Next Article in Journal
Genome-Wide Identification of B-Box Gene Family and Candidate Light-Related Member Analysis of Tung Tree (Vernicia fordii)
Previous Article in Journal
Do DEEPER ROOTING 1 Homologs Regulate the Lateral Root Slope Angle in Cucumber (Cucumis sativus)?
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Endothelial Senescence: From Macro- to Micro-Vasculature and Its Implications on Cardiovascular Health

by
Peichun Wang
1,2,
Daniels Konja
1,2,
Sandeep Singh
1,2,
Beijia Zhang
1,2 and
Yu Wang
1,2,*
1
State Key Laboratory of Pharmaceutical Biotechnology, The University of Hong Kong, Hong Kong SAR, China
2
Department of Pharmacology and Pharmacy, LKS Faculty of Medicine, The University of Hong Kong, Hong Kong SAR, China
*
Author to whom correspondence should be addressed.
Int. J. Mol. Sci. 2024, 25(4), 1978; https://doi.org/10.3390/ijms25041978
Submission received: 27 December 2023 / Revised: 2 February 2024 / Accepted: 3 February 2024 / Published: 6 February 2024
(This article belongs to the Special Issue Novel Insight into Microcirculation and Microvascular Diseases)

Abstract

:
Endothelial cells line at the most inner layer of blood vessels. They act to control hemostasis, arterial tone/reactivity, wound healing, tissue oxygen, and nutrient supply. With age, endothelial cells become senescent, characterized by reduced regeneration capacity, inflammation, and abnormal secretory profile. Endothelial senescence represents one of the earliest features of arterial ageing and contributes to many age-related diseases. Compared to those in arteries and veins, endothelial cells of the microcirculation exhibit a greater extent of heterogeneity. Microcirculatory endothelial senescence leads to a declined capillary density, reduced angiogenic potentials, decreased blood flow, impaired barrier properties, and hypoperfusion in a tissue or organ-dependent manner. The heterogeneous phenotypes of microvascular endothelial cells in a particular vascular bed and across different tissues remain largely unknown. Accordingly, the mechanisms underlying macro- and micro-vascular endothelial senescence vary in different pathophysiological conditions, thus offering specific target(s) for therapeutic development of senolytic drugs.

1. Introduction

Cellular senescence is the cessation of cell division. Senescence is originally defined as the irreversible loss of proliferative potential in somatic cells, which enter a viable and metabolically active state of permanent growth arrest that is distinct from quiescence and terminal differentiation [1,2,3]. It is characterized by a number of distinct phenotypic changes, such as enlarged cell size, elevated senescence-associated beta-galactosidase (SA-β-gal) activity, formation of telomere-associated foci, and increased expression of cyclin-dependent kinase inhibitors p21 and p16 [4,5,6,7,8]. SA-β-gal activity is commonly used as a biomarker for cellular senescence. Lysosomal beta-galactosidase activity is ordinarily detected at low pH (about pH 4) but becomes detectable at a higher pH level (pH 6) in senescent cells due to significant expansion of the lysosomal compartment [9]. Cellular senescence occurs in not only mitotic, but also postmitotic cells, referred to as the replicative and premature types, respectively [10,11]. Premature senescence occurs upon exposure to various stress conditions, including DNA damage [12], reactive oxygen species (ROS) [13], oncogene activation [12,14], telomere attrition [15,16,17], and metabolic dysregulation [18]. Accumulation of senescent cells contributes to age-related tissue degeneration by developing a complex senescence-associated secretory phenotype (SASP) [19,20,21,22,23,24]. By secreting a plethora of factors, including pro-inflammatory cytokines, chemokines, growth modulators, matrix metalloproteinases, and compromised extracellular vesicles which represent senescence-associated phenotype, senescent cells reprogram the surrounding microenvironment and cause tissue damage, thus promoting ageing and the development of age-associated diseases [20,24,25,26,27,28,29]. Intervention experiments have proven that senescent cell accumulation is an important driver of age-associated functional decline, (multi-)morbidity, and mortality [8]. Systemic clearance of senescent cells delays ageing and extend lifespan [8]. Therapeutically targeting cellular senescence, known as senotherapy, to eliminate senescent cells or induce senolysis, represents a rapidly growing and promising strategy for the prevention and/or treatment of ageing-related diseases [8,30,31,32,33]. Targeting senescent cells can improve both health- and life-span in mice [34]. In both pre-clinical and clinical models of geriatric decline and chronic illnesses, the efficacy of senescent cell removal via apoptosis-inducing “senolytic” drugs or therapies that inhibit the senescence-associated secretory phenotype, SASP inhibitors, has been demonstrated. The present review focuses on the senescence of endothelial cells at different anatomic locations of the vasculature and the implications in macro- and micro-circulatory diseases. The prospect and challenges of anti-endothelial senolytic therapy will also be discussed.

2. Microvascular Circulation and CVD

The microvasculature is a broad term used to describe blood vessels with diameters of 100 µm or less. In the peripheral circulation, the cardiovascular system terminates into a network of micro-vessels such as arterioles, capillaries, venules, and other cellular components all of which function to meet the oxygen and nutrient requirements of cells and tissues [35,36]. In addition, these vessels play an important role in peripheral vascular resistance and blood pressure regulation, as well as contributing significantly to immune function by trafficking lymphocytes and leukocytes to target tissues. Thus, these vessels are usually in direct contact with cells of body tissues, making the microvasculature arguably the most important aspect of the cardiovascular system. Vessels that make up the microcirculation are almost entirely lined with endothelial cells which together with smooth muscle layer play an important role in regulating the tone of arterioles [36] via three mechanisms: myogenecity, metabolic control, and neuro-hormonal regulation [37]. Under physiological conditions, these three mechanisms ensure progression and maintenance of proper microcirculatory function. However, under conditions of disease, dysregulation of these control mechanisms results in microvascular dysfunction.

