Skip to main content
GeroScience logoLink to GeroScience
. 2025 Jan 3;47(1):301–337. doi: 10.1007/s11357-024-01489-2

Sex-specific mechanisms in vascular aging: exploring cellular and molecular pathways in the pathogenesis of age-related cardiovascular and cerebrovascular diseases

Anna Ungvari 1,✉,#, Rafal Gulej 2,#, Roland Patai 2,#, Zoltan Papp 3,4, Attila Toth 3,4, Attila Á Szabó 3,4, Bruno K Podesser 5, Péter Sótonyi 6, Zoltán Benyó 7,8, Andriy Yabluchanskiy 2,9,10,11,12,13,14, Stefano Tarantini 2,9,10,11,12, Andrea B Maier 15,16,17,18, Anna Csiszar 2,10,11,12, Zoltan Ungvari 2,9,10,11,12
PMCID: PMC11872871  PMID: 39754010

Abstract

Aging remains the foremost risk factor for cardiovascular and cerebrovascular diseases, surpassing traditional factors in epidemiological significance. This review elucidates the cellular and molecular mechanisms underlying vascular aging, with an emphasis on sex differences that influence disease progression and clinical outcomes in older adults. We discuss the convergence of aging processes at the macro- and microvascular levels and their contributions to the pathogenesis of vascular diseases. Critical analysis of both preclinical and clinical studies reveals significant sex-specific variations in these mechanisms, which could be pivotal in understanding the disparity in disease morbidity and mortality between sexes. The review highlights key molecular pathways, including oxidative stress, inflammation, and autophagy, and their differential roles in the vascular aging of males and females. We argue that recognizing these sex-specific differences is crucial for developing targeted therapeutic strategies aimed at preventing and managing age-related vascular pathologies. The implications for personalized medicine and potential areas for future research are also explored, emphasizing the need for a nuanced approach to the study and treatment of vascular aging.

Keywords: Vascular aging, Cellular senescence, Menopause, Endocrine, Sex differences, Sexual dimorphism, Atherosclerosis, Vascular cognitive impairment, Ischemic heart disease

Introduction

The adage, “a man is as old as his arteries,” attributed to the seventeenth-century physician Thomas Sydenham, succinctly encapsulates the pivotal role of vascular health in determining longevity and quality of life. Modern geroscience affirms this view, placing cardiovascular and cerebrovascular diseases at the forefront of morbidity and mortality in aging populations [1, 2]. While traditional risk factors such as hypertension, diabetes mellitus, obesity, and hypercholesterolemia are significant, advanced age emerges as the paramount predictor of vascular disease prevalence, underscoring the critical role of vascular aging in the pathogenesis of a wide range of cardiovascular and cerebrovascular diseases [13].

Vascular aging involves a multifaceted array of structural and functional alterations within both large vessels and the microcirculation, including pathological remodeling of the vascular wall, increased arterial stiffness and atherogenesis, endothelial dysfunction, pro-inflammatory phenotypic changes, barrier dysfunction, and microvascular rarefaction [1, 2]. These changes are not isolated phenomena but are driven by an intricate interplay of evolutionarily conserved molecular and cellular mechanisms of aging. Keys among these mechanisms are oxidative stress, chronic low-grade inflammation, mitochondrial dysfunction, cellular senescence, and impaired proteostasis. These aging processes not only undermine the structural and functional integrity of the vascular system but also exert widespread effects on other organ systems, influencing overall health span [1, 2]. The systemic impact of vascular aging highlights its role as both a driver and a consequence of the broader aging process, with far-reaching implications for the health and longevity of the entire organism [1, 2].

Emerging research, however, suggests that the impact and progression of vascular aging are not uniformly experienced across sexes [46]. Preclinical and clinical research reveals substantial sex differences in the molecular and cellular mechanisms of aging, which significantly influence the onset, progression, and therapeutic outcomes of age-related diseases in general [7], and cardiovascular and cerebrovascular disorders in particular [46, 8]. These distinctions, evident at the genetic, cellular, and systemic levels, underscore the need for a nuanced understanding of how aging impacts males and females differently, shaping disease manifestation and response to treatment in distinct ways. For instance, differences in hormone levels, gene expression, cellular stress resilience, immune function, endothelial reactivity, metabolic processing, and response to oxidative stress between males and females could explain variations in the prevalence and severity of vascular diseases [5, 6, 8, 9]. Additionally, sex-specific variations in autophagy, inflammatory response, and senescence-associated secretory phenotypes further influence how vascular aging manifests distinctly across sexe [5, 6, 8, 9]. These mechanisms collectively contribute to the differential risk and progression of cardiovascular and cerebrovascular conditions observed between males and females.

This review aims to dissect these sex differences in vascular aging, highlighting how they modulate the risk and progression of age-related vascular diseases. By integrating insights from recent preclinical and clinical studies, this overview explores how these distinctions can guide the development of targeted therapeutic interventions, paving the way for personalized medicine approaches that enhance cardiovascular and cerebrovascular health in older adults. Understanding these sex-specific pathways is not only crucial for advancing treatment but also for designing preventative strategies that accommodate the unique physiological profiles of males and females, ultimately improving health outcomes across the lifespan.

Sex differences in vascular aging

Vascular aging manifests differently between sexes, influencing the prevalence, progression, and outcomes of both age-related macrovascular and microvascular diseases [6, 8, 10, 11]. These sex differences are evident in various vascular conditions, including atherosclerotic diseases such as ischemic heart disease and acute myocardial infarction (AMI) [6, 12, 13], stroke [1417], peripheral artery disease (PAD) [10], and aneurysms [1822]. Sex differences are also apparent in vascular contributions to cognitive impairment and dementia (VCID) [2328] and Alzheimer’s disease (AD) [23, 2933], as well as age-related macular degeneration (AMD) [34]. Understanding these differences is crucial for developing sex-specific prevention strategies and treatments.

Estrogen plays a pivotal role in modulating vascular health and vascular aging processes in women [8, 35]. Its protective effects include promoting a youthful endothelial phenotype by increasing nitric oxide bioavailability and reducing inflammation and oxidative stress [3539]. Before menopause, these effects help delay the progression of atherosclerosis compared to men [35, 40, 41]. However, post-menopause, the decline in estrogen levels leads to an accelerated vascular aging process in women, often surpassing the risk levels seen in men [5, 8, 35, 39, 4251]. Conversely, testosterone in men influences vascular tone and structure but tends to predispose them to earlier vascular stiffening and hypertension [52, 53].

Men typically develop atherosclerosis earlier than women, a disparity largely attributed to protective effects of estrogen in premenopausal women [41, 48, 50, 51]. However, the risk in women increases and may surpass that in men post-menopause, suggesting a role for sex hormones in modulating disease progression. Men have a higher incidence of AMI at a younger age compared to women [54]. However, women tend to have worse outcomes post-AMI, including higher mortality rates [54]. This discrepancy may be due to differences in the pathophysiology of AMI between sexes, with women more likely to experience microvascular dysfunction rather than large vessel obstruction [55, 56]. Stroke also exhibits sex-specific patterns, with men having a higher incidence at younger ages, but women catching up and even surpassing men in older age groups [54]. Women also tend to have poorer recovery and higher mortality rates after a stroke [54]. Differences in risk factors, such as the higher prevalence of atrial fibrillation in elderly women, contribute to these patterns [5658]. PAD is common in both sexes but underdiagnosed in women, partly due to atypical presentation [59, 60]. Women with PAD often experience worse functional impairment and quality of life than men [61]. The reasons behind these differences are not fully understood but may involve sex-specific responses to ischemia [61]. Abdominal aortic aneurysms (AAAs) are more prevalent in men, while women with AAAs have a higher risk of rupture at smaller sizes [22, 62, 63], suggesting differences in the biology of the aneurysm wall or in the distribution of mechanical stress across it [6466]. Differences in body fat distribution, inflammatory responses, and plaque characteristics between sexes [67, 68] likely also influence the progression of PAD and the risk and rupture rates of aortic aneurysms.

Women are at a higher risk of developing AD [32, 33, 69] and VCID [2628]. The link between vascular aging and neurodegenerative conditions such as AD is increasingly recognized as a critical factor in understanding sex differences in cognitive outcomes. Cerebrovascular health, particularly the integrity of the cerebral small vessels and the blood–brain barrier (BBB), plays a central role in maintaining brain homeostasis. Microvascular pathologies, including cerebral small vessel disease (CSVD), play a pivotal role in the development of VCID and AD [7077]. Aging-related cerebrovascular changes, such as endothelial dysfunction, reduced capillary density, and impaired autoregulation of cerebral blood flow, exacerbate neuronal injury and cognitive decline. These microvascular changes can lead to a range of structural and functional abnormalities in the brain, including white matter lesions, microbleeds, and lacunar infarcts, which are closely associated with cognitive decline and dementia [7884]. The disruption of the brain’s microvascular network impairs cerebral blood flow regulation and BBB integrity, leading to chronic hypoperfusion, heightened neuroinflammation, and oxidative stress—conditions that exacerbate neuronal damage and neurodegeneration [71, 77, 85, 86]. In women, these risks may be further elevated due to life-course vascular risk factors such as hypertension and diabetes mellitus [87], which are known to predispose individuals to microvascular damage. Additionally, post-menopausal hormonal changes significantly affect vascular health, potentially increasing susceptibility to CSVD and its cognitive consequences [8891]. Understanding the interplay between microvascular health and neurodegenerative processes is essential for developing targeted therapies that can mitigate the progression of VCID and AD, particularly in populations at increased risk due to sex-specific pathophysiological changes.

AMD is a complex eye condition that primarily affects the macula, the central part of the retina responsible for sharp, detailed vision [9294]. AMD prevalence is slightly higher in women [34, 92, 9597]. While the exact mechanisms underlying AMD are not fully understood, microvascular contributions play a significant role in the progression and severity of the disease [94, 98, 99]. One of the hallmarks of wet AMD (the more severe form of the disease) is the growth of abnormal blood vessels from the choroid into the macular region [99]. These vessels are often fragile and leaky, leading to hemorrhage, fluid accumulation, and rapid vision loss. The integrity of the retinal blood supply is crucial for maintaining photoreceptor health. In AMD, changes in the microvasculature, such as capillary drop-out and reduced perfusion, can lead to ischemic conditions within the retina [99]. This ischemia contributes to the degeneration of photoreceptors and the accumulation of waste products like lipofuscin and drusen, exacerbating the disease process. In AMD, inflammation around the retinal microvasculature can lead to structural changes in the blood-retina barrier, promoting further leakage and damage [99101]. The inflammatory milieu can also accelerate the deposition of extracellular matrix components, contributing to drusen formation, a key feature in the early stages of AMD [99].

Age-related endothelial dysfunction: role of estrogen

Estrogen plays a crucial role in maintaining endothelial function, which is central to vascular health [39, 40]. One of its primary protective mechanisms involves the upregulation of endothelial nitric oxide synthase (eNOS), promoting the production of NO [37, 102, 103]. NO is a critical mediator of vascular tone, anti-inflammatory signaling, and vascular homeostasis. By increasing NO bioavailability, estrogen helps preserve endothelial function, delay arterial stiffening, and reduce oxidative stress and inflammation [104]. Before menopause, the presence of circulating estrogen confers significant vascular protection to women, delaying the onset of endothelial dysfunction and related cardiovascular diseases compared to men of the same age [4, 5, 8, 39, 105]. However, the decline in estrogen levels following menopause marks a pivotal shift in vascular aging for women [105]. Reduced estrogen levels lead to decreased NO production, increased oxidative stress, and an elevated inflammatory burden [5, 8, 105]. This decline accelerates endothelial dysfunction, contributing to a higher risk of developing age-related vascular diseases, including atherosclerosis, hypertension, and VCID [8]. Post-menopausal endothelial dysfunction is also linked to microvascular rarefaction and impaired angiogenesis [106], exacerbating the risk of ischemic diseases and neurovascular complications [5, 8]. Estrogen’s influence extends beyond direct vascular effects. It also modulates epigenetic and transcriptional pathways that regulate endothelial function [104]. For example, estrogen has been shown to influence DNA methylation patterns of vascular genes [107], histone acetylation [104], and the expression of non-coding RNAs such as miRNAs [106, 108] that regulate angiogenesis and inflammation. These molecular effects collectively shape the trajectory of endothelial aging in women. Understanding the role of estrogen in endothelial function underscores the need for targeted therapeutic strategies, such as hormone replacement therapy (HRT), to mitigate post-menopausal vascular aging [4, 5, 8, 35]. However, the risks and benefits of HRT remain an area of active research, necessitating individualized approaches based on cardiovascular risk profiles and other health considerations. Future studies should focus on identifying estrogen-mimetic therapies that preserve endothelial health without the adverse effects associated with traditional HRT, thereby providing a pathway to improve vascular aging outcomes in post-menopausal women.

Genetic predispositions and epigenetic modifications in vascular aging

Sex differences in vascular aging are influenced not only by hormonal factors but also by genetic predispositions and epigenetic modifications [6, 7,]. Genetic variations in genes related to vascular function, such as those encoding antioxidant enzymes and regulators of endothelial nitric oxide production, have been shown to differ between men and women. These genetic differences can predispose individuals to sex-specific patterns of vascular aging and associated disease risks [6}. Epigenetic modifications play a crucial role in mediating sex-specific differences in vascular function and aging [173]. This regulatory effect diminishes post-menopause, leading to an epigenetic shift that contributes to accelerated vascular aging in women. These genetic and epigenetic factors collectively contribute to the observed differences in clinical outcomes and disease progression between sexes. Understanding these mechanisms provides critical insights into the interplay between genetics, epigenetics, and sex hormones in vascular aging and highlights potential targets for personalized therapeutic interventions.

Mechanisms of vascular aging and sex differences

Vascular aging is governed by a complex interplay of evolutionarily conserved molecular and cellular mechanisms [1, 2] that exhibit significant sex differences [6, 8]. These differences influence how aging-related pathologies develop and respond to treatment in males and females [6, 8]. Vascular aging contributes to the pathogenesis and progression of a wide range of cardiovascular and cerebrovascular diseases [1, 2]. This process manifests across a continuum, where shared mechanisms promote age-related pathologies in both large vessels, such as atherosclerotic vascular diseases, and microvascular pathologies [1, 2, 86]. These microvascular issues range from contributions to cognitive decline and AD, to AMD, and microvascular pathologies in ischemic heart disease and intermittent claudication. This continuum underscores the interconnected nature of vascular aging, affecting various organ systems through similar pathological mechanisms [86]. Building on previous insights [1, 2, 71, 109113], this section explores the critical mechanisms involved in vascular aging, highlighting how they differ between sexes.

Oxidative and nitrative stress

Oxidative stress, implicated in endothelial dysfunction associated with both macrovascular and microvascular aging, primarily arises from increased reactive oxygen species (ROS) production by NAD(P)H oxidases and mitochondria [1, 2, 109, 110]. This process results in endothelial dysfunction and arterial stiffening by inactivating nitric oxide (NO), a critical regulator of vascular tone and health [1, 2, 109]. Peroxynitrite, a potent oxidant formed from the interaction of NO with superoxide, exacerbates vascular aging through cytotoxic effects and the activation of inflammatory pathways, further contributing to large artery stiffening and the risk of microvascular pathologies such as cerebral microhemorrhages [114]. Additionally, ROS contributes to vascular aging by causing DNA damage, which promotes cellular senescence, and by activating NF-kB, a key factor in the cascade leading to age-related sterile low-grade inflammation [1, 2, 115118].

Cellular resilience to oxidative stress, critical for maintaining vascular health, is significantly compromised in aging by impaired function of nuclear factor erythroid 2-related factor 2 (Nrf2), a major transcriptional regulator of antioxidant response [117, 119125]. Nrf2 dysfunction contributes to the pathophysiology of vascular aging by diminishing the cellular ability to counteract the damaging effects of ROS [117, 119125]. Under normal conditions, Nrf2 activates the expression of various antioxidant enzymes that help detoxify ROS and protect cells from oxidative damage. However, with aging and in the presence of chronic oxidative stress, Nrf2 signaling is often disrupted, which not only reduces the cellular defenses against oxidative stress but also exacerbates endothelial dysfunction [117, 121, 125]. This impaired oxidative stress resilience due to Nrf2 dysfunction is further linked to several other aging-related vascular issues [117]. The decrease in protective antioxidant responses leads to enhanced oxidative damage to cellular components such as lipids, proteins, and DNA and thereby promotes cellular senescence. The resulting damage accelerates the aging process in vascular cells, promotes the development of atherosclerotic lesions, and increases the susceptibility of the vasculature to inflammatory injuries [117]. Therefore, restoring Nrf2 function and enhancing the cellular antioxidant capacity are promising therapeutic targets for mitigating vascular aging and its associated pathologies [124, 126, 127].

Sex differences in oxidative stress

Oxidative and nitrative stress play crucial roles in vascular aging, with significant differences observed between males and females in how these processes influence vascular health [128131]. These differences are largely mediated by biological variations in hormone levels, antioxidant capacity, and cellular responses to oxidative damage.

Estrogen exerts a protective effect against oxidative stress [39, 132, 133]. It enhances the expression and activity of various antioxidant enzymes [132, 133] and can modulate the expression of NAD(P)H oxidases [134], which are primary sources of ROS in vascular cells. Estrogen’s ability to upregulate eNOS also promotes higher nitric oxide availability [37, 103]. As a result, premenopausal women generally experience lower levels of oxidative and nitrative stress and better endothelial function compared to men of the same age, potentially delaying the onset of vascular aging [39, 135]. However, this protective effect diminishes with menopause, leading to an increase in oxidative stress that accelerates vascular aging in post-menopausal women [105, 136139].

Men and women may also exhibit genetic differences that affect their cellular responses to oxidative stress. These can include variations in genes related to antioxidant defenses such as superoxide dismutase (SOD), catalase, and glutathione peroxidase [131]. Additionally, males may have a higher baseline oxidative status, partially due to the androgen testosterone, which has been linked to higher production of ROS [140]. This intrinsic difference could explain why men generally develop oxidative stress–related vascular diseases at a younger age compared to women.

Understanding these sex-specific pathways in oxidative and nitrative stress can guide more targeted interventions. Antioxidant therapies may have differing efficacies in men and women, particularly considering hormonal influences and genetic predispositions [141]. For instance, antioxidant supplementation that benefits post-menopausal women might not be as effective in men or premenopausal women, underscoring the need for personalized treatment strategies based on sex-specific research [141].

Mitochondrial dysfunction

Mitochondria are central to aging, with declines in respiratory chain efficiency leading to increased ROS production and decreased ATP generation [110, 142, 143]. Vascular aging is associated with impaired mitochondrial biogenesis and enhanced mitochondrial ROS output, which promotes oxidative stress and inflammation via mechanisms such as NF-κB activation [110, 144151]. These changes negatively impact cellular energy metabolism and endothelial NO production, and are key drivers of vascular aging.

Sex differences in mitochondrial dysfunction

Mitochondrial dysfunction is a critical factor in vascular aging, with distinct manifestations observed between males and females [129, 152156]. These differences are shaped by variations in mitochondrial biogenesis, dynamics, and susceptibility to oxidative damage, which are influenced by genetic and hormonal factors. Estrogens play a significant role in modulating mitochondrial function [157161]. They are known to enhance mitochondrial efficiency and promote the expression of genes involved in mitochondrial biogenesis through estrogen receptor–mediated mechanisms [162167]. This results in better maintenance of mitochondrial DNA integrity and more efficient electron transport chain function in females, particularly before menopause [167]. Although one might anticipate premenopausal women to have lower levels of mitochondrial-derived reactive oxygen species (ROS) and a better cellular energy balance compared to men, the current evidence supporting this assumption is not robust. Further research is needed to clarify these potential sex-related differences in mitochondrial function and their implications for health [155]. Sex chromosomes themselves may influence mitochondrial function. The presence of two X chromosomes in females includes genes that impact mitochondrial activity and could provide a genetic buffer against mitochondrial decay that is not present in males [168, 169]. Additionally, epigenetic modifications regulated by sex hormones can alter the expression of mitochondrial and nuclear genes related to oxidative phosphorylation, affecting mitochondrial function differently in males and females [170173]. The sex-specific differences in mitochondrial function may impact the development and progression of various vascular diseases [170]. Recognizing these sex differences in mitochondrial dysfunction is important for designing effective interventions. Therapies aimed at enhancing mitochondrial function, such as coenzyme Q10, resveratrol, and other mitochondria-targeted antioxidants, might be more effective if tailored to the specific mitochondrial health profiles of men and women. Additionally, hormone replacement therapy in post-menopausal women could be considered not only for its traditional benefits but also for its potential to maintain mitochondrial health [174].

Cellular senescence

Cellular senescence is a hallmark of aging that plays a critical role in the progression of various age-related diseases [117, 175183]. Cells of the aging cardiovascular system also often enter a state of senescence, characterized by irreversible cell cycle arrest and adoption of a pro-inflammatory senescence-associated secretory phenotype (SASP) [1, 116, 117, 177, 184189]. This state is particularly detrimental in vascular contexts, where senescent endothelial cells contribute significantly to endothelial dysfunction, impaired angiogenesis, and compromise the structural integrity of the microcirculatory network [187, 188, 190]. The removal of these senescent cells through experimental senolytic interventions has been shown to mitigate vascular aging effects and improve endothelial function, rescue blood–brain barrier, and prevent the genesis of cerebral microhemorrhages in preclinical models [188, 190192].

Sex differences in cellular senescence

Estrogen is known to have a protective effect against the onset and progression of cellular senescence [104, 193, 194]. It enhances DNA repair mechanisms and modulates inflammatory responses, thereby delaying the senescence onset in vascular endothelial cells [195199]. Estrogen also suppresses the activity of pathways commonly associated with senescence [200]. These preclinical studies suggest that as women approach menopause and experience declines in estrogen levels, there may be an acceleration in the rate of cellular senescence, which contributes to diminishing the observed differences in vascular aging between sexes noted in younger demographics [104]. The SASP includes the release of pro-inflammatory cytokines, chemokines, and proteases that can disrupt local tissue environments and promote paracrine senescence [184, 201203]. Men and women may exhibit different SASP profiles due to hormonal influences [204, 205]. Additionally, sex differences in the production of certain inflammatory mediators could potentially exacerbate the local and systemic effects of SASP. Senescent cells likely contribute to a variety of vascular aging outcomes, including endothelial dysfunction [184, 186, 191, 201, 206, 207], barrier dysfunction [115, 207210], atherogenesis [138, 181, 211215], and impaired angiogenesis and capillarization [115, 207, 216218], which are fundamental to the development of AMI, stroke, VCID, and other cardiovascular diseases. The acceleration of senescence in post-menopausal women [219] might lead to a more rapid decline in vascular health compared to men of the same age. Understanding sex differences in cellular senescence is critical for designing effective senolytic therapies—treatments that selectively remove senescent cells. Current research in senolytic agents has shown promise in preclinical models, including the potential to restore endothelial function [184, 186, 190, 201, 207, 220], rescue the integrity of the blood–brain barrier [115, 207], and prevent cerebral microhemorrhages [188]. However, the efficacy and safety of these interventions may vary between sexes, necessitating sex-specific adjustments in therapeutic approaches.

Heightened state of inflammation

Chronic low-grade sterile inflammation is a hallmark of vascular aging and plays a pivotal role in the progression of various age-related vascular diseases [1, 2, 71, 86, 149, 209, 221]. This heightened state of inflammation is primarily driven by increased ROS, cellular senescence, and the activation of key inflammatory pathways such as NF-κB [113, 122, 216, 222224]. These factors lead to endothelial activation and increased expression of pro-inflammatory cytokines, creating a vascular environment that is conducive to atherogenesis and other pathological changes. The process of atherogenesis is significantly influenced by inflammation [225230]. Inflammatory cytokines along with chemokines and adhesion molecules, facilitate the recruitment and adhesion of monocytes to the endothelium. These monocytes then migrate into the intima, differentiate into macrophages, and ingest oxidized LDL, forming foam cells—a key event in the early stages of atherosclerotic plaque formation. Chronic inflammation also contributes to the progression and instability of these plaques [231, 232]. Cytokines and growth factors released in the inflamed vascular environment promote the proliferation of vascular smooth muscle cells, which migrate into the intima, contributing to plaque growth and the fibrous cap formation. Inflammation also impairs endothelial function and disrupts the barrier function of the endothelium, increasing its permeability. Inflammation also affects coagulation pathways, increasing the risk of thrombosis, and contributes to structural changes in the vasculature, which lead to increased arterial stiffness. This stiffening of arteries elevates systolic blood pressure and decreases diastolic pressure, which can impair coronary artery perfusion and contribute to myocardial ischemia. Additionally, the increased arterial stiffness is likely to play a significant role in the genesis of cerebrovascular injury and the pathogenesis of VCID [71, 84].

Sex differences in vascular inflammation

Notable sex differences in chronic low-grade sterile inflammation exist, significantly influencing its impact on vascular health [4, 9, 51, 205, 233]. These differences are crucial for understanding the disparities in the prevalence and progression of vascular diseases between males and females [6, 8, 9, 16, 20, 51, 233235]. Estrogens have anti-inflammatory properties that play a significant role in modulating immune responses in women. These hormones generally suppress the activation of NF-κB and other pro-inflammatory pathways, reducing the production of inflammatory cytokines, chemokines, and adhesion molecules [6, 8, 51]. This mechanism helps premenopausal women maintain a lower inflammatory state within the vascular system, providing a protective buffer against atherogenesis and related vascular pathologies [9, 48, 51]. After menopause, the decline in estrogen levels leads to an increase in inflammatory activity, which can accelerate the progression of atherosclerosis and increase susceptibility to vascular diseases such as stroke and myocardial infarction [51]. Understanding these sex-specific inflammatory responses is vital for developing targeted interventions. Anti-inflammatory treatments may require customization to be effective and safe across different sex and age groups.

Autophagy impairment

Autophagy impairment is a significant factor in vascular aging, affecting the health and functionality of vascular cells [1, 2]. Autophagy is a critical cellular process for recycling damaged or dysfunctional cellular components, helping to maintain cell integrity and function [236239]. As individuals age, the efficiency of the autophagy process diminishes, contributing to the accumulation of cellular debris and dysfunctional organelles. This decline has profound implications for vascular health, particularly in the endothelium and vascular smooth muscle cells.

