Sunteți pe pagina 1din 12

Reviews

Mitochondrial quality control


mechanisms as molecular targets
in cardiac ageing
Anna Picca1,5, Robert T. Mankowski2,5, Jonathon L. Burman2,3,6, Luca Donisi1,2,
Jae-​Sung Kim4, Emanuele Marzetti1* and Christiaan Leeuwenburgh2*
Abstract | Cardiovascular disease is the leading cause of morbidity and mortality worldwide.
Advancing age is a major risk factor for developing cardiovascular disease because of the lifelong
exposure to cardiovascular risk factors and specific alterations affecting the heart and the
vasculature during ageing. Indeed, the ageing heart is characterized by structural and functional
changes that are caused by alterations in fundamental cardiomyocyte functions. In particular,
the myocardium is heavily dependent on mitochondrial oxidative metabolism and is especially
susceptible to mitochondrial dysfunction. Indeed, primary alterations in mitochondrial function,
which are subsequently amplified by defective quality control mechanisms, are considered to be
major contributing factors to cardiac senescence. In this Review , we discuss the mechanisms
linking defective mitochondrial quality control mechanisms (that is, proteostasis, biogenesis,
dynamics, and autophagy) to organelle dysfunction in the context of cardiac ageing. We also
illustrate relevant molecular pathways that might be exploited for the prevention and treatment
of age-​related heart dysfunction.

Cardiovascular disease is the main cause of morbidity to organelle dysfunction in the context of cardiac ageing.
1
Department of Geriatrics, and mortality in the general population and is especially Relevant molecular pathways that might be exploited
Neuroscience and prevalent among older adults1. Approximately 20% of for the prevention and treatment of age-​related heart
Orthopedics, Teaching people aged ≥80 years are at risk of heart failure, and dysfunction are also summarized.
Hospital “Agostino Gemelli”, those aged ≥65 years are at high risk of atrial fibrillation
Catholic University of the
Sacred Heart School of
and related stroke1. Indeed, advancing age is associated Cellular senescence and cardiac ageing
Medicine, Rome, Italy. with the progressive degeneration of blood vessels and An increase in the number of cardiac, muscular, endothe-
2
Department of Aging and the heart, making them more vulnerable to stressors lial, and endothelial progenitor senescent cells occurs in
Geriatric Research, University and contributing to increased morbidity and mor- several disease conditions associated with cardiovascu-
of Florida, Gainesville, FL, tality 2. In particular, the aged heart has increased lar dysfunction, including hypertension, atherosclerosis,
USA.
mass, ventricular wall thickness, and cardiomyocyte heart failure, and stroke9. Cellular senescence is charac-
National Institute of cross-​sectional area despite decreased cell number3–5. terized by genome instability (that is, nuclear DNA dam-
3

Neurological Disorders and


Stroke, National Institutes of
Aged cardiomyocytes show abnormalities in mito- age), telomere attrition, and mitochondrial dysfunction8.
Health, Bethesda, MD, USA. chondrial structure (enlarged organelles, matrix In particular, genome instability and telomere attrition
4
Department of Surgery,
derangement, and loss of cristae) and increased gen- are underlying causes of molecular damage and are indi-
University of Florida, eration of reactive oxygen species (ROS)6. These modi­ cated as primary hallmarks of cell senescence8 (Box 1).
Gainesville, FL, USA. fi­c ations are, in turn, associated with functional Conversely, antagonistic hallmarks in the ageing heart
5
These authors contributed impairments at the organ and whole-​body levels, includ- include mitochondrial dysfunction and cellular senes-
equally: Anna Picca, ing diastolic dysfunction, left ventricular hypertrophy, cence, which exert beneficial or protective effects at low
Robert T. Mankowski.
increased risk of atrial fibrillation, valvular degeneration, levels but are deleterious at high levels8. Finally, when
6
Deceased: Jonathon L. and decreased exercise capacity7. As such, alterations in the accumulating damage cannot be compensated for
Burman.
mitochondrial function, amplified by dysfunctional by homeostatic mechanisms, stem-​cell exhaustion and
*e-​mail: emanuele.marzetti@
quality control mechanisms, are considered to be major altered intercellular communication occur, contributing
policlinicogemelli.it;
cleeuwen@ufl.edu contributing factors to heart senescence8. to ageing (integrative hallmarks)8 (Box 1).
https://doi.org/10.1038/ In this Review, we discuss the mechanisms linking Mitochondrial dysfunction is a core feature of ageing
s41569-018-0059-z defective mitochondrial quality control (MQC) (Fig. 1) and forms the crossroads for several pathways related

NAture RevIeWs | CaRdiology volume 15 | september 2018 | 543


© 2018 Macmillan Publishers Limited, part of Springer Nature. All rights reserved.
Reviews

Key points vascular smooth muscle cell proliferation, and extracel-


lular matrix remodelling are associated with the genesis
• Older adults are especially vulnerable to developing cardiovascular disease owing to and progression of atherosclerosis and hypertension22,23.
long-​term exposure to risk factors and intrinsic cardiovascular alterations occurring Increases in pro-​inflammatory mediators are neces-
during ageing. sary for senescent cell removal. However, accumulation
• Mitochondrial quality control (MQC) operates through the coordination of various of senescent cells is paralleled by stem-​cell exhaus-
processes (proteostasis, biogenesis, dynamics, and mitophagy) to ensure cell tion and loss of function of regenerative cell lineages,
homeostasis.
which contribute to ageing24,25. The senescent state is
• Mitochondrial dysfunction, amplified by failing quality control processes, is believed under the control of two complex pathways (p53–p27
to be a major mechanism underlying cardiac ageing and cardiovascular disease.
(cyclin-​dependent kinase inhibitor 1) and p16INK4A
• Preclinical evidence suggests that modulation of MQC can be harnessed for (cyclin-​dependent kinase inhibitor 2A)–retinoblastoma-​
therapeutic benefit against cardiac ageing and cardiovascular disease.
related protein)24,25, but their role in the cardiovascular
• Current unknowns include the optimal window of MQC functioning to achieve system is currently undetermined.
cardioprotection, the timing and intensity of interventions, and noninvasively
A role for crosstalk between the endoplasmic reticu­
accessible biomarkers of MQC in the heart.
lum (ER) and mitochondria has been proposed in the
context of mitochondrial dysfunction (reviewed pre-
viously26). A major mediator in this crosstalk is Ca2+,
to senescence 10. For instance, genomic instability which is mainly stored in the ER. Following stress,
causes metabolic disarrangements that favour cellular Ca2+ is released from the ER, enters the mitochondria,
senescence and organismal ageing through a complex and increases both oxidative phosphorylation27 and ROS
response to persistent DNA damage11. This response generation28. ROS themselves can alter Ca2+ homeostasis
affects pathways that regulate bioenergetic metabolism by regulating the function of inositol 1,4,5-trisphosphate
and block cell anabolism induced by insulin, insulin-​like receptors and ryanodine receptors29,30.
growth factor I (IGF1), and somatotropin12. Oxidative stress is also modulated by steroidogen-
Similarly, telomere attrition promotes activation esis in the adrenal glands and by energy metabolism
of cellular tumour antigen p53 and inhibits mito- in the heart and brown adipose tissue. This control is
chondrial biogenesis and function in mice deficient exerted through the concerted activity of the antioxidant
in either telomerase reverse transcriptase (Tert−/−) or enzymes sulfiredoxin 1 and mitochondrial thioredoxin-​
telomerase RNA component (Terc−/−)13. Therefore, tel- dependent peroxide reductase (also known as perox-
omere shortening might limit mitochondrial turnover iredoxin III; PRDX3)31. PRDX3 is the most abundant
despite age-​related metabolic perturbations. In humans, and efficient mitochondrial hydrogen peroxide (H2O2)
leuko­c yte telomere length negatively correlates with scavenger in these tissues, and mitochondrial H2O2 is
γ-​glutamyltyrosine and γ-​glutamylphenylalanine, two buffered within the organelle with concomitant accu-
markers of oxidative stress14. Conversely, lysolipids have mulation of its oxidized form, PRDX3–SO2H. When
been found to be associated with phospholipase A2 PRDX3 antioxidant capacity is overwhelmed, H2O2 is
expression, which might indicate poor membrane flu- delivered into the cytosol and triggers the phosphoryl­
idity14. Furthermore, deficiency in the recycling auto- ation of mitogen-​activated protein kinase (MAPK)
phagic machinery contributes to cell senescence through 11–14, collectively known as p38 MAPK, resulting in
the accumulation of intracellular toxic waste15. downregulation of cortisone production31. The levels
Mitochondrial-​derived oxidative stress is thought of both PRDX3–SO2H and phosphorylated p38 MAPK
to have a pivotal role in the development of age-​related have a daily oscillation in several tissues, including the
conditions through irreversible damage to macromol­ heart. This variation suggests that mitochondrial H2O2
ecules and bioenergetic failure16,17. In addition, the acti- is released into the cytosol in a rhythmic manner and
vation of redox-​sensitive mediators, including nuclear supports the concept of a link between mitochondrial
factor-​κB (NF-​κB), modulates the transcription of biology and circadian rhythms31,32.
several pro-​inflammatory cytokines18. Under normal
conditions, transient NF-​κB activation in response to Oxidative stress and mtDNA mutations
oxidative stimuli ceases with resolution of the inflam- Reactive oxygen species. According to the ROS theory
matory reaction18. However, long-​term exposure to of ageing, mitochondria are the primary source and the
high levels of oxidants, as occurs during ageing, results immediate target of ROS within the cell33. However,
in chronic activation of the NF-​κB-mediated inflamma- the extent to which oxidative stress is involved in heart
tory response and cellular damage18. A set of NF-​κB- senescence remains to be conclusively established.
responsive cytokines and chemokines has been identi- Increased mitochondrial size, reduced mitochondrial
fied as part of the senescence-​associated secretory phe- respiratory capacity, and higher ROS production have
notype (SASP)19 (Fig. 2). An increase in inflammatory been reported in cardiac ageing34. Because of their post-
mediators leads to the expression of inflammatory pro- mitotic nature, cardiomyocytes are devoid of replica-
teins involved in extracellular matrix remodelling and tive dilution of damage and are, therefore, particularly
of pro-​inflammatory cytokines (such as tumour necro- vulnerable to ROS-​mediated lesions. Indeed, defective
sis factor, IL-1α, IL-1β, and IL-6). SASP mediators or electron transport chain (ETC) subunits, including sites
their products (such as inducible nitric oxide synthase, in complexes I and III, contribute to ROS generation35,36.
cytochrome c oxidase, and prostaglandin E2) are prom- However, a causative role for ROS in mitochondrial dys-
inent sources of ROS20,21. Indeed, endothelial damage, function is debated. Although ROS have been linked to

544 | september 2018 | volume 15 www.nature.com/nrcardio


© 2018 Macmillan Publishers Limited, part of Springer Nature. All rights reserved.
Reviews