2.1. Ageing and Microvascular Dysfunction

Biological age is reflective of vascular age, and microvascular age is cardinal in the ageing process as it exerts immense influence on every tissue/organ of the body [38]. Thus, a decline in microvascular function is a hallmark of the biological ageing process [39]. Endothelial dysfunction is an indicator of microvascular ageing, and age-related endothelial dysfunction refers to an impairment in vasodilatation or reduction in the production of endothelium-derived nitric oxide, as a result of endothelial cell senescence [40]. As a result of decreased endothelial NO synthase expression and reduced NO production, inducible NO synthase (iNOS) and ROS levels increase significantly, leading to DNA damage and promotion of senescence and apoptosis [39,41]. In addition, aged vascular endothelium have reduced ability to release growth factors [42], reduced regenerative capacity [43], and impaired tissue angiogenesis [44]. Low-grade inflammation is characteristic of aged microvasculature which affects both the endothelial layer as well as the surrounding layers such as the perivascular adipose tissue. The low-grade inflammation further disrupts the homeostatic capacity of the microvasculature and promotes microvascular disease [38]. Decline in mitochondrial function, dynamics, and metabolic activity due to ageing is significantly implicated in many ageing-associated diseases. Mitochondrial dysfunction, classically referring to impaired electron transport and decreased ATP production, typically results in increased mitochondrial ROS (mROS) production [38]. Sustained increased mROS production leads to mitochondrial DNA damage and accelerated ageing. In the vascular bed, endothelial senescence and smooth muscle dysfunction are subsequent to mitochondrial dysfunction and increased mROS. Increasing evidence of the role of mitochondrial dysfunction and oxidative stress in accelerated ageing has opened new paradigms and offered insights into potential therapies for microvascular dysfunction. Thus, targeting oxidative stress in the microvascular bed may represent a promising therapeutic strategy to slow down endothelial senescence and accelerated vascular ageing.

2.2. Microvascular Dysfunction in Obesity

Evidence of microvascular dysfunction in obesity is abound, and these largely point to endothelial dysfunction in the microvasculature of obese subjects. For example, a human study using skin and resistance arteries obtained from obese patients demonstrated that arteries responded poorly to endothelium-dependent vasodilatory agents, including the all-important insulin-induced endothelium-dependent vasodilatation [37]. In addition, in animal models, rarefaction of capillary density and structural remodeling of micro-vessels have been demonstrated [37]. Thus, obesity alters microvascular structure and function, leading to breakdown on the regulation of vascular tone and blood pressure regulation. In the heart, for instance, obesity is detrimental to coronary microcirculation. Development of coronary artery disease as a result of structural and functional changes in the coronary vasculature is often seen among obese patients [45]. Studies have shown that myocardial ischemia often results from coronary microvascular abnormalities in obesity. Although there is controversy surrounding the reduced myocardial perfusion in obesity, there is general consensus of reduction in response to vasodilator agents by coronary resistance arteries. This may either contribute to cardiovascular disease risk or to obesity pathogenesis [45]. The mechanism by which obesity causes reduction in vasodilator activity in coronary micro-vessels, however, remains to be properly elucidated. In addition, increased adiposity around the heart and coronary arteries shifts the balance of adipokine secretion. Many studies have shown that adipokines such as adiponectin, leptin, and resistin have direct effects on the vasodilatory function of coronary arteries and arterioles on obesity. Whereas some of these studies have pointed to endothelial dysfunction and subsequent loss of nitric oxide production as a mediator of adipokine-induced coronary artery dysfunction in obesity, other studies point to increased ROS production in the adipocytes and coronary vasculature [46,47,48,49]. However, the exact mechanism by which adipokines induce increases ROS production still needs to be studied.

2.3. Microvascular Dysfunction in Diabetes Mellitus

Arteriosclerotic lesions are cardinal events in type-2 diabetes mellitus (T2DM) patients. In addition to this, risks of myocardial infarction and cerebrovascular accidents are more likely in T2DM patients compared to in non-diabetic patients [50]. Cardiac and vascular complications of diabetes, including micro- and macro-angiopathy, are the leading causes of death among T2DM patients [51,52]. Dysfunction in the coronary microvasculature and impaired vasodilatation are characteristic of T2DM. Humans with T2DM generally have impaired microvascular endothelial function, reduced NO, and enhanced ROS production [53,54]. There is a consensus that increased mitochondrial ROS production in endothelial cells of T2DM patients promotes microvascular damage [55]. Endothelial cells typically respond to hyperglycemia by increasing ROS production. Subsequently, ROS-induced DNA damage activates DNA repair enzyme poly[ADP ribose]polymerase 1, which initiates a cascade of events, eventually leading to onset and/or progression of vascular injury [56]. Importantly, while ECs in culture respond to hyperglycemia-induced ROS stress within short periods and animal models show microvascular injury within weeks of hyperglycemia, human T2DM subjects require many years to develop microvascular damage. The explanations of the long duration of hyperglycemic exposure leading to anatomical damage to the microvasculature is currently a subject of intense investigation [56].

2.4. Microvascular Dysfunction in COVID-19

CVD risk factors (age, sex, obesity, diabetes mellitus, and hypertension) are also documented risk factors for COVID-19 [57]. Microvascular endothelial cell damage is reportedly a hallmark of COVID-19 progression subsequent to pro-inflammatory cytokine release after host invasion and immune activation [58,59]. Concomitant damage to the microvascular endothelium leads to sepsis, a severe form endothelial dysfunction syndrome, leading to septic shock and death due to irreversible damage to the microcirculation [60]. COVID-19 patients typically have reduced ACE2 levels which result in reduced vasodilatory activity in the microvasculature. Decreased coronary flow reserve and increased microvascular resistance as a result of systemic inflammation in COVID-19 patients result in coronary microthrombi as a result of injuries to the coronary microvasculature [61,62].

