Aging and Senescence Associated Mitochondrial Dysfunction: A Target against Cardiovascular Disorders of the Elderly Individuals
by Lucia-Doina Popov*
Institute of Cellular Biology and Pathology “Nicolae Simionescu” of the Romanian Academy, 8, B.P. Hasdeu Street, 050568, Bucharest, Romania.
*Corresponding authors: Popov LD, Institute of Cellular Biology and Pathology “Nicolae Simionescu” of the Romanian Academy, 8, B.P. Hasdeu Street, 050568, Bucharest, Romania.
Received Date: 08 January, 2025
Accepted Date: 14 January, 2025
Published Date: 16 January, 2025
Citation: Popov LD (2025) Aging and Senescence Associated Mitochondrial Dysfunction: A Target against Cardiovascular Disorders of the Elderly Individuals. Int J Geriatr Gerontol 9: 198. https://doi.org/10.29011/2577-0748.100098
Abstract
The expansion of the world`s elderly population requires the identification of complex mechanisms beyond the continuous decline in cellular functions associated with aging and senescence. Both are risk factors that gradually impede health homeostasis and promote the incidence of cardiovascular, metabolic, neurodegenerative, and immune diseases in elderly people. The review aims to update and discuss the role of mitochondrial dysfunction in cardiovascular aging and senescence. The focus is targeted on molecular mechanisms beyond mitochondrial dysfunction occurring in (i) cardiac aging, (ii) vasculature aging, and (iii) cellular senescence. In line with the ongoing basic cardiovascular research, the review uncovers the promising strategies directed towards alleviating dysregulated and interrelated pathways of mitochondrial dysfunction by (iv) anti-aging therapies, and (v) antisenescence treatments. Ultimately, the open questions and the perspectives of this domain (vi) are underlined. One can safely state that the recent translation of preclinical endeavors and interventions into clinical conduits helps to prevent/delay cardiovascular mitochondrial-dysfunction, and is of benefit to aged people.
Keywords: Cardiomyocytes; Blood vessels; Anti-aging therapy; Anti-senescence therapy
Introduction
Biological aging starts with the intracellular occurrence of molecular damages, followed by their gradual and irreversible accumulation. These changes result in the progressive loss of normal cellular functions, dysfunction of intracellular signaling, and altered intercellular communication. Next, such defects expand to the systemic decline of tissues and organs’ operation and ultimately trigger organismal death. Aging is not a disease, but it significantly increases the susceptibility to the occurrence of a variety of agerelated diseases, including cardiovascular, neurodegenerative, musculoskeletal, and metabolic diseases, macular degeneration, cancer, and many other disabilities with a poor prognosis for the elderly individuals [1-7]. A series of complex and interlinked processes are known as “hallmarks of aging”. These involve (i) systemic alterations (such as deregulated nutrient sensing), (ii) specific cellular hallmarks (cellular senescence, exhaustion of stem cells, and altered intercellular communication), and (iii) molecular hallmarks, such as genomic instability, shortening of telomeres (repetitive DNA sequences found at the terminal loops of linear eukaryotic chromosomes), epigenetic alterations, loss of protein homeostasis (“proteostasis”), metabolome adjustments, low-grade chronic inflammation, compromised autophagy, and mitochondrial dysfunction [4, 8-10].
In cardiovascular aging, the decline of mitochondrial function (known as “mitochondrial dysfunction”) is characterized by reduced ATP generation, impaired oxidative phosphorylation (OXPHOS), diminished mitochondrial biogenesis, depletion of NAD+, overproduction of mitochondrial reactive oxygen species (mROS) correlated with increased oxidative stress, amplified mitochondrial DNA (mtDNA) mutation rate, telomere shortening, compromised quality control processes, and inefficient mitophagy.
These traits of mitochondrial dysfunction are implied in the development and progress of cellular dysfunction [4, 11-16].
Senescence is a pleiotropic process [17]: acute senescence appears to be a normal physiological activity with beneficial roles in embryogenesis, tissue remodeling, and wound healing [18, 19], while the chronic senescence has detrimental effects because the gradual accumulation of senescent cells during aging and agerelated diseases leads to progressive tissular dysfunction [18, 20]. Here, the focus is on chronic age-related senescence (referred to further as “cellular senescence”). This is an adaptative response of cells facing the damage of severe stresses, leading to the irreversible loss of their proliferative potential and the long-term and stable cell cycle arrest. Meanwhile, the up-regulation of the anti-apoptotic pathways imposes cells to remain metabolically active [21]. Senescence is not identical to aging, as cells may become senescent irrespective of organismal age [22]. It should not be confused with quiescence, a condition of reversible proliferative arrest [23, 24]. The cell-cycle arrest pathways are different: activation of cell death inductor p53 and mammalian Target of Rapamycin (mTOR) causes cellular senescence, whereas p53 activation and mTOR inhibition trigger quiescence [25, 26]. Moreover, dependent on the diversity of cells and stressors, the attainment of cell senescence takes longer compared with some other cellular activities (replication, differentiation, apoptosis, or necrosis) [12]. The biomarkers of cellular senescence comprise alteration of morphology (cells become flat and enlarged), augmented reactivity of senescence-associated β-galactosidase, expression of Senescence-Associated Secretory Phenotype (SASP, a collection of factors with pro-inflammatory, proteolytic, extracellular matrix-degrading, complement-activating and procoagulating roles), intensified activity of Cyclin-Dependent Kinase (CDK) inhibitors, and modifications of chromatin and mtDNA [2, 18, 22, 27-31].
Mitochondrial dysfunction plays a key role in the initiation and progression of cellular senescence. The main promoters are excessive mROS generation, the conversion of metabolism from OXPHOS to glycolysis, impaired ATP generation, reduced mitochondrial membrane potential (ΔΨm), diminished NAD+/ NADH ratio, and antioxidant capability. Moreover, released from the ER stores, Ca2+ triggers mitochondria overload and opening of the mitochondrial permeability transition pores (mPTP) located at the inner mitochondrial membrane (IMM). In cellular senescence, the dysfunctional mitochondria accumulate and are not efficiently cleared from the affected cells [13].
This review surveys and updates the molecular mechanisms of mitochondrial dysfunction in (i) cardiac aging, (ii) blood vessel aging, (iii) cardiovascular senescence, (iv) the current antiaging, and (v) anti-senescence therapies targeting cardiovascular mitochondrial dysfunction. The open questions and the perspectives of this age-related essential topic conclude the review.
Mitochondrial dysfunction in cardiac aging
Mitochondria are abundant in the cardiomyocytes. They occupy 30-40% of the cell volume and generate ~90% of the ATP necessary for the normal contractile function [24, 32]. Noteworthy, cardiomyocytes contain spatially and morphologically distinct mitochondrial subpopulations with specific tasks: (i) the subsarcolemmal mitochondria (SSL, 0.4-3.0 μm in length) provide the ATP used in the transport of electrolytes and metabolites across the sarcolemma, (ii) the interfibrillar mitochondria (IF, ~1.5-2.0 μm in length) supply the ATP used in contraction, and (iii) the perinuclear mitochondria (PN, smaller in size, compared to SSL and IF) are relatively mobile during organelle`s fusion/fission dynamics [33-35].
Dysfunctional mitochondria are recognized as central contributors to heart aging, a process that harms the IF electron transport chain (ETC) [36]. Interestingly, an early event linked to cardiac aging is the acute ER stress (sustained by calpain I activation) that occurs earlier than mitochondrial dysfunction [35]. ER stress affects also the mitochondria-endoplasmic reticulum (ER) interacting zones (MERCs or Mitochondria Associated Membranes, MAMs); these function as signaling centers implied in lipid and calcium transfer, mitochondrial dynamics, and autophagy associated with the aging process [37-39].
The prominent features of the aging heart are hypertrophy, diastolic dysfunction, augmented fibrosis of the myocardium, and valvular calcification [32]. Recent knowledge highlights the main triggers of mitochondrial dysfunction in cardiac aging:
(i) Of the utmost importance is oxidative stress, defined as an imbalance between excessive ROS production and reduced scavenging capacity [16, 32, 40, 41]. Chemically, ROS are the superoxide anions (O2•−), hydrogen peroxide (H2O2), and hydroxyl radicals (•OH). mROS are generated as by-products of OXPHOS and are viewed nowadays not only as inductors of oxidative stress but also as signaling molecules [42]. In physiological conditions, the low/moderate ROS levels contribute to cell homeostasis, as ROS are cleared by the cell`s antioxidant defense scavengers, including mitochondrial superoxide dismutase (SOD2 or MnSOD), peroxiredoxin 3, glutathione peroxidase (located in the cytosol, mitochondria, and peroxysomes), and the peroxisomal catalase. Excess of ROS levels suppresses the scavengers` activity, causees oxidative damage to mtDNA, proteins, and lipids, and ultimately activates the apoptotic pathways that induce cell death [1, 36, 41].