Mitochondrial
Mitophagy
biogenesis dynamics

PGC1α PPARGC1A
TF
Depolarization PINK1
DNM1L
PGC1α NRF1/NFE2L2 Parkin
Beclin 1
TF ROS
p62
TFAM LC3 Ubiquitin
NRF1/2 MFN1
TF Lysosome
Fusion Fission
TFAM OPA1
OPA1
mtDNA
MFN2 Phagophore

mtDNA OPA1 OPA1


transcription Autophagosome
mtDNA
replication OPA1
DNM1L
FIS1
Mitochondrion

Degradation

Fig. 1 | Mitochondrial quality control pathways. Mitochondrial content. Changes in mitochondrial morphology are under the control
homeostasis is ensured through the coordination of mitochondrial of fusion (mitofusin 1 (MFN1), MFN2, and mitochondrial dynamin-​like
biogenesis, dynamics, and autophagy. After appropriate stimuli (such as 120 kDa protein (OPA1)) and fission (dynamin 1-like protein (DNM1L)
exercise stimulus), the upregulation of proliferator-​activated receptor-​γ and mito­chondrial fission 1 protein (FIS1)) proteins. These factors regulate
co-activator 1α (PGC1α) and other transcription factors (TFs) activates the mitochondrial turnover by facilitating the dilution and clearance of
transcription of nuclear genes encoding mitochondrial proteins such as damaged organelles. Mitochondrial components are eventually recycled
mitochondrial transcription factor A (TFAM). TFAM is then imported into through a specialized autophagic pathway , known as mitophagy. LC3,
mitochondria by the protein import machinery and reaches its final microtubule-​associated proteins 1A/1B light chain 3; NRF1, nuclear
destination on mitochondrial DNA (mtDNA). Here, TFAM upregulates the respiratory factor 1; NRF2, nuclear factor erythroid 2-related factor 2; p62,
expression of genes encoding electron transport chain subunits, resulting sequestosome 1; parkin, E3 ubiquitin-​protein ligase parkin; PINK1,
in increased oxygen consumption, ATP synthesis, and mitochondrial serine/threonine-​protein kinase PINK1; ROS, reactive oxygen species.

mitochondrial DNA (mtDNA) damage (Fig. 2), a number antioxidant enzymes, in older mice did not affect
of findings indicate mtDNA mutations primarily aris- mitochondrial function44.
ing from errors during mtDNA replication as a major Nevertheless, several findings still support the ROS
factor in cardiac ageing37,38. Indeed, mice expressing a theory of ageing. The implementation of redox-​sensitive
proofreading-​deficient mtDNA polymerase-​γ (mtDNA mass spectrometry approaches allowed the detection
mutator mice) have increased rates of mtDNA mutation of increased production of ROS in mutator mice and
and several features of ageing, including increased risk of indicated the existence of a mitochondrial redox signal-
cardiac disease39–41. Furthermore, long-​lived naked mole ling cascade in response to chronic exposure to ROS40.
rats reach very old age despite oxidative stress through Such a signalling system would operate through the
uncertain cytoprotective mechanisms42. Of note, anti-​ generation of a pro-​inflammatory environment and is
ageing and ROS-​limiting calorie restriction inter- hypothesized to contribute to the accelerated ageing
ventions alone are not sufficient to rescue the ageing of mutator mice45. Indeed, expression of the antioxi-
phenotype of mutator mice, which questions the idea dant enzyme catalase rescues some of the age-​related
that oxidative stress is the primary mechanism deter- features in this model (such as heart enlargement and
mining ageing phenotypes in this experimental model43. cardiomyo­pathy)39,41. Furthermore, superoxide produc-
Collectively, these findings indicate that accumulation of tion correlates with ageing and damage to mitochondrial
mtDNA mutations during ageing might compromise cell lipids and proteins3.
signalling and promote cell impairment independently In further support of a role for ROS-​mediated dam-
of oxidative stress. age and mitochondrial decline in cardiac ageing is the
Most of the concerns raised against this hypothesis finding that supplementation with the natural polyam-
pertain to the extent to which increased ROS produc- ine spermidine counteracts age-​related declines in the
tion causes mtDNA damage40. Deep sequencing and expression and function of ETC complex I in mice46.
PCR-​based mutation-​detection methods have not identi- Although the mechanism of action of spermidine
fied a mutation spectrum consistent with ROS-​mediated might be multifactorial, current evidence indicates that
mutagenesis in mtDNA from flies and humans37,38. spermidine acts as an antioxidant46.
In this regard, a 50% reduction in manganese super- Additional studies showed that B2 bradykinin recep-
oxide dismutase, one of the main mitochondrial tor deficiency exacerbates cardiac dysfunction in aged

NAture RevIeWs | CaRdiology volume 15 | september 2018 | 545


© 2018 Macmillan Publishers Limited, part of Springer Nature. All rights reserved.
Reviews

Box 1 | Hallmarks of senescence in cardiac ageing dysfunction, its relative levels must exceed a certain
threshold that is dependent on the type of mutation and
Primary hallmarks of ageing (the cause of molecular the energy demand of the affected cell type58.
damage): In the setting of cardiac ageing, the accumulation of
• Genome instability heteroplasmic mtDNA mutations can result in a mosaic
• Telomere shortening of cells that can reach a critical threshold of mtDNA
Secondary hallmarks of ageing (which arise as a response mutations59. mtDNA deletions accumulate steadily in
to damage to mitigate the insult, but their persistence cardiomyocytes of Twnk (encoding twinkle mtDNA
has detrimental effects): heli­case) mutant mice during ageing53. Notably, the
• Mitochondrial dysfunction mutational load is paralleled by mitochondrial defi-
• Oxidative stress ciency, as measured by cytochrome c oxidase staining,
• Systemic inflammation and is associated with arrhythmias4. These findings sup-
port the ideas that foci of dysfunctional mitochondria in
cardiomyocytes are sufficient for inducing whole-​organ
mice via increased p53 expression, reduced mitochon- dysfunction and that mtDNA damage contributes to
drial biogenesis through lower proliferator-​activated cardiac ageing.
receptor-​γ co-​a ctivator 1α (PGC1α) expression, However, several findings indicate that the abun-
increased inflammation, and increased oxidative stress47. dance of mtDNA mutations rarely exceeds 1% in aged
The equivocal results about the role of ROS in mito- humans, which is well below the phenotypic expression
chondrial dysfunction during ageing might be explained, threshold60,61. Moreover, the idea of mtDNA deletions
at least in part, by the experimental approaches fol- causing ageing and age-​related diseases is contradicted
lowed by different studies. For instance, variability in by the study of so-​called Mito-​mice that harbour a
time points of measurements, degree of mitochondrial 4,696 bp mtDNA deletion uniformly distributed across
purity, assays and techniques used, data analyses, and various tissues. These mice, despite showing accu-
samples used to determine ROS concentrations might mulation of up to 60% mtDNA deletion in different
all affect the results. Nevertheless, although low levels tissues, have no signs of mitochondrial dysfunction
of mitochondrial-​derived ROS potentially promote or disease manifestation, whereas tissues with >85%
autophagy signalling and organelle clearing, high and mtDNA deletion show mitochondrial dysfunction and
sustained levels of ROS clearly cause mitochondrial disease phenotypes62,63.
and cellular toxicity48. Therefore, low levels of ROS are Another study argued against a central role for
beneficial and instrumental for normal cellular function mtDNA mutations in the ageing process by using a
and cardioprotection through a hormetic response49–51. highly sensitive random mutation capture assay 64.
In particular, the leakage of ROS outside mitochondria In this study, mtDNA mutation frequency in the brain
activates a H2O2-mediated cell-​warning system for oxi- and heart of aged, wild-​type mice was approximately
dative stress that acts as a retrograde signal to nuclear-​ tenfold lower than previously reported. This finding
targeted cytosolic pathways52. When intracellular ROS is in contrast to the 500-fold higher mtDNA mutation
concentrations overwhelm antioxidant defences, the load of heterozygous mutator mice, which have a normal
resulting oxidative stress can induce loss of ROS sig- lifespan and no signs of premature ageing64. However,
nal localization and disruption of cell homeostasis53. a caveat of this study is that large-​scale mtDNA dele-
However, the relationship between endogenous levels of tions were not detected, which instead have been shown
ROS and ‘healthy’ cardiac ageing is an area that requires to accumulate in aged human tissues65. Furthermore,
further clarification for therapeutic exploitation. species-​specific differences that might causally relate
mtDNA mutations to ageing in humans could not be
mtDNA mutations. Cells have hundreds to thousands ruled out66,67. Therefore, further studies are needed to
of copies of mtDNA. The accrual of mtDNA mutations clarify species-​specific differences in somatic mtDNA
exceeding a certain threshold (that is, >60–80% hetero- mutation accumulation during ageing and to deter-
plasmy based on the type of mutation) results in the syn- mine whether the lower frequency of mtDNA mutation
thesis of a critical mass of defective ETC components54. reported in aged mice might be functionally relevant to
This eventually leads to mitochondrial dysfunction and human ageing68.
phenotypic expression54. mtDNA mutations can be pres- Computational approaches support the idea that
ent in only a subset of genome copies (heteroplasmy) only mtDNA mutations occurring early in life have suf-
or in all copies (homoplasmy). These mutations can ficient time to undergo clonal expansion and cause focal
be either inherited or formed de novo in the oocyte or oxidative phosphorylation dysfunction during human
embryo. Unless point mutations affect regions that are ageing56,69. Indeed, focal oxidative phosphorylation dys-
relevant to regulating mtDNA replication55, negative function is observed over the age of 30 years, and the
selection does not seem to occur against low levels of prevalence of oxidative phosphorylation-​deficient cells
heteroplasmic mtDNA point mutations. Under these in the affected tissues (for example, colonic epithelium
circumstances, clonal expansion of mtDNA mutations and skeletal muscle) increases with age70.
in somatic tissues follows the neutral drift principle56,57. Taken together, mitochondrial respiratory deficiency,
As subsequent cycles of mtDNA replication occur, lev- ROS generation, and mtDNA damage are central to
els of mutated mtDNA can either increase or decrease. age-​associated dysfunction of cardiomyocytes (Fig. 2).
For a mutation to cause oxidative phosphorylation Mitochondrial-​t argeted or untargeted antioxidant

546 | september 2018 | volume 15 www.nature.com/nrcardio


© 2018 Macmillan Publishers Limited, part of Springer Nature. All rights reserved.
Reviews

therapies, activation of organelle-​specific autophagy, oxidative phosphorylation is required to sustain


particularly mitophagy, and additional strategies for the the Ca2+-dependent contraction of cardiomyocytes.
selective elimination of mtDNA mutations in the aged Consequently, the maintenance of mitochondrial
heart might, therefore, be promising areas of investigation homeo­stasis is crucial to proper heart function. How­
for developing treatments against cardiovascular disease. ever, mitochondrial function is not limited to energy
provision. This organelle is a hub for many other activ-
Mitochondrial quality control failure ities within the cell, including metabolic signalling,
The heart tissue includes cardiomyocytes, fibroblasts, iron–sulfur cluster and haem biosynthesis, regulation of
smooth muscle cells, and endothelial cells, with a con- programmed cell death, and Ca2+ and iron buffering74.
tractile component accounting for about 30–40% of The intimate dependence of the heart on mitochondrial
these cells in number and 75% in volume71,72. Like neu- function highlights the vulnerability of cardiac tissue to
rons and skeletal myocytes, adult cardiomyocytes are mitochondrial dysfunction in ageing and disease39,40,75.
postmitotic cells with very limited regenerative cap­ MQC mechanisms are, therefore, crucial to preserving
acity73. Therefore, damaged organelle clearing to ensure cardiomyocyte homeostasis by preventing the expan-
organ integrity and maintenance relies on the recycling sion of the primary mitochondrial defect(s). MQC
of cardiomyocyte components. involves the coordinated regulation of a hierarchical
Cardiac muscle cells have a high energy demand network of pathways acting sequentially from individ-
and are, therefore, enriched in mitochondria (35% ual molecules to the whole organelle76. Antioxidant sys-
of cell volume). Most of the ATP generated through tems function as the primary line of defence to prevent