In vascular endothelial and smooth muscle cells, autophagy regulates several key functions that impact overall vascular health [236, 239241]. These include the removal of oxidized and glycated proteins, which accumulate as a result of metabolic and oxidative stress, and the turnover of dysfunctional mitochondria (mitophagy), which is crucial to preventing the release of pro-inflammatory mitochondrial DNA and other DAMPs (damage-associated molecular patterns) into the cytoplasm [236, 239]. By maintaining the quality of mitochondria, autophagy directly influences the production of ROS and the overall oxidative stress burden within vascular cells [237, 242]. Impaired autophagy leads to the accumulation of oxidized proteins and damaged organelles, particularly mitochondria [236, 239]. This accumulation exacerbates oxidative stress within vascular cells, contributing to endothelial dysfunction and atherosclerotic changes. The build-up of cellular debris can activate innate immune responses, leading to chronic inflammation in the vascular wall [236, 239]. This inflammation can accelerate the progression of vascular diseases, including atherosclerosis and hypertension [236, 239241]. The inability to clear dysfunctional components can accelerate the onset of cellular senescence in vascular cells. Enhancing autophagy represents a promising therapeutic strategy for mitigating the impacts of vascular aging [236, 237, 239, 242]. Interventions such as calorie restriction and other anti-aging dietary interventions, exercise, and pharmacological agents like rapamycin [243], which stimulates autophagy via inhibition of the mTOR pathway, have shown potential in restoring autophagic function and improving vascular health [239].

Sex differences in autophagy

Autophagy exhibits notable sex differences, particularly influenced by hormonal regulation [244248]. Estrogen has been shown to enhance autophagic activity. However, post-menopause, as estrogen levels drastically decrease, there is a corresponding and significant decline in autophagic efficiency [161, 249]. Beyond hormonal effects, genetic and cellular differences between sexes also impact autophagy [250]. Research has identified sex-specific variations in the expression of genes integral to the autophagy pathway [246, 251]. These differences suggest that male and female sex hormones might regulate these genes in divergent ways, potentially influencing how cells from each sex initiate and process autophagic activity. Furthermore, the cellular response to metabolic and oxidative stress, which are critical triggers for autophagy, can differ markedly between males and females. Such differences may affect not only the baseline levels of autophagy but also the efficiency and outcomes of these processes under stress conditions.

Impaired proteostasis

Impaired proteostasis, characterized by disruptions in the balance and function of proteins within cells, is a critical factor in vascular aging [1, 2]. Proteostasis encompasses all aspects of protein synthesis, folding, trafficking, and degradation, ensuring that proteins maintain their correct structure and functional states [252]. As the vascular system ages, the mechanisms that support proteostasis become less effective, leading to the accumulation of misfolded proteins and the formation of protein aggregates [252]. This impairment can significantly affect the health and function of vascular cells, contributing to a variety of age-related vascular diseases [253].

With age, the efficiency of protein synthesis machinery declines, and the cellular environment becomes less conducive to correct protein folding [252]. This can be exacerbated by oxidative stress and inflammation, common in aging, which further disrupts protein structure and function [252]. Chaperone proteins, which assist in the folding and refolding of misfolded proteins, show reduced expression and functionality in aging vascular cells. Similarly, the ubiquitin–proteasome system, responsible for degrading unwanted proteins, becomes less efficient. This reduction in quality control contributes to the vascular pathologies associated with aging. As previously discussed, autophagy plays a significant role in removing damaged and dysfunctional proteins and organelles. A decline in autophagic activity with age leads to an accumulation of defective proteins and cellular components, further impairing vascular cell function and contributing to disease processes such as atherosclerosis.

Sex differences in proteostasis

Estrogens have been shown to influence several aspects of proteostasis, including enhancing the expression of molecular chaperones and components of the protein degradation machinery [253256]. This hormonal support is vital in maintaining protein homeostasis, facilitating the proper folding and disposal of misfolded proteins, and preventing the accumulation of harmful protein aggregates that can lead to vascular cell damage. The protective effects of estrogens are particularly prominent in younger women but see a marked reduction as estrogen levels decline with age [257]. This reduction correlates with an increased proteostatic stress in post-menopausal women, potentially accelerating the process of vascular aging by allowing the build-up of damaged and dysfunctional proteins. Moreover, inherent sex differences exist in the baseline expression and activity of critical proteostatic components [256, 258, 259], including chaperones, elements of the ubiquitin–proteasome system, and autophagy-related proteins. These differences can lead to divergent responses to the challenges of maintaining protein homeostasis as both men and women age.

Epigenetic changes

Epigenetic modifications, which include DNA methylation, histone modifications, and the expression of non-coding RNAs, are crucial in regulating gene expression and play a pivotal role in the aging process, including the aging of the vascular system [1, 2, 177, 260, 261]. Changes in these epigenetic regulatory mechanisms can lead to altered cellular functions and are implicated in the development and progression of age-related vascular pathologies [1, 2]. Understanding and manipulating the epigenetic landscape in vascular cells provides a powerful approach to potentially reverse or mitigate the effects of aging on the vascular system, improving overall cardiovascular health and longevity [260, 262].

Methylation of DNA at cytosine bases adjacent to guanine (CpG sites) is a common epigenetic modification that generally represses gene expression [263266]. In vascular aging, DNA methylation patterns change significantly [267]. These changes can lead to the repression of protective genes or the activation of harmful genes involved in processes such as inflammation, cell adhesion, and smooth muscle cell proliferation.

Histones can undergo various modifications, such as acetylation, methylation, and phosphorylation [261, 268, 269]. These modifications can influence chromatin structure and gene expression. In the context of vascular aging, changes in histone acetylation and methylation patterns can significantly affect genes involved in inflammatory pathways and oxidative stress responses [261, 267]. Importantly, pharmacological activation of histone deacetylases, such as SIRT-1, which leads to decreased histone acetylation levels, has been associated with downregulated expression of pro-inflammatory genes and improved mitochondrial function in endothelial cells [222, 270274].

Non-coding RNAs, including miRNAs and long non-coding RNAs (lncRNAs), also play roles in regulating gene expression through epigenetic mechanisms. MiRNAs can degrade target mRNAs or inhibit their translation, affecting many aspects of vascular function, including cell proliferation, apoptosis, and capillary formation. Aging is associated with changes in the expression of specific miRNAs [122, 260, 275277], which have been linked to impaired angiogenesis, atherosclerosis, and hypertension. LncRNAs are involved in the regulation of chromatin dynamics and transcription, influencing vascular aging by modifying the cellular environment to favor aging processes [278281].

Sex differences in epigenetic regulation

Epigenetic modifications play a critical role in vascular aging, with distinct patterns observed between males and females [282284]. In females, estrogens are known to influence the methylation of genes that are crucial for vascular protection [107, 285, 286]. This includes genes involved in anti-inflammatory responses and maintaining endothelial cell function. Sex hormones, including estrogen, are likely responsible for the multifaceted sex differences in DNA methylation [287293]. As estrogen levels decline with menopause, changes in methylation patterns [294297] can lead to the activation of genes that promote inflammation and vascular dysfunction, accelerating vascular aging in women. Histone modifications also show sex-specific variations [298, 299] that may affect vascular aging. For example, histone acetylation, which is generally associated with increased gene expression, can be differentially regulated by sex hormones. These modifications can affect the expression of genes involved in vascular smooth muscle cell proliferation, endothelial function, and inflammation, thereby modulating the progression of vascular aging in a sex-dependent manner. The roles of miRNAs and lncRNAs in regulation of cellular aging processes also differ between sexes [300302]. These small regulatory RNA molecules are crucial in fine-tuning the expression of genes associated with vascular aging, such as those regulating endothelial function, angiogenic processes, oxidative stress responses, and inflammation. Variations in the expression and activity of these miRNAs and lncRNAs between males and females can lead to differential susceptibility to vascular diseases as aging progresses. Understanding these sex-specific epigenetic modifications not only sheds light on the complex dynamics of vascular aging but also highlights potential therapeutic targets. By addressing these epigenetic factors, particularly in the context of the changes induced by menopause, targeted interventions can be developed to mitigate the heightened risk of vascular aging in women, aligning with precision medicine approaches.

Energy sensing pathways

Energy sensing pathways play a critical role in modulating cellular responses to environmental and metabolic cues and are intimately involved in the aging processes of the vascular system [1, 2]. Key pathways include those regulated by the mechanistic target of rapamycin (mTOR), adenosine monophosphate–activated protein kinase (AMPK), and sirtuins [1, 2, 238]. These pathways not only influence cellular metabolism and energy balance but also affect endothelial function and the structural integrity of the vascular system.

The mTOR pathway is a central regulator of cell growth and metabolism, responding to nutrients, growth factors, and cellular energy levels [303306]. In vascular cells, mTOR activity influences endothelial cell proliferation, angiogenesis, and response to stress [238, 306309]. However, overactivation of mTOR has been linked to accelerated aging, including increased endothelial cell senescence, enhanced inflammatory responses, and atherosclerosis [310, 311]. Inhibiting mTOR with compounds like rapamycin has been shown to extend lifespan in various organisms [312316] and to ameliorate age-related vascular dysfunction [310, 311] by reducing oxidative stress, inflammation, and enhancing autophagy.

AMPK acts as a cellular energy sensor, activated under conditions of low energy (high AMP/ATP ratio) to restore energy balance by promoting catabolic processes and inhibiting anabolic ones [238, 317320]. In the vascular system, AMPK activation improves endothelial function by enhancing the bioavailability of nitric oxide, reducing oxidative stress, and inhibiting inflammatory responses [238, 321, 322]. It also promotes vascular smooth muscle cell relaxation, thereby reducing arterial stiffness and hypertension. Drugs like metformin, which activate AMPK, have been used clinically to improve metabolic health and have shown promise in reducing age-related vascular diseases and extending lifespan [239, 323, 324].

Sirtuins are a family of NAD + -dependent deacetylases that play a major role in cellular stress resistance, metabolism, and aging [325, 326]. In vascular aging, sirtuins, particularly SIRT1, are critical for maintaining endothelial homeostasis [104, 273, 325, 327331]. They regulate endothelial nitric oxide synthase (eNOS), protect cells from oxidative stress, and inhibit pro-inflammatory pathways [270, 272, 273, 328334]. Sirtuins also influence the function of vascular smooth muscle cells and the extracellular matrix, affecting vascular elasticity and integrity [327, 335342]. Enhancing sirtuin activity, through diet (such as calorie restriction), exercise, or pharmacological agents like resveratrol, has been shown to improve vascular function and reduce age-related arterial stiffening [124, 126, 127, 149, 223, 270, 273, 331, 332, 343349].

Modulating these energy sensing pathways offers potential therapeutic strategies for combating vascular aging and its associated diseases. By targeting mTOR, AMPK, and sirtuins, it is possible to enhance endothelial function, reduce vascular inflammation and stiffness, and ultimately improve cardiovascular health in aging populations. Developing drugs that specifically and effectively modulate these pathways could provide a means to mitigate the effects of aging on the vascular system, potentially extending vascular health span and overall longevity.

Sex differences in energy sensing pathways

Energy sensing pathways may play differential roles in vascular aging across sexes due to intrinsic hormonal and metabolic differences [303, 350]. The mTOR pathway is influenced by sex due to hormonal interactions [350, 351]. Testosterone in males and estrogens in females can modulate mTOR activity differently [303, 350, 352354]. For example, estrogen is known to inhibit mTOR signaling, which may help delay the onset of mTOR-associated vascular aging phenomena. Additionally, lifespan extension by the mTOR inhibitor rapamycin has been observed to vary somewhat between males and females [312, 355]. Studies suggest that while both sexes benefit from rapamycin treatment, the extent and nature of these benefits can differ, potentially due to variations in baseline mTOR activity influenced by sex-specific hormonal environments [303, 350]. AMPK also exhibits sex-dependent activity variations, potentially due to differences in body composition and hormonal regulation [321, 356, 357]. Females typically have higher baseline levels of AMPK activity, possibly due to estrogen’s effect on enhancing AMPK activation. This could confer a protective effect against the development of metabolic syndromes and their vascular consequences.

NAD+ deficiency in vascular aging

NAD+ is a vital coenzyme essential for redox reactions, playing a pivotal role in metabolizing nutrients to release energy within cells [358360]. Beyond its critical role in metabolic processes, NAD+ also serves as a substrate for several enzymes that are important in the regulation of cellular aging and stress responses, including sirtuins [361363] and poly(ADP-ribose) polymerases (PARPs) [364]. As individuals age, NAD+ levels in the body steadily decline, contributing significantly to the aging process, particularly in the vascular system [359, 360, 365].

The mechanisms of age-related NAD+ decline include increased consumption, reduced biosynthesis, and inefficient recycling [360, 366371]. Aging is associated with increased oxidative-notrative stress–mediated DNA damage, which activates PARPs [372378]. PARPs use NAD+ to repair damaged DNA, thus depleting cellular NAD+ reserves [364, 372378]. The rate of NAD+ biosynthesis also decreases with age due to reduced expression of enzymes involved in the NAD+ biosynthesis pathway, such as nicotinamide phosphoribosyltransferase (NAMPT) [360]. Additionally, NAD+ is recycled in cells through the salvage pathway, which also declines with age, further contributing to reduced overall levels of NAD+ [360, 371].

NAD+ is essential for the function of sirtuins, particularly SIRT1, which plays a key role in maintaining endothelial health by deacetylating and activating eNOS, regulating mitochondrial function and oxidative stress resilience pathways [360, 371, 379, 380]. Reduced NAD + levels lead to diminished SIRT1 activity [371, 381, 382], impaired NO production, and endothelial dysfunction [360, 380], a precursor to VCID, atherosclerosis, and hypertension. The NAD + /sirtuin axis also regulate cellular antioxidant defenses [360]. With lower NAD+ availability, these defenses weaken, increasing oxidative stress and vascular inflammation, which are critical drivers of vascular aging [360]. Caloric restriction and intermittent fasting have been shown to increase NAD+ levels and activate sirtuins, enhancing vascular function and reducing oxidative stress [122, 331, 360, 383, 384]. Supplementing with NAD+ precursors such as nicotinamide riboside (NR) and nicotinamide mononucleotide (NMN) has been shown to effectively restore cellular NAD+ levels [385387]. Inhibition of PARP-1 also increases cellular NAD+ levels in aging [364, 373, 376378]. In preclinical studies, supplementations with NAD+ precursors and lifestyle interventions and pharmacological treatments [388392] that increase NAD+ levels have demonstrated potential in reversing age-related phenotypic and functional changes [388, 389], including enhancing endothelial function, diminishing oxidative stress, and promoting transcriptional rejuvenation in endothelial cells [147, 260, 271, 272, 364, 380].

Sex differences in NAD+ metabolism

The decline in NAD+ levels with age and its impact on aging processes exhibit significant sex differences [393395]. Estrogen has been shown to enhance the expression of enzymes involved in the NAD+ biosynthesis pathway, such as NAMPT [396, 397], and can improve the efficiency of NAD+ recycling mechanisms. This hormonal support helps maintain higher NAD+ levels in premenopausal women, providing them with a protective buffer against early vascular aging [393, 395]. As women enter menopause and estrogen levels decline, this metabolic advantage decreases, leading to a sharper drop in NAD+ levels and accelerated vascular aging compared to men [393, 395]. Understanding these sex differences is key for optimizing NAD+-boosting therapies, such as supplementation with NR or NMN. These interventions can potentially restore NAD+ levels and sirtuin activity more effectively if tailored to the specific metabolic needs and hormonal profiles of men and women. For example, post-menopausal women might benefit significantly from NAD+ supplementation combined with strategies to modulate estrogen levels, thereby synergistically enhancing endothelial function and vascular health.

Systemic factors in vascular aging: insights from heterochronic parabiosis

Aging involves significant alterations in systemic factors that regulate vascular function, including various hormones, cytokines, and metabolites derived from different organs [1, 2, 398404]. Vascular aging is notably influenced by circulating factors that can either accelerate aging (pro-geronic) or counteract it (anti-geronic) [399, 403, 404]. These factors are crucial in modulating cellular metabolism, inflammation, and the structural properties of blood vessels.

Heterochronic parabiosis is a research technique where two animals of different ages are surgically joined, creating a shared circulatory system [399, 403, 404]. This method has provided profound insights into the systemic regulation of aging. Studies using this approach have shown that older animals exhibit signs of rejuvenation when exposed to the blood of younger animals [403, 404]. Conversely, younger animals show signs of accelerated aging when their circulatory systems are combined with those of older animals [399]. These effects are thought to be mediated by various blood-borne factors that differ in concentration between young and old blood. Key findings from heterochronic parabiosis suggest that young blood contains factors that can rejuvenate aged tissue and improve vascular function, potentially reversing aspects of vascular aging such as endothelial dysfunction, oxidative stress, impaired barrier function, impaired angiogenesis, and dysregulation of the vascular mitochondrial transcriptome [398, 399, 403406]. These factors include, but are not limited to, proteins and endocrine factors involved in enhancing blood flow, reducing inflammation, maintaining mitochondrial health, and promoting cellular repair and regeneration [398, 399, 403406]. Circulating pro-geronic and anti-geronic factors, which modulate cellular aging processes throughout the body, are derived from the brain, endocrine system [400, 407413], immune system, adipose tissue [179, 414], and other organs (including the gastrointestinal tract and the symbiotic microbiota). Sex differences in the function and secretory profile of these organs can significantly impact vascular aging, contributing to the observed differences in vascular aging phenotypes between males and females. For example, variations in the production of various endocrine and immune factors, and adipokines as well as microbiota-derived factors between men and women can influence the rate and nature of vascular aging in a sex-specific manner. Understanding these differences is crucial for developing targeted interventions aimed at mitigating the impact of aging on vascular health in both sexes.

Sex differences in endocrine regulation of aging

The endocrine system plays a crucial role in the regulation of aging and lifespan, with significant sex differences that influence how hormones such as estrogen and insulin-like growth factor 1 (IGF-1) impact age-related physiological changes [398, 415419].

Estrogen is well-known for its role in modulating lifespan and the aging process, particularly evident in rodent studies [416421]. In female mice, estrogen has been shown to enhance lifespan and delay the onset of age-related pathologies. This hormone exerts protective effects against oxidative stress, helps maintain mitochondrial function, and influences gene expression related to longevity. Estrogen’s effects are partially mediated through its interaction with estrogen receptors, which can activate pathways that promote cellular health and prevent age-related decline. The decline in estrogen levels during menopause in humans correlates with an acceleration of aging symptoms and an increase in the risk of diseases such as osteoporosis and cardiovascular disorders, highlighting its critical role in aging trajectories.

IGF-1 is another key hormone implicated in the regulation of aging and lifespan [398]. It affects various age-related conditions including cardiovascular diseases, osteoporosis, and neurodegenerative disorders [398, 401, 422, 423]. IGF-1 levels naturally decrease with age, but the patterns of this decline and its physiological impacts vary significantly between the sexes [424, 425]. Research, including findings from the Mayo Clinic Study of Aging, indicates that men experience greater decreases in total IGF-1 levels compared to women [424]. This differential trajectory can be attributed to factors such as baseline hormone levels, body mass index, comorbidities like hypertension and cardiovascular diseases, and lifestyle factors such as physical activity levels [424]. The variations in IGF-1 levels between sexes are also influenced by the use of hormone replacement therapy (HRT) in women [424]. HRT has been shown to modulate the natural decline of IGF-1, potentially mitigating some of the negative effects associated with lower hormone levels post-menopause. This suggests that exogenous hormones could play a beneficial role in managing age-related changes by supporting more stable IGF-1 levels. Recent findings have further highlighted the complex role of IGF-1 in aging, particularly its sex-specific effects on lifespan. Genetic IGF-1 deficiency has been shown to exert differing impacts on longevity in humans, with these effects varying significantly between males and females [426]. Preclinical studies have reinforced this observation, demonstrating that the influences of IGF-1 on lifespan regulation and aging processes are distinctly sex-dependent [422, 423].

In conclusion, the endocrine regulation of aging is profoundly influenced by sex, with estrogen and IGF-1 playing pivotal roles in determining aging trajectories and lifespan. Understanding these hormonal effects not only helps explain the differences in aging patterns between men and women but also offers potential avenues for therapeutic interventions. By targeting these hormonal pathways, possibly through approaches like HRT for post-menopausal women or interventions to stabilize IGF-1 levels, it may be possible to ameliorate age-related declines and improve the quality of life in older adults. Future research should continue to explore these sex differences and their implications for clinical strategies aimed at extending cardiovascular health span and managing age-associated cardiovascular and cerebrovascular diseases.

Impact of sex on therapeutic interventions

Sex differences significantly influence the efficacy and outcomes of therapeutic interventions aimed at mitigating aging processes, necessitating a tailored approach to treatment that considers these biological variations [205, 254, 312, 314, 355, 418, 427432]. As research in this area advances, it is becoming increasingly clear that men and women respond differently to various therapies, including senolytics, antioxidants, and lifestyle modifications [7]. These differences underscore the importance of personalized medicine and the development of sex-specific treatment protocols.

Senolytics, a class of drugs designed to selectively eliminate senescent cells, have shown promise in preclinical studies in reducing vascular aging and improving cardiovascular health [115, 188, 190]. However, the response to senolytic therapies may vary between sexes [433435]. For instance, estrogen may modulate the SASP, potentially altering the effectiveness of senolytic drugs in women, particularly those who are premenopausal or on hormone replacement therapy. Additionally, the expression of senescence markers and the accumulation of senescent cells can differ between men and women, influencing the therapeutic impact of senolytics. These factors highlight the need for sex-specific dosing and treatment strategies to optimize the benefits of senolytic therapies.

Antioxidants and mitochondria-targeted therapies, which aim to reduce oxidative stress and improve cellular energetics may also exhibit sex-dependent efficacy [436]. As women generally have more robust antioxidant defenses, the therapeutic benefits of antioxidant supplementation might differ between men and women, with men potentially deriving greater benefit from these interventions. However, post-menopausal women, who experience a decline in estrogen levels, may see a shift in this dynamic, making them more susceptible to oxidative damage and potentially more responsive to antioxidant therapy.

Lifestyle modifications, including dietary changes, exercise, and stress management, are foundational in managing vascular aging [1, 2, 4, 437]. However, the impact of these interventions can be influenced by sex-specific factors [4, 437442]. For example, women may experience greater benefits from lifestyle interventions that promote estrogen activity, such as those that involve phytoestrogens or specific exercise regimens that modulate hormone levels. Men, on the other hand, may respond differently to calorie restriction or physical activity due to differences in metabolism and muscle mass. These differences necessitate a tailored approach to lifestyle interventions, ensuring that recommendations are aligned with the unique physiological profiles of men and women.

Perspectives

As our understanding of vascular aging continues to evolve, it is clear that significant gaps remain in our knowledge of how sex differences influence this process. Addressing these gaps is crucial for developing more effective, personalized interventions to mitigate the impact of vascular aging on health. Despite advances in geroscience and vascular biology, the specific mechanisms by which sex differences affect vascular aging are not fully understood. Current research often overlooks the influence of sex hormones, genetic factors [443], and the interaction between sex-specific lifestyle factors and vascular aging. Additionally, many studies on vascular aging and its therapeutic interventions have predominantly used male models, leading to a lack of comprehensive data on female-specific pathways and responses. Preclinical research should also encourage the use of both male and female animal models from different species, including non-human primates and dogs [444, 445], to explore the underlying mechanisms of sex differences in vascular aging. These studies should examine how sex hormones, gene expression, and cellular stress responses contribute to the differential aging of the vasculature in males and females. There is also a pressing need for large-scale, longitudinal studies that track and compare vascular aging in both males and females over time. These studies should focus on identifying sex-specific risk factors, progression patterns, and outcomes related to vascular aging. By following individuals across different stages of life, we can better understand how sex influences the trajectory of vascular aging and how complex early interventions might alter these trajectories in a sex-specific manner.

The recognition of sex differences in the response to therapeutic interventions [446448] has profound implications for personalized healthy longevity medicine [449]. It emphasizes the need for developing sex-specific treatment protocols that account for the unique biological and hormonal landscapes of men and women [450]. Personalized medicine approaches should consider these differences when designing clinical trials, interpreting outcomes, and implementing treatment plans. This tailored approach not only enhances the efficacy of interventions but also minimizes the risk of adverse effects, ultimately improving the quality of care for both sexes. Additionally, recruiting a balanced number of male and female participants in clinical trials is essential for generating data that accurately reflects sex differences in treatment outcomes. Further, research should explore the potential benefits of combining lifestyle interventions with pharmacological treatments to optimize outcomes for each sex.

Sex-specific biomarkers represent a crucial, yet underdeveloped, area in the assessment of vascular aging and disease risk. Current diagnostic approaches often fail to account for the distinct biological and hormonal factors that drive differences in vascular aging between men and women, resulting in sub-optimal predictive accuracy and therapeutic targeting. There is a need for more human research and research on non-human primates into the genomic and epigenomic factors that drive sex differences in vascular aging. Studies that explore how DNA methylation, histone modifications, and non-coding RNAs differ between men and women could provide new insights into the molecular underpinnings of vascular aging. Such research could also reveal potential biomarkers for early detection and targeted treatment of vascular aging. Emerging evidence points to the potential utility of sex-specific biomarkers, including inflammatory cytokines, epigenetic markers, and proteomic signatures, in refining diagnostic and prognostic models for vascular diseases. Furthermore, a critical area that requires more attention is the study of sex differences in microvascular aging. Microvascular health is crucial for the function of all organs, and its decline is implicated in a range of age-related conditions, including cognitive impairment [78, 86, 209, 451, 452] and ischemic heart disease. There is an urgent need to investigate how sex influences microvascular aging processes, including endothelial function, capillary density, and microcirculatory blood flow in humans. Additionally, the phenomenon of accelerated vascular aging, such as that observed in conditions like long COVID [453, 454], may manifest differently between sexes, potentially due to varying immune responses, hormonal influences, and underlying vascular health. Understanding these differences could provide vital insights into how to tailor interventions for conditions characterized by accelerated vascular aging, ensuring that therapies are effective for both men and women.

Addressing the gaps in our understanding of sex differences in vascular aging is essential for advancing the field and improving health outcomes for both men and women. By focusing on these key areas for future research, we can develop more effective, personalized strategies to combat vascular aging, ultimately enhancing the quality of life and longevity across populations. The integration of sex-specific insights into clinical practice will mark a significant step forward in the pursuit of personalized healthy longevity medicine and prevention and optimal management of age-related vascular diseases [455].

Acknowledgements

This work was also supported by the EKÖP-2024-2 and EKÖP-2024-9 New National Excellence Program of the Ministry for Culture and Innovation from the source of the National Research, Development and Innovation Fund.