The abnormal accumulation of dicarbonyls in the aged myocardium (caused by the reduced efficiency of the glyoxalase detoxification pathway) was reported recently as an inductor of oxidative stress. The dicarbonyls are α-oxaldehydes (methylglyoxal, glyoxal, 3-deoxyglucosone), intermediates of glycolysis, gluconeogenesis, and lipid metabolism that favor ROS generation [43]. Moreover, excess dicarbonyls alter the formation and assembly of FoF1-ATP synthase monomers, which conduct to aberrant cristae formation, less efficient OXPHOS, and augmented energy dissipation through the opening of the mPTP. The glycation in the 5 subunits of FoF1-ATP synthase favors the opening of the mPTP [41,44]. Notable, the partial mPTP opening releases mtROS and Ca2+ that activate nucleus-associated protective mechanisms such as the nuclear transcription factor E2-related factor (Nrf2) (with antioxidant function) and PGC-1α (implied in mitochondrial biogenesis). When mPTP opening is prolonged, the cytoplasm flows into mitochondria and causes extensive swelling of the IMM; subsequently, the outer mitochondrial membrane (OMM) becomes damaged, cytochrome c is released, and cell apoptosis ocurrs. mPTP opening can be normally stopped by the removal of dysfunctional mitochondria by mitophagy. In aging conditions, two situations may arise: (a) in case of massive mitophagy, the cell will be depleted of mitochondria leading to its death [16], and (b) in case of full opening of mPTP, the matrix metabolites (OXPHOS substrates, mROS, Ca2+, NAD+, and glutathione) will be released and an increased “proton (H+) leak” through the mitochondrial inner membrane Adenine Nucleotide Transporter 1 will be stimulated [45]. In health conditions, a part of protons is pumped from the matrix to the mitochondrial intermembrane space, but some leak back to the matrix and generate ATP via ATP synthase. In the aged heart, the returned protons do not yield ATP, a condition known as “proton leak” [45]. The augmented proton leak is considered the primary bioenergetic change in aged heart mitochondria [46]. This is another example of a pleiotropic process: the mild proton leak occurs in the young heart, mimics caloric restriction, and confers protection against the damaging effects of ROS and oxidative stress, while in the aged heart, the excessive proton leak is detrimental, decreasing the respiratory efficiency [3].
(ii) The modification of Zn2+ transporters (in charge of Zn2+ distribution among cytosol and intracellular organelles) results in mitochondrial Zn2+ overload associated with increased ROS production and dysfunction of aged cardiomyocytes [40]. Earlier, it has been reported that Zn2+ originates from the lysosomes, after permeabilization of their membrane by Ca2+ that entered through the Transient Receptor Potential Melastatin 2 (TRPM2) channel; subsequently, the released Zn2+ stimulates the mitochondrial recruitment of Dynamin-related protein 1 (Drp-1) that triggers the aging-associated mitochondrial fission [47].
(iii) Another feature of mitochondrial dysfunction consists in the low levels of Coenzyme Q10 which transfers electrons from complexes I and II to complex III in the electron transport chain [48, 49].
(iv) The modifications of mTOR complex 1 and 2 signaling pathways occur in age-related cardiac dysfunction and heart failure [50]. It is known that the mTOR pathway regulates both cardiac homeostasis and aging through the adjustment of protein synthesis, autophagy, and mitochondrial function [51].
The recent reports bring evidence that mitochondrial dysfunction is a common attribute shared by both aged cardiomyocytes and blood vessels. The common traits consist in:
(a) the augmented production of ROS and the altered expression of proteins that regulate the redox balance; among the up-regulated proteins are the NADPH oxidase 4 (NOX4), the Src homologouscollagen homolog adaptor (p66Shc), and Arginase II (Arg-II). Opposed, are proteins down-regulated by aging, such as the Silent Information Regulator 1 (SIRT1), the antioxidant Nrf2, and the Nrf2 regulator, Klotho [32],
(b) the failure of the mitochondrial quality control caused by the offset of fission/fusion balance, and by the inefficient mitophagy conduct to the diminishment of endogenous antioxidant defenses. To compensate for the reduced functionality, mitochondrial morphology is affected. The malfunctioning mitochondria split by fission (“hyperfission”) to remove the defective fragments and generate novel robust mitochondria aiming at the covering of energy requirements for growth and division (Figure 1),
Figure 1: Electron microscopic ultrastructure of cardiomyocyte mitochondria in health (a), in aging: (b) smaller mitochondria generated by fission exposing defective cristae, (c) apoptotic mitochondria with a disorganized morphology, and in senescence: (d) fused, elongated mitochondria.
Magnifications: (a, c, d)- 54,600x; (b) 36,450x;
(c) other common traits of mitochondrial dysfunction in aged hearts and vasculature are reduced mitochondrial biogenesis, defective mitochondrial Ca2+ cycling, and impaired autophagy; the latter is caused by the reduced activation of AMP-activated protein kinase (AMPK) and E3 ubiquitin ligase Parkin, along with the involvement of Rho-associated coiled-coil-containing protein kinase (ROCK)1 and ROCK2 [11, 32, 52, 53],
(d) the malfunction of telomeres leads to mitochondrial dysfunction by the downregulation of transformation-related protein 53(TRP53)dependent of peroxisome proliferator-activated receptor gamma coactivator 1α (PGC-1α) and PGC-1β [54, 55],
(e) myocardial and blood vessels aging show activation of the NACHT, LRR, and PYD domains-containing protein 3 (NLRP3) inflammasome, stimulation of the Toll-like receptor 4 (TLR4), of inflammatory cytokines production, and NF-κB signaling [32]; thus, aging is associated with a low-grade, chronic inflammation that augments the local generation of ROS and amplifies the inflammatory response [9, 56-59]. The process was coined “inflammaging” by Franceschi et al. in 2000 and now is generally recognized as a risk factor for cardiovascular diseases [59,60].
The above-dysregulated and interrelated pathways of mitochondrial dysfunction in aged cardiomyocytes and blood vessels [61, 62] are accompanied by specific features developed in aged arteries and microvasculature (discussed in the next section).
Mitochondrial dysfunction in blood vessels aging
The data so far established that all cellular components within the vascular wall of large arteries (aorta), coronary arteries, atherosclerotic arteries, and microvasculature (capillaries included) are affected by age-related mitochondrial dysfunction. The elastic arteries (aorta and carotid artery) remodel in aging their wall; the lamellae become partially split and replaced by collagen, and the endothelial cells (ECs) lose the angiogenic ability and ECsdependent vasodilation (to acetylcholine) [62, 63, 64]. Previously, it was demonstrated that in rat-aged aortas, the O2•− reacts with the vasorelaxant nitric oxide (NO), forms peroxinitrite and nitrosylates mitochondrial manganese superoxide dismutase (MnSOD) [65]. Within the aged aorta, the enhanced Interleukin-6 (IL-6) levels increase mitochondrial dysfunction, and augment mitophagy and Parkin levels; these changes assist atherogenesis development in hyperlipidemia [66].
The high incidence of abdominal aortic aneurysm (AAA) in the aged population attracted attention to mitochondrial dysfunction`s role in this degenerative disease [67]. Recently, Navas-Madroñal et al. [68] showed the involvement of harmful mitochondrial oxidative stress in AAA and discovered the positive effects of mitochondria-targeted tetrapeptide Szeto-Schiller 31 that reduced the occurrence and gravity of AAA. Other studies showed that thoracic aortic aneurysm modified the vessel proteome both quantitatively and qualitatively [69].
Within the aged coronaries, mitochondrial dysfunction is promoted by acyl-coenzyme A: lysocardiolipin acyltransferase-1 (ALCAT-1) involved in cardiolipin remodeling [70]. Additionally, aging is associated with coronaries hyperconstriction, an event in which dysregulated mitochondrial redox homeostasis and the imbalanced fission/fusion dynamics play a role; the consequences consist of impaired physiological perfusion and the installment of several heart pathologies [71]. Such alterations are intensified by the mROS levels exceeding the cell’s antioxidant buffering capacity [72]. Advanced aging produces modifications of the bioenergetic profile of coronary artery ECs and vascular smooth muscle cells (VSMCs) that express in aging lower resting OXPHOS levels, and reduced reserve capacity [73].
Effects of aging on microvasculature implied in-depth studies on coronaries and brain microcirculation. Recently, the group of Mengozzi et al. [56] advanced the idea that microvascular dysfunction might represent a noticeable marker of aging compared to chronological age; this conclusion resulted from the study of age-associated mitochondrial and ECs dysfunction correlated with the irreversible modification in microvascular wall structure, lowgrade inflammation, and oxidative stress. Furthermore, in ECs, the mitochondrial Sirtuin 3 (SIRT3) levels progressively decline with aging, the SIRT3-related ECs metabolism is impaired, and these modifications conduct in the rarefaction of coronary microvasculature [74]. Remarkably, vascular alterations occur earlier in individuals at risk for cardiovascular disease development [64]. Brain microvasculature restrains in aging a decreased number of mitochondria and a reduced efficiency of the remaining ones [75, 76]; the latter trait contributes the impaired OXPHOS, lower ATP generation, diminished glycolysis, and increased glutamine utilization as an energy source [56].
Nowadays, it is established that by affecting blood vessels, mitochondrial dysfunction has a key role in the morbidity and mortality of older individuals [29]. The input of mitochondrial dysfunction on cardiovascular senescence is discussed below.
Mitochondrial dysfunction in cardiovascular senescence
In response to aging and stressors, the cardiomyocytes (terminally differentiated post-mitotic cells) develop a senescent phenotype and accumulate within the myocardium, contributing to the risk of age-related cardiovascular pathologies, such as heart failure, diastolic dysfunction, myocardial infarction, cardiac arrhythmias, and atherosclerosis [24, 77-81].