↑ AMP:ATP
FIS1 DMN1L
↑ AMPK ↓ PGC1α
MFN1 Telomere PGC1α PPARGC1A
OPA1 TF
FOXO1/3 ↓ SIRT1
OPA1 (pFOXO1/3) ROS
MFN2
PGC1α
↓ Fusion ↑ Fission Nucleus TF NRF1/NFE2L2
↓ NAD+ NF-κB

Ubiquitin Depolarized ROS TFAM


mitochondrion DNA damage ↑ PARP Stress response NRF1/2
TF
ROS
Mitochondrion-
↓ PINK1 derived vesicle TFAM
Parkin Telomere erosion
mtDNA
Beclin 1
Phagophore p62 SASP
Telomere mtDNA
transcription
mtDNA
replication
Lysosome
Mitochondrion
↓ Mitochondrial
biogenesis

Autophagosome ↓ Mitophagy DAMP release Senescence

TLR9 CGAS, STING, and TBK1

Inflammageing

Fig. 2 | Nucleus–mitochondrion crosstalk during cardiac ageing. This multi-​pathway derangement leads to cellular stress, induction of
The age-​related surge in generation of reactive oxygen species (ROS) the senescence-​associated secretory pathway (SASP), and senescence.
arising primarily from the accumulation of dysfunctional mitochondria CGAS, cGMP-​A MP synthase; DAMP, damage-​a ssociated molecular
causes nuclear DNA damage and activates 5′-AMP-​activated protein pattern; DNM1L , dynamin 1-like protein; FIS1, mitochondrial fission 1
kinase (AMPK) signalling. The latter, in turn, inhibits NAD-​dependent protein; MFN, mitofusin; mtDNA , mitochondrial DNA ; NRF1, nuclear
protein deacetylase sirtuin 1 (SIRT1) activity. The decrease in NAD+ levels respiratory factor 1; NRF2, nuclear factor erythroid 2-related factor 2;
in the setting of oxidative stress further affects SIRT1, resulting in OPA1, mitochondrial dynamin-​like 120 kDa protein; p62, sequestosome 1;
decreased levels of proliferator-​activated receptor-​γ co-​activator 1α PARP, poly[ADP-​r ibose] polymerase; parkin, E3 ubiquitin-​p rotein
(PGC1α), leading to a decline in mitochondrial biogenesis; upregulation of ligase parkin; PINK1, serine/threonine-​protein kinase PINK1; STING,
nuclear factor-​κ B (NF-​κ B), leading to inflammation; and decreased stimulator of interferon genes protein; TBK1, serine/threonine-​protein
expression and phosphorylation of forkhead box protein O1 (FOXO1) and kinase TBK1; TFAM, mitochondrial transcription factor A ; TLR9, Toll-​like
FOXO3, transcription factors (TFs) that participate in cytoprotection. receptor 9.

NAture RevIeWs | CaRdiology volume 15 | september 2018 | 547


© 2018 Macmillan Publishers Limited, part of Springer Nature. All rights reserved.
Reviews

mitochondrial molecular damage. When damage has is also under circadian control. This observation is par-
occurred, a second battery of MQC pathways is engaged, ticularly relevant during periods of low activity when
which includes mitochondrial repair processes (that is, heart rate and blood pressure are low, creating optimal
mtDNA-​repair systems, reductase systems, and chaper­ conditions for sarcomere repair and regeneration80.
ones) 77. An intramitochondrial proteolytic system Along with the UPS, mitoproteases act as a first line
intervenes to clear irreversibly damaged mitochondrial of defence against mild mitochondrial damage86. In the
proteins for their subsequent replacement. Sustained mitochondrial matrix, protein turnover is controlled by
insults that overwhelm molecular MQC trigger path- three AAA proteases: the soluble mitochondrial Lon
ways acting at the whole organelle level. Indeed, dam- protease homologue (LONP1) and mitochondrial ATP-​
aged mitochondria can fuse with neighbouring, intact dependent Clp protease (CLPP), and the mitochondrial
organelles to dilute focal dysfunction, whereas severely inner membrane-​b ound m-​A AA protease87. In the
damaged mitochondria are segregated from the network intermembrane space, mitochondrial protein quality
through fission and eventually degraded by a specialized is ensured by the membrane-​bound ATP-​dependent
form of autophagy78,79 (Fig. 2). Derangements at any level zinc metalloproteinase YME1L1, the soluble mito-
of the MQC axis can result in amplification of mitochon- chondrial serine protease HTRA2, the mitochondrial
drial dysfunction, energy shortage, and ultimately loss metalloendo­p eptidase OMA1, and the mito­c hon­
of cell viability (Fig. 2). The following sections describe drial presenilins-​associated rhomboid-​l ike protein
mitochondrial proteostasis, biogenesis, dynamics, and (PARL)86. The level and activity of these mitoproteases
autophagy as core MQC mechanisms. change during ageing. For instance, the expression and
function of LONP1 decrease with age88. The relevance
Proteostasis. Cardiac homeostasis and activity are of mitoprotease activity is epitomized by the deletion of
dependent on the tight regulation of protein synthe- genes encoding AFG3-like protein 2, CLPP, and PARL,
sis and degradation, given that cardiomyocytes are which causes severe defects in mice (such as axonal
enriched in the myofibrillar proteins myosin and actin degeneration, multisystem disorder, and cachexia)
that form, in association with other protein components, and ultimately shortens murine lifespan through
the contractile subunit known as the sarcomere80. In the mitochondrial dysfunction89–91.
context of cardiac workload, the demands of high con- A role for ER stress in mitochondrial proteostasis has
tractile activity result in increased protein synthesis, and also been acknowledged. cAMP-​dependent transcrip-
this enhanced rate of synthesis can lead to physiological tion factor ATF4, which is involved in the unfolded pro-
or pathological cardiac hypertrophy. Reduced cardiac tein response, has been shown to induce the expression
mass has been shown to result from either a substan- of E3 ubiquitin-​protein ligase parkin, which regulates
tial decrease in protein synthesis (by as much as 50% mitochondrial fission, bioenergetics, and mitophagy92
in <2 weeks)81 or an elevated activity of the ubiquitin– by favouring transient Ca2+ transfer from the ER to
proteosome system (UPS)82. These responses suggest the mitochondria93. Another mediator of the unfolded pro-
existence of mechanosensors in cardiomyocytes ensur- tein response, eukaryotic translation initiation factor
ing a mechanoproteostasis link between the intensity 2α-​kinase 3, which is enriched at the mitochondria-​
of contraction and the molecular pathways regulating associated ER membranes, favours the propagation of
protein synthesis and degradation82. ROS from the ER to the mitochondria94.
Regulation of protein turnover, from synthesis to deg- Cardiac dysfunction of different aetiologies (including
radation, is also relevant to tissue architecture. Whereas ischaemia, pressure or volume overload, and arrhythmia)
mature myofibrils are confined to the perinuclear region, has been associated with redox imbalance that affects
the Z-​disc is located in a subcellular region where defined several processes, including ROS-​mediated regulatory
sarcomeric structures are assembled80. Through a highly pathways, the expression and/or function of proteins
dynamic process that depends on proper protein con- involved in Ca2+ homeostasis, and structural alterations
formations and interactions, the addition of new sarco- of myofibrillar proteins95,96. In the context of an oxi-
meres to the structure occurs on a short timescale. In dative environment, post-​translational modifications
response to external mechanical stressors (pressure or (such as disulfide bonds and carbonylation) alter the
volume overload), sarcomeric addition occurs either in conformation of myofibrillar proteins and are likely to
parallel (concentric hypertrophy) or in series (eccentric induce functional changes of the sarcomeric contractile
hypertrophy). The production of functional proteins apparatus. Furthermore, specific subunits of the 19S pro-
requires the completion of protein folding and 3D teasome become oxidized, which results in a significant
organiza­tion, which involves transient associations with reduction in the 26S proteasome activity97.
molecular chaperones. An increased oxidative environ- As a countermeasure to the accumulation of dam-
ment in cardiomyocytes might contribute to primary aged macromolecules within the cell, elevated levels
sequence modifications and protein misfolding. of ROS have been reported to trigger autophagy by
Protein synthesis operates only through the ribosomal mechanisms that are not fully understood98,99. A deeper
machinery, but three distinct and complementary systems understanding of the molecular pathways (both dele­
have been identified for degrading and recycling cellu- terious and protective) that are activated by redox
lar components and organelles: the calpain–calpastatin imbalance and that regulate cardiac proteostasis is
system, the UPS, and macroautophagy83–85. These three highly important to develop therapeutic approaches
processes are intimately interconnected and orchestrate aimed at reducing their harmful consequences and to
cardiac sarcomere degradation83–85. Cardiac proteostasis prevent cardiac dysfunction.

548 | september 2018 | volume 15 www.nature.com/nrcardio


© 2018 Macmillan Publishers Limited, part of Springer Nature. All rights reserved.
Reviews