Funding

This work was supported by grants from the National Institute on Aging (R01AG072295, R01AG055395, R01AG068295; R01AG070915), the National Institute of Neurological Disorders and Stroke (R01NS100782), the National Cancer Institute (R01CA255840). AU was supported by TKP2021-NKTA-47, implemented with the support provided by the Ministry of Innovation and Technology of Hungary from the National Research, Development and Innovation Fund, financed under the TKP2021-NKTA funding scheme; by funding through the National Cardiovascular Laboratory Program (RRF-2.3.1–21-2022–00003) provided by the Ministry of Innovation and Technology of Hungary from the National Research, Development and Innovation Fund; Project no. 135784 implemented with the support provided from the National Research, Development and Innovation Fund of Hungary, financed under the K20 funding scheme and the European University for Well-Being (EUniWell) program (grant agreement number: 101004093/ EUniWell/EAC-A02-2019 / EAC-A02-2019–1).

Declarations

Ethics approval

The content is solely the responsibility of the authors and does not necessarily represent the official views of the National Institutes of Health. The 4o version of ChatGPT, developed by OpenAI, was used as a language tool to refine our writing, enhancing the clarity of our work.

Competing interests

Dr. Zoltan Papp, Dr. Andriy Yabluchanskiy, Dr. Andrea Maier, Dr. Stefano Tarantini, and Dr. Anna Csiszar serve as Associate Editors for GeroScience. Dr. Zoltan Ungvari serves as Editor-in-Chief for GeroScience and has personal relationships with individuals involved in the submission of this paper.

Footnotes

Publisher's Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Anna Ungvari, Rafal Gulej, and Roland Patai contributed equally to this manuscript.