Cardiomyocyte senescence is induced by various factors: mitochondrial dysfunction, oxidative stress, activation of the hexosamine biosynthetic pathway, and epigenetic regulation [82, 83]. Several processes explain mitochondrial dysfunction associated with cardiomyocyte senescence: the oxidative stress (counting for 90% of age-related ROS) [26], the overexpression of mitochondrial-membrane flavoenzyme Monoamine
Oxidase-A (MAO-A) (another source for elevated ROS levels), downregulation of genes encoding subunits of mitochondrial ETC complexes, defects in mitochondrial dynamics and quality control [84], and the inefficient removal of damaged mitochondria by mitophagy (as a consequence of Parkin-mediated mitophagy inhibition by cytosolic p53) [85-89]. To assist the replenishment of damaged mitochondrial DNA, extensive mitochondrial fusion takes place, resulting in elongated mitochondria (Figure 1); this process is coordinated by the mitochondrial fusion proteins: Mitofusin 1(Mfn1), Mitofusin 2 (Mfn2), and optic atrophy 1 (OPA1) [90-92]. The cellular senescence is induced also by the clinical doses of chemotherapy that cause cardiotoxicity [93, 94].
The hallmarks of cardiomyocyte senescence are genomic instability, mitochondrial dysfunction, ER stress, contractile dysfunction, hypertrophic growth, β-galactosidase expression, and increased production of pro-inflammatory, pro-fibrotic and pro-hypertrophic SASP factors [24, 77, 79, 95, 96]. The latter is facilitated by minor permeabilization of the OMM (miMOMP) that permits the release of mtDNA into the cytosol (via BAX and BAK macropores), activates the cyclic GMP–AMP synthase (cGAS)–stimulator of interferon genes (STING) pathway, and conducts to the increased expression of inflammatory molecules and of SASP [97]. The SASP generation in aged cardiomyocytes is also stimulated by the length-independent telomere damage that activates the classical senescence-inducing pathways p21CIP and p16 INK4a [77].
Within the myocardial “microenvironment” and under stress conditions, the senescence of resident cardiomyocytes and ECs (representing ~60% of heart noncardiomyocytes) is modulated by paracrine signaling factors released by these two types of cells: (i) the dysfunctional cardiomyocytes secrete angiogenic factors implied in the induction of ECs senescence; examples are the Vascular Endothelial Growth Factor A (VEGF A), angiopoietin-1, Lipoprotein Lipase (LPL, in diabetes), SASP, and extracellular vesicles (EV) [26, 98, 99]; (ii) the malfunctioning cardiac ECs secrete pro-inflammatory factors, such as Transforming Growth Factor- β (TGF-β), Interleukin-6 (IL-6), IL-33, Endothelin-1 (ET-1), Angiotensin II (Ang II), and EV, recognized as players in cardiomyocytes senescence [26, 96].
Among the consequences of cardiomyocyte senescence, studies uncover the increased risk of ventricular arrhythmias [100], the development of cardiomyopathy [101], and telomere dysfunction; ATP deficiency, excessive ROS generation, and chronic inflammation are potential therapeutic targets to improve the associated mitochondrial dysfunction [102]. The senescence of cardiac ECs may lead to reduced vasodilation and atrial fibrillation associated with the expression of senescence effector pathways, p53, and p16 [103]. Aortic stiffness, enhanced inflammation, and dysregulated vascular tone have been acknowledged among the features of senescent vascular ECs [104].
Senescence disturbs also the VSMCs that show increased β-galactosidase expression, short telomeres, up-regulated secretion of inflammatory cytokines, and enhanced DNA damage [105]. In cardiovascular pathophysiology, accumulation of senescent VSMCs increases with age, is regulated by Ang II [106], occurs within atherosclerotic plaques, throughout all stages of the disease [107], and is implicated in the calcification of old aortas, a process associated with the upregulation of transcriptional factor GATA6 [108]. Furthermore, the senescent VSMCs are involved in the activation of pro-ferroptotic signaling, a novel form of regulated cell death associated with arterial stiffness [109]; the secreted SASP factors contribute to the development of vascular diseases, such as atherosclerosis, aneurysm, and hypertension [110].
The knowledge described above allows the selection of the common and individual traits of mitochondrial dysfunction in cardiovascular aging and senescence. A brief synopsis is included in Figure 2.
Figure 2: Mitochondrial dysfunction molecular mechanisms in cardiovascular aging and senescence: the common and the specific features.
The recent preclinical endeavors to alleviate cardiovascular consequences of aging and cellular senescence are discussed in the last part of this review.
Current anti-aging therapies targeting cardiovascular mitochondrial dysfunction
Preclinical studies acknowledge that alleviation of mitochondrial dysfunction in cardiovascular aging is produced by several natural compounds, antioxidants, peptides, pharmacological agents, and hormones; other recent promising approaches are nanomedicine drugs, gene therapy, mitochondrial transfer, and lifestyle changes (Table 1).
Therapeutic approach |
Targeted mitochondrial dysfunction/Effects |
References |
NATURAL COMPOUNDS Flavonoids – Quercetin |
Abnormal mitochondrial dynamics ↓mitochondrial superoxide Protects mitochondrial morphology |
111-116 |
Phenols and polyphenols -Resveratrol (3,5,4’- trihydroxystilbene) Salvinoic acid D |
AMPK-SIRT1-PGC-1α mitochondrial biogenesis pathway SIRT1 activation Sirt1/Sirt3-FoxO pathway activation mitophagy impediment, mPTP opening prevention restores mitochondrial morphology |
115, 117-120 |
Metformin |
↑ autophagy; alleviate aging-associated inflammation ↓ ROS generation; mitochondrial function modulation offsets aging and extend lifespan |
121-123 |
ANTIOXIDANTS Coenzyme Q10 MitoQ (Mitoquinone) Vitamin E MitoTEMPO (2-(2,2,6,6-Tetramethylpiperidin-1oxyl-4-ylamino)-2 -oxoethyl) triphenylphosphonium chloride monohydrate) |
Antioxidant, mROS scavenger improves age-related endothelial dysfunction by ↓oxidized LDL, ↑ NO production and ↓mitochondrial oxidative stress ↓mROS prevents vascular reactivity alterations |
117, 124-127 |
Zn2+ |
Modulation of cardiomyocyte Zn2+transporters |
41 |
PEPTIDES Spermidine (1,8-Diamino-4-azaoctane, N-(3- Aminopropyl)-1,4-diaminobutane MOTS-c |
Attenuates mitochondrial dysfunction, ↓IL-6 and Parkin Restores mitochondrial metabolic imbalance |
66 128 |
PHARMACOLOGICAL AGENTS The SS-31 tetrapeptide (D-Arg-2’,6’-dimethylTyr- Lys-Phe-NH2) (elamipretide or bendavia) |
↓ mitochondrial proton leak & PTP opening, prevents mitochondrial proton leak |
28, 47, 72 |
Rapamycin |
mTOR inhibitor, improves cardiac systolic and diastolic function |
129 |
GENE THERAPY |
Overexpression of PGC-1α SIRT1, TFAM & Parkin |
15, 130 |
NEUROENDOCRINE HORMONE Melatonin (N-acetyl-5-methoxytryptamine) |
Antioxidant, anti-inflammatory ↓Drp1 expression; mitochondrial fission inhibition ↑mitochondrial fusion/mitophagy, activation of AMPK-OPA1 signalling pathways ↓ apoptosis mitochondrial membrane potential restauration ↑myocardial mitochondrial dynamics & Sirt3 expression protective in cardiovascular diseases |
131-141 |
NANOMEDICINE DRUGS Nanocarriers, nanoparticles |
Cyclosporin A nanoparticles conjugated with poly-lactic/glycolic acid or with SS-31 |
142 |
MITOCHONDRIAL TRANSFER |
Re-establishes mitochondrial function |
143 |
LIFE STYLE CHANGES Physical activity/Exercise Calorie restriction; calorie restriction mimetics |
Improve the altered mitochondrial quality control mechanisms; nutritional strategy; the targets include mTOR, sirtuins, diminishment of mitochondrial dysfunction |
2, 5, 33, 144147 |
Table 1: Therapeutic targeting of mitochondrial dysfunction in cardiovascular aging.
The current knowledge ascertains that aging is a remarkably complex process, and mitochondrial dysfunction is only one of its hallmarks. Using experimental models (cell cultures and laboratory animals) a large diversity of compounds with anti-aging effects have been tested/discovered. Moreover, the last decade brought the fast transfer of several promising compounds from preclinical studies to human clinical trials. According to Guarente, Sinclair, and Kroemer [148], the beneficial compounds are metformin (a biguanidine known for its glucose-lowering effects), NAD+ precursors, glucagon-like peptide-1 receptor agonists, TORC1 inhibitors, spermidine, senolytics, probiotics, and antiinflammatory drugs. Large clinical trials are ongoing to check metformin`s effects on health-span extension and cardiovascular advantage [149, 150]. The potential use of peptides in antiaging strategies is facilitated nowadays by the availability of a comprehensive peptide data base (“AagingBase”) [151]. Targeting aging genes is a fast-developing research area, and TERT and ApoE genes are now exploited in clinical trials [152]. In applying caloric restriction, attention is given now to alternative “antiaging “diets (intermittent fasting, protein restriction, ketogenic diets, etc.) [153].