Mitochondrial bioenergetics and biogenesis. Several content and an inducer of mitochondrial biogenesis via
studies have reported a decline in mitochondrial res- a 5ʹ-AMP-​activated protein kinase (AMPK)–PGC1α
piration and decreases in mitochondrial membrane signalling cascade116.
potential during ageing both in laboratory rodents In addition to PGC1α, the mitochondrial transcrip-
and in humans34,41. However, the extent to which ETC tion factor A (TFAM), through its binding to mtDNA,
complex I–IV-​supported respiration is reduced and has been indicated as a relevant modulator of mitochon-
the tissue-​specificity of these reductions are contro- drial biogenesis117 (Fig. 1). This interaction is modulated
versial34,100,101. Furthermore, whether ETC functional by several mechanisms, including regulation of TFAM
decline and increased ROS generation are upstream expression and turnover, post-​translational modifi-
or secondary to age-​related changes in mitochondrial cations and differential affinity of TFAM to specific
energy transfer systems (for example, creatine kinase mtDNA regions, TFAM sliding on mtDNA filaments
(CK) energy exchange) or organelle content variations and cooperative binding among TFAM molecules, and
is uncertain100. modulation of protein–protein interactions117.
A causal link between the age-​dependent decline in Apart from the specific mechanisms responsible
heart muscle performance and the decrease in the CK for mitochondrial functional impairment, differential
transfer system is plausible considering both its role in decline in ETC activity has been found between inter-
supplying high-​energy compounds to the cytosol and fibrillar mitochondria and subsarcolemmal mitochon-
substrate to the ETC and the observation of decreased dria44, suggesting that subclasses of mitochondria are
phosphocreatine:ATP ratios in aged human and rodent differentially susceptible to dysfunction118. Indeed, stud-
tissues100,102–105. However, additional explanations are ies by Hoppel and colleagues reported increased activity
available for the age-​related decline in ETC function, of mitochondrial citrate synthase and ETC complexes,
including changes in diffusion barriers within the myo- state 3 respiration rates, and abundance of respiratory
cardium that limit mitochondrial substrate availability cytochromes in interfibrillar mitochondria isolated
and variations in paracrine signalling103,106. In addition, from the rat heart119,120. This possibility suggests that
the increased susceptibility to heart disease in male ani- distinct therapeutic approaches are necessary to restore
mals suggests a role for age-​related changes in sex hor- bioenergetics in the two mitochondrial subpopulations.
mone levels differentially influencing ETC function in
the two sexes107. Indeed, mitochondria from the heart Mitochondrial dynamics. Mitochondria exist in a retic-
of ovariectomized rats show aberrant mitochondrial ular state in the myocardium, allowing electrochemical
morphology, overproduction of ROS, and impairment conductance of the proton motive force throughout
in basal and stress-​induced mitochondrial fission, which the mitochondrial network to facilitate network-​wide
are prevented by treatment with oestrogens and proges- ATP synthesis121–123 (Fig.  1). This structural organi-
terone107. Oestrogens can also modulate ATP synthe- zation, reminiscent of an electrical power grid, has
sis107. Moreover, results from muscle-​specific oestrogen been revealed from classic electron microscopy-​based
receptor knockout (Esr1−/−) mice suggest a link between studies and more recent fluorescence and 3D focused
nuclear receptor signalling and hormonal control of ion beam scanning electron microscopy structural
mitochondrial function and metabolism107. The role analyses122–124. Mitochondrial–mitochondrial contact
of sex hormones in the bioenergetic decline observed sites — gap junction-​like structures — are thought
during ageing is an attractive area of investigation106,108. to facilitate energy distribution between organelles.
Cardiac mitochondriogenesis is a complex nuclear– However, the molecular structure of these junctions
mitochondrial process that orchestrates both genome and how they change during ageing remain important
transcription and replication109 (Fig. 1). PGC1α is a mas- unanswered aspects. Morphometric analysis of mito-
ter regulator of mitochondrial biogenesis and energy chondrial structure has demonstrated that cardiomyo-
metabolism110,111. In the heart, PGC1α is induced at cyte mitochondrial networks deteriorate with age125,126.
birth to support the metabolic shift in fuel preference In particular, the area of the inner mitochondrial mem-
from glucose and lactate during the fetal period to brane substantially decreases with age, as shown by
fatty acids after birth112. The transcriptional activities morphometric analysis of electron microscopy images of
of a set of factors, including peroxisome proliferator-​ rodent heart muscle sections125. The inner mitochondrial
activated receptor-​α, steroid hormone receptor ERR1, membrane houses the ETC, the site of respiration and
and nuclear respiratory factor 1 (NRF1), are under the ATP production; ETC functional changes can, therefore,
control of PGC1α111. Through this regulation, PGC1α be expected to accompany changes in inner membrane
modulates mitochondrial biogenesis and energy metab- morphology.
olism. Mitochondrial content is substantially reduced An important area of future research will be to deter-
in the failing hearts of both rodents and humans113,114. mine the role of fission factors, such as dynamin 1-like
Moreover, downregulation of PGC1α signalling has been protein (DNM1L) and mitochondrial fission factor
observed in the setting of experimental heart failure115. (MFF)127, and fusion factors, such as mitochondrial
As such, discovering the cardiac mechanisms regulating dynamin-​like 120 kDa protein (OPA1) and mito-
PGC1α signalling might lead to the development of ther- fusin 1 (MFN1) and MFN2, in regulating mitochon-
apies aimed at stimulating mitochondrial biogenesis and drial morphology, Ca2+, and energy conductance inside
increasing energy production in the setting of higher adult cardiomyocytes. Towards this understanding,
contractile demand114. Hydrogen sulfide has been indi- murine knockout of Dnm1l results in dilated cardio-
cated as an important regulator of cardiac mitochondrial myopathy128. Similarly, heart-​specific Mfn1 and Mfn2

NAture RevIeWs | CaRdiology volume 15 | september 2018 | 549


© 2018 Macmillan Publishers Limited, part of Springer Nature. All rights reserved.
Reviews

Energy status Growth and survival Mitochondrial autophagy. Autophagy is an evolu-


tionarily conserved catabolic pathway that selectively
AMPK signalling Insulin signalling or nonselectively eliminates long-​lived, unnecessary, or
damaged proteins and organelles to ensure cell homeo-
• Increased ROS Insulin and IGF1
• Reduced ATP level stasis132. Of the three known types of autophagy (includ-
ing macroautophagy, chaperone-​mediated autophagy,
AKT1 and microautophagy), macroautophagy is the best-​
understood pathway. When autophagy is stimulated
by starvation and other stresses, a double-​membrane
AMPK RHEB phagophore proximal to the cellular cargos expands to
generate the autophagosome, which ultimately fuses
mTORC1 with the late endosome or lysosome to hydrolyse the
engulfed constituents132.
ULK1 TFEB Mitophagy is a form of selective autophagy that not
only prevents the accumulation of abnormal or dam-
aged mitochondria but also promotes the maintenance
BCL-2 and Beclin 1 Lysosomal biogenesis
of a stable number of healthy mitochondria within
Cardiac autophagy
cells (Fig. 1). A study in mito-​Keima-expressing mice
demonstrated that the heart was one of the most robust
Pro-autophagic Anti-autophagic mitophagic organs, substantiating the importance of
mitophagy for normal cardiac function133. The onset
Fig. 3 | Regulation of cardiac autophagy. Cardiomyocyte of mitophagy can be triggered by at least three differ-
energy status regulates autophagy via metabolic signalling. ent mechanisms in the cell134. Type I mitophagy largely
Under substrate deficit conditions or oxidative stress, resembles canonical autophagy and utilizes the classical
decreased ATP levels stimulate 5′-AMP-​activated protein
autophagy machinery, involving phosphatidylinositol
kinase (AMPK) activity and, therefore, autophagy via
activation of serine/threonine-​protein kinase ULK1 and 3-kinase class III (PI3K-​III) and phagophore assembly.
downstream apoptosis regulator BCL-2 and beclin 1 Type II mitophagy is instigated by recruitment and
through inhibition of autophagy suppressors (such as activation of the mitochondrial serine/threonine-​protein
mechanistic target of rapamycin complex 1 (mTORC1)). kinase PINK1 and the E3 ubiquitin-​protein ligase par-
At the same time, growth or cell survival stimuli activate kin to the outer mitochondrial membrane135 (Fig. 1).
insulin or insulin-​like growth factor I (IGF1) signalling in In healthy polarized mitochondria, PINK1 is imported
cardiomyocytes, leading to the induction of the insulin– into the inner mitochondrial membrane and matrix via
RACα serine/threonine-​protein kinase (AKT1) pathway. the translocase of the inner membrane (TIM)–trans-
Activation of GTP-​binding protein RHEB results in locase of the outer membrane (TOM) complex, where
inhibition of autophagy by autophagy suppressors
it is degraded by PARL. Upon depolarization, PINK1
(primarily mTORC1) and transcription factors related to
lysosomal biogenesis (such as transcription factor EB is stabilized within the TOM complex in the outer
(TFEB)). ROS, reactive oxygen species. mitochondrial membrane. Outer membrane stabiliza-
tion prevents matrix-​localized proteases from cleav-
ing PINK1, resulting in its accumulation on the outer
double knockout results in cardiac dysfunction in membrane of mitochondria with compromised mem-
mice128. However, heart-​specific Dnm1l, Mfn1, and brane potential136,137. Under these conditions, PINK1 can
Mfn2 triple-​knockout mice show a partial rescue of phosphorylate ubiquitin138,139, which in turn recruits and
myocardial function, demonstrating that the balance activates parkin. PINK1-induced phosphorylation of
between fission and fusion is an important requirement parkin at its Ser65 residue140 further drives the activation
for maintaining heart health129. of parkin activity, resulting in a feedforward phospho-​
Mechanistically, impairment of mitochondrial ubiquitylation cascade, which drives mitophagy to
dynamics enhances mitophagy128,130,131. Excess mito- completion. Ubiquitylated targets then interact with
phagy might lead to loss of healthy mitochondria and mitophagy receptors, including sequestosome 1 (p62)141,
energy decline128,130. However, because active mitochon- optineurin, tax1-binding protein 1, calcium-​binding and
drial dynamics and mitophagy have not been observed coiled-​coil domain-​containing protein 2 (ref.142), next to
in vivo in the heart, the role of mitochondrial dynamics BRCA1 gene 1 protein, and histone deacetylase 6 (ref.142).
factors in the heart remains unclear. In addition, because In the heart, PINK1 might have a role in phosphorylat-
heart-​specific knockout of essential genes is expected to ing MFN2 at Thr11 and Ser442 (ref.138), and phosphoryl-
result in pathological phenotypes, how more subtle alter- ated MFN2 is a well-​characterized substrate of parkin137.
ations in mitochondrial dynamics factors contribute to However, mitophagy can occur independently of parkin
cardiovascular disease susceptibility and cardiac ageing and PINK1. BCL-2-like protein 13, BCL-2/adenovirus
remains to be shown. When a molecular understand- E1B 19 kDa protein-​interacting protein 3, and FUN14
ing of the role of mitochondrial dynamics in the heart domain-​containing 1 are known to induce mitophagy
is obtained, therapeutic manipulation of mitochondrial in a parkin-​independent manner143–146.
fission and fusion might become a powerful tool to Type III mitophagy refers to unconventional
combat cardiovascular disease and promote ‘healthy’ mitophagy in which damaged regions of an individ-
cardiac ageing. ual mitochondrion selectively bud off as a form of