3. Endothelial Senescence

The endothelium forms an integral layer at the luminal surface of blood vessel and participates in physiologic functions including wound healing, hemostasis, substance transfer, and vascular tone/reactivity regulation [63,64]. The turnover of endothelial cells has been generally considered very slow. However, in areas of bifurcations and branching points of the arteries, the haemodynamic forces of shear and stretch cause chronic injuries to the endothelium; thus, the rate of endothelial cell replication/regeneration is increased [65]. Endothelial regeneration is of pivotal importance for maintaining the integrity of endothelium, in turn promoting vascular repair and health [66]. Regenerating endothelial cells arise from sites adjacent to the injury and exhibit significant proliferative potential [67]. However, the regenerative response is significantly impaired with age in older subjects due to the development of endothelial senescence, which influences cell responses [68,69]. Endothelial progenitor cells (EPCs) mediate repair mechanisms for endothelial regeneration and maintenance, probably by directly differentiating into endothelial cells (ECs) and integrating into injured vessels, or act on cells and blood vessels by releasing paracrine substances [70]. Aging subjects showed negative effects on the EPC number and function, of which subsequently impair endothelial cell repair and regeneration in the aging vasculature [71,72,73,74], thus indicating that the EPCs are involved in endothelial senescence as well. “Senescence” refers to the finite capacity for division in normal diploid cells. Endothelial cell senescence can be attributed to a number of factors on vascular pathologies, especially oxidative stress and sustained cell replication [75]. From a molecular point of view, the telomere hypothesis explains the mechanism of cellular senescence. Telomeres are the physical ends of chromosomes, and the presence of telomerase, a specialized reverse transcriptase, is required for the synthesis of telomeric DNA. The telomere hypothesis suggests that cells lacking telomerase have shortened DNA, and senescence occurs when telomere strength reaches a critical point [76]. Unlike young endothelium, senescent endothelial cells do not respond to growth factors due to defective signaling pathways. With age, the accumulation of dysfunctional senescent cells and the loss of regenerative capacity together contribute to the loss of arterial function and integrity. Depending on different locations and pathophysiological conditions, senescent endothelial cells exhibit upregulation of p53/p21 or p16/retinoblastoma protein (Rb) pathways, markers associated with activation of the DNA damage response (DDR) (p38 mitogen-activated protein kinase (p38MAPK) and phosphorylated histone 2AX (γH2AX)) (Figure 1), senescence-associated heterochromatin foci, SASP, or increased SA-β-Gal activity [77,78,79].
Much of our current knowledge about the properties of senescent endothelial cells is based on experiments in cultured cells. Vascular endothelial cells retain both proliferative potential and a specialized phenotype in a culture dish [80,81,82]. Human umbilical vein ECs (HUVECs) cultured on collagen gels form confluent monolayers that bind silver at their intercellular border similar to cells in vivo [83]. There are intercellular junctional structures present, such as adherents and tight junctions. In contrast, HUVECs grown on plastic surfaces do not stain with silver. The silver-staining characteristic of endothelial monolayers is related to their in vitro maturation and senescence [83]. The endothelial-cell growth factor (ECGF) delays the premature senescence of HUVECs [84]. Typical endothelial cells isolated from aorta or inferior venae cavae are small, round to polygonally shaped with the diameter ranged from 50 to 70 microns and arranged uniformly [85]. Cultured endothelial cells have a finite life-span. Population doubling (PD) time varies as a function of the cell seeding density. After a certain number of PD, the endothelial subculture density rapidly decreases [86]. Addition of the fibroblast growth factor (FGF) shortens PD time but increases the replicative lifespan of endothelial cultures [87]. When the inoculated endothelial cells do not double after three weeks of frequent refeeding with fresh medium and serum, they are considered to be senescent [88]. As cells senesce, their average attachment area increases more than three folds, ranging from 100 to 200 microns in diameter (Figure 2). Giant senescent endothelial cells reach a diameter of more than 250 microns, with the mean areas as high as 3660 μm2 [85]. The protein content also increases dramatically, starting at about 400–600 pg/106 young to 2000–2400 pg/106 senescent endothelial cells. Senescent endothelial cells are usually multinucleated and show no incorporation of [3H] thymidine. The loss of proliferative capacity is accompanied by increased chromosomal aberrations, such as translocations, increased heterochromatin foci, and polyploidy and telomere damage, in senescence endothelial cells [89,90,91]. High-mobility group A (HMGA) proteins or heterochromatin markers, including HP1 and tri-methylated lysine 9 histone H3 (H3K9me3), are recognized as molecular markers of senescence-associated heterochromatin foci and are considered to indicate cellular senescence [92]. In large arteries, as chronological age advances, areas with high endothelial cell turnover are covered by clusters of senescent cells [65]. Senescent cells accumulate in the vessels of patients with atherosclerosis, hypertension, aneurysms, diabetes, and intimal hyperplasia [93]. Endothelial cells that have undergone replicative senescence show an increased size, polymorphic nuclei, flattening, and vacuolization. Compared to their normal counterparts, senescent endothelial cells show increased adhesion to the basement membrane and reduced capacity to align with the direction of blood flow and is causatively implicated in age-related inhibition of endothelium-dependent vasodilatation [94,95]. The presence of multinucleated endothelial cells in aortae in vivo has been confirmed by both scanning and transmission electron microscopy [96]. These senescent endothelial cells exist as colonies in the aortae from elderly subjects with intimal-thickened or advanced atherosclerotic lesions and contribute to further development of atherosclerotic lesions [93,97]. Senescence of the endothelium leads to pathological conditions such as impaired vasodilation, increased vascular stiffness, increased vascular permeability, micro-thrombosis, atherosclerosis, in-stent restenosis, hypertension, ischemic or hemorrhagic disorders, and inflammation [95,98,99,100,101]. However, age-dependent vascular dysfunction caused by senescent endothelial cells is location-specific and mechanism-dependent. The differential rates of vascular aging and diversified organ dysfunction caused by endothelial senescence in different locations will be discussed, particularly focusing on the microvasculature of the brain and the heart.