From the survey of human anti-aging therapies, novel research directions emerged:
(i) geroscience (anti-aging medicine), as a strategy to improve health span free of disabled age-related pathologies [149, 154]; back in 2019, Campisi et al identified compounds currently tested in humans for their geroprotective potential: metformin, rapamycin analogs, sirtuin activators (resveratrol, SRT2) [2], nicotinamide riboside, nicotinamide mononucleotide (NAD+ precursors), exercise, and senolytics (discussed in the next subchapter),
(ii) rejuvenating conduits to delay/reverse aging by epigenetic regulation reprograming [155], and
(iii) interventions targeting the (healthy) longevity pathways like MILES (Metformin in Longevity Study) [3, 5, 53, 123].
Current anti-senescence therapies targeting cardiovascular mitochondrial dysfunction
To challenge senescence, the link between mitochondrial dysfunction and cellular senescence is exploited by strategies that adequately adjust and reprogram the disturbed mitochondrial metabolism [153] (Table 2).
Therapeutic approach |
Targeted mitochondrial dysfunction/Effects |
References |
Mitochondrial metabolic reprogramming for senescence alleviation |
↑ OXPHOS efficiency, ↓mtROS |
153 |
Glycolytic enzymes inhibition |
Cellular glucose metabolism alterations → ECs senescence |
156 |
Mitochondrial-derived peptides (Humanin, MOTS-c) administration |
Mitochondrial function regulation, senolytic effects |
157, 158 |
Senolytic drugs |
Induce the selective apoptosis of senescent cells |
30, 159, 160 |
Flavonoids – Quercetin combined with Dasatinib or with Fisetin (Senolytic cocktail) |
Reduce inflammation, alleviates fraility in humans |
80, 81, 161 |
Naringenin, hesperetin, hesperidin, fisetin, kaempferol, rutin, apigenin, luteolin, nobiletin, tangeretin, genistein, wogonin, epigallocatechin gallate (EGCG), theaflavin-3-gallate (TF2A), procyanidin C1 |
Modulate cellular senescence pathways/interact with molecular targets that regulate ageing-related processes. |
162 |
Bcl-2 family protein inhibitor, Navitoclax (ABT-263) |
Clearance of senescent cardiomyocytes, improves myocardial remodelling, diastolic function, and survival following myocardial infarction |
163 |
Heat-Shock Protein 90 inhibitors |
Reduces age-related symptoms in progeroid mice. |
164 |
Polyphenols - Resveratrol (3,5,4’- trihydroxystilbene) with and without nanocarriers. |
Senotherapeutics used in both preclinical and clinical settings. |
165 |
NAD+ precursors supplementation |
CD38/NAD+/SIRT1 axis for enhanced efficacy of geroprotectors |
166 |
Senolytic vaccination |
Reduces atherosclerosis in apolipoprotein E knockout mice on a high-fat diet. |
167 |
Dietary restriction |
Small adipocyte size and low DNA damage |
168 |
Senomorphic drugs |
Suppress SASP |
30,159 |
mTOR inhibition |
Associated with SGLT2, synergistic benefits on senescence pathways |
169 |
JAK/STAT inhibition |
Augments muscle function in myopathy. |
170 |
NF-kB inhibition by avenanthramide C |
SASP suppression |
171 |
Activation of SIRT1 (a longevity modulator) by Nephelium lappaceum (rambutan) seeds |
SASP selective inhibition |
172 |
SIRT3 activation |
Effects linked to exercise-induced adaptation |
173 |
Rapamycin and its derivatives (rapalogs) |
mTOR inhibitors, improve physiological parameters associated with ageing in cardiovascular system, including. |
174 |
Kaempferol |
Anti-inflammatory, antioxidant, and anti-apoptotic actions |
175 |
Ruxolitinib |
Reduces cytokine release and protects the endothelium from Ischemia/Reperfusion-mediated dysfunction. |
176 |
Table 2: Senotherapeutic interventions targeting mitochondrial dysfunction.
Noteworthy, in the therapeutic alleviation of aging-related senescence two types of drugs are considered: the “senolytics” that selectively remove the senescent cells (diminishing their number), and the “senomorphics” that suppress the production/expression of secreted SASP factors (Table 2). Although the search for specific senescent senolytics and senomorphics is ongoing, these drugs have a series of impediments: they are not directed to a certain intracellular pathway and the dynamics of the senescence process across the lifespan make it impossible to use a single drug to target the diversity of senescent cells (the use of cocktails is preferable); the senotherapeutic-associated adverse effects should be also considered: as an example, the caloric restriction could increase the risk for osteoporosis, and is not be employed at people with body mass index less than 21 kg/m2) [12, 81, 172, 178]. A current trend in senescence treatment is the identification of genes associated with this process (http://Senequest.net) along with the translation of preclinical endeavors on “mitochondrial transplantation” to clinical trials [179, 180].
Conclusions, Open Questions and Perspectives
This review provides an updated outlook on the molecular mechanisms of mitochondrial dysfunction in cardiovascular aging and senescence, and emphasizes the specific alleviation therapies. Based on the significant recent progress in this area, it was possible to delineate not only the common and the individual features of mitochondrial dysfunction in aged cardiomyocytes and blood vessels but also the evaluation of the attributes in aging vs. senescence.
It becomes apparent that several pertinent questions related to mitochondrial dysfunction during aging require fast answers. These are the following: (i) whether oxidative stress is a cause or a consequence of the elderly`s cardiovascular pathology, (ii) the uncovering of adequate aging models, (iii) the establishment of strategies for scavenging the dicarbonyl compounds, (iv) the prevention of aging-related mitochondrial proton leak, and (v) the identification of mechanisms involved in microvascular aging [4, 36, 45, 127, 143, 176]. The “geroscience” area needs reliable biomarkers to prevent/delay the aging process, and to assess the efficacy of anti-aging treatments [144, 181]. The ongoing endeavors are focused on the quantification of vascular aging, and on translational research (mitochondrial transfer included) to promote healthy aging and longevity [123, 143, 181, 182].
In the cellular senescence area, an urgent need is uncovering molecular mechanisms beyond the different types of senescence, and identifying phenotypes that “escape” senescence [18]. The recent literature acknowledged the lack of sensitive and specific markers for senescent cells [183]; the MAMs modulation [184], the metabolic reprogramming [185], and the genetic-related approaches, in terms of identification of senescence-related genes [186], and the genomic repair systems operating in this pathology [187, 188] are ongoing trends in this area. Taken together, the above directions at the horizon emphasize the complex interactions taking place in aging and cardiovascular senescence. Once deciphered, further translational research may pave the way toward people`s healthy aging and longer lives.
References
- Giorgi C, Marchi S, Simoes ICM, Ren Z, Morciano G, et al. (2018) Mitochondria and reactive oxygen species in aging and age-related diseases. Int Rev Cell Mol Biol. 340:209-344.
- Campisi J, Kapahi P, Lithgow GJ, Melov S, Newman JC, et al. (2019) From discoveries in ageing research to therapeutics for healthy ageing. Nature. 571:183-192.
- Klaus S, Ost M (2020) Mitochondrial uncoupling and longevity - A role for mitokines? Exp Gerontol. 130:110796.
- Guo J, Huang X, Dou L, Yan M, Shen T, et al. (2022) Aging and agingrelated diseases: from molecular mechanisms to interventions and treatments. Signal Transduct Target Ther. 7:391.
- Amorim JA, Coppotelli G, Rolo AP, Palmeira CM, Ross JM, et al. (2022) Mitochondrial and metabolic dysfunction in ageing and agerelated diseases. Nat Rev Endocrinol. 18:243-258.
- López-Otín C, Blasco MA, Partridge L, Serrano M, Kroemer G (2023) Hallmarks of aging: An expanding universe. Cell. 186:243-278.
- Protasoni M, Serrano M (2023) Targeting mitochondria to control ageing and senescence. Pharmaceutics. 15:352.
- Jasbi P, Nikolich-Žugich J, Patterson J, Knox KS, Jin Y, et al. (2023) Targeted metabolomics reveals plasma biomarkers and metabolic alterations of the aging process in healthy young and older adults. Geroscience. 45: 3131-3146.
- Pilkington SM, Bulfone-Paus S, Griffiths CEM, Watson REB (2021) Inflammaging and the skin. J Invest Dermatol. 141:1087-1095.
- Di Giosia P, Stamerra CA, Giorgini P, Jamialahamdi T, Butler AE, et al. (2022) The role of nutrition in inflammaging. Ageing Res Rev. 77:101596.
- Wu NN, Zhang Y, Ren J (2019) Mitophagy, mitochondrial dynamics, and homeostasis in cardiovascular aging. Oxid Med Cell Longev. 9825061.
- Kirkland JL, Tchkonia T (2020) Senolytic drugs: from discovery to translation. J Intern Med. 288:518-536.
- Lee YH, Park JY, Lee H, Song ES, Kuk MU, et al. (2021) Targeting mitochondrial metabolism as a strategy to treat senescence. Cells. 10:3003.
- Tracy EP, Nair R, Rowe G, Beare JE, Beyer A, et al. (2022) Adipose stromal vascular fraction reverses mitochondrial dysfunction and hyperfission in aging-induced coronary microvascular disease. Am J Physiol Heart Circ Physiol. 323:H749-H762.
- Bottani E, Brunetti D (2023) Advances in mitochondria-targeted drug delivery. Pharmaceutics. 15:2089.
- Rottenberg H (2023) The reduction in the mitochondrial membrane potential in aging: The role of the mitochondrial permeability transition pore. Int J Mol Sci. 24:12295.