550 | september 2018 | volume 15 www.nature.com/nrcardio


© 2018 Macmillan Publishers Limited, part of Springer Nature. All rights reserved.
Reviews

mitochondrion-​d erived vesicles that subsequently including PGC1α, CREB-​regulated transcription


transit to lysosomes147. Although the onset of this non-​ co-​activator 2, FOXO1 and FOXO3, fibroblast growth
canonical mitophagy does not require mitochondrial factor 21, microtubule-​associated proteins 1A/1B light
depolarization, a subset of mitochondrion-​derived chain 3A, autophagy protein 5, autophagy-​related pro-
vesicles needs parkin and PINK1 for their forma- tein 7, and signal transducer and activator of transcrip-
tion 148. Membrane potential-​independent PINK1– tion 3, which are all closely linked to energy homeostasis
parkin-​mediated piecemeal mitophagy has also been and autophagy154,155. In a rodent model, ageing substan-
demonstrated in cell lines expressing a mitochondrial tially reduced SIRT1 activity in the heart, accompanied
matrix-​localized misfolded protein130,149. Loss of mito- by increased cardiac oxidative injury156,157. Cytoprotective
chondrial fission in Dnm1l−/− cells results in the loss of effects of SIRT1 are further substantiated by a study
the specificity of mitophagy130 and enhanced mitophagic showing that cardiac-​specific deletion of Sirt1 markedly
flux in cell lines and Dnm1l−/− mouse hearts128,131. impairs myocardium contractility together with increased
Autophagy is a central cellular process involved in ER stress and apoptosis158. Therefore, reduced expression
ageing and lifespan in many organisms and progres- or activity of SIRT1 with advancing age might alter the
sively declines in the heart during ageing, leading to status of acetylation or deacetylation of target substrates,
increased susceptibility to stress149. Mechanisms under- ultimately resulting in autophagy defects in aged hearts.
lying this age-​mediated decrease in autophagy include Hoshino and colleagues reported that p53, a tran-
downregulation of important autophagy-​related proteins scription factor known to promote DNA repair and
and autophagy regulators, as well as modification of apoptosis, prevents the onset of mitophagy by binding to
autophagy-​related proteins and altered autophagy sig- the RING0 domain of parkin and consequently inhibit-
nalling. Aged mice have a substantial loss of beclin 1 and ing parkin translocation to mitochondria159. Importantly,
microtubule-​associated proteins 1A/1B light chain 3A the age-​dependent decline in mitochondrial bioener-
and 3B as compared with younger littermates, suggesting getics was substantially alleviated by Tp53 knockdown.
impaired formation of autophagosomes with advancing Furthermore, cardiac function was well maintained in
age150,151. In addition, important regulators of autophagy old Tp53 heterozygous mice, suggesting that p53 and
such as forkhead box protein O1 (FOXO1), transcrip- parkin have a pivotal role in cardiac ageing through their
tion factor EB, PI3K-​III, and glycogen synthase kinase regulation of mitophagy. Indeed, gene-​expression pro-
3α are reduced in the aged heart151,152. In further sup- filing studies of mouse muscles revealed a substantial
port of an integral role of autophagy in cardiac ageing, increase in Tp53 mRNA levels in the old mice160.
heart-​specific overexpression of foxo improves cardiac In the context of cardiac ageing, energy stress modu-
function in aged Drosophila153. lation of autophagy involves AMPK and insulin signal-
Post-​translational modifications markedly influence ling pathways (Fig. 3). Although not fully elucidated, this
the stability and activity of proteins. NAD-​dependent regulation seems to be mediated by the nutrient-​sensing
protein deacetylase sirtuin 1 (SIRT1), located in both mechanistic target of rapamycin complex 1 (mTORC1)
the nucleus and the cytosol, regulates autophagy, that integrates the metabolic signals from both AMPK
mitochondrial biogenesis, and antioxidant defence154. and insulin to induce autophagy161. Indeed, the activa-
Many non-​histone targets are deacetylated by SIRT1, tion of mTORC1 inhibits both serine/threonine-​protein
kinase ULK1 (a key instigator of autophagosome for-
Box 2 | Calorie restriction, autophagy, and cardiac ageing mation)162 and transcription factor EB (a lysosomal
biogenesis modulator)163. In particular, changes in
Exploiting the capacity of cells to clear irreversibly damaged molecules and organelles amino acid availability influence mTORC1 and regulate
is an appealing approach to delay cardiac ageing and to prevent or treat cardiovascular
macrophagic protein breakdown161.
disease. Particularly intriguing is the prospect of preserving cardiac health through
fine-​tuning cardiomyocyte autophagy. Calorie restriction (CR), defined as a reduction
Beclin 1 is another relevant regulator of autophagy
in food intake without malnutrition, is among the most powerful inducers of in the heart164. In particular, beclin 2 and BCL-2-like
autophagy171. Indeed, the modulation of autophagy is a primary mechanism underlying protein 1 are negative regulators of autophagy through
the lifespan-​extending and health-​promoting properties of CR172. Studies in old rodents binding to beclin 1. The modulation of this binding
have shown that CR delays cardiac ageing, possibly via improved mitochondrial quality occurs via phosphorylation and can inhibit or activate
control processes, including autophagy. For instance, lifelong 40% CR increased the cardiomyocyte autophagy depending on the amino acid
protein expression of autophagy-​related protein 7, autophagy-​related protein 9, and residues being phosphorylated165–167.
lipidated microtubule-​associated proteins 1A/1B light chain 3B in the hearts of old rats, Although induction of autophagy is essential to
which was associated with a higher occurrence of autophagic vacuoles173. A similar ensure heart function during fasting168, fasted mice that
degree of CR increased the autophagic flux in the heart of old rats via suppression of
are deficient in IGF1 show cardiac atrophy following
mechanistic target of rapamycin signalling174. This suppression was accompanied by
reduced myocardial lipofuscin accumulation, decreased cardiomyocyte apoptosis, and
overactivation of autophagy169. The insulin signalling
preservation of left ventricular diastolic function. Induction of cardiac autophagy and pathway operates through activation of PI3K, RACα
amelioration of cardiomyocyte contractile performance were also observed in old mice serine/threonine-​protein kinase (AKT1), GTP-​binding
with lifelong 40% CR175. Challenges and concerns associated with long-​term CR protein RHEB, and mechanistic target of rapamycin
implementation in humans have instigated a great deal of research aimed at identifying (mTOR), and results in inhibition of autophagic activity
compounds to recapitulate the autophagy-​inducing properties of CR7. Several agents, (Fig. 3). Emerging evidence suggests that activation of
such as 2-deoxy-​d-glucose, metformin, rapamycin, resveratrol, salicylates, spermidine, autophagy elicits a prosurvival response in cardiomyo-
TEMPOL, and verapamil, have shown promising results in experimental animal models, cytes, but has also been linked to cardiomyocyte death
but whether these agents can exert cardioprotection in humans through modulation of when overactivated. Indeed, glucose deprivation in car-
autophagy remains to be established176.
diomyocytes overexpressing Rheb induces activation

NAture RevIeWs | CaRdiology volume 15 | september 2018 | 551


© 2018 Macmillan Publishers Limited, part of Springer Nature. All rights reserved.
Reviews

Box 3 | Unanswered research questions selective autophagy in the setting of cardiac ageing and


cardiovascular disease make this cellular process highly
• How much of cardiomyocyte age-​related dysfunction attractive for developing targeted therapeutic approaches
is attributable to failing mitochondrial quality control? (Box 2).
• In the context of mitochondrial quality control, are
there pathways that are primarily involved in heart Conclusions
senescence and that might be better targets to achieve Although we are still far from fully understanding the
cardioprotection?
events that trigger cardiomyocyte senescence and under-
• What is the optimal window of mitochondrial quality pin cardiac ageing, wide consensus exists on the central
control functioning to achieve cardioprotection
role of mitochondrial dysfunction. MQC mechanisms
without disrupting cardiomyocyte homeostasis?
operate through an integrated hierarchical network of
• To what extent can data obtained from
pathways, and derangements at any level of the MQC
pharmacological or behavioural modulation of
mitochondrial quality control in model organisms be
machinery can affect the whole system. During ageing
translated to humans? and in the setting of cardiovascular disease, failing MQC
• When should an intervention that modulates cardiac
might allow primary mitochondrial defects to expand
mitochondrial quality control be started and for how until a critical threshold is breached and mitochondrial
long should it be administered? dysfunction becomes phenotypically evident.
• How can mitochondrial quality control adaptations The recently identified SASP has been recognized
elicited by experimental drugs be monitored to establish direct or indirect contacts between cellular
noninvasively in humans? components (including ER, peroxisomes, lysosomes, and
vacuoles) and the extracellular environment through
mitochondrion-​d erived vesicle release. However,
of autophagy via downregulation of the RHEB–mTOR the functional consequences of these interactions in the
signalling pathway and is essential for cardiomyocyte context of cardiac ageing are not fully understood, and
survival. Similarly, in response to cardiac ischaemia several research questions remain unanswered (Box 3).
in vivo, Rheb overexpression prevents ischaemia-​induced Dissecting the interaction between MQC pathways and
autophagy and increases infarct size170. the pattern of circulating mediators associated with car-
Taken together, these results highlight the debate on diac senescence might be exploited to unveil new path-
whether upregulation of autophagy is cardioprotective or ways for the prevention and treatment of age-​related
harmful. Extrapolation of findings in other tissues might heart dysfunction.
be helpful given that large knowledge gaps exist in the
field of cardiac autophagy. The vital roles of general and Published online 24 July 2018