References

  • 1.Ungvari Z, Tarantini S, Sorond F, Merkely B, Csiszar A. Mechanisms of Vascular aging, a geroscience perspective: JACC focus seminar. J Am Coll Cardiol. 2020;75:931–41. 10.1016/j.jacc.2019.11.061. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 2.Ungvari Z, Tarantini S, Donato AJ, Galvan V, Csiszar A. Mechanisms of vascular aging. Circ Res. 2018;123:849–67. 10.1161/CIRCRESAHA.118.311378. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 3.Fekete M, Major D, Feher A, Fazekas-Pongor V, Lehoczki A. Geroscience and pathology: a new frontier in understanding age-related diseases. Pathol Oncol Res. 2024. 10.3389/pore.2024.1611623. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 4.Moreau KL, Clayton ZS, DuBose LE, Rosenberry R, Seals DR. Effects of regular exercise on vascular function with aging: does sex matter? Am J Physiol Heart Circ Physiol. 2024;326:H123–37. 10.1152/ajpheart.00392.2023. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 5.Lopez Laporte MA, Coutinho T. Vascular aging in women. Can J Cardiol. 2024. 10.1016/j.cjca.2024.01.036. [DOI] [PubMed] [Google Scholar]
  • 6.Ji H, Kwan AC, Chen MT, Ouyang D, Ebinger JE, Bell SP, Niiranen TJ, Bello NA, Cheng S. Sex differences in myocardial and vascular aging. Circ Res. 2022;130:566–77. 10.1161/CIRCRESAHA.121.319902. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 7.Austad SN. Sex differences in health and aging: a dialog between the brain and gonad? Geroscience. 2019;41:267–73. 10.1007/s11357-019-00081-3. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 8.Moreau KL. Modulatory influence of sex hormones on vascular aging. Am J Physiol Heart Circ Physiol. 2019;316:H522–6. 10.1152/ajpheart.00745.2017. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 9.Dela Justina V, Miguez JSG, Priviero F, Sullivan JC, Giachini FR, Webb RC. Sex differences in molecular mechanisms of cardiovascular aging. Front Aging. 2021;2:725884. 10.3389/fragi.2021.725884. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 10.Tseng CH. Sex difference in the distribution of atherosclerotic risk factors and their association with peripheral arterial disease in Taiwanese type 2 diabetic patients. Circ J. 2007;71:1131–6. 10.1253/circj.71.1131. [DOI] [PubMed] [Google Scholar]
  • 11.Price JF, Lee AJ, Fowkes FG, Housley E, Riemersma RA, Lowe GD. Influence of high-density lipoprotein cholesterol and rheological factors on the sex difference in cardiovascular disease. J Cardiovasc Risk. 2000;7:49–56. 10.1177/204748730000700109. [DOI] [PubMed] [Google Scholar]
  • 12.Chung JH, Lee KE, Lee JM, Her AY, Kim CH, Choi KH, Song YB, Hahn JY, Kim HY, Choi JH, Garg S, Doh JH, Nam CW, Koo BK, Shin ES. Effect of sex difference of coronary microvascular dysfunction on long-term outcomes in deferred lesions. JACC Cardiovasc Interv. 2020;13:1669–79. 10.1016/j.jcin.2020.04.002. [DOI] [PubMed] [Google Scholar]
  • 13.Woodward M. Cardiovascular disease and the female disadvantage. Int J Environ Res Public Health. 2019;16:1165. 10.3390/ijerph16071165. [DOI] [PMC free article] [PubMed]
  • 14.Carcel C, Wang X, Sandset EC, Delcourt C, Arima H, Lindley R, Hackett ML, Lavados P, Robinson TG, Munoz Venturelli P, Olavarria VV, Brunser A, Berge E, Chalmers J, Woodward M, Anderson CS. Sex differences in treatment and outcome after stroke: pooled analysis including 19,000 participants. Neurology. 2019;93:e2170–80. 10.1212/WNL.0000000000008615. [DOI] [PubMed] [Google Scholar]
  • 15.Ayala C, Croft JB, Greenlund KJ, Keenan NL, Donehoo RS, Malarcher AM, Mensah GA. Sex differences in US mortality rates for stroke and stroke subtypes by race/ethnicity and age, 1995–1998. Stroke. 2002;33:1197–201. [DOI] [PubMed] [Google Scholar]
  • 16.Ryu WS, Chung J, Schellingerhout D, Jeong SW, Kim HR, Park JE, Kim BJ, Kim JT, Hong KS, Lee K, Park TH, Park SS, Park JM, Kang K, Cho YJ, Park HK, Lee BC, Yu KH, Oh MS, Lee SJ, Kim JG, Cha JK, Kim DH, Lee J, Han MK, Park MS, Choi KH, Lee J, Bae HJ, Kim DE. Biological mechanism of sex difference in stroke manifestation and outcomes. Neurology. 2023;100:e2490–503. 10.1212/WNL.0000000000207346. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 17.Maeda K, Toyoda K, Minematsu K, Kobayashi S, Japan Standard Stroke Registry Study G. Effects of sex difference on clinical features of acute ischemic stroke in Japan. J Stroke Cerebrovasc Dis. 2013;22:1070–5. 10.1016/j.jstrokecerebrovasdis.2012.07.006. [DOI] [PubMed] [Google Scholar]
  • 18.Rehman S, Chandra RV, Zhou K, Tan D, Lai L, Asadi H, Froelich J, Thani N, Nichols L, Blizzard L, Smith K, Thrift AG, Stirling C, Callisaya ML, Breslin M, Reeves MJ, Gall S. Sex differences in aneurysmal subarachnoid haemorrhage (aSAH): aneurysm characteristics, neurological complications, and outcome. Acta Neurochir (Wien). 2020;162:2271–82. 10.1007/s00701-020-04469-5. [DOI] [PubMed] [Google Scholar]
  • 19.Korfer D, Grond-Ginsbach C, Hakimi M, Bockler D, Erhart P. Arterial aneurysm localization is sex-dependent. J Clin Med. 2022;11:2450. 10.3390/jcm11092450. [DOI] [PMC free article] [PubMed]
  • 20.Zuurbier CCM, Molenberg R, Mensing LA, Wermer MJH, Juvela S, Lindgren AE, Jaaskelainen JE, Koivisto T, Yamazaki T, Uyttenboogaart M, van Dijk JMC, Aalbers MW, Morita A, Tominari S, Arai H, Nozaki K, Murayama Y, Ishibashi T, Takao H, Gondar R, Bijlenga P, Rinkel GJE, Greving JP, Ruigrok YM. Sex difference and rupture rate of intracranial aneurysms: an individual patient data meta-analysis. Stroke. 2022;53:362–9. 10.1161/STROKEAHA.121.035187. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 21.Teng A, Sun A, Chen Z, Deng X, Fan Y. Sex different abdominal artery anatomy may induce different displacement force on stent-graft after endovascular aneurysm repair. Int J Numer Method Biomed Eng. 2023;39:e3739. 10.1002/cnm.3739. [DOI] [PubMed] [Google Scholar]
  • 22.Makrygiannis G, Courtois A, Drion P, Defraigne JO, Kuivaniemi H, Sakalihasan N. Sex differences in abdominal aortic aneurysm: the role of sex hormones. Ann Vasc Surg. 2014;28:1946–58. 10.1016/j.avsg.2014.07.008. [DOI] [PubMed] [Google Scholar]
  • 23.Fitzpatrick AL, Kuller LH, Ives DG, Lopez OL, Jagust W, Breitner JC, Jones B, Lyketsos C, Dulberg C. Incidence and prevalence of dementia in the Cardiovascular Health Study. J Am Geriatr Soc. 2004;52:195–204. 10.1111/j.1532-5415.2004.52058.x. [DOI] [PubMed] [Google Scholar]
  • 24.Zinman J, Kapoor A, Si K, Sujanthan S, Southwell A, Cayley ML, Sicard MN, Lien K, Murray BJ, Lanctot K, Herrmann N, Dowlatshahi D, Sahlas DJ, Saposnik G, Mandzia JL, Casaubon LK, Hassan A, Perez Y, Swartz RH. Men are at higher risk of screening positive for vascular cognitive impairment compared to women after stroke and transient ischemic attack. J Alzheimers Dis. 2023;94:89–94. 10.3233/JAD-230021. [DOI] [PubMed] [Google Scholar]
  • 25.Nguyen DH, Cunningham JT, Sumien N. Estrogen receptor involvement in vascular cognitive impairment and vascular dementia pathogenesis and treatment. Geroscience. 2021;43:159–66. 10.1007/s11357-020-00263-4. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 26.Gannon OJ, Robison LS, Custozzo AJ, Zuloaga KL. Sex differences in risk factors for vascular contributions to cognitive impairment & dementia. Neurochem Int. 2019;127:38–55. 10.1016/j.neuint.2018.11.014. [DOI] [PubMed] [Google Scholar]
  • 27.Exalto LG, Boomsma JMF, Babapour Mofrad R, Barkhof F, Groeneveld ON, Heinen R, Kuijf HJ, Leeuwis AE, Prins ND, Biessels GJ, Vd Flier WM, group T-Vs. Sex differences in memory clinic patients with possible vascular cognitive impairment. Alzheimers Dement (Amst). 2020;12:e12090. 10.1002/dad2.12090. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 28.Chang YT, Chen YL, Kang HY. Revealing the influences of sex hormones and sex differences in atrial fibrillation and vascular cognitive impairment. Int J Mol Sci. 2021;22:8776. 10.3390/ijms22168776. [DOI] [PMC free article] [PubMed]
  • 29.Cantone M, Fisicaro F, Ferri R, Bella R, Pennisi G, Lanza G, Pennisi M. Sex differences in mild vascular cognitive impairment: a multimodal transcranial magnetic stimulation study. PLoS One. 2023;18:e0282751. 10.1371/journal.pone.0282751. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 30.Tang AS, Rankin KP, Cerono G, Miramontes S, Mills H, Roger J, Zeng B, Nelson C, Soman K, Woldemariam S, Li Y, Lee A, Bove R, Glymour M, Aghaeepour N, Oskotsky TT, Miller Z, Allen IE, Sanders SJ, Baranzini S, Sirota M. Leveraging electronic health records and knowledge networks for Alzheimer’s disease prediction and sex-specific biological insights. Nat Aging. 2024;4:379–95. 10.1038/s43587-024-00573-8. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 31.Reed EG, Keller-Norrell PR. Minding the gap: exploring neuroinflammatory and microglial sex differences in Alzheimer’s disease. Int J Mol Sci. 2023;24:17377. 10.3390/ijms242417377. [DOI] [PMC free article] [PubMed]
  • 32.Lopez-Lee C, Torres ERS, Carling G, Gan L. Mechanisms of sex differences in Alzheimer’s disease. Neuron. 2024;112:1208–21. 10.1016/j.neuron.2024.01.024. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 33.Fernandez A, Cuesta P, Marcos A, Montenegro-Pena M, Yus M, Rodriguez-Rojo IC, Bruna R, Maestu F, Lopez ME. Sex differences in the progression to Alzheimer’s disease: a combination of functional and structural markers. Geroscience. 2024;46:2619–40. 10.1007/s11357-023-01020-z. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 34.Defay R, Pinchinat S, Lumbroso S, Sutan C, Delcourt C, Pola Study G. Sex steroids and age-related macular degeneration in older French women: the POLA study. Ann Epidemiol. 2004;14:202–8. 10.1016/S1047-2797(03)00130-3. [DOI] [PubMed] [Google Scholar]
  • 35.Davezac M, Buscato M, Zahreddine R, Lacolley P, Henrion D, Lenfant F, Arnal JF, Fontaine C. Estrogen receptor and vascular aging. Front Aging. 2021;2:727380. 10.3389/fragi.2021.727380. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 36.Meyer MC, Cummings K, Osol G. Estrogen replacement attenuates resistance artery adrenergic sensitivity via endothelial vasodilators. Am J Physiol. 1997;272:H2264–2270. [DOI] [PubMed] [Google Scholar]
  • 37.McNeill AM, Kim N, Duckles SP, Krause DN, Kontos HA. Chronic estrogen treatment increases levels of endothelial nitric oxide synthase protein in rat cerebral microvessels. Stroke. 1999;30:2186–90. [DOI] [PubMed] [Google Scholar]
  • 38.Kleinert H, Wallerath T, Euchenhofer C, Ihrig-Biedert I, Li H, Forstermann U. Estrogens increase transcription of the human endothelial NO synthase gene: analysis of the transcription factors involved. Hypertension. 1998;31:582–8. [DOI] [PubMed] [Google Scholar]
  • 39.Somani YB, Pawelczyk JA, De Souza MJ, Kris-Etherton PM, Proctor DN. Aging women and their endothelium: probing the relative role of estrogen on vasodilator function. Am J Physiol Heart Circ Physiol. 2019;317:H395–404. 10.1152/ajpheart.00430.2018. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 40.Zahreddine R, Davezac M, Buscato M, Smirnova N, Laffargue M, Henrion D, Adlanmerini M, Lenfant F, Arnal JF, Fontaine C. A historical view of estrogen effect on arterial endothelial healing: from animal models to medical implication. Atherosclerosis. 2021;338:30–8. 10.1016/j.atherosclerosis.2021.10.013. [DOI] [PubMed] [Google Scholar]
  • 41.Boese AC, Kim SC, Yin KJ, Lee JP, Hamblin MH. Sex differences in vascular physiology and pathophysiology: estrogen and androgen signaling in health and disease. Am J Physiol Heart Circ Physiol. 2017;313:H524–45. 10.1152/ajpheart.00217.2016. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 42.Tong W, Lai H, Yang C, Ren S, Dai S, Lai S. Age, gender and metabolic syndrome-related coronary heart disease in U.S. adults. Int J Cardiol. 2005;104:288–91. 10.1016/j.ijcard.2004.10.035. [DOI] [PubMed] [Google Scholar]
  • 43.Stanhewicz AE, Wenner MM, Stachenfeld NS. Sex differences in endothelial function important to vascular health and overall cardiovascular disease risk across the lifespan. Am J Physiol Heart Circ Physiol. 2018;315:H1569–88. 10.1152/ajpheart.00396.2018. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 44.Rexrode K. Sex differences in sex hormones, carotid atherosclerosis, and stroke. Circ Res. 2018;122:17–9. 10.1161/CIRCRESAHA.117.312336. [DOI] [PubMed] [Google Scholar]
  • 45.Mathur P, Ostadal B, Romeo F, Mehta JL. Gender-related differences in atherosclerosis. Cardiovasc Drugs Ther. 2015;29:319–27. 10.1007/s10557-015-6596-3. [DOI] [PubMed] [Google Scholar]
  • 46.Harman SM, Black DM, Naftolin F, Brinton EA, Budoff MJ, Cedars MI, Hopkins PN, Lobo RA, Manson JE, Merriam GR, Miller VM, Neal-Perry G, Santoro N, Taylor HS, Vittinghoff E, Yan M, Hodis HN. Arterial imaging outcomes and cardiovascular risk factors in recently menopausal women: a randomized trial. Ann Intern Med. 2014;161:249–60. 10.7326/M14-0353. [DOI] [PubMed] [Google Scholar]
  • 47.Arnold AP, Cassis LA, Eghbali M, Reue K, Sandberg K. Sex hormones and sex chromosomes cause sex differences in the development of cardiovascular diseases. Arterioscler Thromb Vasc Biol. 2017;37:746–56. 10.1161/ATVBAHA.116.307301. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 48.Yerly A, van der Vorst EPC, Baumgartner I, Bernhard SM, Schindewolf M, Doring Y. Sex-specific and hormone-related differences in vascular remodelling in atherosclerosis. Eur J Clin Invest. 2023;53:e13885. 10.1111/eci.13885. [DOI] [PubMed] [Google Scholar]
  • 49.Nour J, Bonacina F, Norata GD. Gonadal sex vs genetic sex in experimental atherosclerosis. Atherosclerosis. 2023;384:117277. 10.1016/j.atherosclerosis.2023.117277. [DOI] [PubMed] [Google Scholar]
  • 50.Hakamaa E, Goebeler S, Martiskainen M, Louhelainen AM, Ahinko K, Lehtimaki T, Karhunen P. Sex differences in coronary atherosclerosis during the pre- and postmenopausal period: the Tampere Sudden Death Study. Atherosclerosis. 2024;390:117459. 10.1016/j.atherosclerosis.2024.117459. [DOI] [PubMed] [Google Scholar]
  • 51.Cignarella A, Bolego C, Barton M. Sex and sex steroids as determinants of cardiovascular risk. Steroids. 2024;206:109423. 10.1016/j.steroids.2024.109423. [DOI] [PubMed] [Google Scholar]
  • 52.Colafella KMM, Denton KM. Sex-specific differences in hypertension and associated cardiovascular disease. Nat Rev Nephrol. 2018;14:185–201. 10.1038/nrneph.2017.189. [DOI] [PubMed] [Google Scholar]
  • 53.Ogola BO, Zimmerman MA, Clark GL, Abshire CM, Gentry KM, Miller KS, Lindsey SH. New insights into arterial stiffening: does sex matter? Am J Physiol Heart Circ Physiol. 2018;315:H1073–87. 10.1152/ajpheart.00132.2018. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 54.Benjamin EJ, Virani SS, Callaway CW, Chamberlain AM, Chang AR, Cheng S, Chiuve SE, Cushman M, Delling FN, Deo R, de Ferranti SD, Ferguson JF, Fornage M, Gillespie C, Isasi CR, Jimenez MC, Jordan LC, Judd SE, Lackland D, Lichtman JH, Lisabeth L, Liu S, Longenecker CT, Lutsey PL, Mackey JS, Matchar DB, Matsushita K, Mussolino ME, Nasir K, O’Flaherty M, et al. Heart disease and stroke statistics-2018 update: a report from the American Heart Association. Circulation. 2018;137:e67–492. 10.1161/CIR.0000000000000558. [DOI] [PubMed] [Google Scholar]
  • 55.Vaccarino V, Badimon L, Corti R, de Wit C, Dorobantu M, Manfrini O, Koller A, Pries A, Cenko E, Bugiardini R. Presentation, management, and outcomes of ischaemic heart disease in women. Nat Rev Cardiol. 2013;10:508–18. 10.1038/nrcardio.2013.93. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 56.Vaccarino V, Badimon L, Corti R, de Wit C, Dorobantu M, Hall A, Koller A, Marzilli M, Pries A, Bugiardini R, Working Group on Coronary P, Microcirculation. Ischaemic heart disease in women: are there sex differences in pathophysiology and risk factors? Position paper from the working group on coronary pathophysiology and microcirculation of the European Society of Cardiology. Cardiovasc Res. 2011;90:9–17. 10.1093/cvr/cvq394. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 57.Mosca L, Benjamin EJ, Berra K, Bezanson JL, Dolor RJ, Lloyd-Jones DM, Newby LK, Pina IL, Roger VL, Shaw LJ, Zhao D, Beckie TM, Bushnell C, D’Armiento J, Kris-Etherton PM, Fang J, Ganiats TG, Gomes AS, Gracia CR, Haan CK, Jackson EA, Judelson DR, Kelepouris E, Lavie CJ, Moore A, Nussmeier NA, Ofili E, Oparil S, Ouyang P, Pinn VW, et al. Effectiveness-based guidelines for the prevention of cardiovascular disease in women–2011 update: a guideline from the american heart association. Circulation. 2011;123:1243–62. 10.1161/CIR.0b013e31820faaf8. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 58.Siddiqi HK, Vinayagamoorthy M, Gencer B, Ng C, Pester J, Cook NR, Lee IM, Buring J, Manson JE, Albert CM. Sex differences in atrial fibrillation risk: the VITAL rhythm study. JAMA Cardiol. 2022;7:1027–35. 10.1001/jamacardio.2022.2825. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 59.Whipple MO. Sex differences in the clinical presentation and management of peripheral artery disease. J Vasc Nurs. 2023;41:36–7. 10.1016/j.jvn.2023.01.005. [DOI] [PubMed] [Google Scholar]
  • 60.Pabon M, Cheng S, Altin SE, Sethi SS, Nelson MD, Moreau KL, Hamburg N, Hess CN. Sex differences in peripheral artery disease. Circ Res. 2022;130:496–511. 10.1161/CIRCRESAHA.121.320702. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 61.Roumia M, Aronow HD, Soukas P, Gosch K, Smolderen KG, Spertus JA, Abbott JD. Sex differences in disease-specific health status measures in patients with symptomatic peripheral artery disease: Ddata from the PORTRAIT study. Vasc Med. 2017;22:103–9. 10.1177/1358863X16686408. [DOI] [PubMed] [Google Scholar]
  • 62.Talvitie M, Stenman M, Roy J, Leander K, Hultgren R. Sex differences in rupture risk and mortality in untreated patients with intact abdominal aortic aneurysms. J Am Heart Assoc. 2021;10:e019592. 10.1161/JAHA.120.019592. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 63.Grootenboer N, Hunink MG, Hendriks JM, van Sambeek MR, Buth J, collaborators E. Sex differences in 30-day and 5-year outcomes after endovascular repair of abdominal aortic aneurysms in the EUROSTAR study. J Vasc Surg. 2013;58:42–49 e41. 10.1016/j.jvs.2013.01.028. [DOI] [PubMed] [Google Scholar]
  • 64.Cullen JM, Shannon AH, Lu G, Su G, Spinosa MD, Montgomery WG, Ailawadi G, Salmon M, Upchurch GR Jr. Sex-based differences among experimental swine abdominal aortic aneurysms. J Surg Res. 2021;260:488–98. 10.1016/j.jss.2020.11.061. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 65.Boese AC, Chang L, Yin KJ, Chen YE, Lee JP, Hamblin MH. Sex differences in abdominal aortic aneurysms. Am J Physiol Heart Circ Physiol. 2018;314:H1137–52. 10.1152/ajpheart.00519.2017. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 66.Hultgren R. The role of sex hormones in abdominal aortic aneurysms: a topical review. Ann Cardiothorac Surg. 2023;12:536–48. 10.21037/acs-2023-adw-17. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 67.Thorand B, Baumert J, Doring A, Herder C, Kolb H, Rathmann W, Giani G, Koenig W, Group K. Sex differences in the relation of body composition to markers of inflammation. Atherosclerosis. 2006;184:216–24. 10.1016/j.atherosclerosis.2005.04.011. [DOI] [PubMed] [Google Scholar]
  • 68.Mongraw-Chaffin ML, Anderson CA, Allison MA, Ouyang P, Szklo M, Vaidya D, Woodward M, Golden SH. Association between sex hormones and adiposity: qualitative differences in women and men in the multi-ethnic study of atherosclerosis. J Clin Endocrinol Metab. 2015;100:E596–600. 10.1210/jc.2014-2934. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 69.Nicoletti A, Baschi R, Cicero CE, Iacono S, Re VL, Luca A, Schiro G, Monastero R, Gender Neurology Study Group of the Italian Society of N. Sex and gender differences in Alzheimer’s disease, Parkinson’s disease, and amyotrophic lateral sclerosis: a narrative review. Mech Ageing Dev. 2023;212:111821. 10.1016/j.mad.2023.111821. [DOI] [PubMed] [Google Scholar]
  • 70.Fang X, Crumpler RF, Thomas KN, Mazique JN, Roman RJ, Fan F. Contribution of cerebral microvascular mechanisms to age-related cognitive impairment and dementia. Physiol Int. 2022. 10.1556/2060.2022.00020. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 71.Toth P, Tarantini S, Csiszar A, Ungvari Z. Functional vascular contributions to cognitive impairment and dementia: mechanisms and consequences of cerebral autoregulatory dysfunction, endothelial impairment, and neurovascular uncoupling in aging. Am J Physiol Heart Circ Physiol. 2017;312:H1–20. 10.1152/ajpheart.00581.2016. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 72.Iadecola C, Duering M, Hachinski V, Joutel A, Pendlebury ST, Schneider JA, Dichgans M. Vascular cognitive impairment and dementia: JACC Scientific Expert Panel. J Am Coll Cardiol. 2019;73:3326–44. 10.1016/j.jacc.2019.04.034. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 73.Gorelick PB, Scuteri A, Black SE, Decarli C, Greenberg SM, Iadecola C, Launer LJ, Laurent S, Lopez OL, Nyenhuis D, Petersen RC, Schneider JA, Tzourio C, Arnett DK, Bennett DA, Chui HC, Higashida RT, Lindquist R, Nilsson PM, Roman GC, Sellke FW, Seshadri S. Vascular contributions to cognitive impairment and dementia: a statement for healthcare professionals from the American Heart Association/American Stroke Association. Stroke. 2011;42:2672–713. 10.1161/STR.0b013e3182299496. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 74.Zlokovic BV, Gottesman RF, Bernstein KE, Seshadri S, McKee A, Snyder H, Greenberg SM, Yaffe K, Schaffer CB, Yuan C, Hughes TM, Daemen MJ, Williamson JD, Gonzalez HM, Schneider J, Wellington CL, Katusic ZS, Stoeckel L, Koenig JI, Corriveau RA, Fine L, Galis ZS, Reis J, Wright JD, Chen J. Vascular contributions to cognitive impairment and dementia (VCID): a report from the 2018 National Heart, Lung, and Blood Institute and National Institute of Neurological Disorders and Stroke Workshop. Alzheimers Dement. 2020. 10.1002/alz.12157. [DOI] [PubMed] [Google Scholar]
  • 75.Kisler K, Nelson AR, Montagne A, Zlokovic BV. Cerebral blood flow regulation and neurovascular dysfunction in Alzheimer disease. Nat Rev Neurosci. 2017;18:419–34. 10.1038/nrn.2017.48. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 76.Sweeney MD, Zhao Z, Montagne A, Nelson AR, Zlokovic BV. Blood-brain barrier: from physiology to disease and back. Physiol Rev. 2019;99:21–78. 10.1152/physrev.00050.2017. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 77.Sweeney MD, Sagare AP, Zlokovic BV. Blood-brain barrier breakdown in Alzheimer disease and other neurodegenerative disorders. Nat Rev Neurol. 2018;14:133–50. 10.1038/nrneurol.2017.188. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 78.Pinter D, Enzinger C, Fazekas F. Cerebral small vessel disease, cognitive reserve and cognitive dysfunction. J Neurol. 2015;262:2411–9. 10.1007/s00415-015-7776-6. [DOI] [PubMed] [Google Scholar]
  • 79.Ren B, Tan L, Song Y, Li D, Xue B, Lai X, Gao Y. Cerebral small vessel disease: neuroimaging features, biochemical markers, influencing factors, pathological mechanism and treatment. Front Neurol. 2022;13:843953. 10.3389/fneur.2022.843953. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 80.Chen X, Wang J, Shan Y, Cai W, Liu S, Hu M, Liao S, Huang X, Zhang B, Wang Y, Lu Z. Cerebral small vessel disease: neuroimaging markers and clinical implication. J Neurol. 2019;266:2347–62. 10.1007/s00415-018-9077-3. [DOI] [PubMed] [Google Scholar]
  • 81.Chojdak-Lukasiewicz J, Dziadkowiak E, Zimny A, Paradowski B. Cerebral small vessel disease: a review. Adv Clin Exp Med. 2021;30:349–56. 10.17219/acem/131216. [DOI] [PubMed] [Google Scholar]
  • 82.Cannistraro RJ, Badi M, Eidelman BH, Dickson DW, Middlebrooks EH, Meschia JF. CNS small vessel disease: a clinical review. Neurology. 2019;92:1146–56. 10.1212/WNL.0000000000007654. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 83.Ungvari Z, Toth P, Tarantini S, Prodan CI, Sorond F, Merkely B, Csiszar A. Hypertension-induced cognitive impairment: from pathophysiology to public health. Nat Rev Nephrol. 2021;17:639–54. 10.1038/s41581-021-00430-6. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 84.Ungvari Z, Tarantini S, Kirkpatrick AC, Csiszar A, Prodan CI. Cerebral microhemorrhages: mechanisms, consequences, and prevention. Am J Physiol Heart Circ Physiol. 2017;312:H1128–43. 10.1152/ajpheart.00780.2016. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 85.Sweeney MD, Kisler K, Montagne A, Toga AW, Zlokovic BV. The role of brain vasculature in neurodegenerative disorders. Nat Neurosci. 2018;21:1318–31. 10.1038/s41593-018-0234-x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 86.Csiszar A, Ungvari A, Patai R, Gulej R, Yabluchanskiy A, Benyo Z, Kovacs I, Sotonyi P, Kirkpartrick AC, Prodan CI, Liotta EM, Zhang XA, Toth P, Tarantini S, Sorond FA, Ungvari Z. Atherosclerotic burden and cerebral small vessel disease: exploring the link through microvascular aging and cerebral microhemorrhages. Geroscience. 2024. 10.1007/s11357-024-01139-7. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 87.Yoshida Y, Chen Z, Fonseca VA, Mauvais-Jarvis F. Sex differences in cardiovascular risk associated with prediabetes and undiagnosed diabetes. Am J Prev Med. 2023;65:854–62. 10.1016/j.amepre.2023.05.011. [DOI] [PubMed] [Google Scholar]
  • 88.Yoon CW, Rha JH, Park HK, Park SH, Kwon S, Kim BC, Youn YC, Jeong JH, Han HJ, Choi SH. Sex differences in the progression of cerebral microbleeds in patients with concomitant cerebral small vessel disease. Front Neurol. 2022;13:1054624. 10.3389/fneur.2022.1054624. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 89.Jimenez-Sanchez L, Hamilton OKL, Clancy U, Backhouse EV, Stewart CR, Stringer MS, Doubal FN, Wardlaw JM. Sex differences in cerebral small vessel disease: a systematic review and meta-analysis. Front Neurol. 2021;12:756887. 10.3389/fneur.2021.756887. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 90.Lohner V, Pehlivan G, Sanroma G, Miloschewski A, Schirmer MD, Stocker T, Reuter M, Breteler MMB. Relation between sex, menopause, and white matter hyperintensities: the Rhineland study. Neurology. 2022;99:e935–43. 10.1212/WNL.0000000000200782. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 91.Pavlovic A, Pekmezovic T, Mijajlovic M, Tomic G, Zidverc TJ. Is the female sex associated with an increased risk for long-term cognitive decline after the first-ever lacunar stroke? Prospective study on small vessel disease cohort. Front Neurol. 2022;13:1052401. 10.3389/fneur.2022.1052401. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 92.Fleckenstein M, Schmitz-Valckenberg S, Chakravarthy U. Age-related macular degeneration: a review. JAMA. 2024;331:147–57. 10.1001/jama.2023.26074. [DOI] [PubMed] [Google Scholar]
  • 93.Buonfiglio F, Korb CA, Stoffelns B, Pfeiffer N, Gericke A. Recent advances in our understanding of age-related macular degeneration: mitochondrial dysfunction, redox signaling, and the complement system. Aging Dis. 2024. 10.14336/AD.2024.0124. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 94.Mullins RF, Khanna A, Schoo DP, Tucker BA, Sohn EH, Drack AV, Stone EM. Is age-related macular degeneration a microvascular disease? Adv Exp Med Biol. 2014;801:283–9. 10.1007/978-1-4614-3209-8_36. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 95.Marin AI, Poppelaars F, Wagner BD, Palestine AG, Patnaik JL, Holers VM, Frazer-Abel AA, Mathias MT, Manoharan N, Fonteh CN, Mandava N, Lynch AM, University of Colorado Retina Research G. Sex and age-related differences in complement factors among patients with intermediate age-related macular degeneration. Transl Vis Sci Technol. 2022;11:22. 10.1167/tvst.11.5.22. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 96.Joslin CE, Vajaranant TS, Ulanski LJ 2nd. Age-related macular degeneration and mortality in the Age-Related Eye Disease Study (AREDS): the effect of sex and time. Ophthalmol Retina. 2017;1:49–58. 10.1016/j.oret.2016.09.004. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 97.Brandl C, Breinlich V, Stark KJ, Enzinger S, Assenmacher M, Olden M, Grassmann F, Graw J, Heier M, Peters A, Helbig H, Kuchenhoff H, Weber BH, Heid IM. Features of age-related macular degeneration in the general adults and their dependency on age, sex, and smoking: results from the German KORA study. PLoS One. 2016;11:e0167181. 10.1371/journal.pone.0167181. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 98.Toto L, Borrelli E, Mastropasqua R, Di Antonio L, Doronzo E, Carpineto P, Mastropasqua L. Association between outer retinal alterations and microvascular changes in intermediate stage age-related macular degeneration: an optical coherence tomography angiography study. Br J Ophthalmol. 2017;101:774–9. 10.1136/bjophthalmol-2016-309160. [DOI] [PubMed] [Google Scholar]