- Gorgoulis V, Adams PD, Alimonti A, Bennett DC, Bischof O, et al. (2019) Cellular senescence: Defining a path forward. Cell. 179:813827.
- Sreekumar PG, Hinton DR, Kannan R (2020) The emerging role of senescence in ocular disease. Oxid Med Cell Longev. 2583601.
- Chu X, Wen J, Raju RP (2020) Rapid senescence-like response after acute injury. Aging Cell. 19:e13201.
- Regulski MJ (2017) Cellular senescence: What, why, and how. Wounds. 29:168-174.
- Booth LK, Redgrave RE, Tual-Chalot S, Spyridopoulos I, Phillips HM, et al. (2023) Heart disease and ageing: The roles of senescence, mitochondria, and telomerase in cardiovascular disease. Subcell Biochem. 103:45-78.
- Rodier F, Campisi J (2011) Four faces of cellular senescence. J Cell Biol. 192:547-56.
- Marescal O, Cheeseman IM (2020) Cellular mechanisms and regulation of quiescence. Dev Cell. 55:259-271.
- Jung SH, Hwang HJ, Kang D, Park HA, Lee HC, et al. (2019) mTOR kinase leads to PTEN-loss-induced cellular senescence by phosphorylating p53. Oncogene. 38:1639-1650.
- Luan Y, Zhu X, Jiao Y, Liu H, Huang Z, et al. (2024) Cardiac cell senescence: molecular mechanisms, key proteins and therapeutic targets. Cell Death Discov. 10:78.
- Birch J, Gil J (2020) Senescence and the SASP: many therapeutic avenues. Genes Dev. 34:1565-1576.
- Roger L, Tomas F, Gire V (2021) Mechanisms and regulation of cellular senescence. Int J Mol Sci. 22:13173.
- Qiu L, Liu X, Xia H, Xu C (2022) Downregulation of P300/CBP-associated factor protects from vascular aging via Nrf2 signal pathway activation. Int J Mol Sci. 23:12574.
- Schmitt CA, Tchkonia T, Niedernhofer LJ, Robbins PD, Kirkland JL, et al. (2023) COVID-19 and cellular senescence. Nat Rev Immunol. 23:251-263.
- Zhang L, Pitcher LE, Prahalad V, Niedernhofer LJ, Robbins PD (2023) Targeting cellular senescence with senotherapeutics: senolytics and senomorphics. FEBS J. 290:1362-1383.
- Ziegler DV, Martin N, Bernard D (2021) Cellular senescence links mitochondria-ER contacts and aging. Commun Biol. 4:1323.
- Lin R, Kerkelä R (2020) Regulatory mechanisms of mitochondrial function and cardiac aging. Int J Mol Sci. 21:1359.
- Vakka A, Warren JS, Drosatos K (2023) Cardiovascular aging: from cellular and molecular changes to therapeutic interventions. J Cardiovasc Aging. 3:23.
- Lu X, Thai PN, Lu S, Pu J, Bers DM (2019) Intrafibrillar and perinuclear mitochondrial heterogeneity in adult cardiac myocytes. J Mol Cell Cardiol. 136:72-84.
- Voglhuber J, Holzer M, Radulović S, Thai PN, Djalinac N, et al (2022) Functional remodeling of perinuclear mitochondria alters nucleoplasmic Ca2+ signaling in heart failure. Philos Trans R Soc Lond B Biol Sci. 377:20210320.
- Chen Q, Thompson J, Hu Y, Lesnefsky EJ (2022) Reversing mitochondrial defects in aged hearts: role of mitochondrial calpain activation. Am J Physiol Cell Physiol. 322:C296-C310.
- Sagar S, Gustafsson AB (2023) Cardiovascular aging: the mitochondrial influence. J Cardiovasc Aging. 3:33.
- Salazar G, Cullen A, Huang J, Zhao Y, Serino A, et al. (2020) SQSTM1/ p62 and PPARGC1A/PGC-1alpha at the interface of autophagy and vascular senescence. Autophagy. 16:1092-1110.
- Janikiewicz J, Szymański J, Malinska D, Patalas-Krawczyk P, Michalska B, et al. (2018) Mitochondria-associated membranes in aging and senescence: structure, function, and dynamics. Cell Death Dis. 9:332.
- Moltedo O, Remondelli P, Amodio G (2019) The mitochondria-endoplasmic reticulum contacts and their critical role in aging and age-associated diseases. Front Cell Dev Biol. 7:172.
- Dabravolski SA, Sadykhov NK, Kartuesov AG, Borisov EE, Sukhorukov VN, et al. (2022) Interplay between Zn2+ homeostasis and mitochondrial functions in cardiovascular diseases and heart ageing. Int J Mol Sci. 23:6890.
- Murphy E, Liu JC (2023) Mitochondrial calcium and reactive oxygen species in cardiovascular disease. Cardiovasc Res. 119:1105-1116.
- Andrés CMC, Pérez de la Lastra JM, Andrés Juan C, Plou FJ, PérezLebeña E (2023) Superoxide anion chemistry-its role at the core of the innate immunity. Int J Mol Sci. 24:1841.
- Bou-Teen D, Miro-Casas E, Ruiz-Meana M (2023) Dicarbonyl stress and mitochondrial dysfunction in the aged heart. Aging (Albany NY). 15:3223-3225.
- Bou-Teen D, Fernandez-Sanz C, Miro-Casas E, Nichtova Z, BonzonKulichenko E et al. (2022) Defective dimerization of FoF1-ATP synthase secondary to glycation favors mitochondrial energy deficiency in cardiomyocytes during aging. Aging Cell. 21:e13564.
- Qi X, Rusch NJ, Fan J, Mora CJ, Xie L, et al. (2023) Mitochondrial proton leak in cardiac aging. Geroscience. 45:2135-2143.
- Zhang H, Alder NN, Wang W, Szeto H, Marcinek DJ, et al. (2020) Reduction of elevated proton leak rejuvenates mitochondria in the aged cardiomyocyte. Elife. 9:e60827.
- Abuarab N, Munsey TS, Jiang LH, Li J, Sivaprasadarao A (2017) High glucose-induced ROS activates TRPM2 to trigger lysosomal membrane permeabilization and Zn2+-mediated mitochondrial fission. Sci Signal. 10: eaal4161.
- Marcheggiani F, Cirilli I, Orlando P, Silvestri S, Vogelsang A, et al. (2019) Modulation of Coenzyme Q10 content and oxidative status in human dermal fibroblasts using HMG-CoA reductase inhibitor over a broad range of concentrations. From mitohormesis to mitochondrial dysfunction and accelerated aging. Aging (Albany NY). 11:2565-2582.
- Barcelos IP, Haas RH (2019) CoQ10 and aging. Biology (Basel). 8:28.
- Dai DF, Kang P, Bai H (2023) The mTOR signaling pathway in cardiac aging. J Cardiovasc Aging. 3:24.
- Daneshgar N, Rabinovitch PS, Dai DF (2021) TOR signaling pathway in cardiac aging and heart failure. Biomolecules. 11:168.
- Chen G, Kroemer G, Kepp O (2020) Mitophagy: An emerging role in aging and age-associated diseases. Front Cell Dev Biol. 8:200.
- Zheng Q, Huang J, Wang G (2019) Mitochondria, telomeres and telomerase subunits. Front Cell Dev Biol. 7:274.
- Salazar G, Cullen A, Huang J, Zhao Y, Serino A, et al. (2020) SQSTM1/ p62 and PPARGC1A/PGC-1alpha at the interface of autophagy and vascular senescence. Autophagy. 16:1092-1110.
- Mengozzi A, Pugliese NR, Chiriacò M, Masi S, Virdis A, et al. (2021) Microvascular ageing links metabolic disease to age-related disorders: The role of oxidative stress and inflammation in promoting microvascular dysfunction. J Cardiovasc Pharmacol. 78: S78-S87.
- Liberale L, Badimon L, Montecucco F, Lüscher TF, Libby P, et al. (2022) Inflammation, aging, and cardiovascular disease: JACC Review Topic of the Week. J Am Coll Cardiol. 79:837-847.
- Fulop T, Larbi A, Pawelec G, Khalil A, Cohen AA, et al. (2023) Immunology of aging: the birth of inflammaging. Clin Rev Allergy Immunol. 64:109-122.
- Franceschi C, Bonafè M, Valensin S, Olivieri F, De Luca M, et al. (2000) Infammaging. an evolutionary perspective on immunosenescence. Ann N Y Acad Sci 908:244–254.
- Ferrucci L, Fabbri E (2018) Inflammageing: chronic inflammation in ageing, cardiovascular disease, and frailty. Nat Rev Cardiol. 15:505522.
- Rossman MJ, Gioscia-Ryan RA, Clayton ZS, Murphy MP, Seals DR (2020) Targeting mitochondrial fitness as a strategy for healthy vascular aging. Clin Sci (Lond). 134:1491-1519.
- Kirkman DL, Robinson AT, Rossman MJ, Seals DR, Edwards DG (2021) Mitochondrial contributions to vascular endothelial dysfunction, arterial stiffness, and cardiovascular diseases. Am J Physiol Heart Circ Physiol. 320: H2080-H2100.
- Olgar Y, Degirmenci S, Durak A, Billur D, Can B, et al. (2018) Aging related functional and structural changes in the heart and aorta: MitoTEMPO improves aged-cardiovascular performance. Exp Gerontol. 110:172-181.