1. Mozaffarian, D. et al. Heart disease and stroke 13. Sahin, E. et al. Telomere dysfunction induces 25. Muñoz-​Espín, D. & Serrano, M. Cellular senescence:
statistics—2016 update. Circulation 133, e38–e360 metabolic and mitochondrial compromise. Nature from physiology to pathology. Nat. Rev. Mol. Cell Biol.
(2016). 470, 359–365 (2011). 15, 482–496 (2014).
2. Chiao, Y. A. & Rabinovitch, P. S. The aging heart. 14. Zierer, J. et al. Metabolomics profiling reveals novel 26. Senft, D. & Ronai, Z. A. UPR, autophagy, and
Cold Spring Harb. Perspect. Med. 5, a025148 markers for leukocyte telomere length. Aging (Albany, mitochondria crosstalk underlies the ER stress
(2015). NY) 8, 77–94 (2016). response. Trends Biochem. Sci. 40, 141–148 (2015).
3. Zhang, Y. et al. Mitochondrial aldehyde dehydrogenase 2 15. Yen, W.-L. & Klionsky, D. J. How to live long and 27. Sano, R. et al. Endoplasmic reticulum protein BI-1
accentuates aging-​induced cardiac remodeling prosper: autophagy, mitochondria, and aging. regulates Ca2+-mediated bioenergetics to promote
and contractile dysfunction: role of AMPK, Sirt1, and Physiology 23, 248–262 (2008). autophagy. Genes Dev. 26, 1041–1054 (2012).
mitochondrial function. Free Radic. Biol. Med. 71, 16. Marzetti, E. et al. Role of mitochondrial dysfunction 28. Adam-​Vizi, V. & Starkov, A. A. Calcium and
208–220 (2014). and altered autophagy in cardiovascular aging and mitochondrial reactive oxygen species generation:
4. Baris, O. R. et al. Mosaic deficiency in mitochondrial disease: from mechanisms to therapeutics. Am. J. how to read the facts. J. Alzheimers Dis. 20 (Suppl. 2),
oxidative metabolism promotes cardiac arrhythmia Physiol. Heart Circ. Physiol. 305, H459–H476 S413–S426 (2010).
during aging. Cell Metab. 21, 667–677 (2015). (2013). 29. Andersson, D. C. et al. Ryanodine receptor oxidation
5. Lok, N. S. & Lau, C. P. Prevalence of palpitations, 17. Wohlgemuth, S. E., Calvani, R. & Marzetti, E. The causes intracellular calcium leak and muscle weakness
cardiac arrhythmias and their associated risk factors interplay between autophagy and mitochondrial in aging. Cell Metab. 14, 196–207 (2011).
in ambulant elderly. Int. J. Cardiol. 54, 231–236 dysfunction in oxidative stress-​induced cardiac aging 30. Bánsághi, S. et al. Isoform- and species-​specific
(1996). and pathology. J. Mol. Cell. Cardiol. 71, 62–70 control of inositol 1,4,5-trisphosphate (IP3) receptors
6. Dutta, D., Calvani, R., Bernabei, R., Leeuwenburgh, C. (2014). by reactive oxygen species. J. Biol. Chem. 289,
& Marzetti, E. Contribution of impaired mitochondrial 18. Chung, H. Y. et al. Molecular inflammation: 8170–8181 (2014).
autophagy to cardiac aging: mechanisms and underpinnings of aging and age-​related diseases. 31. Rhee, S. G. & Kil, I. S. Mitochondrial H2O2 signaling is
therapeutic opportunities. Circ. Res. 110, 1125–1138 Ageing Res. Rev. 8, 18–30 (2009). controlled by the concerted action of peroxiredoxin III
(2012). 19. Fougère, B., Boulanger, E., Nourhashémi, F., and sulfiredoxin: linking mitochondrial function to
7. Marzetti, E. et al. Cellular mechanisms of Guyonnet, S. & Cesari, M. Chronic inflammation: circadian rhythm. Free Radic. Biol. Med. 100, 73–80
cardioprotection by calorie restriction: state of the accelerator of biological aging. J. Gerontol. A Biol. Sci. (2016).
science and future perspectives. Clin. Geriatr. Med. 25, Med. Sci. 72, 1218–1225 (2017). 32. Manella, G. & Asher, G. The circadian nature of
715–732 (2009). 20. Lin, C.-C. et al. NADPH oxidase/ROS-​dependent mitochondrial biology. Front. Endocrinol. (Lausanne)
8. López-​Otín, C., Blasco, M. A., Partridge, L., Serrano, M. VCAM-1 induction on TNF-​α-challenged human 7, 162 (2016).
& Kroemer, G. The hallmarks of aging. Cell 153, cardiac fibroblasts enhances monocyte adhesion. 33. Harman, D. Aging: a theory based on free radical
1194–1217 (2013). Front. Pharmacol. 6, 310 (2015). and radiation chemistry. J. Gerontol. 11, 298–300
9. North, B. J. & Sinclair, D. A. The intersection between 21. Sallam, N. & Laher, I. Exercise modulates oxidative (1956).
aging and cardiovascular disease. Circ. Res. 110, stress and inflammation in aging and cardiovascular 34. Duicu, O. M. et al. Ageing-​induced decrease in cardiac
1097–1108 (2012). diseases. Oxid. Med. Cell. Longev. 2016, 7239639 mitochondrial function in healthy rats. Can. J. Physiol.
10. Theurey, P. & Pizzo, P. The aging mitochondria. (2016). Pharmacol. 91, 593–600 (2013).
Genes (Basel) 9, 22 (2018). 22. Bennett, M. R., Sinha, S. & Owens, G. K. Vascular 35. Kuka, S. et al. Effect of aging on formation of
11. Hernandez-​Segura, A., Nehme, J. & Demaria, M. smooth muscle cells in atherosclerosis. Circ. Res. 118, reactive oxygen species by mitochondria of rat heart.
Hallmarks of cellular senescence. Trends Cell Biol. 28, 692–702 (2016). Gen. Physiol. Biophys. 32, 415–420 (2014).
436–453 (2018). 23. Gimbrone, M. A. & García-​Cardeña, G. Endothelial cell 36. Wong, H.-S., Dighe, P. A., Mezera, V., Monternier, P.-A.
12. Garinis, G. A., van der Horst, G. T. J., Vijg, J. & dysfunction and the pathobiology of atherosclerosis. & Brand, M. D. Production of superoxide and
Hoeijmakers, J. H. J. DNA damage and ageing: Circ. Res. 118, 620–636 (2016). hydrogen peroxide from specific mitochondrial sites
new-​age ideas for an age-​old problem. Nat. Cell Biol. 24. Campisi, J. Aging, cellular senescence, and cancer. under different bioenergetic conditions. J. Biol. Chem.
10, 1241–1247 (2008). Annu. Rev. Physiol. 75, 685–705 (2013). 292, 16804–16809 (2017).

552 | september 2018 | volume 15 www.nature.com/nrcardio


© 2018 Macmillan Publishers Limited, part of Springer Nature. All rights reserved.
Reviews