4. Microvascular Endothelial Senescence

Endothelial cells differentiate from a common precursor, angioblast, and present many common morphological and functional features [103]. During different stages of differentiation, they express the angiogenic growth factor receptor VEGFR2 (flk1/KDR), one of the receptors for VEGF, and PECAM-1, followed by Tie-2, Tie-1, and vascular endothelial cadherin. Thus, endothelial cells may exhibit intermediate phenotype(s) in vivo. Heterogeneity is a characteristic feature of endothelial cells [104,105]. For example, Weibel−Palade bodies (WPB) are organelles specifically present in endothelial cells to store the von Willebrand factor (VWF). However, there is a heterogeneous distribution of WPB along the vascular tree: absent from the thoracic aorta, rare in the abdominal aorta, present in myocardial capillaries, and numerous in the inferior vena cava and pulmonary artery [106]. The VWF labeling exhibits the same variation in its distribution along the vascular tree as for its storage organelle. Note that even in the same organ, the endothelia of large and small vessels, veins, and arteries exhibit a complex array of specialized functions and molecular signatures. In brain, the expression of VWF is increased in venous compared to arterial and capillary endothelial cells, whereas the major facilitator superfamily domain containing protein 2a (Mfsd2a) protein is highly abundant in capillary compared to arterial and venous endothelium [107]. The microvasculature is comprosed of arterioles, capillaries, and venules [108,109]. The microcirculation allows the delivery of oxygen and nutrients to meet the energetic demands of local tissues, mainly through regulation of vascular tone, angiogenesis, regulation of hemostasis, inflammation, and vascular permeability [55,110]. The tissue rather than the vessel type contributes to microvascular endothelial cell heterogeneity [111]. Depending on the context and the surrounding microenvironment, microvascular endothelial cells display remarkable heterogeneity and acquire organ-specific properties [112,113]. Endothelial cells from the microvasculature of different organs display specialized properties both in vivo and in vitro. Understanding the heterogeneity in microvascular endothelial senescence is of fundamental importance for developing targeted approaches to prevent age-associated functional decline in different organs.

4.1. Brain

The endothelial cells of the brain microvasculature serves as the interface between blood and the central nervous system (CNS), so-called blood−brain barrier (BBB) [114]. The BBB primarily consists of brain microvascular endothelial cells (BMECs) that are sealed together by tight junction proteins. Beyond the vagal afferents and choroid plexus, BMECs are the first CNS cell type exposed to the systemic harmful stimuli. BMECs are embedded in a basement membrane and surrounded by pericytes and astrocytes to restrict transcytosis. Endothelial cells of the BBB exhibit unique biochemical and morphological features such as tight intercellular junctions, few pinocytic vesicles, and no fenestra but abundant mitochondria [115]. The communications of BMECs with other cells of the neurovascular unit, such as pericytes, glial cells, and neurons, play a critical role in neurogenesis and maintenance of normal cognitive function. Age-induced alterations of the transcellular transport machinery and increased senescence in BMECs are potential underlying mechanisms of the endothelial dysfunction that causes the BBB breakdown [116]. With age, the BBB dysfunction is characterized by increased leakiness and impaired transport of molecules such as glucose, amyloid-beta peptide, and xenobiotics. There is an increased ratio of senescent endothelial cells (~10%) in the mouse cerebral microcirculation [117]. These changes at the BBB may be drivers of cognitive dysfunction during aging and may be further impaired in neurodegenerative diseases [118].
When compared to endothelial cells of the peripheral vasculature, tissue-nonspecific alkaline phosphatase (TNAP) activity is highly upregulated in BMECs and shows a continuous and uniform layer of distribution across the plasma membrane, thus being used as a histological marker to detect changes in cerebral microvessel function or morphology [119,120]. TNAP is absent in the endothelial cells of the liver sinusoids. However, skeletal endothelial cells contain a strong TNAP activity, the pattern of which is different from BMECs, showing discontinuous or irregularly scattered distribution across the plasma membrane [120]. BMECs also exhibit a typical perinuclear staining of factor VIII. Heparin is included in cultures to retard growth of factor VIII negative cell types (e.g., pericytes and smooth muscle cells). With passage, markers typically used to identify the BBB, including alkaline phosphatase and γ-glutamyl transpeptidase, declines and increases, respectively. While the permeability of BMECs does not change with passage, receptor-mediated transcytosis is altered [121]. Alterations in the specialized transport systems across the cerebral capillary lead to adverse changes in cerebral and neurotransmitter metabolism.
Cultured BMECs are frequently used to study the function of the BBB [122,123]. They develop confluent, contact-inhibited monolayers with a characteristic polygonal appearance [124]. Primary mouse BMECs become senescent (as defined by SA-β-gal staining, upregulation of p21, DNA damage marked by γH2A.X, and heterochromatin foci) after six complete PD. Senescence can also be induced in vitro through physical or chemical stressors, such as ionizing radiation, chemotherapeutics, and oxidative stress [125]. Upon induction of cellular senescence in culture, BMECs undergo cell cycle arrest and acquire SASP, characterized by increased secretion of pro-inflammatory mediators [126]. Note that senescence is a slow process, taking about three to seven days to develop in response to stressors. Senescent mouse BMECs show increased BBB leakage as evidenced by reduced transendothelial electrical resistance and increased permeability to albumin in comparison to non-senescent/low passage cells [118].
With advanced aging, there is an increased ratio of senescent endothelial cells in the cerebral microcirculation [117,127]. Compared to young animals, the percentage of senescent BMECs in old mouse brain increases from 5.23% to 10.06% as revealed by single-cell RNAseq [117]. The total protein in the brain endothelium has been shown to reduce with increasing age [128]. Senescence of cerebromicrovascular endothelial cells leads to cerebral blood flow dysregulation and the BBB disruption, promoting the pathogenesis of vascular cognitive impairment [117,129,130,131]. Since senescent cells elicit a secretory phenotype, senescence of BMECs within the neurovascular unit contributes to the inflammation associated with neurodegenerative diseases [132]. However, evaluation of senescence-associated beta-galactosidase (SA-β-gal) is not enough to consistently detect senescent endothelial cells within the brain tissue. This is because the BBB, in particular BMECs, primarily exist in a quiescent state, which shares some features of senescent cells, such as increased lysosome content/SA-β-gal activity [133,134]. Other senescence markers are notoriously non-specific to detect using antibodies. Multiple senescence biomarkers within the same cells are usually difficult to detect. As a result, there is an urgent need for novel approaches to identifying, quantifying and characterizing senescent BMECs. Furthermore, it is unknown if aging has specific or global effects on different subtypes of BMECs [111]. A deeper understanding of the heterogeneous nature of BMECs will provide cell type-specific vascular therapies targeting endothelial senescence for neurological disorders.