- Li A, Yan J, Zhao Y, Yu Z, Tian S, et al. (2023) Vascular aging: Assessment and intervention. Clin Interv Aging. 18:1373-1395.
- an der Loo B, Labugger R, Skepper JN, Bachschmid M, Kilo J, et al. (2000) Enhanced peroxynitrite formation is associated with vascular aging. J Exp Med. 192:1731-1744.
- Tyrrell DJ, Blin MG, Song J, Wood SC, Zhang M, et al. (2020) Ageassociated mitochondrial dysfunction accelerates atherogenesis. Circ Res. 126:298-314.
- Wu JQ, Wang W, Zheng YH (2021) Role of vascular aging in the pathogenesis of abdominal aortic aneurysm and potential therapeutic targets. Zhongguo Yi Xue Ke Xue Yuan Xue Bao. 43:962-968.
- Navas-Madroñal M, Almendra-Pegueros R, Puertas-Umbert L, Jiménez-Altayó F, Julve J, et al. (2023) Targeting mitochondrial stress with Szeto-Schiller 31 prevents experimental abdominal aortic aneurysm: Crosstalk with endoplasmic reticulum stress. Br J Pharmacol. 180:2230-2249.
- Tyrrell DJ, Chen J, Li BY, Wood SC, Rosebury-Smith W, et al. (2022) Aging alters the aortic proteome in health and thoracic aortic aneurysm. Arterioscler Thromb Vasc Biol. 42:1060-1076.
- Jia D, Zhang J, Nie J, Andersen JP, Rendon S, et al. (2021) Cardiolipin remodeling by ALCAT1 links hypoxia to coronary artery disease by promoting mitochondrial dysfunction. Mol Ther. 29:3498-3511.
- Tracy EP, Hughes W, Beare JE, Rowe G, Beyer A, et al. (2021) Aginginduced impairment of vascular function: mitochondrial redox contributions and physiological/clinical implications. Antioxid Redox Signal. 35:974-1015.
- Lee WE, Genetzakis E, Figtree GA (2023) Novel strategies in the early detection and treatment of endothelial cell-specific mitochondrial dysfunction in coronary artery disease. Antioxidants (Basel).12:1359.
- Yang M, Chadwick AE, Dart C, Kamishima T, Quayle JM (2017) Bioenergetic profile of human coronary artery smooth muscle cells and effect of metabolic intervention. PLoS One. 12: e0177951.
- He X, Zeng H, Chen ST, Roman RJ, Aschner JL, et al. (2017) Endothelial specific SIRT3 deletion impairs glycolysis and angiogenesis and causes diastolic dysfunction. J Mol Cell Cardiol. 112:104-113.
- Frías-Anaya E, Gromnicova R, Kraev I, Rogachevsky V, Male DK, et al. (2021) Age-related ultrastructural neurovascular changes in the female mouse cortex and hippocampus. Neurobiol Aging. 101:273-284.
- Seman A, Chandra PK, Byrum SD, Mackintosh SG, Gies AJ, et al. (2023) Targeting mitochondria in the aged cerebral vasculature with SS-31, a proteomic study of brain microvessels. Geroscience. 45:2951-2965.
- Anderson R, Lagnado A, Maggiorani D, Walaszczyk A, Dookun E, et al. (2019) Length-independent telomere damage drives post-mitotic cardiomyocyte senescence. EMBO J. 38: e100492.
- Chiao YA, Zhang H, Sweetwyne M, Whitson J, Ting YS, et al. (2020) Late-life restoration of mitochondrial function reverses cardiac dysfunction in old mice. Elife. 9: e55513.
- Dookun E, Passos JF, Arthur HM, Richardson GD (2022) Therapeutic potential of senolytics in cardiovascular disease. Cardiovasc Drugs Ther. 36:187-196.
- Nieto M, Könisgberg M, Silva-Palacios A (2024) Quercetin and dasatinib, two powerful senolytics in age-related cardiovascular disease. Biogerontology 25:71-82.
- Sweeney M, Cook SA, Gil J (2023) Therapeutic opportunities for senolysis in cardiovascular disease. FEBS J. 290:1235-1255.
- Chen MS, Lee RT, Garbern JC (2022) Senescence mechanisms and targets in the heart. Cardiovasc Res. 118:1173-1187.
- Grilo LF, Zimmerman KD, Puppala S, Chan J, Huber HF, et al. (2024) Cardiac molecular analysis reveals aging-associated metabolic alterations promoting glycosaminoglycans accumulation via hexosamine biosynthetic pathway. Adv Sci (Weinh). 11: e2309211.
- Kaarniranta K, Kajdanek J, Morawiec J, Pawlowska E, Blasiak J (2018) PGC-1α protects RPE Cells of the aging retina against oxidative stress-induced degeneration through the regulation of senescence and mitochondrial quality control. The significance for AMD pathogenesis. Int J Mol Sci.19:2317.
- Hoshino A, Mita Y, Okawa Y, Ariyoshi M, Iwai-Kanai E, et al. (2013) Cytosolic p53 inhibits Parkin-mediated mitophagy and promotes mitochondrial dysfunction in the mouse heart. Nat Commun.4:2308.
- Manzella N, Santin Y, Maggiorani D, Martini H, Douin-Echinard V, et al. (2018) Monoamine oxidase-A is a novel driver of stress-induced premature senescence through inhibition of Parkin-mediated mitophagy. Aging Cell. 17: e12811.
- Iacovino LG, Manzella N, Resta J, Vanoni MA, Rotilio L, et al. (2020) Rational redesign of Monoamine Oxidase A into a dehydrogenase to probe ROS in cardiac aging. ACS Chem Biol. 15:1795-1800.
- Rizvi F, Preston CC, Emelyanova L, Yousufuddin M, Viqar M, et al. (2021) Effects of aging on cardiac oxidative stress and transcriptional changes in pathways of reactive oxygen species generation and clearance. J Am Heart Assoc. 10: e019948.
- Luan Y, Ren KD, Luan Y, Chen X, Yang Y (2021) Mitochondrial dynamics: pathogenesis and therapeutic targets of vascular diseases. Front Cardiovasc Med. 8:770574.
- Ong SB, Kalkhoran SB, Hernández-Reséndiz S, Samangouei P, Ong SG, et al. (2017) Mitochondrial-shaping proteins in cardiac health and disease - the long and the short of It! Cardiovasc Drugs Ther. 31:87107.
- Picca A, Mankowski RT, Burman JL, Donisi L, Kim JS, et al. (2018) Mitochondrial quality control mechanisms as molecular targets in cardiac ageing. Nat Rev Cardiol. 15:543-554.
- Uchikado Y, Ikeda Y, Ohishi M (2022) Current understanding of the pivotal role of mitochondrial dynamics in cardiovascular diseases and senescence. Front Cardiovasc Med. 9:905072.
- Booth LK, Redgrave RE, Folaranmi O, Gill JH, Richardson GD (2022) Anthracycline-induced cardiotoxicity and senescence. Front Aging. 3:1058435.
- Mehdizadeh M, Aguilar M, Thorin E, Ferbeyre G, Nattel S (2022) The role of cellular senescence in cardiac disease: basic biology and clinical relevance. Nat Rev Cardiol. 19:250-264.
- Hernandez-Segura A, Nehme J, Demaria M (2018) Hallmarks of cellular senescence. Trends Cell Biol. 28:436-453.
- Tang X, Li PH, Chen HZ (2020) Cardiomyocyte senescence and cellular communications within myocardial microenvironments. Front Endocrinol (Lausanne). 11:280.
- Victorelli S, Salmonowicz H, Chapman J, Martini H, Vizioli MG, et al. (2023) Apoptotic stress causes mtDNA release during senescence and drives the SASP. Nature. 622: 627-636.
- Latorre E, Pilling LC, Lee BP, Bandinelli S, Melzer D, et al. (2018) The VEGFA156b isoform is dysregulated in senescent endothelial cells and may be associated with prevalent and incident coronary heart disease. Clin Sci (Lond). 132:313-325.
- Suda M, Paul KH, Minamino T, Miller JD, Lerman A, et al. (2023) Senescent cells: a therapeutic target in cardiovascular diseases. Cells. 12:1296.
- Chadda KR, Ajijola OA, Vaseghi M, Shivkumar K, Huang CL, et al. (2018) Ageing, the autonomic nervous system and arrhythmia: From brain to heart. Ageing Res Rev. 48:40-50.
- Rouhi L, Cheedipudi SM, Chen SN, Fan S, Lombardi R, et al. (2021) Haploinsufficiency of Tmem43 in cardiac myocytes activates the DNA damage response pathway leading to a late-onset senescence-associated pro-fibrotic cardiomyopathy. Cardiovasc Res. 117:2377-2394.
- Camacho-Encina M, Booth LK, Redgrave RE, Folaranmi O, Spyridopoulos I, et al. (2024) Cellular senescence, mitochondrial dysfunction, and their link to cardiovascular disease. Cells. 13:353.
- Jesel L, Abbas M, Park SH, Matsushita K, Kindo M, et al. (2019) Atrial fibrillation progression is associated with cell senescence burden as determined by p53 and p16 expression. J Clin Med. 9:36.