37. Kennedy, S. R., Salk, J. J., Schmitt, M. W. & Loeb, L. A. involved in natural aging? Aging Cell 5, 279–282 88. Ngo, J. K. & Davies, K. J. A. Importance of the lon
Ultra-​sensitive sequencing reveals an age-​related (2006). protease in mitochondrial maintenance and the
increase in somatic mitochondrial mutations that are 62. Inoue, K. et al. Generation of mice with mitochondrial significance of declining lon in aging. Ann. NY Acad. Sci.
inconsistent with oxidative damage. PLoS Genet. 9, dysfunction by introducing mouse mtDNA carrying 1119, 78–87 (2007).
e1003794 (2013). a deletion into zygotes. Nat. Genet. 26, 176–181 89. Gispert, S. et al. Loss of mitochondrial peptidase Clpp
38. Itsara, L. S. et al. Oxidative stress is not a major (2000). leads to infertility, hearing loss plus growth retardation
contributor to somatic mitochondrial DNA mutations. 63. Nakada, K. et al. Inter-​mitochondrial complementation: via accumulation of CLPX, mtDNA and inflammatory
PLoS Genet. 10, e1003974 (2014). mitochondria-​specific system preventing mice from factors. Hum. Mol. Genet. 22, 4871–4887 (2013).
39. Trifunovic, A. et al. Premature ageing in mice expression of disease phenotypes by mutant mtDNA. 90. Cipolat, S. et al. Mitochondrial rhomboid PARL
expressing defective mitochondrial DNA polymerase. Nat. Med. 7, 934–940 (2001). regulates cytochrome c release during apoptosis via
Nature 429, 417–423 (2004). 64. Vermulst, M. et al. Mitochondrial point mutations do OPA1-dependent cristae remodeling. Cell 126,
40. Kujoth, G. C. et al. Mitochondrial DNA mutations, not limit the natural lifespan of mice. Nat. Genet. 39, 163–175 (2006).
oxidative stress, and apoptosis in mammalian aging. 540–543 (2007). 91. Maltecca, F. et al. The mitochondrial protease AFG3L2
Science 309, 481–484 (2005). 65. Meissner, C. et al. The 4977 bp deletion of is essential for axonal development. J. Neurosci. 28,
41. Dai, D.-F. et al. Age-​dependent cardiomyopathy in mitochondrial DNA in human skeletal muscle, heart 2827–2836 (2008).
mitochondrial mutator mice is attenuated by and different areas of the brain: a useful biomarker 92. Narendra, D., Tanaka, A., Suen, D.-F. & Youle, R. J.
overexpression of catalase targeted to mitochondria. or more? Exp. Gerontol. 43, 645–652 (2008). Parkin is recruited selectively to impaired mitochondria
Aging Cell 9, 536–544 (2010). 66. Kraytsberg, Y. et al. Mitochondrial DNA deletions are and promotes their autophagy. J. Cell Biol. 183,
42. Lewis, K. N., Andziak, B., Yang, T. & Buffenstein, R. abundant and cause functional impairment in aged 795–803 (2008).
The naked mole-​rat response to oxidative stress: just human substantia nigra neurons. Nat. Genet. 38, 93. Calì, T., Ottolini, D., Negro, A. & Brini, M. Enhanced
deal with it. Antioxid. Redox Signal. 19, 1388–1399 518–520 (2006). parkin levels favor ER-​mitochondria crosstalk and
(2013). 67. Khrapko, K. & Vijg, J. Mitochondrial DNA mutations guarantee Ca2+ transfer to sustain cell bioenergetics.
43. Someya, S. et al. Effects of calorie restriction on the and aging: a case closed? Nat. Genet. 39, 445–446 Biochim. Biophys. Acta 1832, 495–508 (2013).
lifespan and healthspan of POLG mitochondrial (2007). 94. Verfaillie, T. et al. PERK is required at the
mutator mice. PLoS ONE 12, e0171159 (2017). 68. Srivastava, S. The mitochondrial basis of aging ER-​mitochondrial contact sites to convey apoptosis
44. Das, K. C. & Muniyappa, H. Age-​dependent and age-​related disorders. Genes (Basel) 8, 398 after ROS-​based ER stress. Cell Death Differ. 19,
mitochondrial energy dynamics in the mice heart: (2017). 1880–1891 (2012).
role of superoxide dismutase-2. Exp. Gerontol. 48, 69. Taylor, S. D. et al. Targeted enrichment and high-​ 95. Santos, C. X. C., Anilkumar, N., Zhang, M., Brewer, A. C.
947–959 (2013). resolution digital profiling of mitochondrial DNA & Shah, A. M. Redox signaling in cardiac myocytes.
45. Logan, A. et al. In vivo levels of mitochondrial deletions in human brain. Aging Cell 13, 29–38 Free Radic. Biol. Med. 50, 777–793 (2011).
hydrogen peroxide increase with age in mtDNA (2014). 96. Sumandea, M. P. & Steinberg, S. F. Redox signaling
mutator mice. Aging Cell 13, 765–768 (2014). 70. Taylor, R. W. et al. Mitochondrial DNA mutations in and cardiac sarcomeres. J. Biol. Chem. 286,
46. Eisenberg, T. et al. Cardioprotection and lifespan human colonic crypt stem cells. J. Clin. Invest. 112, 9921–9927 (2011).
extension by the natural polyamine spermidine. 1351–1360 (2003). 97. Divald, A. et al. Myocardial ischemic preconditioning
Nat. Med. 22, 1428–1438 (2016). 71. Nag, A. C. Study of non-​muscle cells of the adult preserves postischemic function of the 26S
47. Feng, W. et al. Increased age-​related cardiac mammalian heart: a fine structural analysis and proteasome through diminished oxidative damage
dysfunction in bradykinin B2 receptor-​deficient mice. distribution. Cytobios 28, 41–61 (1980). to 19S regulatory particle subunits. Circ. Res. 106,
J. Gerontol. A Biol. Sci. Med. Sci. 71, 178–187 72. Vliegen, H. W., van der Laarse, A., Cornelisse, C. J. & 1829–1838 (2010).
(2016). Eulderink, F. Myocardial changes in pressure overload-​ 98. Yuan, H. et al. LPS-​induced autophagy is mediated by
48. Marzetti, E. et al. Shorter telomeres in peripheral induced left ventricular hypertrophy. A study on tissue oxidative signaling in cardiomyocytes and is
blood mononuclear cells from older persons with composition, polyploidization and multinucleation. associated with cytoprotection. Am. J. Physiol. Heart
sarcopenia: results from an exploratory study. Eur. Heart J. 12, 488–494 (1991). Circ. Physiol. 296, H470–H479 (2009).
Front. Aging Neurosci. 6, 233 (2014). 73. Bergmann, O. et al. Evidence for cardiomyocyte 99. Haberland, M., Montgomery, R. L. & Olson, E. N. The
49. Tocchi, A., Quarles, E. K., Basisty, N., Gitari, L. & renewal in humans. Science 324, 98–102 (2009). many roles of histone deacetylases in development
Rabinovitch, P. S. Mitochondrial dysfunction in cardiac 74. Nunnari, J. & Suomalainen, A. Mitochondria: in and physiology: implications for disease and therapy.
aging. Biochim. Biophys. Acta 1847, 1424–1433 sickness and in health. Cell 148, 1145–1159 (2012). Nat. Rev. Genet. 10, 32–42 (2009).
(2015). 75. Bates, M. G. D. et al. Cardiac involvement in 100. Tepp, K. et al. Changes in the mitochondrial function
50. Sena, L. A. & Chandel, N. S. Physiological roles of mitochondrial DNA disease: clinical spectrum, and in the efficiency of energy transfer pathways
mitochondrial reactive oxygen species. Mol. Cell 48, diagnosis, and management. Eur. Heart J. 33, during cardiomyocyte aging. Mol. Cell. Biochem. 432,
158–167 (2012). 3023–3033 (2012). 141–158 (2017).
51. Wojtovich, A. P., Nadtochiy, S. M., Brookes, P. S. & 76. Fischer, F., Hamann, A. & Osiewacz, H. D. 101. Tatarková, Z. et al. Effects of aging on activities of
Nehrke, K. Ischemic preconditioning: the role of Mitochondrial quality control: an integrated network mitochondrial electron transport chain complexes
mitochondria and aging. Exp. Gerontol. 47, 1–7 of pathways. Trends Biochem. Sci. 37, 284–292 and oxidative damage in rat heart. Physiol. Res. 60,
(2012). (2012). 281–289 (2011).
52. Mishra, P. & Chan, D. C. Metabolic regulation of 77. Szklarczyk, R., Nooteboom, M. & Osiewacz, H. D. 102. Esterhammer, R. et al. Cardiac high-​energy phosphate
mitochondrial dynamics. J. Cell Biol. 212, 379–387 Control of mitochondrial integrity in ageing and metabolism alters with age as studied in 196 healthy
(2016). disease. Phil. Trans. R. Soc. B Biol. Sci. 369, males with the help of 31-phosphorus 2-dimensional
53. Wu, H., Wei, H., Sehgal, S. A., Liu, L. & Chen, Q. 20130439 (2014). chemical shift imaging. PLoS ONE 9, e97368 (2014).
Mitophagy receptors sense stress signals and couple 78. Twig, G., Hyde, B. & Shirihai, O. S. Mitochondrial 103. Yaniv, Y., Juhaszova, M. & Sollott, S. J. Age-​related
mitochondrial dynamic machinery for mitochondrial fusion, fission and autophagy as a quality control axis: changes of myocardial ATP supply and demand
quality control. Free Radic. Biol. Med. 100, 199–209 the bioenergetic view. Biochim. Biophys. Acta 1777, mechanisms. Trends Endocrinol. Metab. 24, 495–505
(2016). 1092–1097 (2008). (2013).
54. Rossignol, R. et al. Mitochondrial threshold effects. 79. Calvani, R. et al. Mitochondrial pathways in sarcopenia 104. Nathania, M. et al. Impact of age on the association
Biochem. J. 370, 751–762 (2003). of aging and disuse muscle atrophy. Biol. Chem. 394, between cardiac high-​energy phosphate metabolism
55. Wanrooij, S. et al. In vivo mutagenesis reveals that 393–414 (2013). and cardiac power in women. Heart 104, 111–118
OriL is essential for mitochondrial DNA replication. 80. Boateng, S. Y. & Goldspink, P. H. Assembly and (2018).
EMBO Rep. 13, 1130–1137 (2012). maintenance of the sarcomere night and day. 105. Klepinin, A. et al. Simple oxygraphic analysis for the
56. Elson, J. L., Samuels, D. C., Turnbull, D. M. & Cardiovasc. Res. 77, 667–675 (2008). presence of adenylate kinase 1 and 2 in normal and
Chinnery, P. F. Random intracellular drift explains the 81. Klein, I., Samarel, A. M., Welikson, R. & Hong, C. tumor cells. J. Bioenerg. Biomembr. 48, 531–548
clonal expansion of mitochondrial DNA mutations with Heterotopic cardiac transplantation decreases the (2016).
age. Am. J. Hum. Genet. 68, 802–806 (2001). capacity for rat myocardial protein synthesis. Circ. Res. 106. Huss, J. M. & Kelly, D. P. Nuclear receptor signaling
57. Greaves, L. C. et al. Comparison of mitochondrial 68, 1100–1107 (1991). and cardiac energetics. Circ. Res. 95, 568–578
mutation spectra in ageing human colonic epithelium 82. Razeghi, P. et al. Atrophic remodeling of the heart (2004).
and disease: absence of evidence for purifying in vivo simultaneously activates pathways of protein 107. Rattanasopa, C., Phungphong, S., Wattanapermpool, J.
selection in somatic mitochondrial DNA point synthesis and degradation. Circulation 108, & Bupha-​Intr, T. Significant role of estrogen in
mutations. PLoS Genet. 8, e1003082 (2012). 2536–2541 (2003). maintaining cardiac mitochondrial functions. J. Steroid
58. Kauppila, T. E. S., Kauppila, J. H. K. & Larsson, N.-G. 83. Patterson, C., Portbury, A. L., Schisler, J. C. & Willis, M. S. Biochem. Mol. Biol. 147, 1–9 (2015).
Mammalian mitochondria and aging: an update. Tear me down: role of calpain in the development of 108. Huss, J. M., Torra, I. P., Staels, B., Giguère, V. &
Cell Metab. 25, 57–71 (2017). cardiac ventricular hypertrophy. Circ. Res. 109, Kelly, D. P. Estrogen-​related receptor alpha directs
59. Müller-​Höcker, J., Droste, M., Kadenbach, B., 453–462 (2011). peroxisome proliferator-​activated receptor alpha
Pongratz, D. & Hübner, G. Fatal mitochondrial 84. Portbury, A. L., Willis, M. S. & Patterson, C. Tearin’ up signaling in the transcriptional control of energy
myopathy with cytochrome-​c-oxidase deficiency and my heart: proteolysis in the cardiac sarcomere. J. Biol. metabolism in cardiac and skeletal muscle. Mol. Cell.
subunit-​restricted reduction of enzyme protein in Chem. 286, 9929–9934 (2011). Biol. 24, 9079–9091 (2004).
two siblings: an autopsy-​immunocytochemical study. 85. Powell, S. R. The ubiquitin-​proteasome system in 109. Dorn, G. W., Vega, R. B. & Kelly, D. P. Mitochondrial
Hum. Pathol. 20, 666–672 (1989). cardiac physiology and pathology. Am. J. Physiol. biogenesis and dynamics in the developing and
60. Cottrell, D. A. et al. Cytochrome c oxidase deficient Circ. Physiol. 291, H1–H19 (2006). diseased heart. Genes Dev. 29, 1981–1991 (2015).
cells accumulate in the hippocampus and choroid 86. Quirós, P. M., Langer, T. & López-​Otín, C. New roles for 110. Kubli, D. A. & Gustafsson, Å. B. Mitochondria and
plexus with age. Neurobiol. Aging 22, 265–272 mitochondrial proteases in health, ageing and disease. mitophagy: the yin and yang of cell death control.
(2001). Nat. Rev. Mol. Cell Biol. 16, 345–359 (2015). Circ. Res. 111, 1208–1221 (2012).
61. Khrapko, K., Kraytsberg, Y., de Grey, A. D. N. J., Vijg, J. 87. Voos, W. Chaperone-​protease networks in 111. Fernandez-​Marcos, P. J. & Auwerx, J. Regulation of
& Schon, E. A. Does premature aging of the mtDNA mitochondrial protein homeostasis. Biochim. PGC-1α, a nodal regulator of mitochondrial biogenesis.
mutator mouse prove that mtDNA mutations are Biophys. Acta 1833, 388–399 (2013). Am. J. Clin. Nutr. 93, 884S–890S (2011).

NAture RevIeWs | CaRdiology volume 15 | september 2018 | 553


© 2018 Macmillan Publishers Limited, part of Springer Nature. All rights reserved.
Reviews

