all ACE inhibitors are not equivalent. Pharmacol Res
C1240–6. 2007;56:42–8.
51. Wong JC, Fiscus RR. Essential roles of the nitric oxide (no)/cGMP/ 72. Ferrari R, Fox K. Insight into the mode of action of ACE inhibition in
protein kinase G type-Ia (PKG-Ia) signaling pathway and the atrial coronary artery disease: the ultimate ‘EUROPA’ story. Drugs 2009;69:
natriuretic peptide (ANP)/cGMP/PKG-Ia autocrine loop in promoting 265–77.
proliferation and cell survival of OP9 bone marrow stromal cells. J Cell 73. Li H, Förstermann U. Prevention of atherosclerosis by interference
Biochem 2011;112:829–39. with the vascular nitric oxide system. Curr Pharm Des 2009;15:
52. Leri A, Kajstura J, Anversa P. Role of cardiac stem cells in cardiac 3133–45.
pathophysiology: a paradigm shift in human myocardial biology. Circ 74. Napoli C, Ackah E, De Nigris F, et al. Chronic treatment with nitric
Res 2011;109:941–61. oxide-releasing aspirin reduces plasma low-density lipoprotein oxida-
53. Napoli C, Hayashi T, Cacciatore F, et al. Endothelial progenitor cells tion and oxidative stress, arterial oxidation-specific epitopes, and
as therapeutic agents in the microcirculation: an update. Atherosclerosis atherogenesis in hypercholesterolemic mice. Proc Natl Acad Sci U S A
2011;215:9–22. 2002;99:12467–70.
54. Mujoo K, Sharin VG, Bryan NS, et al. Role of nitric oxide signaling 75. de Nigris F, Mancini FP, Balestrieri ML, et al. Therapeutic dose of
components in differentiation of embryonic stem cells into myocardial nebivolol, a nitric oxide-releasing beta-blocker, reduces atherosclerosis
cells. Proc Natl Acad Sci U S A 2008;105:18924–9. in cholesterol-fed rabbits. Nitric Oxide 2008;19:57–63.
55. Ybarra N, del Castillo JR, Troncy E. Involvement of the nitric oxide- 76. Napoli C, Loscalzo J. Nitric oxide and other novel therapies for
soluble guanylyl cyclase pathway in the oxytocin-mediated differentia- pulmonary hypertension. J Cardiovasc Pharmacol Ther 2004;9:1–8.
tion of porcine bone marrow stem cells into cardiomyocytes. Nitric 77. Zuckerbraun BS, Shiva S, Ifedigbo E, et al. Nitrite potently inhibits
Oxide 2011;24:25–33. hypoxic and inflammatory pulmonary arterial hypertension and smooth
56. Napoli C, Williams-Ignarro S, de Nigris F, et al. Beneficial effects of muscle proliferation via xanthine oxidoreductase-dependent nitric oxide
concurrent autologous bone marrow cell therapy and metabolic inter- generation. Circulation 2010;121:98–109.
vention in ischemia-induced angiogenesis in the mouse hindlimb. Proc 78. Broderick KE, Alvarez L, Balasubramanian M, et al. Nitrosyl-
Natl Acad Sci U S A 2005;102:17202–6. cobinamide, a new and direct nitric oxide releasing drug effective
57. de Nigris F, Balestrieri ML, Williams-Ignarro S, et al. Therapeutic in vivo. Exp Biol Med (Maywood) 2007;232:1432–40.
effects of autologous bone marrow cells and metabolic intervention in 79. Cabrales P, Han G, Roche C, Nacharaju P, Friedman AJ,
the ischemic hindlimb of spontaneously hypertensive rats involve Friedman JM. Sustained release nitric oxide from long-lived circulating
reduced cell senescence and CXCR4/Akt/eNOS pathways. nanoparticles. Free Radic Biol Med 2010;49:530–8.
J Cardiovasc Pharmacol 2007;50:424–33. 80. Smith RA, Murphy MP. Mitochondria-targeted antioxidants as ther-
58. Balestrieri ML, Lu SJ, de Nigris F, et al. Therapeutic angiogenesis in apies. Discov Med 2011;11:106–14.
diabetic apolipoprotein E-deficient mice using bone marrow cells, 81. Terai T, Nagano T. Fluorescent probes for bioimaging applications.
functional hemangioblasts and metabolic intervention. Atherosclerosis Curr Opin Chem Biol 2008;12:515–21.
2010;209:403–14. 82. Nagano T. Bioimaging probes for reactive oxygen species and reactive
59. Napoli C, Farzati B, Sica V, et al. Beneficial effects of autologous bone nitrogen species. J Clin Biochem Nutr 2009;45:111–24.
marrow cell infusion and antioxidants/L-arginine in patients with chronic 83. McQuade LE, Lippard SJ. Fluorescent probes to investigate nitric
critical limb ischemia. Eur J Cardiovasc Prev Rehabil 2008;15:709–18. oxide and other reactive nitrogen species in biology (truncated form:
60. Casamassimi A, Grimaldi V, Infante T, et al. Adult stem cells and the fluorescent probes of reactive nitrogen species). Curr Opin Chem Biol
clinical arena: are we able to widely use this therapy in patients with chronic 2010;14:43–9.
limbs arteriopathy and ischemic ulcers without possibility of revasculari- 84. Zhang J, Hong H. Selective fluorescent activation for bioimaging the
zation? Cardiovasc Hematol Agents Med Chem 2012;10:99–108. expression of nitric oxide in cellular and in vivo systems. Methods Mol
61. Hamed S, Brenner B, Roguin A. Nitric oxide: a key factor behind the Biol 2011;704:57–71.
dysfunctionality of endothelial progenitor cells in diabetes mellitus
type-2. Cardiovasc Res 2011;91:9–15.
62. Fadini GP, Miorin M, Facco M, et al. Circulating endothelial Key Words: cardiovascular diseases - cell cycle - cell proliferation -

progenitor cells are reduced in peripheral vascular complications of diagnostic and therapeutic tools - imaging - nitric oxide -
type 2 diabetes mellitus. J Am Coll Cardiol 2005;45:1449–57. type 2 diabetes.

S-ar putea să vă placă și