- Hwang HJ, Kim N, Herman AB, Gorospe M, Lee JS (2022) Factors and pathways modulating endothelial cell senescence in vascular aging. Int J Mol Sci. 23:10135.
- Xu Q, Mojiri A, Boulahouache L, Morales E, Walther BK, et al. (2022) Vascular senescence in progeria: role of endothelial dysfunction. Eur Heart J Open. 2: oeac047.
- Okuno K, Cicalese S, Elliott KJ, Kawai T, Hashimoto T, et al. (2020) Targeting molecular mechanism of vascular smooth muscle senescence induced by angiotensin II, A Potential Therapy via senolytics and senomorphics. Int J Mol Sci. 21:6579.
- Hu D, Yin C, Luo S, Habenicht AJR, Mohanta SK. (2019) Vascular smooth muscle cells contribute to atherosclerosis immunity. Front Immunol. 10:1101.
- Li X, Liu A, Xie C, Chen Y, Zeng K, et al. (2024) The transcription factor GATA6 accelerates vascular smooth muscle cell senescencerelated arterial calcification by counteracting the role of anti-aging factor SIRT6 and impeding DNA damage repair. Kidney Int. 105:115-131.
- Sun DY, Wu WB, Wu JJ, Shi Y, Xu JJ, et al. (2024) Pro-ferroptotic signaling promotes arterial aging via vascular smooth muscle cell senescence. Nat Commun. 15:1429.
- Lin MJ, Hu SL, Tian Y, Zhang J, Liang N, et al. (2023) Targeting vascular smooth muscle cell senescence: A novel strategy for vascular diseases. J Cardiovasc Transl Res. 16:1010-1020.
- Cui L, Li Z, Chang X, Cong G, Hao L (2017) Quercetin attenuates vascular calcification by inhibiting oxidative stress and mitochondrial fission. Vascul Pharmacol. 88:21-29.
- Chen WJ, Cheng Y, Li W, Dong XK, Wei JL, et al. (2021) Quercetin attenuates cardiac hypertrophy by inhibiting mitochondrial dysfunction through SIRT3/PARP-1 Pathway. Front Pharmacol. 12:739615.
- Cui Z, Zhao X, Amevor FK, Du X, Wang Y, et al. (2022) Therapeutic application of quercetin in aging-related diseases: SIRT1 as a potential mechanism. Front Immunol. 13:943321.
- Chen C, Huang J, Shen J, Bai Q (2019) Quercetin improves endothelial insulin sensitivity in obese mice by inhibiting Drp1 phosphorylation at serine 616 and mitochondrial fragmentation. Acta Biochim Biophys Sin (Shanghai). 51:1250-1257.
- Chen Q, Ruan D, Shi J, Du D, Bian C (2023) The multifaceted roles of natural products in mitochondrial dysfunction. Front Pharmacol. 14:1093038.
- Chang X, Zhang Q, Huang Y, Liu J, Wang Y, et al. (2024) Quercetin inhibits necroptosis in cardiomyocytes after ischemia-reperfusion via DNA-PKcs-SIRT5-orchestrated mitochondrial quality control. Phytother Res. 38:2496-2517.
- Gherardi G, Corbioli G, Ruzza F, Rizzuto R (2022) CoQ10 and Resveratrol effects to ameliorate aged-related mitochondrial dysfunctions. Nutrients. 14:4326.
- Pyo IS, Yun S, Yoon YE, Choi JW, Lee SJ (2020) Mechanisms of aging and the preventive effects of Resveratrol on age-related diseases. Molecules. 25:4649.
- Zheng M, Bai Y, Sun X, Fu R, Liu L, et al. (2022) Resveratrol reestablishes mitochondrial quality control in myocardial ischemia/reperfusion injury through Sirt1/Sirt3-Mfn2-Parkin-PGC-1α pathway. Molecules. 27:5545.
- Wang P, Yang Y, Guo J, Ma T, Hu Y, et al. (2024) Resveratrol inhibits Zinc deficiency-induced mitophagy and exerts cardiac cytoprotective effects. Biol Trace Elem Res. 202:1669-1682.
- Bharath LP, Agrawal M, McCambridge G, Nicholas DA, Hasturk H, et al. (2020) Metformin enhances autophagy and normalizes mitochondrial function to alleviate aging-associated inflammation. Cell Metab. 32:44-55.e6.
- Kulkarni AS, Gubbi S, Barzilai N (2020) Benefits of metformin in attenuating the hallmarks of aging. Cell Metab. 32:15-30.
- Triggle CR, Mohammed I, Bshesh K, Marei I, Ye K, et al. (2022) Metformin: Is it a drug for all reasons and diseases? Metabolism. 133:155223
- Murray KO, Ludwig KR, Darvish S, Coppock ME, Seals DR, et al. (2023) Chronic mitochondria antioxidant treatment in older adults alters the circulating milieu to improve endothelial cell function and mitochondrial oxidative stress. Am J Physiol Heart Circ Physiol. 325:H187H194.
- Beyer AM, Zinkevich N, Miller B, Liu Y, Wittenburg AL, et al. (2017) Transition in the mechanism of flow-mediated dilation with aging and development of coronary artery disease. Basic Res Cardiol. 112:5.
- Olgar Y, Billur D, Tuncay E, Turan B (2020) MitoTEMPO provides an antiarrhythmic effect in aged-rats through attenuation of mitochondrial reactive oxygen species. Exp Gerontol. 136:110961.
- Owada T, Yamauchi H, Saitoh SI, Miura S, Machii H, et al. (2017) Resolution of mitochondrial oxidant stress improves aged-cardiovascular performance. Coron Artery Dis. 28:33-43.
- Mohtashami Z, Singh MK, Salimiaghdam N, Ozgul M, Kenney MC (2022) MOTS-c, the most recent mitochondrial derived peptide in human aging and age-related diseases. Int J Mol Sci. 23:11991.
- Gao G, Chen W, Yan M, Liu J, Luo H, et al. (2020) Rapamycin regulates the balance between cardiomyocyte apoptosis and autophagy in chronic heart failure by inhibiting mTOR signaling. Int J Mol Med. 45:195-209.
- Zong Y, Li H, Liao P, Chen L, Pan Y, et al. (2024) Mitochondrial dysfunction: mechanisms and advances in therapy. Signal Transduct Target Ther. 9:124.
- Agil A, Chayah M, Visiedo L, Navarro-Alarcon M, Rodríguez Ferrer JM, et al. (2020) Melatonin improves mitochondrial dynamics and function in the kidney of Zücker dabetic fatty rats. J Clin Med. 9:2916.
- Arinno A, Maneechote C, Khuanjing T, Ongnok B, Prathumsap N, et al. (2021) Cardioprotective effects of melatonin and metformin against doxorubicin-induced cardiotoxicity in rats are through preserving mitochondrial function and dynamics. Biochem Pharmacol. 192:114743
- Fu Z, Jiao Y, Wang J, Zhang Y, Shen M, et al. (2020) Cardioprotective role of melatonin in acute myocardial infarction. Front Physiol. 11:366.
- Ding M, Feng N, Tang D, Feng J, Li Z, et al. (2018) Melatonin prevents Drp1-mediated mitochondrial fission in diabetic hearts through SIRT1PGC1α pathway. J Pineal Res. 65: e12491.
- Ma WY, Song RJ, Xu BB, Xu Y, Wang XX, et al. (2021) Melatonin promotes cardiomyocyte proliferation and heart repair in mice with myocardial infarction via miR-143-3p/Yap/Ctnnd1 signaling pathway. Acta Pharmacol Sin. 42:921-931.
- Zhang Y, Wang Y, Xu J, Tian F, Hu S, et al. (2019) Melatonin attenuates myocardial ischemia-reperfusion injury via improving mitochondrial fusion/mitophagy and activating the AMPK-OPA1 signaling pathways. J Pineal Res. 66: e12542.
- Wang Z, Gao Z, Zheng Y, Kou J, Song D, et al. (2023) Melatonin inhibits atherosclerosis progression via galectin-3 downregulation to enhance autophagy and inhibit inflammation. J Pineal Res. 74: e12855.
- Hosseini L, Vafaee MS, Badalzadeh R (2020) Melatonin and Nicotinamide Mononucleotide attenuate myocardial ischemia/reperfusion injury via modulation of mitochondrial function and hemodynamic parameters in aged rats. J Cardiovasc Pharmacol Ther. 25:240-250.
- Zhi W, Li K, Wang H, Lei M, Guo Y (2020) Melatonin elicits protective effects on OGD/R insulted H9c2 cells by activating PGC 1α/Nrf2 signaling. Int J Mol Med. 45:1294-1304.
- El-Sayed SF, Abdelhamid AM, ZeinElabdeen SG, El-Wafaey DI, Moursi SMM (2024) Melatonin enhances captopril mediated cardioprotective effects and improves mitochondrial dynamics in male Wistar rats with chronic heart failure. Sci Rep. 14:575.
- Huang X, Zeng Z, Li S, Xie Y, Tong X (2022) The therapeutic strategies targeting mitochondrial metabolism in cardiovascular disease. Pharmaceutics. 14:2760.
- Zhang CX, Cheng Y, Liu DZ, Liu M, Cui H, et al. (2019) Mitochondriatargeted cyclosporin A delivery system to treat myocardial ischemia reperfusion injury of rats. J Nanobiotechnology. 17:18.
- Headley CA, Tsao PS (2023) Building the case for mitochondrial transplantation as an anti-aging cardiovascular therapy. Front Cardiovasc Med. 10:1141124.