112. Leone, T. C. & Kelly, D. P. Transcriptional control of 137. Chen, Y. & Dorn, G. W. PINK1-phosphorylated 161. Tan, V. P. & Miyamoto, S. Nutrient-​sensing mTORC1:
cardiac fuel metabolism and mitochondrial function. mitofusin 2 is a Parkin receptor for culling damaged Integration of metabolic and autophagic signals.
Cold Spring Harb. Symp. Quant. Biol. 76, 175–182 mitochondria. Science 340, 471–475 (2013). J. Mol. Cell. Cardiol. 95, 31–41 (2016).
(2011). 138. Okatsu, K. et al. Phosphorylated ubiquitin chain is the 162. Egan, D., Kim, J., Shaw, R. J. & Guan, K.-L. The
113. Karamanlidis, G. et al. Defective DNA replication genuine Parkin receptor. J. Cell Biol. 209, 111–128 autophagy initiating kinase ULK1 is regulated via
impairs mitochondrial biogenesis in human failing (2015). opposing phosphorylation by AMPK and mTOR.
hearts. Circ. Res. 106, 1541–1548 (2010). 139. Koyano, F. et al. Ubiquitin is phosphorylated by PINK1 Autophagy 7, 643–644 (2011).
114. Bayeva, M., Gheorghiade, M. & Ardehali, H. to activate parkin. Nature 510, 162–166 (2014). 163. Martina, J. A., Chen, Y., Gucek, M. & Puertollano, R.
Mitochondria as a therapeutic target in heart failure. 140. Sarraf, S. A. et al. Landscape of the PARKIN-​ MTORC1 functions as a transcriptional regulator of
J. Am. Coll. Cardiol. 61, 599–610 (2013). dependent ubiquitylome in response to mitochondrial autophagy by preventing nuclear transport of TFEB.
115. Faerber, G. et al. Induction of heart failure by minimally depolarization. Nature 496, 372–376 (2013). Autophagy 8, 903–914 (2012).
invasive aortic constriction in mice: reduced peroxisome 141. Pankiv, S. et al. p62/SQSTM1 binds directly to 164. Maejima, Y., Isobe, M. & Sadoshima, J. Regulation
proliferator-​activated receptor γ coactivator levels and Atg8/LC3 to facilitate degradation of ubiquitinated of autophagy by Beclin 1 in the heart. J. Mol.
mitochondrial dysfunction. J. Thorac. Cardiovasc. Surg. protein aggregates by autophagy. J. Biol. Chem. 282, Cell. Cardiol. 95, 19–25 (2016).
141, 492–500.e1 (2011). 24131–24145 (2007). 165. Zalckvar, E. et al. DAP-​kinase-mediated
116. Shimizu, Y. et al. Hydrogen sulfide regulates cardiac 142. Lazarou, M. et al. The ubiquitin kinase PINK1 phosphorylation on the BH3 domain of beclin 1
mitochondrial biogenesis via the activation of AMPK. recruits autophagy receptors to induce mitophagy. promotes dissociation of beclin 1 from Bcl-​XL and
J. Mol. Cell. Cardiol. 116, 29–40 (2018). Nature 524, 309–314 (2015). induction of autophagy. EMBO Rep. 10, 285–292
117. Picca, A. & Lezza, A. M. S. Regulation of mitochondrial 143. Murakawa, T. et al. Bcl-2-like protein 13 is a (2009).
biogenesis through TFAM-​mitochondrial DNA mammalian Atg32 homologue that mediates 166. Gurkar, A. U. et al. Identification of ROCK1 kinase
interactions. Useful insights from aging and calorie mitophagy and mitochondrial fragmentation. as a critical regulator of Beclin1-mediated autophagy
restriction studies. Mitochondrion 25, 67–75 (2015). Nat. Commun. 6, 7527 (2015). during metabolic stress. Nat. Commun. 4, 2189
118. Anmann, T. et al. Formation of highly organized 144. Chen, Y. et al. Dual autonomous mitochondrial cell (2013).
intracellular structure and energy metabolism in death pathways are activated by Nix/BNip3L and 167. Wei, Y., Pattingre, S., Sinha, S., Bassik, M. & Levine, B.
cardiac muscle cells during postnatal development of induce cardiomyopathy. Proc. Natl Acad. Sci. USA JNK1-mediated phosphorylation of Bcl-2 regulates
rat heart. Biochim. Biophys. Acta 1837, 1350–1361 107, 9035–9042 (2010). starvation-​induced autophagy. Mol. Cell 30, 678–688
(2014). 145. Hanna, R. A. et al. Microtubule-​associated protein 1 (2008).
119. Palmer, J. W., Tandler, B. & Hoppel, C. L. Biochemical light chain 3 (LC3) interacts with Bnip3 protein to 168. Ikeda, Y. et al. Endogenous Drp1 mediates
properties of subsarcolemmal and interfibrillar selectively remove endoplasmic reticulum and mitochondrial autophagy and protects the heart
mitochondria isolated from rat cardiac muscle. J. Biol. mitochondria via autophagy. J. Biol. Chem. 287, against energy stress. Circ. Res. 116, 264–278 (2015).
Chem. 252, 8731–8739 (1977). 19094–19104 (2012). 169. Troncoso, R. et al. Energy-​preserving effects of IGF-1
120. Palmer, J. W., Tandler, B. & Hoppel, C. L. Biochemical 146. Liu, L. et al. Mitochondrial outer-​membrane protein antagonize starvation-​induced cardiac autophagy.
differences between subsarcolemmal and interfibrillar FUNDC1 mediates hypoxia-​induced mitophagy Cardiovasc. Res. 93, 320–329 (2012).
mitochondria from rat cardiac muscle: effects of in mammalian cells. Nat. Cell Biol. 14, 177–185 170. Sciarretta, S. et al. Rheb is a critical regulator of
procedural manipulations. Arch. Biochem. Biophys. (2012). autophagy during myocardial ischemia:
236, 691–702 (1985). 147. Soubannier, V. et al. A vesicular transport pathway pathophysiological implications in obesity and
121. Ichas, F., Jouaville, L. S. & Mazat, J. P. Mitochondria shuttles cargo from mitochondria to lysosomes. metabolic syndrome. Circulation 125, 1134–1146
are excitable organelles capable of generating and Curr. Biol. 22, 135–141 (2012). (2012).
conveying electrical and calcium signals. Cell 89, 148. McLelland, G.-L., Soubannier, V., Chen, C. X., 171. Cuervo, A. M. Calorie restriction and aging: the
1145–1153 (1997). McBride, H. M. & Fon, E. A. Parkin and PINK1 function ultimate “cleansing diet”. J. Gerontol. A Biol. Sci.
122. Glancy, B. et al. Mitochondrial reticulum for cellular in a vesicular trafficking pathway regulating mito­ Med. Sci. 63, 547–549 (2008).
energy distribution in muscle. Nature 523, 617–620 chondrial quality control. EMBO J. 33, 282–295 (2014). 172. Tóth, M. L. et al. Longevity pathways converge
(2015). 149. Zhou, J. et al. Changes in macroautophagy, on autophagy genes to regulate life span in
123. Amchenkova, A. A., Bakeeva, L. E., Chentsov, Y. S., chaperone-​mediated autophagy, and mitochondrial Caenorhabditis elegans. Autophagy 4, 330–338
Skulachev, V. P. & Zorov, D. B. Coupling membranes metabolism in murine skeletal and cardiac muscle (2008).
as energy-​transmitting cables. I. Filamentous during aging. Aging (Albany, NY) 9, 583–599 (2017). 173. Wohlgemuth, S. E. et al. Autophagy in the heart and
mitochondria in fibroblasts and mitochondrial clusters 150. Peng, L. et al. Changes in cell autophagy and liver during normal aging and calorie restriction.
in cardiomyocytes. J. Cell Biol. 107, 481–495 (1988). apoptosis during age-​related left ventricular Rejuvenation Res. 10, 281–292 (2007).
124. Glancy, B. et al. Power grid protection of the muscle remodeling in mice and their potential mechanisms. 174. Shinmura, K. et al. Impact of long-​term caloric
mitochondrial reticulum. Cell Rep. 19, 487–496 Biochem. Biophys. Res. Commun. 430, 822–826 restriction on cardiac senescence: caloric restriction
(2017). (2013). ameliorates cardiac diastolic dysfunction associated
125. El’darov, C. M., Vays, V. B., Vangeli, I. M., Kolosova, N. G. 151. Zhang, Y. et al. Complex inhibition of autophagy by with aging. J. Mol. Cell. Cardiol. 50, 117–127 (2011).
& Bakeeva, L. E. Morphometric examination of mitochondrial aldehyde dehydrogenase shortens 175. Han, X. et al. Influence of long-​term caloric restriction
mitochondrial ultrastructure in aging cardiomyocytes. lifespan and exacerbates cardiac aging. on myocardial and cardiomyocyte contractile function
Biochemistry (Mosc.) 80, 604–609 (2015). Biochim. Biophys. Acta 1863, 1919–1932 (2017). and autophagy in mice. J. Nutr. Biochem. 23,
126. Tate, E. L. & Herbener, G. H. A morphometric study of 152. Shirakabe, A., Ikeda, Y., Sciarretta, S., Zablocki, D. K. 1592–1599 (2012).
the density of mitochondrial cristae in heart and liver & Sadoshima, J. Aging and autophagy in the heart. 176. Delbridge, L. M. D., Mellor, K. M., Taylor, D. J. &
of aging mice. J. Gerontol. 31, 129–134 (1976). Circ. Res. 118, 1563–1576 (2016). Gottlieb, R. A. Myocardial stress and autophagy:
127. Chen, H. et al. Titration of mitochondrial fusion 153. Blice-​Baum, A. C. et al. Modest overexpression of mechanisms and potential therapies. Nat. Rev.
rescues Mff-​deficient cardiomyopathy. J. Cell Biol. 211, FOXO maintains cardiac proteostasis and ameliorates Cardiol. 14, 412–425 (2017).
795–805 (2015). age-​associated functional decline. Aging Cell 16,
128. Song, M., Mihara, K., Chen, Y., Scorrano, L. & 93–103 (2017). Acknowledgements
Dorn, G. W. Mitochondrial fission and fusion factors 154. Chun, S. K. et al. Autophagy in ischemic livers: The authors acknowledge support from Fondazione Roma
reciprocally orchestrate mitophagic culling in mouse a critical role of sirtuin 1/mitofusin 2 axis in autophagy (NCDs Call for Proposals 2013), Innovative Medicine
hearts and cultured fibroblasts. Cell Metab. 21, induction. Toxicol. Res. 32, 35–46 (2016). Initiative-​Joint Undertaking (IMI-​JU 115621), intramural
273–286 (2015). 155. Huang, R. et al. Deacetylation of nuclear LC3 drives research grants from the Catholic University of the Sacred
129. Song, M., Franco, A., Fleischer, J. A., Zhang, L. & autophagy initiation under starvation. Mol. Cell 57, Heart (D3.2 2013 and D3.2 2015), the non-​profit research
Dorn, G. W. Abrogating mitochondrial dynamics in 456–466 (2015). foundation “Centro Studi Achille e Linda Lorenzon”, and the
mouse hearts accelerates mitochondrial senescence. 156. Ferrara, N. et al. Exercise training promotes SIRT1 Claude D. Pepper Older Americans Independence Center at
Cell Metab. 26, 872–883.e5 (2017). activity in aged rats. Rejuvenation Res. 11, 139–150 the University of Florida’s Institute on Aging (NIA
130. Burman, J. L. et al. Mitochondrial fission facilitates the (2008). 1P30AG028740).
selective mitophagy of protein aggregates. J. Cell Biol. 157. Ren, J. et al. Akt2 ablation prolongs life span and
216, 3231–3247 (2017). improves myocardial contractile function with adaptive Author contributions
131. Parone, P. A. et al. Preventing mitochondrial fission cardiac remodeling: role of Sirt1-mediated autophagy A.P. and R.T.M. researched data for the article. A.P., J.L.B.,
impairs mitochondrial function and leads to loss of regulation. Aging Cell 16, 976–987 (2017). and J.-S.K. discussed the content of the article. A.P., R.T.M.,
mitochondrial DNA. PLoS ONE 3, e3257 (2008). 158. Hsu, Y.-J. et al. Sirtuin 1 protects the aging heart L.D., and E.M. wrote the manuscript. E.M. and C.L. revised
132. Mizushima, N., Levine, B., Cuervo, A. M. & Klionsky, D. J. from contractile dysfunction mediated through and edited the manuscript before submission.
Autophagy fights disease through cellular self-​digestion. the inhibition of endoplasmic reticulum stress-​
Nature 451, 1069–1075 (2008). mediated apoptosis in cardiac-​specific Sirtuin 1 Competing interests
133. Sun, N. et al. Measuring in vivo mitophagy. Mol. Cell knockout mouse model. Int. J. Cardiol. 228, The authors declare no competing interests.
60, 685–696 (2015). 543–552 (2017).
134. Lemasters, J. J. Variants of mitochondrial autophagy: 159. Hoshino, A. et al. Cytosolic p53 inhibits Parkin-​ Publisher’s note
types 1 and 2 mitophagy and micromitophagy mediated mitophagy and promotes mitochondrial Springer Nature remains neutral with regard to jurisdictional
(type 3). Redox Biol. 2, 749–754 (2014). dysfunction in the mouse heart. Nat. Commun. 4, claims in published maps and institutional affiliations.
135. Youle, R. J. & Narendra, D. P. Mechanisms of 2308 (2013).
mitophagy. Nat. Rev. Mol. Cell Biol. 12, 9–14 (2011). 160. Edwards, M. G. et al. Gene expression profiling Reviewer information
136. Jin, S. M. et al. Mitochondrial membrane potential of aging reveals activation of a p53-mediated Nature Reviews Cardiology thanks E. Lesnefsky and the other
regulates PINK1 import and proteolytic destabilization transcriptional program. BMC Genomics 8, 80 anonymous reviewers for their contribution to the peer review
by PARL. J. Cell Biol. 191, 933–942 (2010). (2007). of this work.

554 | september 2018 | volume 15 www.nature.com/nrcardio


© 2018 Macmillan Publishers Limited, part of Springer Nature. All rights reserved.

S-ar putea să vă placă și