  • 99.Lipecz A, Miller L, Kovacs I, Czako C, Csipo T, Baffi J, Csiszar A, Tarantini S, Ungvari Z, Yabluchanskiy A, Conley S. Microvascular contributions to age-related macular degeneration (AMD): from mechanisms of choriocapillaris aging to novel interventions. Geroscience. 2019;41:813–45. 10.1007/s11357-019-00138-3. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 100.Tisi A, Feligioni M, Passacantando M, Ciancaglini M, Maccarone R. The impact of oxidative stress on blood-retinal barrier physiology in age-related macular degeneration. Cells. 2021;10:64. 10.3390/cells10010064. [DOI] [PMC free article] [PubMed]
  • 101.O’Leary F, Campbell M. The blood-retina barrier in health and disease. FEBS J. 2023;290:878–91. 10.1111/febs.16330. [DOI] [PubMed] [Google Scholar]
  • 102.Huang A, Sun D, Koller A, Kaley G. 17beta-estradiol restores endothelial nitric oxide release to shear stress in arterioles of male hypertensive rats. Circulation. 2000;101:94–100. [DOI] [PubMed] [Google Scholar]
  • 103.Haynes MP, Sinha D, Russell KS, Collinge M, Fulton D, Morales-Ruiz M, Sessa WC, Bender JR. Membrane estrogen receptor engagement activates endothelial nitric oxide synthase via the PI3-kinase-Akt pathway in human endothelial cells. Circ Res. 2000;87:677–82. [DOI] [PubMed] [Google Scholar]
  • 104.Sasaki Y, Ikeda Y, Miyauchi T, Uchikado Y, Akasaki Y, Ohishi M. Estrogen-SIRT1 axis plays a pivotal role in protecting arteries against menopause-induced senescence and atherosclerosis. J Atheroscler Thromb. 2020;27:47–59. 10.5551/jat.47993. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 105.Nappi RE, Chedraui P, Lambrinoudaki I, Simoncini T. Menopause: a cardiometabolic transition. Lancet Diabetes Endocrinol. 2022;10:442–56. 10.1016/S2213-8587(22)00076-6. [DOI] [PubMed] [Google Scholar]
  • 106.Barnabas O, Wang H, Gao XM. Role of estrogen in angiogenesis in cardiovascular diseases. J Geriatr Cardiol. 2013;10:377–82. 10.3969/j.issn.1671-5411.2013.04.008. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 107.Yang YM, Sun D, Kandhi S, Froogh G, Zhuge J, Huang W, Hammock BD, Huang A. Estrogen-dependent epigenetic regulation of soluble epoxide hydrolase via DNA methylation. Proc Natl Acad Sci U S A. 2018;115:613–8. 10.1073/pnas.1716016115. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 108.Perez-Cremades D, Mompeon A, Vidal-Gomez X, Hermenegildo C, Novella S. miRNA as a new regulatory mechanism of estrogen vascular action. Int J Mol Sci. 2018;19:473. 10.3390/ijms19020473. [DOI] [PMC free article] [PubMed]
  • 109.Ungvari Z, Tarantini S, Kiss T, Wren JD, Giles CB, Griffin CT, Murfee WL, Pacher P, Csiszar A. Endothelial dysfunction and angiogenesis impairment in the ageing vasculature. Nat Rev Cardiol. 2018;15:555–65. 10.1038/s41569-018-0030-z. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 110.Dai DF, Rabinovitch PS, Ungvari Z. Mitochondria and cardiovascular aging. Circ Res. 2012;110:1109–24. 10.1161/CIRCRESAHA.111.246140. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 111.Ungvari Z, Kaley G, de Cabo R, Sonntag WE, Csiszar A. Mechanisms of vascular aging: new perspectives. J Gerontol A Biol Sci Med Sci. 2010;65:1028–41. 10.1093/gerona/glq113. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 112.Ungvari Z, Parrado-Fernandez C, Csiszar A, de Cabo R. Mechanisms underlying caloric restriction and lifespan regulation: implications for vascular aging. Circ Res. 2008;102:519–28. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 113.Csiszar A, Wang M, Lakatta EG, Ungvari ZI. Inflammation and endothelial dysfunction during aging: role of NF-{kappa}B. J Appl Physiol. 2008;105:1333–41. [DOI] [PMC free article] [PubMed]
  • 114.Pacher P, Beckman JS, Liaudet L. Nitric oxide and peroxynitrite in health and disease. Physiol Rev. 2007;87:315–424. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 115.Gulej R, Nyul-Toth A, Ahire C, DelFavero J, Balasubramanian P, Kiss T, Tarantini S, Benyo Z, Pacher P, Csik B, Yabluchanskiy A, Mukli P, Kuan-Celarier A, Krizbai IA, Campisi J, Sonntag WE, Csiszar A, Ungvari Z. Elimination of senescent cells by treatment with Navitoclax/ABT263 reverses whole brain irradiation-induced blood-brain barrier disruption in the mouse brain. Geroscience. 2023;45:2983–3002. 10.1007/s11357-023-00870-x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 116.Kiss T, Nyul-Toth A, DelFavero J, Balasubramanian P, Tarantini S, Faakye J, Gulej R, Ahire C, Ungvari A, Yabluchanskiy A, Wiley G, Garman L, Ungvari Z, Csiszar A. Spatial transcriptomic analysis reveals inflammatory foci defined by senescent cells in the white matter, hippocampi and cortical grey matter in the aged mouse brain. Geroscience. 2022;44:661–81. 10.1007/s11357-022-00521-7. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 117.Ungvari Z, Tarantini S, Nyul-Toth A, Kiss T, Yabluchanskiy A, Csipo T, Balasubramanian P, Lipecz A, Benyo Z, Csiszar A. Nrf2 dysfunction and impaired cellular resilience to oxidative stressors in the aged vasculature: from increased cellular senescence to the pathogenesis of age-related vascular diseases. Geroscience. 2019;41:727–38. 10.1007/s11357-019-00107-w. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 118.Csiszar A, Wang M, Lakatta EG, Ungvari ZI. Inflammation and endothelial dysfunction during aging: role of NF-{kappa}B. J Appl Physiol. 2008;105:1333–41. 10.1152/japplphysiol.90470.2008. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 119.Balasubramanian P, Kiss T, Gulej R, Nyul Toth A, Tarantini S, Yabluchanskiy A, Ungvari Z, Csiszar A. Accelerated aging induced by an unhealthy high-fat diet: initial evidence for the role of Nrf2 deficiency and impaired stress resilience in cellular senescence. Nutrients. 2024;16:952. 10.3390/nu16070952. [DOI] [PMC free article] [PubMed]
  • 120.Tarantini S, Valcarcel-Ares MN, Yabluchanskiy A, Tucsek Z, Hertelendy P, Kiss T, Gautam T, Zhang XA, Sonntag WE, de Cabo R, Farkas E, Elliott ME, Kinter MT, Deak F, Ungvari Z, Csiszar A. Nrf2 deficiency exacerbates obesity-induced oxidative stress, neurovascular dysfunction, blood brain barrier disruption, neuroinflammation, amyloidogenic gene expression and cognitive decline in mice, mimicking the aging phenotype. J Gerontol A Biol Sci Med Sci. 2018;73:853–63. [DOI] [PMC free article] [PubMed]
  • 121.Fulop GA, Kiss T, Tarantini S, Balasubramanian P, Yabluchanskiy A, Farkas E, Bari F, Ungvari Z, Csiszar A. Nrf2 deficiency in aged mice exacerbates cellular senescence promoting cerebrovascular inflammation. Geroscience. 2018;40:513–21. 10.1007/s11357-018-0047-6. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 122.Csiszar A, Gautam T, Sosnowska D, Tarantini S, Banki E, Tucsek Z, Toth P, Losonczy G, Koller A, Reglodi D, Giles CB, Wren JD, Sonntag WE, Ungvari Z. Caloric restriction confers persistent anti-oxidative, pro-angiogenic, and anti-inflammatory effects and promotes anti-aging miRNA expression profile in cerebromicrovascular endothelial cells of aged rats. Am J Physiol Heart Circ Physiol. 2014;307:H292–306. 10.1152/ajpheart.00307.2014. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 123.Valcarcel-Ares MN, Gautam T, Warrington JP, Bailey-Downs L, Sosnowska D, de Cabo R, Losonczy G, Sonntag WE, Ungvari Z, Csiszar A. Disruption of Nrf2 signaling impairs angiogenic capacity of endothelial cells: implications for microvascular aging. J Gerontol A Biol Sci Med Sci. 2012;67:821–9. 10.1093/gerona/glr229. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 124.Csiszar A, Sosnowska D, Wang M, Lakatta EG, Sonntag WE, Ungvari Z. Age-associated proinflammatory secretory phenotype in vascular smooth muscle cells from the non-human primate Macaca mulatta: reversal by resveratrol treatment. J Gerontol A Biol Sci Med Sci. 2012;67:811–20. 10.1093/gerona/glr228. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 125.Ungvari Z, Bailey-Downs L, Sosnowska D, Gautam T, Koncz P, Losonczy G, Ballabh P, de Cabo R, Sonntag WE, Csiszar A. Vascular oxidative stress in aging: a homeostatic failure due to dysregulation of Nrf2-mediated antioxidant response. Am J Physiol Heart Circ Physiol. 2011;301:H363–372. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 126.Csiszar A, Pinto JT, Gautam T, Kleusch C, Hoffmann B, Tucsek Z, Toth P, Sonntag WE, Ungvari Z. Resveratrol encapsulated in novel fusogenic liposomes activates Nrf2 and Attenuates oxidative stress in cerebromicrovascular endothelial cells from aged rats. J Gerontol A Biol Sci Med Sci. 2014. 10.1093/gerona/glu029. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 127.Ungvari Z, Bagi Z, Feher A, Recchia FA, Sonntag WE, Pearson K, de Cabo R, Csiszar A. Resveratrol confers endothelial protection via activation of the antioxidant transcription factor Nrf2. Am J Physiol Heart Circ Physiol. 2010;299:H18–24. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 128.Roman RJ, Lombard JH. Does 20-hydroxyeicosatetraenoic acid contribute to sex differences in cardiovascular risk by increasing oxidative stress? Hypertension. 2007;50:37–8. 10.1161/HYPERTENSIONAHA.107.090803. [DOI] [PubMed] [Google Scholar]
  • 129.Norton CE, Shaw RL, Safa, Dockery B, Domeier TL, Segal SS. Advanced age and female sex protect cerebral arteries from mitochondrial depolarization and apoptosis during acute oxidative stress. Aging Cell. 2024;23:e14110. 10.1111/acel.14110. [DOI] [PMC free article] [PubMed]
  • 130.Norton CE, Jacobsen NL, Sinkler SY, Manrique-Acevedo C, Segal SS. Female sex and Western-style diet protect mouse resistance arteries during acute oxidative stress. Am J Physiol Cell Physiol. 2020;318:C627–39. 10.1152/ajpcell.00342.2019. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 131.Morales RC, Bahnson ES, Havelka GE, Cantu-Medellin N, Kelley EE, Kibbe MR. Sex-based differential regulation of oxidative stress in the vasculature by nitric oxide. Redox Biol. 2015;4:226–33. 10.1016/j.redox.2015.01.007. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 132.Wassmann K, Wassmann S, Nickenig G. Progesterone antagonizes the vasoprotective effect of estrogen on antioxidant enzyme expression and function. Circ Res. 2005;97:1046–54. 10.1161/01.RES.0000188212.57180.55. [DOI] [PubMed] [Google Scholar]
  • 133.Strehlow K, Rotter S, Wassmann S, Adam O, Grohe C, Laufs K, Bohm M, Nickenig G. Modulation of antioxidant enzyme expression and function by estrogen. Circ Res. 2003;93:170–7. 10.1161/01.RES.0000082334.17947.11. [DOI] [PubMed] [Google Scholar]
  • 134.Wing LY, Chen YC, Shih YY, Cheng JC, Lin YJ, Jiang MJ. Effects of oral estrogen on aortic ROS-generating and -scavenging enzymes and atherosclerosis in apoE-deficient mice. Exp Biol Med (Maywood). 2009;234:1037–46. 10.3181/0811-RM-332. [DOI] [PubMed] [Google Scholar]
  • 135.Hashimoto M, Akishita M, Eto M, Ishikawa M, Kozaki K, Toba K, Sagara Y, Taketani Y, Orimo H, Ouchi Y. Modulation of endothelium-dependent flow-mediated dilatation of the brachial artery by sex and menstrual cycle. Circulation. 1995;92:3431–5. 10.1161/01.cir.92.12.3431. [DOI] [PubMed] [Google Scholar]
  • 136.O’Brien MW, Johns JA, Petterson JL, Mekary S, Kimmerly DS. The impact of age and sex on popliteal artery endothelial-dependent vasodilator and vasoconstrictor function. Exp Gerontol. 2021;145:111221. 10.1016/j.exger.2020.111221. [DOI] [PubMed] [Google Scholar]
  • 137.Moreau KL, Hildreth KL, Klawitter J, Blatchford P, Kohrt WM. Decline in endothelial function across the menopause transition in healthy women is related to decreased estradiol and increased oxidative stress. Geroscience. 2020. 10.1007/s11357-020-00236-7. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 138.Hayashi T, Matsui-Hirai H, Miyazaki-Akita A, Fukatsu A, Funami J, Ding QF, Kamalanathan S, Hattori Y, Ignarro LJ, Iguchi A. Endothelial cellular senescence is inhibited by nitric oxide: implications in atherosclerosis associated with menopause and diabetes. Proc Natl Acad Sci USA. 2006;103:17018–23. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 139.Nair AR, Pillai AJ, Nair N. Cardiovascular Changes in Menopause. Curr Cardiol Rev. 2021;17:e230421187681. 10.2174/1573403X16666201106141811. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 140.Koukoulis GN, Filiponi M, Gougoura S, Befani C, Liakos P, Bargiota A. Testosterone and dihydrotestosterone modulate the redox homeostasis of endothelium. Cell Biol Int. 2022;46:660–70. 10.1002/cbin.11768. [DOI] [PubMed] [Google Scholar]
  • 141.Decker KP, Feliciano PG, Kimmel MT, Hogwood AC, Weggen JB, Darling AM, Richardson JW, Garten RS. Examining sex differences in sitting-induced microvascular dysfunction: insight from acute vitamin C supplementation. Microvasc Res. 2021;135:104147. 10.1016/j.mvr.2021.104147. [DOI] [PubMed] [Google Scholar]
  • 142.Dai DF, Chiao YA, Marcinek DJ, Szeto HH, Rabinovitch PS. Mitochondrial oxidative stress in aging and healthspan. Longev Healthspan. 2014;3:6. 10.1186/2046-2395-3-6. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 143.Dai DF, Santana LF, Vermulst M, Tomazela DM, Emond MJ, MacCoss MJ, Gollahon K, Martin GM, Loeb LA, Ladiges WC, Rabinovitch PS. Overexpression of catalase targeted to mitochondria attenuates murine cardiac aging. Circulation. 2009;119:2789–97. 10.1161/CIRCULATIONAHA.108.822403. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 144.Kiss T, Nyul-Toth A, Balasubramanian P, Tarantini S, Ahire C, Yabluchanskiy A, Csipo T, Farkas E, Wren JD, Garman L, Csiszar A, Ungvari Z. Nicotinamide mononucleotide (NMN) supplementation promotes neurovascular rejuvenation in aged mice: transcriptional footprint of SIRT1 activation, mitochondrial protection, anti-inflammatory, and anti-apoptotic effects. Geroscience. 2020. 10.1007/s11357-020-00165-5. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 145.Kiss T, Tarantini S, Csipo T, Balasubramanian P, Nyul-Toth A, Yabluchanskiy A, Wren JD, Garman L, Huffman DM, Csiszar A, Ungvari Z. Circulating anti-geronic factors from heterochonic parabionts promote vascular rejuvenation in aged mice: transcriptional footprint of mitochondrial protection, attenuation of oxidative stress, and rescue of endothelial function by young blood. Geroscience. 2020;42:727–48. 10.1007/s11357-020-00180-6. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 146.Springo Z, Tarantini S, Toth P, Tucsek Z, Koller A, Sonntag WE, Csiszar A, Ungvari Z. Aging exacerbates pressure-induced mitochondrial oxidative stress in mouse cerebral arteries. J Gerontol A Biol Sci Med Sci. 2015. 10.1093/gerona/glu244. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 147.Tarantini S, Valcarcel-Ares MN, Toth P, Yabluchanskiy A, Tucsek Z, Kiss T, Hertelendy P, Kinter M, Ballabh P, Sule Z, Farkas E, Baur JA, Sinclair DA, Csiszar A, Ungvari Z. Nicotinamide mononucleotide (NMN) supplementation rescues cerebromicrovascular endothelial function and neurovascular coupling responses and improves cognitive function in aged mice. Redox Biol. 2019;24:101192. 10.1016/j.redox.2019.101192. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 148.Tarantini S, Valcarcel-Ares NM, Yabluchanskiy A, Fulop GA, Hertelendy P, Gautam T, Farkas E, Perz A, Rabinovitch PS, Sonntag WE, Csiszar A, Ungvari Z. Treatment with the mitochondrial-targeted antioxidant peptide SS-31 rescues neurovascular coupling responses and cerebrovascular endothelial function and improves cognition in aged mice. Aging Cell. 2018;17:e12731. 10.1111/acel.12731. [DOI] [PMC free article] [PubMed]
  • 149.Ungvari Z, Orosz Z, Labinskyy N, Rivera A, Xiangmin Z, Smith K, Csiszar A. Increased mitochondrial H2O2 production promotes endothelial NF-kappaB activation in aged rat arteries. Am J Physiol Heart Circ Physiol. 2007;293:H37–47. [DOI] [PubMed] [Google Scholar]
  • 150.Ungvari Z, Sonntag WE, Csiszar A. Mitochondria and aging in the vascular system. J Mol Med (Berl). 2010;88:1021–7. 10.1007/s00109-010-0667-5. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 151.Ungvari ZI, Labinskyy N, Gupte SA, Chander PN, Edwards JG, Csiszar A. Dysregulation of mitochondrial biogenesis in vascular endothelial and smooth muscle cells of aged rats. Am J Physiol Heart Circ Physiol. 2008;294:H2121–2128. [DOI] [PubMed] [Google Scholar]
  • 152.Golob MJ, Tian L, Wang Z, Zimmerman TA, Caneba CA, Hacker TA, Song G, Chesler NC. Mitochondria DNA mutations cause sex-dependent development of hypertension and alterations in cardiovascular function. J Biomech. 2015;48:405–12. 10.1016/j.jbiomech.2014.12.044. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 153.Sarver DC, Saqib M, Chen F, Wong GW. Mitochondrial respiration atlas reveals differential changes in mitochondrial function across sex and age. bioRxiv. 2024. 10.1101/2024.03.26.586781. [DOI] [PMC free article] [PubMed]
  • 154.Kane MS, Benavides GA, Osuma E, Johnson MS, Collins HE, He Y, Westbrook D, Litovsky SH, Mitra K, Chatham JC, Darley-Usmar V, Young ME, Zhang J. The interplay between sex, time of day, fasting status, and their impact on cardiac mitochondrial structure, function, and dynamics. Sci Rep. 2023;13:21638. 10.1038/s41598-023-49018-z. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 155.Junker A, Wang J, Gouspillou G, Ehinger JK, Elmer E, Sjovall F, Fisher-Wellman KH, Neufer PD, Molina AJA, Ferrucci L, Picard M. Human studies of mitochondrial biology demonstrate an overall lack of binary sex differences: a multivariate meta-analysis. FASEB J. 2022;36:e22146. 10.1096/fj.202101628R. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 156.Goedeke L, Murt KN, Di Francesco A, Camporez JP, Nasiri AR, Wang Y, Zhang XM, Cline GW, de Cabo R, Shulman GI. Sex- and strain-specific effects of mitochondrial uncoupling on age-related metabolic diseases in high-fat diet-fed mice. Aging Cell. 2022;21:e13539. 10.1111/acel.13539. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 157.Hsieh YC, Yu HP, Suzuki T, Choudhry MA, Schwacha MG, Bland KI, Chaudry IH. Upregulation of mitochondrial respiratory complex IV by estrogen receptor-beta is critical for inhibiting mitochondrial apoptotic signaling and restoring cardiac functions following trauma-hemorrhage. J Mol Cell Cardiol. 2006;41:511–21. [DOI] [PubMed] [Google Scholar]
  • 158.Razmara A, Sunday L, Stirone C, Wang XB, Krause DN, Duckles SP, Procaccio V. Mitochondrial effects of estrogen are mediated by estrogen receptor alpha in brain endothelial cells. J Pharmacol Exp Ther. 2008;325:782–90. 10.1124/jpet.107.134072. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 159.O’Lone R, Knorr K, Jaffe IZ, Schaffer ME, Martini PG, Karas RH, Bienkowska J, Mendelsohn ME, Hansen U. Estrogen receptors alpha and beta mediate distinct pathways of vascular gene expression, including genes involved in mitochondrial electron transport and generation of reactive oxygen species. Mol Endocrinol. 2007;21:1281–96. 10.1210/me.2006-0497. [DOI] [PubMed] [Google Scholar]
  • 160.Guo J, Krause DN, Horne J, Weiss JH, Li X, Duckles SP. Estrogen-receptor-mediated protection of cerebral endothelial cell viability and mitochondrial function after ischemic insult in vitro. J Cereb Blood Flow Metab. 2010;30:545–54. 10.1038/jcbfm.2009.226. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 161.Sasaki Y, Ikeda Y, Uchikado Y, Akasaki Y, Sadoshima J, Ohishi M. Estrogen plays a crucial role in Rab9-dependent mitochondrial autophagy, delaying arterial senescence. J Am Heart Assoc. 2021;10:e019310. 10.1161/JAHA.120.019310. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 162.Schreiber SN, Emter R, Hock MB, Knutti D, Cardenas J, Podvinec M, Oakeley EJ, Kralli A. The estrogen-related receptor alpha (ERRalpha) functions in PPARgamma coactivator 1alpha (PGC-1alpha)-induced mitochondrial biogenesis. Proc Natl Acad Sci U S A. 2004;101:6472–7. 10.1073/pnas.0308686101. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 163.Rangwala SM, Li X, Lindsley L, Wang X, Shaughnessy S, Daniels TG, Szustakowski J, Nirmala NR, Wu Z, Stevenson SC. Estrogen-related receptor alpha is essential for the expression of antioxidant protection genes and mitochondrial function. Biochem Biophys Res Commun. 2007;357:231–6. 10.1016/j.bbrc.2007.03.126. [DOI] [PubMed] [Google Scholar]
  • 164.Irwin RW, Yao J, Hamilton RT, Cadenas E, Brinton RD, Nilsen J. Progesterone and estrogen regulate oxidative metabolism in brain mitochondria. Endocrinology. 2008;149:3167–75. 10.1210/en.2007-1227. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 165.Hsieh YC, Yu HP, Suzuki T, Choudhry MA, Schwacha MG, Bland KI, Chaudry IH. Upregulation of mitochondrial respiratory complex IV by estrogen receptor-beta is critical for inhibiting mitochondrial apoptotic signaling and restoring cardiac functions following trauma-hemorrhage. J Mol Cell Cardiol. 2006;41:511–21. 10.1016/j.yjmcc.2006.06.001. [DOI] [PubMed] [Google Scholar]
  • 166.Chen JQ, Yager JD, Russo J. Regulation of mitochondrial respiratory chain structure and function by estrogens/estrogen receptors and potential physiological/pathophysiological implications. Biochim Biophys Acta. 2005;1746:1–17. 10.1016/j.bbamcr.2005.08.001. [DOI] [PubMed] [Google Scholar]
  • 167.Lynch S, Boyett JE, Smith MR, Giordano-Mooga S. Sex hormone regulation of proteins modulating mitochondrial metabolism, dynamics and inter-organellar cross talk in cardiovascular disease. Front Cell Dev Biol. 2020;8:610516. 10.3389/fcell.2020.610516. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 168.Marais GAB, Gaillard JM, Vieira C, Plotton I, Sanlaville D, Gueyffier F, Lemaitre JF. Sex gap in aging and longevity: can sex chromosomes play a role? Biol Sex Differ. 2018;9:33. 10.1186/s13293-018-0181-y. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 169.Austad SN, Fischer KE. Sex differences in lifespan. Cell Metab. 2016;23:1022–33. 10.1016/j.cmet.2016.05.019. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 170.Ventura-Clapier R, Moulin M, Piquereau J, Lemaire C, Mericskay M, Veksler V, Garnier A. Mitochondria: a central target for sex differences in pathologies. Clin Sci (Lond). 2017;131:803–22. 10.1042/CS20160485. [DOI] [PubMed] [Google Scholar]
  • 171.Sommer AJ, Okonkwo J, Monteiro J, Bind MC. A permutation-based approach using a rank-based statistic to identify sex differences in epigenetics. Sci Rep. 2023;13:14838. 10.1038/s41598-023-41360-6. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 172.Shen EY, Ahern TH, Cheung I, Straubhaar J, Dincer A, Houston I, de Vries GJ, Akbarian S, Forger NG. Epigenetics and sex differences in the brain: a genome-wide comparison of histone-3 lysine-4 trimethylation (H3K4me3) in male and female mice. Exp Neurol. 2015;268:21–9. 10.1016/j.expneurol.2014.08.006. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 173.Hartman RJG, Huisman SE, den Ruijter HM. Sex differences in cardiovascular epigenetics-a systematic review. Biol Sex Differ. 2018;9:19. 10.1186/s13293-018-0180-z. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 174.Kleis-Olsen AS, Farlov JE, Petersen EA, Schmucker M, Flensted-Jensen M, Blom I, Ingersen A, Hansen M, Helge JW, Dela F, Larsen S. Metabolic flexibility in postmenopausal women: hormone replacement therapy is associated with higher mitochondrial content, respiratory capacity, and lower total fat mass. Acta Physiol (Oxf). 2024;240:e14117. 10.1111/apha.14117. [DOI] [PubMed] [Google Scholar]
  • 175.Rebelo-Marques A, De Sousa LA, Andrade R, Ribeiro CF, Mota-Pinto A, Carrilho F, Espregueira-Mendes J. Aging hallmarks: the benefits of physical exercise. Front Endocrinol (Lausanne). 2018;9:258. 10.3389/fendo.2018.00258. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 176.Terao R, Ahmed T, Suzumura A, Terasaki H. Oxidative stress-induced cellular senescence in aging retina and age-related macular degeneration. Antioxidants (Basel). 2022;11:2189. 10.3390/antiox11112189. [DOI] [PMC free article] [PubMed]
  • 177.Wang JC, Bennett M. Aging and atherosclerosis: mechanisms, functional consequences, and potential therapeutics for cellular senescence. Circ Res. 2012;111:245–59. 10.1161/CIRCRESAHA.111.261388. [DOI] [PubMed] [Google Scholar]
  • 178.Tuttle CSL, Waaijer MEC, Slee-Valentijn MS, Stijnen T, Westendorp R, Maier AB. Cellular senescence and chronological age in various human tissues: a systematic review and meta-analysis. Aging Cell. 2020;19:e13083. 10.1111/acel.13083. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 179.Tchkonia T, Zhu Y, van Deursen J, Campisi J, Kirkland JL. Cellular senescence and the senescent secretory phenotype: therapeutic opportunities. J Clin Invest. 2013;123:966–72. 10.1172/JCI64098. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 180.Regina C, Panatta E, Candi E, Melino G, Amelio I, Balistreri CR, Annicchiarico-Petruzzelli M, Di Daniele N, Ruvolo G. Vascular ageing and endothelial cell senescence: molecular mechanisms of physiology and diseases. Mech Ageing Dev. 2016;159:14–21. 10.1016/j.mad.2016.05.003. [DOI] [PubMed] [Google Scholar]
  • 181.Minamino T, Komuro I. Vascular cell senescence: contribution to atherosclerosis. Circ Res. 2007;100:15–26. 10.1161/01.RES.0000256837.40544.4a. [DOI] [PubMed] [Google Scholar]
  • 182.Childs BG, Durik M, Baker DJ, van Deursen JM. Cellular senescence in aging and age-related disease: from mechanisms to therapy. Nat Med. 2015;21:1424–35. 10.1038/nm.4000. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 183.Chaib S, Tchkonia T, Kirkland JL. Cellular senescence and senolytics: the path to the clinic. Nat Med. 2022;28:1556–68. 10.1038/s41591-022-01923-y. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 184.Clayton ZS, Rossman MJ, Mahoney SA, Venkatasubramanian R, Maurer GS, Hutton DA, VanDongen NS, Greenberg NT, Longtine AG, Ludwig KR, Brunt VE, LaRocca TJ, Campisi J, Melov S, Seals DR. Cellular senescence contributes to large elastic artery stiffening and endothelial dysfunction with aging: amelioration with senolytic treatment. Hypertension. 2023;80:2072–87. 10.1161/HYPERTENSIONAHA.123.21392. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 185.Voghel G, Thorin-Trescases N, Farhat N, Nguyen A, Villeneuve L, Mamarbachi AM, Fortier A, Perrault LP, Carrier M, Thorin E. Cellular senescence in endothelial cells from atherosclerotic patients is accelerated by oxidative stress associated with cardiovascular risk factors. Mech Ageing Dev. 2007;128:662–71. 10.1016/j.mad.2007.09.006. [DOI] [PubMed] [Google Scholar]