- Hood DA, Memme JM, Oliveira AN, Triolo M (2019) Maintenance of skeletal muscle mitochondria in health, exercise, and aging. Annu Rev Physiol. 81:19-41.
- Zhang Y, Oliveira AN, Hood DA (2020) The intersection of exercise and aging on mitochondrial protein quality control. Exp Gerontol. 131:110824.
- Kirkman DL, Chavez DA (2024) Exercise for chronic kidney disease: effects on vascular and cardiopulmonary function. Am J Physiol Heart Circ Physiol. 326:H138-H147.
- Duregon E, Pomatto-Watson LCDD, Bernier M, Price NL, de Cabo R (2021) Intermittent fasting: from calories to time restriction. Geroscience. 43:1083-1092.
- Guarente L, Sinclair DA, Kroemer G (2024) Human trials exploring anti-aging medicines. Cell Metab. 36:354-376.
- Espinoza SE, Khosla S, Baur JA, de Cabo R, Musi N (2023) Drugs targeting mechanisms of aging to delay age-related disease and promote healthspan: Proceedings of a National Institute on Aging Workshop. J Gerontol A Biol Sci Med Sci. 78:53-60.
- Dihoum A, Rena G, Pearson ER, Lang CC, Mordi IR (2023) Metformin: evidence from preclinical and clinical studies for potential novel applications in cardiovascular disease. Expert Opin Investig Drugs. 32:291-299.
- Kunjulakshmi R , Kumar A, Kumar KV, Sengupta A, Kundal K, et al. (2024) AagingBase: a comprehensive data base of anti-aging peptides. Data base (Oxford). baae016.
- Yu J, Li T, Zhu J (2023) Gene therapy strategies targeting aging-related diseases. Aging Dis. 14:398-417.
- Lee MB, Hill CM, Bitto A, Kaeberlein M (2021) Antiaging diets: Separating fact from fiction. Science. 374: eabe7365.
- Mishra M, Wu J, Kane AE, Howlett SE (2024) The intersection of frailty and metabolism. Cell Metab. 36:893-911.
- Ji S, Xiong M, Chen H, Liu Y, Zhou L, et al. (2023) Cellular rejuvenation: molecular mechanisms and potential therapeutic interventions for diseases. Signal Transduct Target Ther. 8:116.
- Stabenow LK, Zibrova D, Ender C, Helbing DL, Spengler K, et al. (2022) Oxidative glucose metabolism promotes senescence in vascular endothelial cells. Cells. 11:2213.
- Kim SJ, Mehta HH, Wan J, Kuehnemann C, Chen J, et al. (2018) Mitochondrial peptides modulate mitochondrial function during cellular senescence. Aging (Albany NY). 10:1239-1256.
- Kong BS, Lee C, Cho YM (2023) Mitochondrial-encoded peptide MOTS-c, diabetes, and aging-related diseases. Diabetes Metab J. 47:315-324.
- Lagoumtzi SM, Chondrogianni N (2021) Senolytics and senomorphics: Natural and synthetic therapeutics in the treatment of aging and chronic diseases. Free Radic Biol Med. 171:169-190.
- Owens WA, Walaszczyk A, Spyridopoulos I, Dookun E, Richardson GD (2021) Senescence and senolytics in cardiovascular disease: Promise and potential pitfalls. Mech Ageing Dev. 198:111540.
- Lee E, Carreras-Gallo N, Lopez L, Turner L, Lin A, et al. (2024) Exploring the effects of Dasatinib, Quercetin, and Fisetin on DNA methylation clocks: a longitudinal study on senolytic interventions. Aging (Albany NY). 16:3088-3106.
- Mbara KC, Devnarain N, Owira PMO (2022) Potential role of polyphenolic flavonoids as senotherapeutic agents in degenerative diseases and geroprotection. Pharmaceut Med. 36:331-352.
- Walaszczyk A, Dookun E, Redgrave R, Tual-Chalot S, Victorelli S, et al. (2019) Pharmacological clearance of senescent cells improves survival and recovery in aged mice following acute myocardial infarction. Aging Cell. 18: e12945.
- Fuhrmann-Stroissnigg H, Ling YY, Zhao J, McGowan SJ, Zhu Y, et al. (2017) Identification of HSP90 inhibitors as a novel class of senolytics. Nat Commun. 8:422.
- Joma N, Bielawski PB, Saini A, Kakkar A, Maysinger D (2024) Nanocarriers for natural polyphenol senotherapeutics. Aging Cell. 23: e14178.
- Sharma A, Chabloz S, Lapides RA, Roider E, Ewald CY (2023) Potential synergistic supplementation of NAD+ promoting compounds as a strategy for increasing healthspan. Nutrients. 15:445.
- Suda M, Shimizu I, Katsuumi G, Yoshida Y, Hayashi Y, et al. (2021) Senolytic vaccination improves normal and pathological age-related phenotypes and increases lifespan in progeroid mice. Nat Aging. 1:1117-1126.
- Ishaq A, Dufour D, Cameron K, von Zglinicki T, Saretzki G (2018) Metabolic memory of dietary restriction ameliorates DNA damage and adipocyte size in mouse visceral adipose tissue. Exp Gerontol. 113:228-236.
- Troise D, Mercuri S, Infante B, Losappio V, Cirolla L, et al. (2024) mTOR and SGLT-2 inhibitors: their synergistic effect on age-related processes. Int J Mol Sci. 25:8676.
- Addinsall AB, Cacciani N, Akkad H, Salah H, Tchkonia T, et al. (2021) JAK/STAT inhibition augments soleus muscle function in a rat model of critical illness myopathy via regulation of complement C3/3R. J Physiol. 599:2869-2886.
- Lim JS, Lee DY, Kim HS, Park SC, Park JT, et al. (2020) Identification of a novel senomorphic agent, avenanthramide C, via the suppression of the senescence-associated secretory phenotype. Mech Ageing Dev. 192:111355.
- Lee YR, Cho HM, Park EJ, Zhang M, Doan TP, et al. (2020) Metabolite profiling of rambutan (Nephelium lappaceum L.) seeds using UPLCqTOF-MS/MS and senomorphic effects in aged human dermal fibroblasts. Nutrients. 12:1430.
- Zhou L, Pinho R, Gu Y, Radak Z (2022) The role of SIRT3 in exercise and aging. Cells. 11:2596.
- Lee DJW, Hodzic Kuerec A, Maier AB (2024) Targeting ageing with rapamycin and its derivatives in humans: a systematic review. Lancet Healthy Longev. 5: e152-e162.
- Hussain MS, Altamimi ASA, Afzal M, Almalki WH, Kazmi I, et al. (2024) Kaempferol: Paving the path for advanced treatments in aging-related diseases. Exp Gerontol. 188:112389.
- Saemann L, Naujoks P, Hartrumpf L, Pohl S, Simm A, et al. (2023) Sex-specific protection of endothelial function after vascular Ischemia/ Reperfusion Injury by the senomorphic agent Ruxolitinib. Int J Mol Sci. 24:11727.
- Li Z, Zhang Z, Ren Y, Wang Y, Fang J, et al. (2021) Aging and age-related diseases: from mechanisms to therapeutic strategies. Biogerontology. 22:165-187.
- Suda M, Paul KH, Tripathi U, Minamino T, Tchkonia T, et al. (2024) Targeting cell senescence and senolytics: Novel interventions for agerelated endocrine dysfunction. Endocr Rev. 45:655-675.
- Zhao R, Dong C, Liang Q, Gao J, Sun C, et al. (2024) Engineered mitochondrial transplantation as an anti-aging therapy. Aging Dis.
- Zhai P, Sadoshima J (2024) Cardiomyocyte senescence and the potential therapeutic role of senolytics in the heart. J Cardiovasc Aging. 4:18.
- Maier JA, Andrés V, Castiglioni S, Giudici A, Lau ES, et al. (2023) Aging and vascular disease: A multidisciplinary overview. J Clin Med. 12:5512.
- Zhong G, Venkatesan JK, Madry H, Cucchiarini M (2022) Advances in human mitochondria-based therapies. Int J Mol Sci. 24:608.
- Kritsilis M, V Rizou S, Koutsoudaki PN, Evangelou K, Gorgoulis VG, et al. (2018) Ageing, cellular senescence and neurodegenerative disease. Int J Mol Sci. 19:2937.
- Jiang T, Ruan N, Luo P, Wang Q, Wei X, et al. (2024) Modulation of ER-mitochondria tethering complex VAPB-PTPIP51: Novel therapeutic targets for aging-associated diseases. Ageing Res Rev. 98:102320.
- Xie S, Xu SC, Deng W, Tang Q (2023) Metabolic landscape in cardiac aging: insights into molecular biology and therapeutic implications. Signal Transduct Target Ther. 8:114.
- Xiang J, Shen J, Zhang L, Tang B (2022) Identification and validation of senescence-related genes in circulating endothelial cells of patients with acute myocardial infarction. Front Cardiovasc Med. 9:1057985.
- Lee SY, Kim JJ, Miller KM (2021) Bromodomain proteins: protectors against endogenous DNA damage and facilitators of genome integrity. Exp Mol Med. 53:1268-1277.
- Yousefzadeh M, Henpita C, Vyas R, Soto-Palma C, Robbins P, et al. (2021) DNA damage-how and why we age? Elife.10:e62852.