4.2. Heart

The most prevalent cardiovascular disease (CVD) is heart failure (HF), which has a high prevalence among the elderly (https://www-who-int.eproxy.lib.hku.hk/en/news-room/fact-sheets/detail/cardiovascular-diseases-(cvds); accessed on 3 September 2022). The prognosis of HF is unacceptably poor, and there is an urgent need to find better therapies for this condition. In particular, heart failure with preserved ejection fraction (HFpEF), which is characterized by diastolic dysfunction, altered ventricular relaxation and filling, increased stiffness, and concentric cardiac remodeling (fibrosis, inflammation, and hypertrophy), represents one of the most complex and prevalent cardiometabolic diseases in the aging population [135,136,137]. HFpEF leads to pressure, but not volume overload, and is present frequently with comorbidities, such as advanced age, obesity, diabetes, and hypertension [138]. Most currently available drugs for HF show little or no effect on mortality rates in patients with HFpEF, confirmed by numerous clinical trials. Instead, guidelines suggest pharmacological approaches to fluid removal, symptomatic relief (e.g., edema), and management of associated comorbidities (e.g., hypertension and chronic obstructive pulmonary disease (COPD)). Agents used include beta blockers and angiotensin-converting enzyme inhibitors (ACEIs’, angiotensin II receptor blockers (ARBs), mineralocorticoid receptor antagonists (MRAs), etc.) [139].
Vascular endothelial cells comprise ~60% of non-cardiomyocytes in the healthy heart and have a major impact on cardiac heath [140]. Based on the common markers PECAM1, CDH5, and VWF, a large-scale single-cell and single-nucleus RNA-seq study reveals 10 populations including arterial, capillary, capillary-like, venous, endocardial, and lymphatic endothelial cells in the heart [141]. The microvascular capillary represents 57.4% of all endothelial cells in the heart. With age or under pathological conditions, the portions of different cell types in the heart change on an individual basis [140,142,143]. Microvascular dysfunction, rarefaction, and chronic low-grade inflammation have been proposed to play major roles in HFpEF development [144,145,146]. Chronic systemic inflammation, in particular, affects other organs such as kidneys and lungs, leading to sodium retention and pulmonary hypertension, respectively. One of the major mechanisms of systemic inflammation causing HFpEF is that it leads to altered paracrine communication between endothelial cells and surrounding cardiomyocytes [147]. Endothelium-dependent coronary microvascular dysfunction, measured by basal myocardial blood flow or coronary flow reserve (CFR), is present in 29% of HFpEF patients [148]. Microvascular rarefaction increases coronary resistance and contributes to insufficient cardiac perfusion, leading to impairment of myocardial blood flow and oxygen delivery [149]. Patients with HFpEF show elevated inflammatory markers such as interleukin-1 type I receptor (IL-1R), tumor necrosis factor α (TNFα), C-reactive protein (CRP), vascular cell adhesion molecule-1 (VCAM-1), and IL-6 [150]. Coronary microvascular inflammation and stiffening lead to ventricular−vascular uncoupling [145].
Senescent endothelial cells are observed in failing hearts, particularly those with diastolic dysfunction, and contribute to microvascular information and cardiac remodeling [93,151,152,153]. Hutchinson−Gilford progeria syndrome (HGPS) is an accelerated aging syndrome associated with premature vascular disease and death due to heart attack and stroke. Endothelial cells (ECs) differentiated from induced pluripotent stem cells (iPSCs) derived from these patients exhibit senescent hallmarks including replication arrest, increased expression of inflammatory markers, DNA damage, and telomere erosion [154]. The expression of p53 is increased in hearts of patients with heart failure and plays a pathological role in causing cellular senescence [152]. Endothelial p53 signaling suppresses angiogenesis and promotes capillary rarefaction in the failing heart. Activation of p53 in microvascular endothelial cells of the left ventricle induces cardiac inflammation and remodeling [155]. Expression of p53 leads to elevated expression of intercellular adhesion molecule (ICAM)-1 by capillary endothelial cells, which enhances macrophage infiltration and cardiac inflammation. Endothelial p53 depletion improves capillary rarefaction and cardiac function while suppressing cardiac fibrosis and remodeling [156].
Female senescence-accelerated mice on a high-fat and high-salt diet develop HFpEF, characterized by diastolic dysfunction, left ventricular (LV) hypertrophy, left atrial dilatation, and interstitial fibrosis, with reduced exercise capacity and increased lung weight. Endothelial senescence contributes to HFpEF in these animals [157]. Aging seems to cause greater DNA damage and telomere dysfunction in vascular endothelial cells than in smooth muscle cells [158]. Excision repair cross-complementation group 1 (ERCC1), an endonuclease non-catalytic subunit, is an essential component in the DNA damage repairing system [101]. Genetic removal of ERCC1 selectively in endothelial cells causes impaired vasodilatation in coronary arteries and decreased cardiac output, but no microvascular inflammation in the heart [99]. Although genomic instability represents a common feature of aging, the accumulation of mutations varies substantially between tissues. Thus, apart from distinct mechanisms, different microenvironments surrounding senescent endothelial cells may also contribute to variations in the pace of vascular aging among individuals, as well as organs within the same human subject or animal model [159]. Regardless, the available evidence suggests that suppression of endothelial senescence represents a promising therapeutic option for HFpEF, although the inter- and intra-individual heterogeneity should be taken into consideration [160,161].