  • 186.Roos CM, Zhang B, Palmer AK, Ogrodnik MB, Pirtskhalava T, Thalji NM, Hagler M, Jurk D, Smith LA, Casaclang-Verzosa G, Zhu Y, Schafer MJ, Tchkonia T, Kirkland JL, Miller JD. Chronic senolytic treatment alleviates established vasomotor dysfunction in aged or atherosclerotic mice. Aging Cell. 2016;15:973–7. 10.1111/acel.12458. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 187.Gulej R, Csik B, Faakye J, Tarantini S, Shanmugarama S, Chandragiri SS, Mukli P, Conley S, Csiszar A, Ungvari Z, Yabluchanskiy A, Nyul-Toth A. Endothelial deficiency of insulin-like growth factor-1 receptor leads to blood-brain barrier disruption and accelerated endothelial senescence in mice, mimicking aspects of the brain aging phenotype. Microcirculation. 2024;31:e12840. 10.1111/micc.12840. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 188.Faakye J, Nyul-Toth A, Muranyi M, Gulej R, Csik B, Shanmugarama S, Tarantini S, Negri S, Prodan C, Mukli P, Yabluchanskiy A, Conley S, Toth P, Csiszar A, Ungvari Z. Preventing spontaneous cerebral microhemorrhages in aging mice: a novel approach targeting cellular senescence with ABT263/navitoclax. Geroscience. 2023. 10.1007/s11357-023-01024-9. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 189.Kiss T, Nyul-Toth A, Balasubramanian P, Tarantini S, Ahire C, DelFavero J, Yabluchanskiy A, Csipo T, Farkas E, Wiley G, Garman L, Csiszar A, Ungvari Z. Single-cell RNA sequencing identifies senescent cerebromicrovascular endothelial cells in the aged mouse brain. Geroscience. 2020;42:429–44. 10.1007/s11357-020-00177-1. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 190.Tarantini S, Balasubramanian P, Delfavero J, Csipo T, Yabluchanskiy A, Kiss T, Nyul-Toth A, Mukli P, Toth P, Ahire C, Ungvari A, Benyo Z, Csiszar A, Ungvari Z. Treatment with the BCL-2/BCL-xL inhibitor senolytic drug ABT263/Navitoclax improves functional hyperemia in aged mice. Geroscience. 2021;43:2427–40. 10.1007/s11357-021-00440-z. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 191.Kiss T, Ungvari A, Gulej R, Nyul-Toth A, Tarantini S, Benyo Z, Csik B, Yabluchanskiy A, Mukli P, Csiszar A, Ungvari Z. Whole brain irradiation-induced endothelial dysfunction in the mouse brain. Geroscience. 2024;46:531–41. 10.1007/s11357-023-00990-4. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 192.Yabluchanskiy A, Tarantini S, Balasubramanian P, Kiss T, Csipo T, Fulop GA, Lipecz A, Ahire C, DelFavero J, Nyul-Toth A, Sonntag WE, Schwartzman ML, Campisi J, Csiszar A, Ungvari Z. Pharmacological or genetic depletion of senescent astrocytes prevents whole brain irradiation-induced impairment of neurovascular coupling responses protecting cognitive function in mice. Geroscience. 2020;42:409–28. 10.1007/s11357-020-00154-8. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 193.Xiang X, Xie L, Lin J, Pare R, Huang G, Huang J, Wang Y, Song S, Ruan Y. Estrogen receptor alpha mediates 17beta-estradiol, up-regulates autophagy and alleviates hydrogen peroxide-induced vascular senescence. Biogerontology. 2023;24:783–99. 10.1007/s10522-023-10015-4. [DOI] [PubMed] [Google Scholar]
  • 194.Imanishi T, Hano T, Nishio I. Estrogen reduces angiotensin II-induced acceleration of senescence in endothelial progenitor cells. Hypertens Res. 2005;28:263–71. 10.1291/hypres.28.263. [DOI] [PubMed] [Google Scholar]
  • 195.Ruan Y, Wu S, Zhang L, Chen G, Lai W. Retarding the senescence of human vascular endothelial cells induced by hydrogen peroxide: effects of 17beta-estradiol (E2) mediated mitochondria protection. Biogerontology. 2014;15:367–75. 10.1007/s10522-014-9507-2. [DOI] [PubMed] [Google Scholar]
  • 196.Li P, Gan Y, Xu Y, Wang L, Ouyang B, Zhang C, Luo L, Zhao C, Zhou Q. 17beta-estradiol attenuates TNF-alpha-induced premature senescence of nucleus pulposus cells through regulating the ROS/NF-kappaB pathway. Int J Biol Sci. 2017;13:145–56. 10.7150/ijbs.16770. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 197.Song S, Wu S, Wang Y, Wang Z, Ye C, Song R, Song D, Ruan Y. 17beta-estradiol inhibits human umbilical vascular endothelial cell senescence by regulating autophagy via p53. Exp Gerontol. 2018;114:57–66. 10.1016/j.exger.2018.10.021. [DOI] [PubMed] [Google Scholar]
  • 198.Xiang X, Huang J, Song S, Wang Y, Zeng Y, Wu S, Ruan Y. 17beta-estradiol inhibits H(2)O(2)-induced senescence in HUVEC cells through upregulating SIRT3 expression and promoting autophagy. Biogerontology. 2020;21:549–57. 10.1007/s10522-020-09868-w. [DOI] [PubMed] [Google Scholar]
  • 199.Lv Y, Huang Y, Fan H, Zhao Y, Ma L, Lan Y, Li C, Chen P, Lou Z, Zhou J. 17beta-Estradiol inhibits hydrogen peroxide-induced senescence and apoptosis in human umbilical vein endothelial cells by regulating the THBS1/TGF-beta/Smad axis. Mol Cell Endocrinol. 2024;580:112111. 10.1016/j.mce.2023.112111. [DOI] [PubMed] [Google Scholar]
  • 200.Zhu C, Zhang L, Zheng Y, Xu J, Song J, Rolfe BE, Campbell JH. Effects of estrogen on stress-induced premature senescence of vascular smooth muscle cells: a novel mechanism for the “time window theory” of menopausal hormone therapy. Atherosclerosis. 2011;215:294–300. 10.1016/j.atherosclerosis.2010.12.025. [DOI] [PubMed] [Google Scholar]
  • 201.Mahoney SA, Venkatasubramanian R, Darrah MA, Ludwig KR, VanDongen NS, Greenberg NT, Longtine AG, Hutton DA, Brunt VE, Campisi J, Melov S, Seals DR, Rossman MJ, Clayton ZS. Intermittent supplementation with fisetin improves arterial function in old mice by decreasing cellular senescence. Aging Cell. 2024;23:e14060. 10.1111/acel.14060. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 202.Schafer MJ, Zhang X, Kumar A, Atkinson EJ, Zhu Y, Jachim S, Mazula DL, Brown AK, Berning M, Aversa Z, Kotajarvi B, Bruce CJ, Greason KL, Suri RM, Tracy RP, Cummings SR, White TA, LeBrasseur NK. The senescence-associated secretome as an indicator of age and medical risk. JCI Insight. 2020;5:e133668. 10.1172/jci.insight.133668. [DOI] [PMC free article] [PubMed]
  • 203.Bautista-Nino PK, Portilla-Fernandez E, Vaughan DE, Danser AH, Roks AJ. DNA damage: a main determinant of vascular aging. Int J Mol Sci. 2016;17:748. 10.3390/ijms17050748. [DOI] [PMC free article] [PubMed]
  • 204.Justice JN, Leng XI, LeBrasseur NK, Tchkonia T, Kirkland JL, Mitin N, Liu Y, Kritchevsky SB, Nicklas BJ, Ding J. Caloric restriction intervention alters specific circulating biomarkers of the senescence-associated secretome in middle-aged and older adults with obesity and prediabetes in an 18-week randomized controlled trial. J Gerontol A Biol Sci Med Sci. 2024;79:glad214. 10.1093/gerona/glad214. [DOI] [PMC free article] [PubMed]
  • 205.Faubion L, White TA, Peterson BJ, Geske JR, Lebrasseur NK, Schafer MJ, Mielke MM, Miller VM. Effect of menopausal hormone therapy on proteins associated with senescence and inflammation. Physiol Rep. 2020;8:e14535. 10.14814/phy2.14535. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 206.Rossman MJ, Kaplon RE, Hill SD, McNamara MN, Santos-Parker JR, Pierce GL, Seals DR, Donato AJ. Endothelial cell senescence with aging in healthy humans: prevention by habitual exercise and relation to vascular endothelial function. Am J Physiol Heart Circ Physiol. 2017;313:H890–5. 10.1152/ajpheart.00416.2017. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 207.Ahire C, Nyul-Toth A, DelFavero J, Gulej R, Faakye JA, Tarantini S, Kiss T, Kuan-Celarier A, Balasubramanian P, Ungvari A, Tarantini A, Nagaraja R, Yan F, Tang Q, Mukli P, Csipo T, Yabluchanskiy A, Campisi J, Ungvari Z, Csiszar A. Accelerated cerebromicrovascular senescence contributes to cognitive decline in a mouse model of paclitaxel (Taxol)-induced chemobrain. Aging Cell. 2023;22:e13832. 10.1111/acel.13832. [DOI] [PMC free article] [PubMed]
  • 208.Krouwer VJ, Hekking LH, Langelaar-Makkinje M, Regan-Klapisz E, Post JA. Endothelial cell senescence is associated with disrupted cell-cell junctions and increased monolayer permeability. Vasc Cell. 2012;4:12. 10.1186/2045-824X-4-12. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 209.Nyul-Toth A, Patai R, Csiszar A, Ungvari A, Gulej R, Mukli P, Yabluchanskiy A, Benyo Z, Sotonyi P, Prodan CI, Liotta EM, Toth P, Elahi F, Barsi P, Maurovich-Horvat P, Sorond FA, Tarantini S, Ungvari Z. Linking peripheral atherosclerosis to blood-brain barrier disruption: elucidating its role as a manifestation of cerebral small vessel disease in vascular cognitive impairment. Geroscience. 2024. 10.1007/s11357-024-01194-0. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 210.Yamazaki Y, Baker DJ, Tachibana M, Liu CC, van Deursen JM, Brott TG, Bu G, Kanekiyo T. Vascular cell senescence contributes to blood-brain barrier breakdown. Stroke. 2016;47:1068–77. 10.1161/STROKEAHA.115.010835. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 211.Childs BG, Baker DJ, Wijshake T, Conover CA, Campisi J, van Deursen JM. Senescent intimal foam cells are deleterious at all stages of atherosclerosis. Science. 2016;354:472–7. 10.1126/science.aaf6659. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 212.Gardner SE, Humphry M, Bennett MR, Clarke MC. Senescent vascular smooth muscle cells drive inflammation through an interleukin-1alpha-dependent senescence-associated secretory phenotype. Arterioscler Thromb Vasc Biol. 2015;35:1963–74. 10.1161/ATVBAHA.115.305896. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 213.Gray K, Kumar S, Figg N, Harrison J, Baker L, Mercer J, Littlewood T, Bennett M. Effects of DNA damage in smooth muscle cells in atherosclerosis. Circ Res. 2015;116:816–26. 10.1161/CIRCRESAHA.116.304921. [DOI] [PubMed] [Google Scholar]
  • 214.Hayashi T, Kotani H, Yamaguchi T, Taguchi K, Iida M, Ina K, Maeda M, Kuzuya M, Hattori Y, Ignarro LJ. Endothelial cellular senescence is inhibited by liver X receptor activation with an additional mechanism for its atheroprotection in diabetes. Proc Natl Acad Sci U S A. 2014;111:1168–73. 10.1073/pnas.1322153111. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 215.Matthews C, Gorenne I, Scott S, Figg N, Kirkpatrick P, Ritchie A, Goddard M, Bennett M. Vascular smooth muscle cells undergo telomere-based senescence in human atherosclerosis: effects of telomerase and oxidative stress. Circ Res. 2006;99:156–64. 10.1161/01.RES.0000233315.38086.bc. [DOI] [PubMed] [Google Scholar]
  • 216.Balasubramanian P, Kiss T, Tarantini S, Nyul-Toth A, Ahire C, Yabluchanskiy A, Csipo T, Lipecz A, Tabak A, Institoris A, Csiszar A, Ungvari Z. Obesity-induced cognitive impairment in older adults: a microvascular perspective. Am J Physiol Heart Circ Physiol. 2021;320:H740–61. 10.1152/ajpheart.00736.2020. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 217.Clements ME, Chaber CJ, Ledbetter SR, Zuk A. Increased cellular senescence and vascular rarefaction exacerbate the progression of kidney fibrosis in aged mice following transient ischemic injury. PLoS One. 2013;8:e70464. 10.1371/journal.pone.0070464. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 218.Ungvari Z, Podlutsky A, Sosnowska D, Tucsek Z, Toth P, Deak F, Gautam T, Csiszar A, Sonntag WE. Ionizing radiation promotes the acquisition of a senescence-associated secretory phenotype and impairs angiogenic capacity in cerebromicrovascular endothelial cells: role of increased DNA damage and decreased DNA repair capacity in microvascular radiosensitivity. J Gerontol A Biol Sci Med Sci. 2013;68:1443–57. 10.1093/gerona/glt057. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 219.Walker AE, Morgan RG, Ives SJ, Cawthon RM, Andtbacka RH, Noyes D, Lesniewski LA, Richardson RS, Donato AJ. Age-related arterial telomere uncapping and senescence is greater in women compared with men. Exp Gerontol. 2016;73:65–71. 10.1016/j.exger.2015.11.009. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 220.Yabluchanskiy A, Tarantini S, Balasubramanian P, Kiss T, Csipo T, Fulop GA, Lipecz A, Ahire C, DelFavero J, Nyul-Toth A, Sonntag WE, Schwartzman ML, Campisi J, Csiszar A, Ungvari Z. Pharmacological or genetic depletion of senescent astrocytes prevents whole brain irradiation-induced impairment of neurovascular coupling responses protecting cognitive function in mice. Geroscience. 2020. 10.1007/s11357-020-00154-8. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 221.Ungvari Z, Csiszar A, Kaley G. Vascular Inflammation in Aging. Herz. 2004;29:733–40. [DOI] [PubMed] [Google Scholar]
  • 222.Csiszar A, Labinskyy N, Jimenez R, Pinto JT, Ballabh P, Losonczy G, Pearson KJ, de Cabo R, Ungvari Z. Anti-oxidative and anti-inflammatory vasoprotective effects of caloric restriction in aging: role of circulating factors and SIRT1. Mech Ageing Dev. 2009. 10.1016/j.mad.2009.06.004. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 223.Ungvari ZI, Orosz Z, Labinskyy N, Rivera A, Xiangmin Z, Smith KE, Csiszar A. Increased mitochondrial H2O2 production promotes endothelial NF-kB activation in aged rat arteries. Am J Physiol Heart Circ Physiol. 2007;293:H37–47. [DOI] [PubMed] [Google Scholar]
  • 224.Ungvari Z, Bailey-Downs L, Gautam T, Sosnowska D, Wang M, Monticone RE, Telljohann R, Pinto JT, de Cabo R, Sonntag WE, Lakatta E, Csiszar A. Age-associated vascular oxidative stress, Nrf2 dysfunction and NF-kB activation in the non-human primate Macaca mulatta. J Gerontol A Biol Sci Med Sci. 2011;66:866–75. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 225.Engelen SE, Robinson AJB, Zurke YX, Monaco C. Therapeutic strategies targeting inflammation and immunity in atherosclerosis: how to proceed? Nat Rev Cardiol. 2022;19:522–42. 10.1038/s41569-021-00668-4. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 226.Lusis AJ. Y-chromosome genetic variation associated with atherosclerosis and inflammation. Arterioscler Thromb Vasc Biol. 2019;39:2201–2. 10.1161/ATVBAHA.119.313369. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 227.Lavin Plaza B, Phinikaridou A, Andia ME, Potter M, Lorrio S, Rashid I, Botnar RM. Sustained focal vascular inflammation accelerates atherosclerosis in remote arteries. Arterioscler Thromb Vasc Biol. 2020;40:2159–70. 10.1161/ATVBAHA.120.314387. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 228.Chait A, Wang S, Goodspeed L, Gomes D, Turk KE, Wietecha T, Tang J, Storey C, O’Brien KD, Rubinow KB, Tang C, Vaisar T, Gharib SA, Lusis AJ, Den Hartigh LJ. Sexually Dimorphic relationships among Saa3 (serum amyloid A3), inflammation, and cholesterol metabolism modulate atherosclerosis in mice. Arterioscler Thromb Vasc Biol. 2021;41:e299–313. 10.1161/ATVBAHA.121.316066. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 229.Wolf D, Ley K. Immunity and inflammation in atherosclerosis. Circ Res. 2019;124:315–27. 10.1161/CIRCRESAHA.118.313591. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 230.Doran AC. Inflammation resolution: implications for atherosclerosis. Circ Res. 2022;130:130–48. 10.1161/CIRCRESAHA.121.319822. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 231.Lombardo A, Biasucci LM, Lanza GA, Coli S, Silvestri P, Cianflone D, Liuzzo G, Burzotta F, Crea F, Maseri A. Inflammation as a possible link between coronary and carotid plaque instability. Circulation. 2004;109:3158–63. 10.1161/01.CIR.0000130786.28008.56. [DOI] [PubMed] [Google Scholar]
  • 232.Ammirati E, Moroni F, Norata GD, Magnoni M, Camici PG. Markers of inflammation associated with plaque progression and instability in patients with carotid atherosclerosis. Mediators Inflamm. 2015;2015:718329. 10.1155/2015/718329. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 233.Chen BA, Lee WJ, Meng LC, Lin YC, Chung CP, Hsiao FY, Chen LK. Sex-specific implications of inflammation in covert cerebral small vessel disease. BMC Neurol. 2024;24:220. 10.1186/s12883-024-03730-z. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 234.Boczar KE, Cheung K, Boodhwani M, Beauchesne L, Dennie C, Nagpal S, Chan K, Coutinho T. Sex differences in thoracic aortic aneurysm growth. Hypertension. 2019;73:190–6. 10.1161/HYPERTENSIONAHA.118.11851. [DOI] [PubMed] [Google Scholar]
  • 235.DuPont JJ, Kim SK, Kenney RM, Jaffe IZ. Sex differences in the time course and mechanisms of vascular and cardiac aging in mice: role of the smooth muscle cell mineralocorticoid receptor. Am J Physiol Heart Circ Physiol. 2021;320:H169–80. 10.1152/ajpheart.00262.2020. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 236.Nussenzweig SC, Verma S, Finkel T. The role of autophagy in vascular biology. Circ Res. 2015;116:480–8. 10.1161/CIRCRESAHA.116.303805. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 237.Marzetti E, Csiszar A, Dutta D, Balagopal G, Calvani R, Leeuwenburgh C. Role of mitochondrial dysfunction and altered autophagy in cardiovascular aging and disease: from mechanisms to therapeutics. Am J Physiol Heart Circ Physiol. 2013. 10.1152/ajpheart.00936.2012. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 238.Donato AJ, Machin DR, Lesniewski LA. Mechanisms of dysfunction in the aging vasculature and role in age-related disease. Circ Res. 2018;123:825–48. 10.1161/CIRCRESAHA.118.312563. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 239.Abdellatif M, Sedej S, Carmona-Gutierrez D, Madeo F, Kroemer G. Autophagy in cardiovascular aging. Circ Res. 2018;123:803–24. 10.1161/CIRCRESAHA.118.312208. [DOI] [PubMed] [Google Scholar]
  • 240.Madrigal-Matute J, de Bruijn J, van Kuijk K, Riascos-Bernal DF, Diaz A, Tasset I, Martin-Segura A, Gijbels MJJ, Sander B, Kaushik S, Biessen EAL, Tiano S, Bourdenx M, Krause GJ, McCracken I, Baker AH, Jin H, Sibinga NES, Bravo-Cordero JJ, Macian F, Singh R, Rensen PCN, Berbee JFP, Pasterkamp G, Sluimer JC, Cuervo AM. Protective role of chaperone-mediated autophagy against atherosclerosis. Proc Natl Acad Sci U S A. 2022;119:e2121133119. 10.1073/pnas.2121133119. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 241.Madrigal-Matute J, Cuervo AM, Sluimer JC. Chaperone-mediated autophagy protects against atherosclerosis. Autophagy. 2022;18:2505–7. 10.1080/15548627.2022.2096397. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 242.Ma L, Li K, Wei W, Zhou J, Li Z, Zhang T, Wangsun Y, Tian F, Dong Q, Zhang H, Xing W. Exercise protects aged mice against coronary endothelial senescence via FUNDC1-dependent mitophagy. Redox Biol. 2023;62:102693. 10.1016/j.redox.2023.102693. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 243.Kim Y, Park JK, Seo JH, Ryu HS, Lim KS, Jeong MH, Kang DH, Kang SW. A rapamycin derivative, biolimus, preferentially activates autophagy in vascular smooth muscle cells. Sci Rep. 2018;8:16551. 10.1038/s41598-018-34877-8. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 244.Triolo M, Oliveira AN, Kumari R, Hood DA. The influence of age, sex, and exercise on autophagy, mitophagy, and lysosome biogenesis in skeletal muscle. Skelet Muscle. 2022;12:13. 10.1186/s13395-022-00296-7. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 245.Olivan S, Calvo AC, Manzano R, Zaragoza P, Osta R. Sex differences in constitutive autophagy. Biomed Res Int. 2014;2014:652817. 10.1155/2014/652817. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 246.Luk HY, Appell C, Levitt DE, Jiwan NC, Vingren JL. Differential autophagy response in men and women after muscle damage. Front Physiol. 2021;12:752347. 10.3389/fphys.2021.752347. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 247.Congdon EE. Sex differences in autophagy contribute to female vulnerability in Alzheimer’s disease. Front Neurosci. 2018;12:372. 10.3389/fnins.2018.00372. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 248.Du L, Hickey RW, Bayir H, Watkins SC, Tyurin VA, Guo F, Kochanek PM, Jenkins LW, Ren J, Gibson G, Chu CT, Kagan VE, Clark RS. Starving neurons show sex difference in autophagy. J Biol Chem. 2009;284:2383–96. 10.1074/jbc.M804396200. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 249.Meng Q, Li Y, Ji T, Chao Y, Li J, Fu Y, Wang S, Chen Q, Chen W, Huang F, Wang Y, Zhang Q, Wang X, Bian H. Estrogen prevent atherosclerosis by attenuating endothelial cell pyroptosis via activation of estrogen receptor alpha-mediated autophagy. J Adv Res. 2021;28:149–64. 10.1016/j.jare.2020.08.010. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 250.Franconi F, Capobianco G, Diana G, Lodde V, De Donno A, Idda ML, Montella A, Campesi I. Sex influence on autophagy markers and miRNAs in basal and angiotensin II-treated human umbilical vein endothelial cells. Int J Mol Sci. 2023;24:14929. 10.3390/ijms241914929. [DOI] [PMC free article] [PubMed]
  • 251.Piekarski A, Khaldi S, Greene E, Lassiter K, Mason JG, Anthony N, Bottje W, Dridi S. Tissue distribution, gender- and genotype-dependent expression of autophagy-related genes in avian species. PLoS One. 2014;9:e112449. 10.1371/journal.pone.0112449. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 252.Kaushik S, Cuervo AM. Proteostasis and aging. Nat Med. 2015;21:1406–15. 10.1038/nm.4001. [DOI] [PubMed] [Google Scholar]
  • 253.Hwang HV, Lin Y, Rebuffatti MN, Tran DT, Lee L, Gomes AV, Li CS, Knowlton AA. Impaired proteostasis in senescent vascular endothelial cells: a perspective on estrogen and oxidative stress in the aging vasculature. Am J Physiol Heart Circ Physiol. 2019;316:H421–9. 10.1152/ajpheart.00318.2018. [DOI] [PubMed] [Google Scholar]
  • 254.Miller BF, Pharaoh GA, Hamilton KL, Peelor FF, Kirkland JL, Freeman WM, Mann SN, Kinter M, Price JC, Stout MB. Short-term calorie restriction and 17alpha-estradiol administration elicit divergent effects on proteostatic processes and protein content in metabolically active tissues. J Gerontol A Biol Sci Med Sci. 2020;75:849–57. 10.1093/gerona/glz113. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 255.Tower J, Pomatto LCD, Davies KJA. Sex differences in the response to oxidative and proteolytic stress. Redox Biol. 2020;31:101488. 10.1016/j.redox.2020.101488. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 256.Jenkins EC, Shah N, Gomez M, Casalena G, Zhao D, Kenny TC, Guariglia SR, Manfredi G, Germain D. Proteasome mapping reveals sexual dimorphism in tissue-specific sensitivity to protein aggregations. EMBO Rep. 2020;21:e48978. 10.15252/embr.201948978. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 257.Gueugneau M, Coudy-Gandilhon C, Gourbeyre O, Chambon C, Combaret L, Polge C, Taillandier D, Attaix D, Friguet B, Maier AB, Butler-Browne G, Bechet D. Proteomics of muscle chronological ageing in post-menopausal women. BMC Genomics. 2014;15:1165. 10.1186/1471-2164-15-1165. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 258.Rao NR, Upadhyay A, Savas JN. Derailed protein turnover in the aging mammalian brain. Mol Syst Biol. 2024;20:120–39. 10.1038/s44320-023-00009-2. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 259.Bellavista E, Martucci M, Vasuri F, Santoro A, Mishto M, Kloss A, Capizzi E, Degiovanni A, Lanzarini C, Remondini D, Dazzi A, Pellegrini S, Cescon M, Capri M, Salvioli S, D’Errico-Grigioni A, Dahlmann B, Grazi GL, Franceschi C. Lifelong maintenance of composition, function and cellular/subcellular distribution of proteasomes in human liver. Mech Ageing Dev. 2014;141–142:26–34. 10.1016/j.mad.2014.09.003. [DOI] [PubMed] [Google Scholar]
  • 260.Kiss T, Giles CB, Tarantini S, Yabluchanskiy A, Balasubramanian P, Gautam T, Csipo T, Nyul-Toth A, Lipecz A, Szabo C, Farkas E, Wren JD, Csiszar A, Ungvari Z. Nicotinamide mononucleotide (NMN) supplementation promotes anti-aging miRNA expression profile in the aorta of aged mice, predicting epigenetic rejuvenation and anti-atherogenic effects. Geroscience. 2019;41:419–39. 10.1007/s11357-019-00095-x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 261.Yang D, Wei G, Long F, Nie H, Tian X, Qu L, Wang S, Li P, Qiu Y, Wang Y, Hong W, Ni T, Liu X, Zhu YZ. Histone methyltransferase Smyd3 is a new regulator for vascular senescence. Aging Cell. 2020;19:e13212. 10.1111/acel.13212. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 262.Panduga S, Vasishta S, Subramani R, Vincent S, Mutalik S, Joshi MB. Epidrugs in the clinical management of atherosclerosis: mechanisms, challenges and promises. Eur J Pharmacol. 2024;980:176827. 10.1016/j.ejphar.2024.176827. [DOI] [PubMed] [Google Scholar]
  • 263.Jokai M, Torma F, McGreevy KM, Koltai E, Bori Z, Babszki G, Bakonyi P, Gombos Z, Gyorgy B, Aczel D, Toth L, Osvath P, Fridvalszky M, Teglas T, Posa A, Kujach S, Olek R, Kawamura T, Seki Y, Suzuki K, Tanisawa K, Goto S, Kerepesi C, Boldogh I, Ba X, Davies KJA, Horvath S, Radak Z. DNA methylation clock DNAmFitAge shows regular exercise is associated with slower aging and systemic adaptation. Geroscience. 2023;45:2805–17. 10.1007/s11357-023-00826-1. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 264.Masser DR, Hadad N, Porter HL, Mangold CA, Unnikrishnan A, Ford MM, Giles CB, Georgescu C, Dozmorov MG, Wren JD, Richardson A, Stanford DR, Freeman WM. Sexually divergent DNA methylation patterns with hippocampal aging. Aging Cell. 2017;16:1342–52. 10.1111/acel.12681. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 265.Jung M, Pfeifer GP. Aging and DNA methylation. BMC Biol. 2015;13:7. 10.1186/s12915-015-0118-4. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 266.Jones MJ, Goodman SJ, Kobor MS. DNA methylation and healthy human aging. Aging Cell. 2015;14:924–32. 10.1111/acel.12349. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 267.Xu H, Li S, Liu YS. Roles and mechanisms of DNA methylation in vascular aging and related diseases. Front Cell Dev Biol. 2021;9:699374. 10.3389/fcell.2021.699374. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 268.Imai S, Johnson FB, Marciniak RA, McVey M, Park PU, Guarente L. Sir2: an NAD-dependent histone deacetylase that connects chromatin silencing, metabolism, and aging. Cold Spring Harb Symp Quant Biol. 2000;65:297–302. [DOI] [PubMed] [Google Scholar]
  • 269.Das C, Tyler JK. Histone exchange and histone modifications during transcription and aging. Biochim Biophys Acta. 2013;1819:332–42. 10.1016/j.bbagrm.2011.08.001. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 270.Ungvari Z, Labinskyy N, Mukhopadhyay P, Pinto JT, Bagi Z, Ballabh P, Zhang C, Pacher P, Csiszar A. Resveratrol attenuates mitochondrial oxidative stress in coronary arterial endothelial cells. Am J Physiol Heart Circ Physiol. 2009;297:H1876–1881. 10.1152/ajpheart.00375.2009. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 271.Kiss T, Nyul-Toth A, Balasubramanian P, Tarantini S, Ahire C, Yabluchanskiy A, Csipo T, Farkas E, Wren JD, Garman L, Csiszar A, Ungvari Z. Nicotinamide mononucleotide (NMN) supplementation promotes neurovascular rejuvenation in aged mice: transcriptional footprint of SIRT1 activation, mitochondrial protection, anti-inflammatory, and anti-apoptotic effects. Geroscience. 2020;42:527–46. 10.1007/s11357-020-00165-5. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 272.Kiss T, Balasubramanian P, Valcarcel-Ares MN, Tarantini S, Yabluchanskiy A, Csipo T, Lipecz A, Reglodi D, Zhang XA, Bari F, Farkas E, Csiszar A, Ungvari Z. Nicotinamide mononucleotide (NMN) treatment attenuates oxidative stress and rescues angiogenic capacity in aged cerebromicrovascular endothelial cells: a potential mechanism for the prevention of vascular cognitive impairment. Geroscience. 2019;41:619–30. 10.1007/s11357-019-00074-2. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 273.Csiszar A, Labinskyy N, Podlutsky A, Kaminski PM, Wolin MS, Zhang C, Mukhopadhyay P, Pacher P, Hu F, de Cabo R, Ballabh P, Ungvari Z. Vasoprotective effects of resveratrol and SIRT1: attenuation of cigarette smoke-induced oxidative stress and proinflammatory phenotypic alterations. Am J Physiol Heart Circ Physiol. 2008;294:H2721–2735. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 274.Csiszar A, Labinskyy N, Pinto JT, Ballabh P, Zhang H, Losonczy G, Pearson K, de Cabo R, Pacher P, Zhang C, Ungvari Z. Resveratrol induces mitochondrial biogenesis in endothelial cells. Am J Physiol Heart Circ Physiol. 2009;297:H13–20. 10.1152/ajpheart.00368.2009. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 275.Ungvari Z, Tucsek Z, Sosnowska D, Toth P, Gautam T, Podlutsky A, Csiszar A, Losonczy G, Valcarcel-Ares MN, Sonntag WE. Aging-induced dysregulation of dicer1-dependent microRNA expression impairs angiogenic capacity of rat cerebromicrovascular endothelial cells. J Gerontol A Biol Sci Med Sci. 2013;68:877–91. 10.1093/gerona/gls242. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 276.Tarantini S, Giles CB, Wren JD, Ashpole NM, Valcarcel-Ares MN, Wei JY, Sonntag WE, Ungvari Z, Csiszar A. IGF-1 deficiency in a critical period early in life influences the vascular aging phenotype in mice by altering miRNA-mediated post-transcriptional gene regulation: implications for the developmental origins of health and disease hypothesis. Age (Dordr). 2016;38:239–58. 10.1007/s11357-016-9943-9. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 277.Menghini R, Stohr R, Federici M. MicroRNAs in vascular aging and atherosclerosis. Ageing Res Rev. 2014;17:68–78. 10.1016/j.arr.2014.03.005. [DOI] [PubMed] [Google Scholar]