5. Endothelial Senescence and Human Diseases

Endothelial cells are the first group of cells to be directly affected at the onset of senescence due to their location in the vasculature. Senescence affects endothelial cells mainly by decrease in endothelium-associated vasodilators (nitric oxide, hydrogen sulfide, and endothelium-dependent hyperpolarizing factors) and an increase in endothelium-associated contractile factors (e.g., reactive oxygen species, endothelin-1, and angiotensin II). Physiologically, endothelial cells in the vasculature are in a constant process of damage and repair to maintain vascular homeostasis. To maintain this important function, vascular endothelial cells replenish themselves to remove damaged cells, when damage occurs. However, endothelial cell senescence significantly reduces the capacity of the vascular endothelium to self-repair, leading to overall decline in endothelial cell number and/or function. Accumulation of senescent endothelial cells in the vascular bed results in increased expression of senescence-associated secretory phenotypes (SASPs), which also results in the senescence of adjacent cells in the vasculature. Eventually, vascular rarefaction and dysfunction occurs as risk factors and/or progression of many human diseases [162].
Atherosclerosis is a complex, chronic inflammatory disease affecting large- and medium-sized arteries. Chief among the factors responsible for the pathogenesis of atherosclerosis (including inflammation, hyperlipidemia, and lipid peroxidation) is cellular senescence. Factors such as hyperglycemia and oxidative stress cause premature cell senescence due to reduction in telomerase activity and consequently telomere shortening, mitochondrial dysfunction, exacerbated ROS production, and DNA damage. Atherosclosis due to cellular senescence is a leading cause of cardiovascular disease-related morbidity and mortality. In the human vasculature, senescent endothelial cells and smooth muscle cells and macrophages are critically involved in atherosclerosis development [163,164]. Senescence-associated endothelial dysfunction is known to be the first stage of atherosclerosis development due to changes in the functional phenotype and/or secretome of senescent endothelial cells. Typically, this shift results in reduced production of vasodilatory factors such as nitric oxide, prostacyclin, hydrogen sulfide, and increased production of inflammatory and vasoconstrictive factors such as reactive oxygen species, endothelin-1, and angiotensin II. In addition, senescent endothelial cells have altered metabolic activities such as glycolysis, oxidative phosphorylation, and fatty acid oxidation. Altogether, these shifts in the functional and metabolic phenotypes of aged endothelial cells contribute significantly to vascular aging and/or atherosclerosis [93,165].
A shift in functional and secretory phenotypes of senescent endothelial cells favors excessive vasoconstriction and hypertension. Oxidative stress, basically referring to an imbalance between nitric oxide production and reactive oxygen species generation, is a key contributor to endothelial dysfunction. An increase in ROS inhibits eNOS mRNA expression and eNOS activity via activation of the PI3K/Ras/Akt/MAPK pathway. Reduction in nitric oxide bioavailability due to aged and dysfunctional endothelial cells, as well as inhibition by ROS, is a major contributor to endothelial dysfunction in aging models of both murine models and human subjects. Thus, the eventual outcome of senescence-associated endothelial dysfunction during aging is typically hypertension. Paradoxically, aging, as well as aging-associated hypertension and hypertension per se, either independently or collectively, impairs endothelial function, leading to atherosclerosis, resulting in cardiovascular and cerebrovascular outcomes [166].
The blood−brain barrier (BBB) is the physical barrier and regulator of transport of molecules between the brain and the rest of the body. Disruption of the BBB and uncoupling of the neuro-vasculature, especially in aged subjects, are hallmarks of cerebrovascular diseases and cognitive decline. Cerebral endothelial cells form the core of the BBB. Thus, senescence-associated dysfunctional cerebral endothelial cells are a direct cause of neuro-vascular disease and cognitive decline among the elderly. Changes to endothelial nitric oxide synthase (eNOS)/nitric oxide (NO) signaling, tight junctions, angiogenesis, and neuroinflammation in cerebral endothelial cells have the potential to drive cerebrovascular dysfunction. NO bioavailability is a crucial regulator of normal cerebral endothelial cell function and regulates cerebral blood flow in response to changes in cellular energy demands. Decreased NO bioavailability via decreased synthesis or accumulation of ROS leads to ineffective cerebral blood flow regulation and hypoperfusion of the brain and ultimately contributes to neuronal cell death and cognitive dysfunction. Decreased angiogenesis occurs with aging and in cerebrovascular disease, which can lead to hypoperfusion, impaired adaptation to hypoxia, compromised recovery to tissue damage, and exacerbated ischemic tissue injury [167].

6. Senolytic Therapy Targeting Endothelial Senescence

Accumulation of senescent endothelial cells leads to diseases associated with aging. Available evidence underpins a central role of endothelial senescence in vascular aging and suggests that targeting various senescent pathways in endothelial cells is a viable entry point to promote healthy aging. Different therapeutic strategies can be used to prevent endothelial senescence or eliminate senescent endothelial cells, thus enabling rejuvenation. Senescent endothelial cells exhibit specific functional abnormalities, such as loss of proteostasis and genomic stability, reduced nitric oxide (NO) production, increased expression of pro-inflammatory cytokines and oxidative stress, abnormal autophagic flux, mitochondrial dysfunction, and deregulation of sirtuins [7,168,169,170,171]. Experimental interventions specifically targeting senescent endothelial cells and vascular aging have shown promising results in model organisms. Telomere uncapping or telomere shortening, for example, induces an early-onset cardiovascular aging phenotype in mice, characterized by premature myocardial hypertrophy, fibrosis, and diastolic dysfunction, as well as vascular oxidative stress, endothelial dysfunction, and increased blood pressure [172,173]. Overexpression of human telomerase reverse transcriptase (hTERT) in HGPS endothelial cells extends telomere length, restores endothelial function and gene expression, increases sirtuin 1 (SIRT1) expression, reduces markers of DNA damage and extends life span [174,175]. Similarly, targeting the vascular endothelial growth factor (VEGF) signaling prevents age-related microvascular attrition and effectively delays age-related pathologies across various organ systems, resulting in a prolonged lifespan of mice [176]. Endothelial overexpression of the longevity regulator SIRT1 attenuates age-related decline in endothelium-dependent vasodilatation, hypertension, atherosclerosis, and various organ dysfunctions [177,178,179,180].
Vascular endothelial cells, particularly those in the cerebral microcirculation, are exposed to senescence-inducing stimuli such as biochemical and hemodynamic factors, as well as numerous senolytic drugs. Long-term treatment with senolytic drugs, dasatinib (a Src/tyrosine kinase inhibitor)/quercetin (a natural flavonoid that binds to BCL-2 and BCL-XL) delays vascular aging as indicated by improved endothelium-dependent vasodilatation, as well as reduced aortic calcification and osteogenic signaling in hypercholesterolemic mice [181]. Dasatinib eliminates senescent human fat cell progenitors, while quercetin is more effective against senescent human endothelial cells. The combination treatment can slow endothelial cell senescence in humans [182]. Senescent endothelial cells are particularly susceptible to apoptosis induced by ABT263/Navitoclax, an experimental orally active anti-cancer drug which inhibits the apoptosis regulator proteins Bcl-2 and Bcl-XL [183]. Treatment with ABT263/Navitoclax enhances neurovascular coupling and improves hippocampus-encoded functions of learning and memory in aged mice [184]. Similar improvements in cognitive function have been observed in elderly INK-ATTAC mice treated with AP20187, a drug that eliminates p16Ink4a-positive senescent cells, or the senolytic treatment combination dasatinib/quercetin [185]. Senescent cell burden can be safely reduced in a clinical context [186]. Aerobic exercise prevents endothelial senescence and protects against endothelial dysfunction in older adults [187]. In fact, the effects of cardiac glycosides are partially attributable to their senolytic activity [188,189,190]. On the other hand, rapamycin, a macrolide antibiotic, inhibits the mammalian target of rapamycin (mTOR), a protein kinase which nucleates two distinct signaling complexes, known as mTORC1 and mTORC2 [191]. Rapamycin suppresses the mammalian TORC1 complex, which regulates translation, and extends lifespan in diverse species, including mice [192]. Treatment of rapamycin has persistent effects that can robustly delay aging, influence cancer prevalence and enhance longevity and healthspan [193,194]. Dietary intake of rapamycin was shown to reverse age-related vascular dysfunction and oxidative stress, in association with reduced arterial expression of the senescence marker p19 [92]. Rapamycin was also found to decrease IL6 and other cytokine mRNA levels while selectively suppressing translation of the membrane-bound cytokine IL1A. Reduced IL1A diminishes NF-κB transcriptional activity, which regulates much of the SASP; exogenous IL1A restores IL6 secretion to rapamycin-treated cells [195].
Considering that senescent cells take up to seven days or longer to accumulate and develop SASP, most therapeutics are adopting a “hit-and-run” strategy that does not require daily or weekly administration. The first generation of senolytic drugs was developed, aiming to disrupt the senescent cell anti-apoptotic pathways (SCAPs) and other pro-survival signaling network [182,196,197,198,199]. Currently, senolytics have expanded and include a broad range of senescent features as targets. However, these senolytic strategies may elicit off-target effects or interfere with beneficial populations [200,201,202]. There is an urgent demand for innovative senolytic drugs devoid of side effects for clinical applications. In general, the phenotype of endothelial senescence depends on the context of stress and the surrounding microenvironment, thus varied by cell location and tissue of origin and/or residence. As a result, the functional impact of senolytic therapy is influenced by how and where senescence is induced. For example, treatment with the BCL-2/BCL-xL inhibitor senolytic drug ABT263/Navitoclax improves functional hyperemia in the brain of aged mice but has no effects on endothelium-dependent acetylcholine-induced relaxation of aorta rings [184]. The differential effects of senolytic treatment may be attributable to the presence of more senescent endothelial cells in the aged cerebral microvasculature than in the conduit arteries [203]. Thus, organ specificity needs to be considered for the treatment of diseases, such as HFpEF. In summary, since senescent cells are highly heterogeneous, targeted strategies are needed that ideally preserve senescent cells in beneficial contexts while eliminating effects that are detrimental [204].

7. Challenges and Perspectives

Senescent endothelial cells promote the development of age-related disorders, including cardiometabolic and neurodegenerative diseases, whereas suppression of endothelial senescence ameliorates phenotypic features of aging in various models. However, the occurrence, location, function, and impact of senescent endothelial cells are very heterogeneous and depend on the tissue environment as well as the type and duration of stress/injury [205]. SASP also varies, depending on the type of endothelial cells, surrounding tissue, and triggers of senescence. Moreover, transient presence of senescent cells is beneficial for conditions such as normal development, wound healing, tissue regeneration, and cancer prevention [206]. Thus, targeted approaches are needed to preserve senescent cells in beneficial contexts while eliminating those that are detrimental. Specific depletion of senescent endothelial cells in a context-dependent manner may be a more promising strategy for the treatment of age-related diseases [207]. To this end, understanding the heterogeneity in both the molecular biology and the pathophysiological function of senescent endothelial cells facilitates the identification of specific biomarkers for targeted senolytic therapy [19,94,95,204,208]. Another issue remaining to be explored is the potential side effects of senolytic treatment [209]. Without a transcriptional analysis at the single-cell level, it is difficult to distinguish between different types of senescent endothelial cells. To identify the most suitable senolytic agents, optimize the dosage for administration and determine the combinations for treatment of various conditions, new mouse models to induce genetic elimination of specific senescent cell types or test cell type-specific senolytics should be generated. Potential gender differences are another critical consideration. The goal is to explore and develop senolytic agents that act on specific senescent mechanism(s) and types of endothelial cells or tissues for selective depletion of pathological senescent cells and reduce the “bystander effect” and “off-target toxicities”.

Funding

This work was supported by grants from Seeding Funds for Basic Research of the University of Hong Kong, the General Research Funds (17124718) and Collaborative Research Funds (C7037-17W) of Research Grant Council, the Areas of Excellence Scheme (AoE/M-707/18) of University Grants Committee, and Health and Medical Research Fund (08192306) of the Food and Health Bureau, The Government of the Hong Kong Special Administrative Region.

Conflicts of Interest

The authors declare no conflict of interest.

Abbreviations

BBBblood−brain barrier
BMECbrain microvascular endothelial cell
CNScentral nervous system
CVDcardiovascular diseases
EBSSEarle’s balanced salt solution
ECendothelial cell
ECGFendothelial-cell growth factor
EPCEndothelial progenitor cell
FBSfetal bovine serum
FGFfibroblast growth factor
HFHeart failure
HFpEFheart failure with preserved ejection fraction
HUVEChuman umbilical vein endothelial cells
ICAMintercellular adhesion molecule
ILinterleukin
NOnitric oxide
PECAMplatelet endothelial CAM
PSFpenicillin−streptomycin−fungizone
ROSreactive oxygen species
SASPsenescence-associated secretory phenotype
SCAPssenescent cell anti-apoptotic pathways
T2DMtype-2 diabetes mellitus
TNAPtissue-nonspecific alkaline phosphatase
VCAMvascular CAM
VEGFvascular endothelial growth factor
VWFvon Willebrand factor