  • 278.Yang M, Zhang M, Li Z, Liu J, Li Y, Yang Z, Wang X, Huang X, Yu B, Hou J, Liu Q. A landscape of long non-coding RNAs reveals the leading transcriptome alterations in murine aorta during aging. Genomics. 2023;115:110573. 10.1016/j.ygeno.2023.110573. [DOI] [PubMed] [Google Scholar]
  • 279.Lapikova-Bryhinska T, Ministrini S, Puspitasari YM, Kraler S, Mohamed SA, Costantino S, Paneni F, Khetsuriani M, Bengs S, Liberale L, Montecucco F, Krampla W, Riederer P, Hinterberger M, Fischer P, Luscher TF, Grunblatt E, Akhmedov A, Camici GG. Long non-coding RNAs H19 and NKILA are associated with the risk of death and lacunar stroke in the elderly population. Eur J Intern Med. 2024;123:94–101. 10.1016/j.ejim.2023.11.013. [DOI] [PubMed] [Google Scholar]
  • 280.Hofmann P, Sommer J, Theodorou K, Kirchhof L, Fischer A, Li Y, Perisic L, Hedin U, Maegdefessel L, Dimmeler S, Boon RA. Long non-coding RNA H19 regulates endothelial cell aging via inhibition of STAT3 signalling. Cardiovasc Res. 2019;115:230–42. 10.1093/cvr/cvy206. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 281.Bink DI, Lozano-Vidal N, Boon RA. Long non-coding RNA in vascular disease and aging. Noncoding RNA. 2019;5:26. 10.3390/ncrna5010026. [DOI] [PMC free article] [PubMed]
  • 282.Pellegrini C, Pirazzini C, Sala C, Sambati L, Yusipov I, Kalyakulina A, Ravaioli F, Kwiatkowska KM, Durso DF, Ivanchenko M, Monti D, Lodi R, Franceschi C, Cortelli P, Garagnani P, Bacalini MG. A meta-analysis of brain DNA methylation across sex, age, and Alzheimer’s disease points for accelerated epigenetic aging in neurodegeneration. Front Aging Neurosci. 2021;13:639428. 10.3389/fnagi.2021.639428. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 283.Oblak L, van der Zaag J, Higgins-Chen AT, Levine ME, Boks MP. A systematic review of biological, social and environmental factors associated with epigenetic clock acceleration. Ageing Res Rev. 2021;69:101348. 10.1016/j.arr.2021.101348. [DOI] [PubMed]
  • 284.Engelbrecht HR, Merrill SM, Gladish N, MacIsaac JL, Lin DTS, Ecker S, Chrysohoou CA, Pes GM, Kobor MS, Rehkopf DH. Sex differences in epigenetic age in Mediterranean high longevity regions. Front Aging. 2022;3:1007098. 10.3389/fragi.2022.1007098. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 285.Kim J, Kim JY, Song KS, Lee YH, Seo JS, Jelinek J, Goldschmidt-Clermont PJ, Issa JP. Epigenetic changes in estrogen receptor beta gene in atherosclerotic cardiovascular tissues and in-vitro vascular senescence. Biochim Biophys Acta. 2007;1772:72–80. 10.1016/j.bbadis.2006.10.004. [DOI] [PubMed] [Google Scholar]
  • 286.Xu Z, Yu Z, Chen M, Zhang M, Chen R, Yu H, Lin Y, Wang D, Li S, Huang L, Li Y, Yuan J, Yin P. Mechanisms of estrogen deficiency-induced osteoporosis based on transcriptome and DNA methylation. Front Cell Dev Biol. 2022;10:1011725. 10.3389/fcell.2022.1011725. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 287.Zhang L, Young JI, Gomez L, Silva TC, Schmidt MA, Cai J, Chen X, Martin ER, Wang L. Sex-specific DNA methylation differences in Alzheimer’s disease pathology. Acta Neuropathol Commun. 2021;9:77. 10.1186/s40478-021-01177-8. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 288.CS T, Zhang W, Young JI, Gomez L, Schmidt MA, Varma A, Chen XS, Martin ER, Wang L. Distinct sex-specific DNA methylation differences in Alzheimer’s disease. Alzheimers Res Ther. 2022;14:133. 10.1186/s13195-022-01070-z. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 289.O’Shea DM, Maynard T, Tremont G. DNA methylation “GrimAge” acceleration mediates sex/gender differences in verbal memory and processing speed: findings from the health and retirement study. J Gerontol A Biol Sci Med Sci. 2022;77:2402–12. 10.1093/gerona/glac133. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 290.Landen S, Jacques M, Hiam D, Alvarez-Romero J, Schittenhelm RB, Shah AD, Huang C, Steele JR, Harvey NR, Haupt LM, Griffiths LR, Ashton KJ, Lamon S, Voisin S, Eynon N. Sex differences in muscle protein expression and DNA methylation in response to exercise training. Biol Sex Differ. 2023;14:56. 10.1186/s13293-023-00539-2. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 291.Hall E, Volkov P, Dayeh T, Esguerra JL, Salo S, Eliasson L, Ronn T, Bacos K, Ling C. Sex differences in the genome-wide DNA methylation pattern and impact on gene expression, microRNA levels and insulin secretion in human pancreatic islets. Genome Biol. 2014;15:522. 10.1186/s13059-014-0522-z. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 292.Grant OA, Wang Y, Kumari M, Zabet NR, Schalkwyk L. Characterising sex differences of autosomal DNA methylation in whole blood using the Illumina EPIC array. Clin Epigenetics. 2022;14:62. 10.1186/s13148-022-01279-7. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 293.Tan Q, Li S, Sorensen M, Nygaard M, Mengel-From J, Christensen K. Age patterns of intra-pair DNA methylation discordance in twins: sex difference in epigenomic instability and implication on survival. Aging Cell. 2021;20:e13460. 10.1111/acel.13460. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 294.Nissen E, Reiner A, Liu S, Wallace RB, Molinaro AM, Salas LA, Christensen BC, Wiencke JK, Koestler DC, Kelsey KT. Assessment of immune cell profiles among post-menopausal women in the Women’s Health Initiative using DNA methylation-based methods. Clin Epigenetics. 2023;15:69. 10.1186/s13148-023-01488-8. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 295.Bahl A, Pollanen E, Ismail K, Sipila S, Mikkola TM, Berglund E, Lindqvist CM, Syvanen AC, Rantanen T, Kaprio J, Kovanen V, Ollikainen M. Hormone replacement therapy associated white blood cell DNA methylation and gene expression are associated with within-pair differences of body adiposity and bone mass. Twin Res Hum Genet. 2015;18:647–61. 10.1017/thg.2015.82. [DOI] [PubMed] [Google Scholar]
  • 296.Boyne DJ, Friedenreich CM, McIntyre JB, Stanczyk FZ, Courneya KS, King WD. Endogenous sex hormone exposure and repetitive element DNA methylation in healthy postmenopausal women. Cancer Causes Control. 2017;28:1369–79. 10.1007/s10552-017-0958-z. [DOI] [PubMed] [Google Scholar]
  • 297.Lu S, Niu Z, Chen Y, Tu Q, Zhang Y, Chen W, Tong W, Zhang Z. Repetitive element DNA methylation is associated with menopausal age. Aging Dis. 2018;9:435–43. 10.14336/AD.2017.0810. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 298.Xu J, Deng X, Watkins R, Disteche CM. Sex-specific differences in expression of histone demethylases Utx and Uty in mouse brain and neurons. J Neurosci. 2008;28:4521–7. 10.1523/JNEUROSCI.5382-07.2008. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 299.Tsai HW, Grant PA, Rissman EF. Sex differences in histone modifications in the neonatal mouse brain. Epigenetics. 2009;4:47–53. 10.4161/epi.4.1.7288. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 300.Zou Y, Wu Z, Fan Z, Liang D, Wang L, Song Z, You F. Analyses of mRNA-seq and miRNA-seq of the brain reveal the sex differences of gene expression and regulation before and during gonadal differentiation in 17beta-estradiol or 17alpha-methyltestosterone-induced olive flounder (Paralichthys olivaceus). Mol Reprod Dev. 2020;87:78–90. 10.1002/mrd.23303. [DOI] [PubMed] [Google Scholar]
  • 301.Tomeva E, Krammer UDB, Switzeny OJ, Haslberger AG, Hippe B. Sex-specific miRNA differences in liquid biopsies from subjects with solid tumors and healthy controls. Epigenomes. 2023;7:2. 10.3390/epigenomes7010002. [DOI] [PMC free article] [PubMed]
  • 302.Lopez-Royo T, Moreno-Martinez L, Moreno-Garcia L, Calvo AC, Manzano R, Osta R. Sex differences on constitutive long non-coding RNA expression: modulatory effect of estradiol and testosterone in muscle cells. Andrology. 2024. 10.1111/andr.13624. [DOI] [PubMed] [Google Scholar]
  • 303.Baar EL, Carbajal KA, Ong IM, Lamming DW. Sex- and tissue-specific changes in mTOR signaling with age in C57BL/6J mice. Aging Cell. 2016;15:155–66. 10.1111/acel.12425. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 304.Ballesteros-Alvarez J, Andersen JK. mTORC2: the other mTOR in autophagy regulation. Aging Cell. 2021;20:e13431. 10.1111/acel.13431. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 305.Tabibzadeh S. Role of autophagy in aging: the good, the bad, and the ugly. Aging Cell. 2023;22:e13753. 10.1111/acel.13753. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 306.Van Skike CE, Lin AL, Roberts Burbank R, Halloran JJ, Hernandez SF, Cuvillier J, Soto VY, Hussong SA, Jahrling JB, Javors MA, Hart MJ, Fischer KE, Austad SN, Galvan V. mTOR drives cerebrovascular, synaptic, and cognitive dysfunction in normative aging. Aging Cell. 2020;19:e13057. 10.1111/acel.13057. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 307.Van Skike CE, Jahrling JB, Olson AB, Sayre NL, Hussong SA, Ungvari Z, Lechleiter JD, Galvan V. Inhibition of mTOR protects the blood-brain barrier in models of Alzheimer’s disease and vascular cognitive impairment. Am J Physiol Heart Circ Physiol. 2018;314:H693–703. 10.1152/ajpheart.00570.2017. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 308.Quarles E, Basisty N, Chiao YA, Merrihew G, Gu H, Sweetwyne MT, Fredrickson J, Nguyen NH, Razumova M, Kooiker K, Moussavi-Harami F, Regnier M, Quarles C, MacCoss M, Rabinovitch PS. Rapamycin persistently improves cardiac function in aged, male and female mice, even following cessation of treatment. Aging Cell. 2020;19:e13086. 10.1111/acel.13086. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 309.Galvan V, Hart MJ. Vascular mTOR-dependent mechanisms linking the control of aging to Alzheimer’s disease. Biochim Biophys Acta. 2016;1862:992–1007. 10.1016/j.bbadis.2015.11.010. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 310.Lin AL, Zheng W, Halloran JJ, Burbank RR, Hussong SA, Hart MJ, Javors M, Shih YY, Muir E, Solano Fonseca R, Strong R, Richardson AG, Lechleiter JD, Fox PT, Galvan V. Chronic rapamycin restores brain vascular integrity and function through NO synthase activation and improves memory in symptomatic mice modeling Alzheimer’s disease. J Cereb Blood Flow Metab. 2013;33:1412–21. 10.1038/jcbfm.2013.82. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 311.Lesniewski LA, Seals DR, Walker AE, Henson GD, Blimline MW, Trott DW, Bosshardt GC, LaRocca TJ, Lawson BR, Zigler MC, Donato AJ. Dietary rapamycin supplementation reverses age-related vascular dysfunction and oxidative stress, while modulating nutrient-sensing, cell cycle, and senescence pathways. Aging Cell. 2017;16:17–26. 10.1111/acel.12524. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 312.Strong R, Miller RA, Bogue M, Fernandez E, Javors MA, Libert S, Marinez PA, Murphy MP, Musi N, Nelson JF, Petrascheck M, Reifsnyder P, Richardson A, Salmon AB, Macchiarini F, Harrison DE. Rapamycin-mediated mouse lifespan extension: late-life dosage regimes with sex-specific effects. Aging Cell. 2020;19:e13269. 10.1111/acel.13269. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 313.Selvarani R, Mohammed S, Richardson A. Effect of rapamycin on aging and age-related diseases-past and future. Geroscience. 2020. 10.1007/s11357-020-00274-1. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 314.Harrison DE, Strong R, Sharp ZD, Nelson JF, Astle CM, Flurkey K, Nadon NL, Wilkinson JE, Frenkel K, Carter CS, Pahor M, Javors MA, Fernandez E, Miller RA. Rapamycin fed late in life extends lifespan in genetically heterogeneous mice. Nature. 2009;460:392–5. 10.1038/nature08221. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 315.Fok WC, Bokov A, Gelfond J, Yu Z, Zhang Y, Doderer M, Chen Y, Javors M, Wood WH 3rd, Becker KG, Richardson A, Perez VI. Combined treatment of rapamycin and dietary restriction has a larger effect on the transcriptome and metabolome of liver. Aging Cell. 2014;13:311–9. 10.1111/acel.12175. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 316.Anisimov VN, Zabezhinski MA, Popovich IG, Piskunova TS, Semenchenko AV, Tyndyk ML, Yurova MN, Antoch MP, Blagosklonny MV. Rapamycin extends maximal lifespan in cancer-prone mice. Am J Pathol. 2010;176:2092–7. 10.2353/ajpath.2010.091050. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 317.Salminen A, Kaarniranta K, Kauppinen A. Age-related changes in AMPK activation: role for AMPK phosphatases and inhibitory phosphorylation by upstream signaling pathways. Ageing Res Rev. 2016;28:15–26. 10.1016/j.arr.2016.04.003. [DOI] [PubMed] [Google Scholar]
  • 318.Salminen A, Kaarniranta K, Kauppinen A. AMPK and HIF signaling pathways regulate both longevity and cancer growth: the good news and the bad news about survival mechanisms. Biogerontology. 2016;17:655–80. 10.1007/s10522-016-9655-7. [DOI] [PubMed] [Google Scholar]
  • 319.Rakshe PS, Dutta BJ, Chib S, Maurya N, Singh S. Unveiling the interplay of AMPK/SIRT1/PGC-1alpha axis in brain health: promising targets against aging and NDDs. Ageing Res Rev. 2024;96:102255. 10.1016/j.arr.2024.102255. [DOI] [PubMed] [Google Scholar]
  • 320.Burkewitz K, Zhang Y, Mair WB. AMPK at the nexus of energetics and aging. Cell Metab. 2014;20:10–25. 10.1016/j.cmet.2014.03.002. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 321.Barcena ML, Tonini G, Haritonow N, Breiter P, Milting H, Baczko I, Muller-Werdan U, Ladilov Y, Regitz-Zagrosek V. Sex and age differences in AMPK phosphorylation, mitochondrial homeostasis, and inflammation in hearts from inflammatory cardiomyopathy patients. Aging Cell. 2023;22:e13894. 10.1111/acel.13894. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 322.Lesniewski LA, Zigler MC, Durrant JR, Donato AJ, Seals DR. Sustained activation of AMPK ameliorates age-associated vascular endothelial dysfunction via a nitric oxide-independent mechanism. Mech Ageing Dev. 2012;133:368–71. 10.1016/j.mad.2012.03.011. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 323.Sardu C, Paolisso P, Sacra C, Mauro C, Minicucci F, Portoghese M, Rizzo MR, Barbieri M, Sasso FC, D’Onofrio N, Balestrieri ML, Calabro P, Paolisso G, Marfella R. Effects of metformin therapy on coronary endothelial dysfunction in patients with prediabetes with stable angina and nonobstructive coronary artery stenosis: the CODYCE multicenter prospective study. Diabetes Care. 2019;42:1946–55. 10.2337/dc18-2356. [DOI] [PubMed] [Google Scholar]
  • 324.Li C, Mu N, Gu C, Liu M, Yang Z, Yin Y, Chen M, Wang Y, Han Y, Yu L, Ma H. Metformin mediates cardioprotection against aging-induced ischemic necroptosis. Aging Cell. 2020;19:e13096. 10.1111/acel.13096. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 325.Begum MK, Konja D, Singh S, Chlopicki S, Wang Y. Endothelial SIRT1 as a target for the prevention of arterial aging: promises and challenges. J Cardiovasc Pharmacol. 2021;78:S63–77. 10.1097/FJC.0000000000001154. [DOI] [PubMed] [Google Scholar]
  • 326.Rogina B, Tissenbaum HA. SIRT1, resveratrol and aging. Front Genet. 2024;15:1393181. 10.3389/fgene.2024.1393181. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 327.Kitada M, Ogura Y, Koya D. The protective role of Sirt1 in vascular tissue: its relationship to vascular aging and atherosclerosis. Aging (Albany NY). 2016;8:2290–307. 10.18632/aging.101068. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 328.Zarzuelo MJ, Lopez-Sepulveda R, Sanchez M, Romero M, Gomez-Guzman M, Ungvary Z, Perez-Vizcaino F, Jimenez R, Duarte J. SIRT1 inhibits NADPH oxidase activation and protects endothelial function in the rat aorta: implications for vascular aging. Biochem Pharmacol. 2013;85:1288–96. 10.1016/j.bcp.2013.02.015. [DOI] [PubMed] [Google Scholar]
  • 329.Ota H, Eto M, Ogawa S, Iijima K, Akishita M, Ouchi Y. SIRT1/eNOS axis as a potential target against vascular senescence, dysfunction and atherosclerosis. J Atheroscler Thromb. 2010;17:431–5. [DOI] [PubMed] [Google Scholar]
  • 330.Gano LB, Donato AJ, Pasha HM, Hearon CM Jr, Sindler AL, Seals DR. The SIRT1 activator SRT1720 reverses vascular endothelial dysfunction, excessive superoxide production, and inflammation with aging in mice. Am J Physiol Heart Circ Physiol. 2014;307:H1754–1763. 10.1152/ajpheart.00377.2014. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 331.Csiszar A, Labinskyy N, Jimenez R, Pinto JT, Ballabh P, Losonczy G, Pearson KJ, de Cabo R, Ungvari Z. Anti-oxidative and anti-inflammatory vasoprotective effects of caloric restriction in aging: role of circulating factors and SIRT1. Mech Ageing Dev. 2009;130:518–27. 10.1016/j.mad.2009.06.004. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 332.Csiszar A, Labinskyy N, Pinto JT, Ballabh P, Zhang H, Losonczy G, Pearson KJ, de Cabo R, Pacher P, Zhang C, Ungvari ZI. Resveratrol induces mitochondrial biogenesis in endothelial cells. Am J Physiol Heart Circ Physiol. 2009. 10.1152/ajpheart.00368.2009. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 333.Stein S, Schafer N, Breitenstein A, Besler C, Winnik S, Lohmann C, Heinrich K, Brokopp CE, Handschin C, Landmesser U, Tanner FC, Luscher TF, Matter CM. SIRT1 reduces endothelial activation without affecting vascular function in ApoE-/- mice. Aging (Albany NY). 2010;2:353–60. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 334.Mattagajasingh I, Kim CS, Naqvi A, Yamamori T, Hoffman TA, Jung SB, DeRicco J, Kasuno K, Irani K. SIRT1 promotes endothelium-dependent vascular relaxation by activating endothelial nitric oxide synthase. Proc Natl Acad Sci USA. 2007;104:14855–60. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 335.Thompson AM, Wagner R, Rzucidlo EM. Age-related loss of SirT1 expression results in dysregulated human vascular smooth muscle cell function. Am J Physiol Heart Circ Physiol. 2014;307:H533–541. 10.1152/ajpheart.00871.2013. [DOI] [PubMed] [Google Scholar]
  • 336.Takemura A, Iijima K, Ota H, Son BK, Ito Y, Ogawa S, Eto M, Akishita M, Ouchi Y. Sirtuin 1 retards hyperphosphatemia-induced calcification of vascular smooth muscle cells. Arterioscler Thromb Vasc Biol. 2011;31:2054–62. 10.1161/ATVBAHA.110.216739. [DOI] [PubMed] [Google Scholar]
  • 337.Miyazaki R, Ichiki T, Hashimoto T, Inanaga K, Imayama I, Sadoshima J, Sunagawa K. SIRT1, a longevity gene, downregulates angiotensin II type 1 receptor expression in vascular smooth muscle cells. Arterioscler Thromb Vasc Biol. 2008;28:1263–9. 10.1161/ATVBAHA.108.166991. [DOI] [PubMed] [Google Scholar]
  • 338.Liu Y, Wang TT, Zhang R, Fu WY, Wang X, Wang F, Gao P, Ding YN, Xie Y, Hao DL, Chen HZ, Liu DP. Calorie restriction protects against experimental abdominal aortic aneurysms in mice. J Exp Med. 2016;213:2473–88. 10.1084/jem.20151794. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 339.Gorenne I, Kumar S, Gray K, Figg N, Yu H, Mercer J, Bennett M. Vascular smooth muscle cell sirtuin 1 protects against DNA damage and inhibits atherosclerosis. Circulation. 2013;127:386–96. 10.1161/CIRCULATIONAHA.112.124404. [DOI] [PubMed] [Google Scholar]
  • 340.Gao P, Xu TT, Lu J, Li L, Xu J, Hao DL, Chen HZ, Liu DP. Overexpression of SIRT1 in vascular smooth muscle cells attenuates angiotensin II-induced vascular remodeling and hypertension in mice. J Mol Med (Berl). 2014;92:347–57. 10.1007/s00109-013-1111-4. [DOI] [PubMed] [Google Scholar]
  • 341.Fry JL, Al Sayah L, Weisbrod RM, Van Roy I, Weng X, Cohen RA, Bachschmid MM, Seta F. Vascular smooth muscle sirtuin-1 protects against diet-induced aortic stiffness. Hypertension. 2016;68:775–84. 10.1161/HYPERTENSIONAHA.116.07622. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 342.Chen HZ, Wang F, Gao P, Pei JF, Liu Y, Xu TT, Tang X, Fu WY, Lu J, Yan YF, Wang XM, Han L, Zhang ZQ, Zhang R, Zou MH, Liu DP. Age-associated sirtuin 1 reduction in vascular smooth muscle links vascular senescence and inflammation to abdominal aortic aneurysm. Circ Res. 2016;119:1076–88. 10.1161/CIRCRESAHA.116.308895. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 343.Wiedenhoeft T, Tarantini S, Nyul-Toth A, Yabluchanskiy A, Csipo T, Balasubramanian P, Lipecz A, Kiss T, Csiszar A, Csiszar A, Ungvari Z. Fusogenic liposomes effectively deliver resveratrol to the cerebral microcirculation and improve endothelium-dependent neurovascular coupling responses in aged mice. Geroscience. 2019;41:711–25. 10.1007/s11357-019-00102-1. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 344.Ungvari Z, Orosz Z, Rivera A, Labinskyy N, Xiangmin Z, Olson S, Podlutsky A, Csiszar A. Resveratrol increases vascular oxidative stress resistance. Am J Physiol. 2007;292:H2417–2424. [DOI] [PubMed] [Google Scholar]
  • 345.Toth P, Tarantini S, Tucsek Z, Ashpole NM, Sosnowska D, Gautam T, Ballabh P, Koller A, Sonntag WE, Csiszar A, Ungvari ZI. Resveratrol treatment rescues neurovascular coupling in aged mice:role of improved cerebromicrovascular endothelial function and down-regulation of NADPH oxidas. Am J Physiol Heart Circ Physiol. 2014;306:H299–308. 10.1152/ajpheart.00744.2013. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 346.Toth P, Tarantini S, Springo Z, Tucsek Z, Gautam T, Giles CB, Wren JD, Koller A, Sonntag WE, Csiszar A, Ungvari Z. Aging exacerbates hypertension-induced cerebral microhemorrhages in mice: role of resveratrol treatment in vasoprotection. Aging Cell. 2015;14:400–8. 10.1111/acel.12315. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 347.Pearson KJ, Baur JA, Lewis KN, Peshkin L, Price NL, Labinskyy N, Swindell WR, Kamara D, Minor RK, Perez E, Jamieson HA, Zhang Y, Dunn SR, Sharma K, Pleshko N, Woollett LA, Csiszar A, Ikeno Y, Le Couteur D, Elliott PJ, Becker KG, Navas P, Ingram DK, Wolf NS, Ungvari Z, Sinclair DA, de Cabo R. Resveratrol delays age-related deterioration and mimics transcriptional aspects of dietary restriction without extending life span. Cell Metab. 2008;8:157–68. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 348.Mattison JA, Wang M, Bernier M, Zhang J, Park SS, Maudsley S, An SS, Santhanam L, Martin B, Faulkner S, Morrell C, Baur JA, Peshkin L, Sosnowska D, Csiszar A, Herbert RL, Tilmont EM, Ungvari Z, Pearson KJ, Lakatta EG, de Cabo R. Resveratrol prevents high fat/sucrose diet-induced central arterial wall inflammation and stiffening in nonhuman primates. Cell Metab. 2014;20:183–90. 10.1016/j.cmet.2014.04.018. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 349.Csiszar A, Smith K, Labinskyy N, Orosz Z, Rivera A, Ungvari Z. Resveratrol attenuates TNF-{alpha}-induced activation of coronary arterial endothelial cells: role of NF-{kappa}B inhibition. Am J Physiol. 2006;291:H1694–1699. [DOI] [PubMed] [Google Scholar]
  • 350.Gurgen D, Kusch A, Klewitz R, Hoff U, Catar R, Hegner B, Kintscher U, Luft FC, Dragun D. Sex-specific mTOR signaling determines sexual dimorphism in myocardial adaptation in normotensive DOCA-salt model. Hypertension. 2013;61:730–6. 10.1161/HYPERTENSIONAHA.111.00276. [DOI] [PubMed] [Google Scholar]
  • 351.Ge Y, Zhou S, Li Y, Wang Z, Chen S, Xia T, Shen J, Teng H, Jiang Q. Estrogen prevents articular cartilage destruction in a mouse model of AMPK deficiency via ERK-mTOR pathway. Ann Transl Med. 2019;7:336. 10.21037/atm.2019.06.77. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 352.Tao Y, Sun H, Sun H, Qiu X, Xu C, Shi C, Du J. 17beta-estradiol activates mTOR in chondrocytes by AKT-dependent and AKT-independent signaling pathways. Int J Clin Exp Pathol. 2015;8:15911–8. [PMC free article] [PubMed] [Google Scholar]
  • 353.Koh PO, Cho JH, Won CK, Lee HJ, Sung JH, Kim MO. Estradiol attenuates the focal cerebral ischemic injury through mTOR/p70S6 kinase signaling pathway. Neurosci Lett. 2008;436:62–6. 10.1016/j.neulet.2008.02.061. [DOI] [PubMed] [Google Scholar]
  • 354.Bian C, Bai B, Gao Q, Li S, Zhao Y. 17beta-estradiol regulates glucose metabolism and insulin secretion in rat islet beta cells through GPER and Akt/mTOR/GLUT2 pathway. Front Endocrinol (Lausanne). 2019;10:531. 10.3389/fendo.2019.00531. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 355.Wolff CA, Lawrence MM, Porter H, Zhang Q, Reid JJ, Laurin JL, Musci RV, Linden MA, Peelor FF 3rd, Wren JD, Creery JS, Cutler KJ, Carson RH, Price JC, Hamilton KL, Miller BF. Sex differences in changes of protein synthesis with rapamycin treatment are minimized when metformin is added to rapamycin. Geroscience. 2020. 10.1007/s11357-020-00243-8. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 356.Juszczak F, Pierre L, Decarnoncle M, Jadot I, Martin B, Botton O, Caron N, Dehairs J, Swinnen JV, Decleves AE. Sex differences in obesity-induced renal lipid accumulation revealed by lipidomics: a role of adiponectin/AMPK axis. Biol Sex Differ. 2023;14:63. 10.1186/s13293-023-00543-6. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 357.Arhen BB, Renwick JRM, Zedic AK, Menezes ES, Preobrazenski N, Simpson CA, Stokes T, McGlory C, Gurd BJ. AMPK and PGC-alpha following maximal and supramaximal exercise in men and women: a randomized cross-over study. Appl Physiol Nutr Metab. 2024;49:526–38. 10.1139/apnm-2023-0256. [DOI] [PubMed] [Google Scholar]
  • 358.Yoshino J, Baur JA, Imai SI. NAD(+) Intermediates: the biology and therapeutic potential of NMN and NR. Cell Metab. 2018;27:513–28. 10.1016/j.cmet.2017.11.002. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 359.Schultz MB, Sinclair DA. Why NAD(+) Declines during aging: it’s destroyed. Cell Metab. 2016;23:965–6. 10.1016/j.cmet.2016.05.022. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 360.Csiszar A, Tarantini S, Yabluchanskiy A, Balasubramanian P, Kiss T, Farkas E, Baur JA, Ungvari ZI. Role of endothelial NAD+ deficiency in age-related vascular dysfunction. Am J Physiol Heart Circ Physiol. 2019;316:H1253–66. 10.1152/ajpheart.00039.2019. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 361.Vaziri H, Dessain SK, Ng Eaton E, Imai SI, Frye RA, Pandita TK, Guarente L, Weinberg RA. hSIR2(SIRT1) functions as an NAD-dependent p53 deacetylase. Cell. 2001;107:149–59. [DOI] [PubMed] [Google Scholar]
  • 362.Revollo JR, Grimm AA, Imai S. The NAD biosynthesis pathway mediated by nicotinamide phosphoribosyltransferase regulates Sir2 activity in mammalian cells. J Biol Chem. 2004;279:50754–63. 10.1074/jbc.M408388200. [DOI] [PubMed] [Google Scholar]
  • 363.Lin SJ, Defossez PA, Guarente L. Requirement of NAD and SIR2 for life-span extension by calorie restriction in Saccharomyces cerevisiae. Science (New York, NY. 2000;289:2126–8. [DOI] [PubMed] [Google Scholar]
  • 364.Tarantini S, Yabluchanskiy A, Csipo T, Fulop G, Kiss T, Balasubramanian P, DelFavero J, Ahire C, Ungvari A, Nyul-Toth A, Farkas E, Benyo Z, Toth A, Csiszar A, Ungvari Z. Treatment with the poly(ADP-ribose) polymerase inhibitor PJ-34 improves cerebromicrovascular endothelial function, neurovascular coupling responses and cognitive performance in aged mice, supporting the NAD+ depletion hypothesis of neurovascular aging. Geroscience. 2019;41:533–42. 10.1007/s11357-019-00101-2. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 365.Lautrup S, Sinclair DA, Mattson MP, Fang EF. NAD(+) in brain aging and neurodegenerative disorders. Cell Metab. 2019;30:630–55. 10.1016/j.cmet.2019.09.001. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 366.Frederick DW, Loro E, Liu L, Davila A Jr, Chellappa K, Silverman IM, Quinn WJ 3rd, Gosai SJ, Tichy ED, Davis JG, Mourkioti F, Gregory BD, Dellinger RW, Redpath P, Migaud ME, Nakamaru-Ogiso E, Rabinowitz JD, Khurana TS, Baur JA. Loss of NAD homeostasis leads to progressive and reversible degeneration of skeletal muscle. Cell Metab. 2016;24:269–82. 10.1016/j.cmet.2016.07.005. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 367.Covarrubias AJ, Kale A, Perrone R, Lopez-Dominguez JA, Pisco AO, Kasler HG, Schmidt MS, Heckenbach I, Kwok R, Wiley CD, Wong HS, Gibbs E, Iyer SS, Basisty N, Wu Q, Kim IJ, Silva E, Vitangcol K, Shin KO, Lee YM, Riley R, Ben-Sahra I, Ott M, Schilling B, Scheibye-Knudsen M, Ishihara K, Quake SR, Newman J, Brenner C, Campisi J, Verdin E. Senescent cells promote tissue NAD(+) decline during ageing via the activation of CD38(+) macrophages. Nat Metab. 2020;2:1265–83. 10.1038/s42255-020-00305-3. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 368.Canto C, Menzies KJ, Auwerx J. NAD(+) metabolism and the control of energy homeostasis: a balancing act between mitochondria and the nucleus. Cell Metab. 2015;22:31–53. 10.1016/j.cmet.2015.05.023. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 369.Camacho-Pereira J, Tarrago MG, Chini CCS, Nin V, Escande C, Warner GM, Puranik AS, Schoon RA, Reid JM, Galina A, Chini EN. CD38 dictates age-related NAD decline and mitochondrial dysfunction through an SIRT3-dependent mechanism. Cell Metab. 2016;23:1127–39. 10.1016/j.cmet.2016.05.006. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 370.Braidy N, Guillemin GJ, Mansour H, Chan-Ling T, Poljak A, Grant R. Age related changes in NAD+ metabolism oxidative stress and Sirt1 activity in wistar rats. PLoS One. 2011;6:e19194. 10.1371/journal.pone.0019194. [DOI] [PMC free article] [PubMed] [Google Scholar] [Retracted]
  • 371.Imai S, Guarente L. NAD+ and sirtuins in aging and disease. Trends Cell Biol. 2014;24:464–71. 10.1016/j.tcb.2014.04.002. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 372.Radovits T, Seres L, Gero D, Lin LN, Beller CJ, Chen SH, Zotkina J, Berger I, Groves JT, Szabo C, Szabo G. The peroxynitrite decomposition catalyst FP15 improves ageing-associated cardiac and vascular dysfunction. Mech Ageing Dev. 2007;128:173–81. 10.1016/j.mad.2006.09.005. [DOI] [PubMed] [Google Scholar]
  • 373.Radovits T, Seres L, Gero D, Berger I, Szabo C, Karck M, Szabo G. Single dose treatment with PARP-inhibitor INO-1001 improves aging-associated cardiac and vascular dysfunction. Exp Gerontol. 2007;42:676–85. 10.1016/j.exger.2007.01.013. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 374.Pacher P, Vaslin A, Benko R, Mabley JG, Liaudet L, Hasko G, Marton A, Batkai S, Kollai M, Szabo C. A new, potent poly(ADP-ribose) polymerase inhibitor improves cardiac and vascular dysfunction associated with advanced aging. J Pharmacol Exp Ther. 2004;311:485–91. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 375.Pacher P, Szabo C. Role of poly(ADP-ribose) polymerase 1 (PARP-1) in cardiovascular diseases: the therapeutic potential of PARP inhibitors. Cardiovasc Drug Rev. 2007;25:235–60. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 376.Pacher P, Mabley JG, Soriano FG, Liaudet L, Szabo C. Activation of poly(ADP-ribose) polymerase contributes to the endothelial dysfunction associated with hypertension and aging. Int J Mol Med. 2002;9:659–64. [PubMed] [Google Scholar]
  • 377.Pacher P, Mabley JG, Soriano FG, Liaudet L, Komjati K, Szabo C. Endothelial dysfunction in aging animals: the role of poly(ADP-ribose) polymerase activation. Br J Pharmacol. 2002;135:1347–50. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 378.Jagtap P, Szabo C. Poly(ADP-ribose) polymerase and the therapeutic effects of its inhibitors. Nat Rev Drug Discov. 2005;4:421–40. [DOI] [PubMed] [Google Scholar]
  • 379.Gomes AP, Price NL, Ling AJ, Moslehi JJ, Montgomery MK, Rajman L, White JP, Teodoro JS, Wrann CD, Hubbard BP, Mercken EM, Palmeira CM, de Cabo R, Rolo AP, Turner N, Bell EL, Sinclair DA. Declining NAD(+) induces a pseudohypoxic state disrupting nuclear-mitochondrial communication during aging. Cell. 2013;155:1624–38. 10.1016/j.cell.2013.11.037. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 380.Das A, Huang GX, Bonkowski MS, Longchamp A, Li C, Schultz MB, Kim LJ, Osborne B, Joshi S, Lu Y, Trevino-Villarreal JH, Kang MJ, Hung TT, Lee B, Williams EO, Igarashi M, Mitchell JR, Wu LE, Turner N, Arany Z, Guarente L, Sinclair DA. Impairment of an endothelial NAD(+)-H2S signaling network is a reversible cause of vascular aging. Cell. 2018;173:74–89 e20. 10.1016/j.cell.2018.02.008. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 381.Imai SI, Guarente L. It takes two to tango: NAD(+) and sirtuins in aging/longevity control. NPJ Aging Mech Dis. 2016;2:16017. 10.1038/npjamd.2016.17. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 382.Imai SI. The NAD World 2.0: the importance of the inter-tissue communication mediated by NAMPT/NAD(+)/SIRT1 in mammalian aging and longevity control. NPJ Syst Biol Appl. 2016;2:16018. 10.1038/npjsba.2016.18. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 383.Csiszar A, Sosnowska D, Tucsek Z, Gautam T, Toth P, Losonczy G, Colman RJ, Weindruch R, Anderson RM, Sonntag WE, Ungvari Z. Circulating factors induced by caloric restriction in the nonhuman primate Macaca mulatta activate angiogenic processes in endothelial cells. J Gerontol A Biol Sci Med Sci. 2013;68:235–49. 10.1093/gerona/gls158. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 384.Moroz N, Carmona JJ, Anderson E, Hart AC, Sinclair DA, Blackwell TK. Dietary restriction involves NAD(+)-dependent mechanisms and a shift toward oxidative metabolism. Aging Cell. 2014;13:1075–85. 10.1111/acel.12273. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 385.Rajman L, Chwalek K, Sinclair DA. Therapeutic potential of NAD-boosting molecules: the in vivo evidence. Cell Metab. 2018;27:529–47. 10.1016/j.cmet.2018.02.011. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 386.Liu L, Su X, Quinn WJ 3rd, Hui S, Krukenberg K, Frederick DW, Redpath P, Zhan L, Chellappa K, White E, Migaud M, Mitchison TJ, Baur JA, Rabinowitz JD. Quantitative analysis of NAD synthesis-breakdown fluxes. Cell Metab. 2018;27:1067–1080 e1065. 10.1016/j.cmet.2018.03.018. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 387.Dellinger RW, Santos SR, Morris M, Evans M, Alminana D, Guarente L, Marcotulli E. Repeat dose NRPT (nicotinamide riboside and pterostilbene) increases NAD(+) levels in humans safely and sustainably: a randomized, double-blind, placebo-controlled study. NPJ Aging Mech Dis. 2017;3:17. 10.1038/s41514-017-0016-9. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 388.Koltai E, Szabo Z, Atalay M, Boldogh I, Naito H, Goto S, Nyakas C, Radak Z. Exercise alters SIRT1, SIRT6, NAD and NAMPT levels in skeletal muscle of aged rats. Mech Ageing Dev. 2010;131:21–8. 10.1016/j.mad.2009.11.002. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 389.Kim HJ, Cao W, Oh GS, Lee S, Shen A, Khadka D, Lee SB, Sharma S, Kim SY, Choe SK, Kwak TH, Kim JM, Park R, So HS. Augmentation of cellular NAD(+) by NQO1 enzymatic action improves age-related hearing impairment. Aging Cell. 2019;18:e13016. 10.1111/acel.13016. [DOI] [PMC free article] [PubMed]
  • 390.Katsyuba E, Auwerx J. Modulating NAD(+) metabolism, from bench to bedside. EMBO J. 2017;36:2670–83. 10.15252/embj.201797135. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 391.Imai S. A possibility of nutriceuticals as an anti-aging intervention: activation of sirtuins by promoting mammalian NAD biosynthesis. Pharmacol Res. 2010;62:42–7. 10.1016/j.phrs.2010.01.006. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 392.Diaz-Ruiz A, Lanasa M, Garcia J, Mora H, Fan F, Martin-Montalvo A, Di Francesco A, Calvo-Rubio M, Salvador-Pascual A, Aon MA, Fishbein KW, Pearson KJ, Villalba JM, Navas P, Bernier M, de Cabo R. Overexpression of CYB5R3 and NQO1, two NAD(+) -producing enzymes, mimics aspects of caloric restriction. Aging Cell. 2018;17:e12767. 10.1111/acel.12767. [DOI] [PMC free article] [PubMed]
  • 393.Yang F, Deng X, Yu Y, Luo L, Chen X, Zheng J, Qiu Y, Xiao F, Xie X, Zhao Y, Guo J, Hu F, Zhang X, Ju Z, Zhou Y. Association of human whole blood NAD(+) contents with aging. Front Endocrinol (Lausanne). 2022;13:829658. 10.3389/fendo.2022.829658. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 394.van der Velpen V, Rosenberg N, Maillard V, Teav T, Chatton JY, Gallart-Ayala H, Ivanisevic J. Sex-specific alterations in NAD+ metabolism in 3xTg Alzheimer’s disease mouse brain assessed by quantitative targeted LC-MS. J Neurochem. 2021;159:378–88. 10.1111/jnc.15362. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 395.Schwarzmann L, Pliquett RU, Simm A, Bartling B. Sex-related differences in human plasma NAD+/NADH levels depend on age. Biosci Rep. 2021;41:BSR20200340. 10.1042/BSR20200340. [DOI] [PMC free article] [PubMed]
  • 396.Park J, Hu R, Xiong S, Qian Y, El-Sabbagh AS, Ibrahim M, Song Q, Yan G, Song Z, Mahmoud AM, He Y, Layden BT, Chen J, Ong SG, Xu P, Jiang Y. Estrogen prevents age-dependent beige adipogenesis failure through NAMPT-controlled ER stress pathway. bioRxiv. 2023. 10.1101/2023.08.31.555821.
  • 397.Zhou SJ, Bi TQ, Qin CX, Yang XQ, Pang K. Expression of NAMPT is associated with breast invasive ductal carcinoma development and prognosis. Oncol Lett. 2018;15:6648–54. 10.3892/ol.2018.8164. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 398.Bickel MA, Csik B, Gulej R, Ungvari A, Nyul-Toth A, Conley SM. Cell non-autonomous regulation of cerebrovascular aging processes by the somatotropic axis. Front Endocrinol (Lausanne). 2023;14:1087053. 10.3389/fendo.2023.1087053. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 399.Kiss T, Nyul-Toth A, Gulej R, Tarantini S, Csipo T, Mukli P, Ungvari A, Balasubramanian P, Yabluchanskiy A, Benyo Z, Conley SM, Wren JD, Garman L, Huffman DM, Csiszar A, Ungvari Z. Old blood from heterochronic parabionts accelerates vascular aging in young mice: transcriptomic signature of pathologic smooth muscle remodeling. Geroscience. 2022;44:953–81. 10.1007/s11357-022-00519-1. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 400.Toth P, Tarantini S, Ashpole NM, Tucsek Z, Milne GL, Valcarcel-Ares NM, Menyhart A, Farkas E, Sonntag WE, Csiszar A, Ungvari Z. IGF-1 deficiency impairs neurovascular coupling in mice: implications for cerebromicrovascular aging. Aging Cell. 2015;14:1034–44. 10.1111/acel.12372. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 401.Sonntag WE, Deak F, Ashpole N, Toth P, Csiszar A, Freeman W, Ungvari Z. Insulin-like growth factor-1 in CNS and cerebrovascular aging. Front Aging Neurosci. 2013;5:27. 10.3389/fnagi.2013.00027. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 402.Csiszar A, Labinskyy N, Smith K, Rivera A, Orosz Z, Ungvari Z. Vasculoprotective effects of anti-TNFalfa treatment in aging. Am J Pathol. 2007;170:388–698. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 403.Gulej R, Nyul-Toth A, Csik B, Petersen B, Faakye J, Negri S, Chandragiri SS, Mukli P, Yabluchanskiy A, Conley S, Huffman DM, Csiszar A, Tarantini S, Ungvari Z. Rejuvenation of cerebromicrovascular function in aged mice through heterochronic parabiosis: insights into neurovascular coupling and the impact of young blood factors. Geroscience. 2023. 10.1007/s11357-023-01039-2. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 404.Gulej R, Nyul-Toth A, Csik B, Petersen B, Faakye J, Negri S, Chandragiri SS, Mukli P, Yabluchanskiy A, Conley S, Huffman DM, Csiszar A, Tarantini S, Ungvari Z. Rejuvenation of cerebromicrovascular function in aged mice through heterochronic parabiosis: insights into neurovascular coupling and the impact of young blood factors. Geroscience. 2024;46:327–47. 10.1007/s11357-023-01039-2. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 405.Gulej R, Nyul-Toth A, Csik B, Patai R, Petersen B, Negri S, Chandragiri SS, Shanmugarama S, Mukli P, Yabluchanskiy A, Conley S, Huffman D, Tarantini S, Csiszar A, Ungvari Z. Young blood-mediated cerebromicrovascular rejuvenation through heterochronic parabiosis: enhancing blood-brain barrier integrity and capillarization in the aged mouse brain. Geroscience. 2024. 10.1007/s11357-024-01154-8. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 406.Khan AS, Lynch CD, Sane DC, Willingham MC, Sonntag WE. Growth hormone increases regional coronary blood flow and capillary density in aged rats. J Gerontol A Biol Sci Med Sci. 2001;56:B364–371. [DOI] [PubMed] [Google Scholar]
  • 407.Tarantini S, Tucsek Z, Valcarcel-Ares MN, Toth P, Gautam T, Giles CB, Ballabh P, Wei JY, Wren JD, Ashpole NM, Sonntag WE, Ungvari Z, Csiszar A. Circulating IGF-1 deficiency exacerbates hypertension-induced microvascular rarefaction in the mouse hippocampus and retrosplenial cortex: implications for cerebromicrovascular and brain aging. Age (Dordr). 2016;38:273–89. 10.1007/s11357-016-9931-0. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 408.Toth P, Tucsek Z, Tarantini S, Sosnowska D, Gautam T, Mitschelen M, Koller A, Sonntag WE, Csiszar A, Ungvari Z. IGF-1 deficiency impairs cerebral myogenic autoregulation in hypertensive mice. J Cereb Blood Flow Metab. 2014;34:1887–97. 10.1038/jcbfm.2014.156. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 409.Higashi Y, Sukhanov S, Anwar A, Shai SY, Delafontaine P. Aging, atherosclerosis, and IGF-1. J Gerontol A Biol Sci Med Sci. 2012;67:626–39. 10.1093/gerona/gls102.gls102[pii]. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 410.Bailey-Downs LC, Sosnowska D, Toth P, Mitschelen M, Gautam T, Henthorn JC, Ballabh P, Koller A, Farley JA, Sonntag WE, Csiszar A, Ungvari Z. Growth hormone and IGF-1 deficiency exacerbate high-fat diet-induced endothelial impairment in obese lewis dwarf rats: implications for vascular aging. J Gerontol A Biol Sci Med Sci. 2012;67:553–64. 10.1093/gerona/glr197. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 411.Bailey-Downs LC, Mitschelen M, Sosnowska D, Toth P, Pinto JT, Ballabh P, Valcarcel-Ares MN, Farley J, Koller A, Henthorn JC, Bass C, Sonntag WE, Ungvari Z, Csiszar A. Liver-specific knockdown of IGF-1 decreases vascular oxidative stress resistance by impairing the Nrf2-dependent antioxidant response: a novel model of vascular aging. J Gerontol A Biol Sci Med Sci. 2012;67:313–29. 10.1093/gerona/glr164. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 412.Lahteenvuo J, Rosenzweig A. Effects of aging on angiogenesis. Circ Res. 2012;110:1252–64. 10.1161/CIRCRESAHA.111.246116. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 413.Baker DJ, Wijshake T, Tchkonia T, LeBrasseur NK, Childs BG, van de Sluis B, Kirkland JL, van Deursen JM. Clearance of p16Ink4a-positive senescent cells delays ageing-associated disorders. Nature. 2011;479:232–6. 10.1038/nature10600.nature10600[pii]. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 414.Petersen B, Negri S, Milan M, Reyff Z, Ballard C, Ihuoma J, Ungvari Z, Tarantini S. Editorial: Endocrine regulation of aging: impacts of humoral factors and circulating mediators. Front Endocrinol (Lausanne). 2024;15:1387435. 10.3389/fendo.2024.1387435. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 415.Jett S, Malviya N, Schelbaum E, Jang G, Jahan E, Clancy K, Hristov H, Pahlajani S, Niotis K, Loeb-Zeitlin S, Havryliuk Y, Isaacson R, Brinton RD, Mosconi L. Endogenous and exogenous estrogen exposures: how women’s reproductive health can drive brain aging and inform Alzheimer’s prevention. Front Aging Neurosci. 2022;14:831807. 10.3389/fnagi.2022.831807. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 416.Said SA, Isedowo R, Guerin C, Nar NN, Lillie L, Bukovac S, Simone JJ, Green MR, McCormick CM, Stuart JA. Effects of long-term dietary administration of estrogen receptor-beta agonist diarylpropionitrile on ovariectomized female ICR (CD-1) mice. Geroscience. 2018;40:393–403. 10.1007/s11357-018-0038-7. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 417.Strong R, Miller RA, Antebi A, Astle CM, Bogue M, Denzel MS, Fernandez E, Flurkey K, Hamilton KL, Lamming DW, Javors MA, de Magalhaes JP, Martinez PA, McCord JM, Miller BF, Muller M, Nelson JF, Ndukum J, Rainger GE, Richardson A, Sabatini DM, Salmon AB, Simpkins JW, Steegenga WT, Nadon NL, Harrison DE. Longer lifespan in male mice treated with a weakly estrogenic agonist, an antioxidant, an alpha-glucosidase inhibitor or a Nrf2-inducer. Aging Cell. 2016;15:872–84. 10.1111/acel.12496. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 418.Labinskyy N, Csiszar A, Veress G, Stef G, Pacher P, Oroszi G, Wu J, Ungvari Z. Vascular dysfunction in aging: potential effects of resveratrol, an anti-inflammatory phytoestrogen. Curr Med Chem. 2006;13:989–96. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 419.Mann SN, Hadad N, Nelson Holte M, Rothman AR, Sathiaseelan R, Ali Mondal S, Agbaga MP, Unnikrishnan A, Subramaniam M, Hawse J, Huffman DM, Freeman WM, Stout MB. Health benefits attributed to 17alpha-estradiol, a lifespan-extending compound, are mediated through estrogen receptor alpha. Elife. 2020;9:e59616. 10.7554/eLife.59616. [DOI] [PMC free article] [PubMed]
  • 420.Choi CI, Lee YD, Gwag BJ, Cho SI, Kim SS, Suh-Kim H. Effects of estrogen on lifespan and motor functions in female hSOD1 G93A transgenic mice. J Neurol Sci. 2008;268:40–7. 10.1016/j.jns.2007.10.024. [DOI] [PubMed] [Google Scholar]
  • 421.Ashpole NM, Logan S, Yabluchanskiy A, Mitschelen MC, Yan H, Farley JA, Hodges EL, Ungvari Z, Csiszar A, Chen S, Georgescu C, Hubbard GB, Ikeno Y, Sonntag WE. IGF-1 has sexually dimorphic, pleiotropic, and time-dependent effects on healthspan, pathology, and lifespan. Geroscience. 2017;39:129–45. 10.1007/s11357-017-9971-0. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 422.Ashpole NM, Herron JC, Mitschelen MC, Farley JA, Logan S, Yan H, Ungvari Z, Hodges EL, Csiszar A, Ikeno Y, Humphrey MB, Sonntag WE. IGF-1 regulates vertebral bone aging through sex-specific and time-dependent mechanisms. J Bone Miner Res. 2015. 10.1002/jbmr.2689. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 423.Wennberg AMV, Hagen CE, Petersen RC, Mielke MM. Trajectories of plasma IGF-1, IGFBP-3, and their ratio in the Mayo Clinic Study of Aging. Exp Gerontol. 2018;106:67–73. 10.1016/j.exger.2018.02.015. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 424.Hochberg MC, Lethbridge-Cejku M, Scott WW Jr, Reichle R, Plato CC, Tobin JD. Serum levels of insulin-like growth factor in subjects with osteoarthritis of the knee. Data from the Baltimore Longitudinal Study of Aging. Arthritis Rheum. 1994;37:1177–80. [DOI] [PubMed] [Google Scholar]
  • 425.Fazekas-Pongor V, Peterfi A, Major D, Szarvas Z, Fekete M, Tabak AG, Csiszar A, Sonntag WE, Austad SN, Ungvari ZI. Decreased lifespan in female “Munchkin” actors from the cast of the 1939 film version of The Wizard of Oz does not support the hypothesis linking hypopituitary dwarfism to longevity. Geroscience. 2022;44:2527–39. 10.1007/s11357-022-00680-7. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 426.Strong R, Miller RA, Astle CM, Baur JA, de Cabo R, Fernandez E, Guo W, Javors M, Kirkland JL, Nelson JF, Sinclair DA, Teter B, Williams D, Zaveri N, Nadon NL, Harrison DE. Evaluation of resveratrol, green tea extract, curcumin, oxaloacetic acid, and medium-chain triglyceride oil on life span of genetically heterogeneous mice. J Gerontol A Biol Sci Med Sci. 2013;68:6–16. 10.1093/gerona/gls070. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 427.Strong R, Miller RA, Astle CM, Floyd RA, Flurkey K, Hensley KL, Javors MA, Leeuwenburgh C, Nelson JF, Ongini E, Nadon NL, Warner HR, Harrison DE. Nordihydroguaiaretic acid and aspirin increase lifespan of genetically heterogeneous male mice. Aging Cell. 2008;7:641–50. 10.1111/j.1474-9726.2008.00414.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 428.Palmer AK, Xu M, Zhu Y, Pirtskhalava T, Weivoda MM, Hachfeld CM, Prata LG, van Dijk TH, Verkade E, Casaclang-Verzosa G, Johnson KO, Cubro H, Doornebal EJ, Ogrodnik M, Jurk D, Jensen MD, Chini EN, Miller JD, Matveyenko A, Stout MB, Schafer MJ, White TA, Hickson LJ, Demaria M, Garovic V, Grande J, Arriaga EA, Kuipers F, von Zglinicki T, LeBrasseur NK, et al. Targeting senescent cells alleviates obesity-induced metabolic dysfunction. Aging Cell. 2019;18:e12950. 10.1111/acel.12950. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 429.Miller RA, Harrison DE, Astle CM, Baur JA, Boyd AR, de Cabo R, Fernandez E, Flurkey K, Javors MA, Nelson JF, Orihuela CJ, Pletcher S, Sharp ZD, Sinclair D, Starnes JW, Wilkinson JE, Nadon NL, Strong R. Rapamycin, but not resveratrol or simvastatin, extends life span of genetically heterogeneous mice. J Gerontol A Biol Sci Med Sci. 2011;66:191–201. 10.1093/gerona/glq178. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 430.Harrison DE, Strong R, Allison DB, Ames BN, Astle CM, Atamna H, Fernandez E, Flurkey K, Javors MA, Nadon NL, Nelson JF, Pletcher S, Simpkins JW, Smith D, Wilkinson JE, Miller RA. Acarbose, 17-alpha-estradiol, and nordihydroguaiaretic acid extend mouse lifespan preferentially in males. Aging Cell. 2014;13:273–82. 10.1111/acel.12170. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 431.Baker DJ, Childs BG, Durik M, Wijers ME, Sieben CJ, Zhong J, Saltness RA, Jeganathan KB, Verzosa GC, Pezeshki A, Khazaie K, Miller JD, van Deursen JM. Naturally occurring p16(Ink4a)-positive cells shorten healthy lifespan. Nature. 2016;530:184–9. 10.1038/nature16932. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 432.Salerno N, Marino F, Scalise M, Salerno L, Molinaro C, Filardo A, Chiefalo A, Panuccio G, De Angelis A, Urbanek K, Torella D, Cianflone E. Pharmacological clearance of senescent cells improves cardiac remodeling and function after myocardial infarction in female aged mice. Mech Ageing Dev. 2022;208:111740. 10.1016/j.mad.2022.111740. [DOI] [PubMed] [Google Scholar]
  • 433.Rani A, Bean L, Budamagunta V, Kumar A, Foster TC. Failure of senolytic treatment to prevent cognitive decline in a female rodent model of aging. Front Aging Neurosci. 2024;16:1384554. 10.3389/fnagi.2024.1384554. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 434.Ng M, Hazrati LN. Evidence of sex differences in cellular senescence. Neurobiol Aging. 2022;120:88–104. 10.1016/j.neurobiolaging.2022.08.014. [DOI] [PubMed] [Google Scholar]
  • 435.Nickel K, Zhu L, Mangalindan R, Snyder JM, Tucker M, Whitson J, Sweetwyne M, Valencia AP, Klug J, Jiang Z, Marcinek DJ, Rabinovitch P, Ladiges W. Long-term treatment with Elamipretide enhances healthy aging phenotypes in mice. Aging Pathobiol Ther. 2022;4:76–83. 10.31491/apt.2022.09.089. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 436.Seals DR, Nagy EE, Moreau KL. Aerobic exercise training and vascular function with ageing in healthy men and women. J Physiol. 2019;597:4901–14. 10.1113/JP277764. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 437.Torrens-Mas M, Navas-Enamorado C, Wahl D, Sanchez-Polo A, Picca A, Oliver J, Roca P, Gonzalez-Freire M. Sex specific differences in response to calorie restriction in skeletal muscle of young rats. Nutrients. 2022;14:4535. 10.3390/nu14214535. [DOI] [PMC free article] [PubMed]
  • 438.Zhao M, Vaartjes I, Graham I, Grobbee D, Spiering W, Klipstein-Grobusch K, Woodward M, Peters SA. Sex differences in risk factor management of coronary heart disease across three regions. Heart. 2017;103:1587–94. 10.1136/heartjnl-2017-311429. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 439.Barha CK, Liu-Ambrose T. Sex differences in exercise efficacy: is midlife a critical window for promoting healthy cognitive aging? FASEB J. 2020;34:11329–36. 10.1096/fj.202000857R. [DOI] [PubMed] [Google Scholar]
  • 440.Sindi S, Kareholt I, Ngandu T, Rosenberg A, Kulmala J, Johansson L, Wetterberg H, Skoog J, Sjoberg L, Wang HX, Fratiglioni L, Skoog I, Kivipelto M. Sex differences in dementia and response to a lifestyle intervention: evidence from Nordic population-based studies and a prevention trial. Alzheimers Dement. 2021;17:1166–78. 10.1002/alz.12279. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 441.Bailey RL, Dog TL, Smith-Ryan AE, Das SK, Baker FC, Madak-Erdogan Z, Hammond BR, Sesso HD, Eapen A, Mitmesser SH, Wong A, Nguyen H. Sex differences across the life course: a focus on unique nutritional and health considerations among women. J Nutr. 2022;152:1597–610. 10.1093/jn/nxac059. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 442.Osmanagic-Myers S, Kiss A, Manakanatas C, Hamza O, Sedlmayer F, Szabo PL, Fischer I, Fichtinger P, Podesser BK, Eriksson M, Foisner R. Endothelial progerin expression causes cardiovascular pathology through an impaired mechanoresponse. J Clin Invest. 2019;129:531–45. 10.1172/JCI121297. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 443.Nam Y, White M, Karlsson EK, Creevy KE, Promislow DEL, McClelland RL, Dog Aging Project C. Dog size and patterns of disease history across the canine age spectrum: results from the Dog Aging Project. PLoS One. 2024;19:e0295840. 10.1371/journal.pone.0295840. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 444.Urfer SR, Kaeberlein M, Promislow DEL, Creevy KE. Lifespan of companion dogs seen in three independent primary care veterinary clinics in the United States. Canine Med Genet. 2020;7:7. 10.1186/s40575-020-00086-8. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 445.Fekete M, Szarvas Z, Fazekas-Pongor V, Feher A, Csipo T, Forrai J, Dosa N, Peterfi A, Lehoczki A, Tarantini S, Varga JT. Nutrition strategies promoting healthy aging: from improvement of cardiovascular and brain health to prevention of age-associated diseases. Nutrients. 2022;15:47. 10.3390/nu15010047. [DOI] [PMC free article] [PubMed]
  • 446.Fekete M, Lehoczki A, Major D, Fazekas-Pongor V, Csipo T, Tarantini S, Csizmadia Z, Varga JT. Exploring the influence of gut-brain axis modulation on cognitive health: a comprehensive review of prebiotics, probiotics, and symbiotics. Nutrients. 2024;16:789. 10.3390/nu16060789. [DOI] [PMC free article] [PubMed]
  • 447.Fekete M, Lehoczki A, Tarantini S, Fazekas-Pongor V, Csipo T, Csizmadia Z, Varga JT. Improving cognitive function with nutritional supplements in aging: a comprehensive narrative review of clinical studies investigating the effects of vitamins, minerals, antioxidants, and other dietary supplements. Nutrients. 2023;15:5116. 10.3390/nu15245116. [DOI] [PMC free article] [PubMed]
  • 448.Bonnes SLR, Strauss T, Palmer AK, Hurt RT, Island L, Goshen A, Wang LYT, Kirkland JL, Bischof E, Maier AB. Establishing healthy longevity clinics in publicly funded hospitals. Geroscience. 2024;46:4217–23. 10.1007/s11357-024-01132-0. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 449.Selb J, Cvetko F, Deutsch L, Bedrac L, Kuscer E, Maier AB. Personalization matters: the effect of sex in multivitamin-multimineral-based cancer prevention. Geroscience. 2024;46:1351–6. 10.1007/s11357-023-00882-7. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 450.Freeze WM, Jacobs HIL, de Jong JJ, Verheggen ICM, Gronenschild E, Palm WM, Hoff EI, Wardlaw JM, Jansen JFA, Verhey FR, Backes WH. White matter hyperintensities mediate the association between blood-brain barrier leakage and information processing speed. Neurobiol Aging. 2020;85:113–22. 10.1016/j.neurobiolaging.2019.09.017. [DOI] [PubMed] [Google Scholar]
  • 451.van Dinther M, Schram MT, Jansen JFA, Backes WH, Houben A, Berendschot T, Schalkwijk CG, Stehouwer CDA, van Oostenbrugge RJ, Staals J. Extracerebral microvascular dysfunction is related to brain MRI markers of cerebral small vessel disease: the Maastricht study. Geroscience. 2022;44:147–57. 10.1007/s11357-021-00493-0. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 452.Russell SJ, Parker K, Lehoczki A, Lieberman D, Partha IS, Scott SJ, Phillips LR, Fain MJ, Nikolich JZ. Post-acute sequelae of SARS-CoV-2 infection (Long COVID) in older adults. Geroscience. 2024. 10.1007/s11357-024-01227-8. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 453.Molnar T, Lehoczki A, Fekete M, Varnai R, Zavori L, Erdo-Bonyar S, Simon D, Berki T, Csecsei P, Ezer E. Mitochondrial dysfunction in long COVID: mechanisms, consequences, and potential therapeutic approaches. Geroscience. 2024. 10.1007/s11357-024-01165-5. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 454.Kuerec AH, Wang W, Yi L, Tao R, Lin Z, Vaidya A, Pendse S, Thasma S, Andhalkar N, Avhad G, Kumbhar V, Maier AB. Towards personalized nicotinamide mononucleotide (NMN) supplementation: nicotinamide adenine dinucleotide (NAD) concentration. Mech Ageing Dev. 2024;218:111917. 10.1016/j.mad.2024.111917. [DOI] [PubMed] [Google Scholar]
  • 455.Bonnes SLR, Strauss T, Palmer AK, Hurt RT, Island L, Goshen A, Wang LYT, Kirkland JL, Bischof E, Maier AB. Establishing healthy longevity clinics in publicly funded hospitals. Geroscience. 2024. 10.1007/s11357-024-01132-0. [DOI] [PMC free article] [PubMed] [Google Scholar]

Articles from GeroScience are provided here courtesy of Springer

RESOURCES