References

  1. Bergmann, K. Handbook of the biology of aging: Edited by Caleb E. Finch and Leonard Hayflick. Van Nostrand Reinhold, New York, London, Toronto, Melbourne. pp. 771. Price £27.15. J. Psychosom. Res. 1979, 23, 226. [Google Scholar] [CrossRef]
  2. Hayflick, L. The Limited In Vitro Lifetime of Human Diploid Cell Strains. Exp. Cell Res. 1965, 37, 614–636. [Google Scholar] [CrossRef]
  3. Hayflick, L.; Moorhead, P.S. The serial cultivation of human diploid cell strains. Exp. Cell Res. 1961, 25, 585–621. [Google Scholar] [CrossRef]
  4. Liu, J.-Y.; Souroullas, G.P.; Diekman, B.O.; Krishnamurthy, J.; Hall, B.M.; Sorrentino, J.A.; Parker, J.S.; Sessions, G.A.; Gudkov, A.V.; Sharpless, N.E. Cells exhibiting strong p16INK4a promoter activation in vivo display features of senescence. Proc. Natl. Acad. Sci. USA 2019, 116, 2603–2611. [Google Scholar] [CrossRef]
  5. Herbig, U.; Ferreira, M.; Condel, L.; Carey, D.; Sedivy, J.M. Cellular senescence in aging primates. Science 2006, 311, 1257. [Google Scholar] [CrossRef]
  6. Campisi, J.; d’Adda di Fagagna, F. Cellular senescence: When bad things happen to good cells. Nat. Rev. Mol. Cell Biol. 2007, 8, 729–740. [Google Scholar] [CrossRef] [PubMed]
  7. Hernandez-Segura, A.; Nehme, J.; Demaria, M. Hallmarks of Cellular Senescence. Trends Cell Biol. 2018, 28, 436–453. [Google Scholar] [CrossRef] [PubMed]
  8. Baker, D.J.; Childs, B.G.; Durik, M.; Wijers, M.E.; Sieben, C.J.; Zhong, J.; Saltness, R.A.; Jeganathan, K.B.; Verzosa, G.C.; Pezeshki, A.; et al. Naturally occurring p16(Ink4a)-positive cells shorten healthy lifespan. Nature 2016, 530, 184–189. [Google Scholar] [CrossRef] [PubMed]
  9. Dimri, G.P.; Lee, X.; Basile, G.; Acosta, M.; Scott, G.; Roskelley, C.; Medrano, E.E.; Linskens, M.; Rubelj, I.; Pereira-Smith, O. A biomarker that identifies senescent human cells in culture and in aging skin in vivo. Proc. Natl. Acad. Sci. USA 1995, 92, 9363–9367. [Google Scholar] [CrossRef] [PubMed]
  10. Sapieha, P.; Mallette, F.A. Cellular Senescence in Postmitotic Cells: Beyond Growth Arrest. Trends Cell Biol. 2018, 28, 595–607. [Google Scholar] [CrossRef]
  11. von Zglinicki, T.; Wan, T.; Miwa, S. Senescence in Post-Mitotic Cells: A Driver of Aging? Antioxid. Redox Signal. 2021, 34, 308–323. [Google Scholar] [CrossRef] [PubMed]
  12. Di Micco, R.; Fumagalli, M.; Cicalese, A.; Piccinin, S.; Gasparini, P.; Luise, C.; Schurra, C.; Garre, M.; Nuciforo, P.G.; Bensimon, A.; et al. Oncogene-induced senescence is a DNA damage response triggered by DNA hyper-replication. Nature 2006, 444, 638–642. [Google Scholar] [CrossRef] [PubMed]
  13. Passos, J.F.; Nelson, G.; Wang, C.; Richter, T.; Simillion, C.; Proctor, C.J.; Miwa, S.; Olijslagers, S.; Hallinan, J.; Wipat, A.; et al. Feedback between p21 and reactive oxygen production is necessary for cell senescence. Mol. Syst. Biol. 2010, 6, 347. [Google Scholar] [CrossRef] [PubMed]
  14. Serrano, M.; Lin, A.W.; McCurrach, M.E.; Beach, D.; Lowe, S.W. Oncogenic ras provokes premature cell senescence associated with accumulation of p53 and p16INK4a. Cell 1997, 88, 593–602. [Google Scholar] [CrossRef] [PubMed]
  15. Sedivy, J.M. Can ends justify the means?: Telomeres and the mechanisms of replicative senescence and immortalization in mammalian cells. Proc. Natl. Acad. Sci. USA 1998, 95, 9078–9081. [Google Scholar] [CrossRef] [PubMed]
  16. Jurk, D.; Wilson, C.; Passos, J.F.; Oakley, F.; Correia-Melo, C.; Greaves, L.; Saretzki, G.; Fox, C.; Lawless, C.; Anderson, R.; et al. Chronic inflammation induces telomere dysfunction and accelerates ageing in mice. Nat. Commun. 2014, 2, 4172. [Google Scholar] [CrossRef]
  17. Shay, J.W.; Wright, W.E. Telomeres and telomerase: Three decades of progress. Nat. Rev. Genet. 2019, 20, 299–309. [Google Scholar] [CrossRef]
  18. Carroll, B.; Nelson, G.; Rabanal-Ruiz, Y.; Kucheryavenko, O.; Dunhill-Turner, N.A.; Chesterman, C.C.; Zahari, Q.; Zhang, T.; Conduit, S.E.; Mitchell, C.A.; et al. Persistent mTORC1 signaling in cell senescence results from defects in amino acid and growth factor sensing. J. Cell Biol. 2017, 216, 1949–1957. [Google Scholar] [CrossRef]
  19. Gorgoulis, V.; Adams, P.D.; Alimonti, A.; Bennett, D.C.; Bischof, O.; Bishop, C.; Campisi, J.; Collado, M.; Evangelou, K.; Ferbeyre, G.; et al. Cellular Senescence: Defining a Path Forward. Cell 2019, 179, 813–827. [Google Scholar] [CrossRef]
  20. Acosta, J.C.; Banito, A.; Wuestefeld, T.; Georgilis, A.; Janich, P.; Morton, J.P.; Athineos, D.; Kang, T.-W.; Lasitschka, F.; Andrulis, M.; et al. A complex secretory program orchestrated by the inflammasome controls paracrine senescence. Nat. Cell Biol. 2013, 15, 978–990. [Google Scholar] [CrossRef]
  21. De Cecco, M.; Ito, T.; Petrashen, A.P.; Elias, A.E.; Skvir, N.J.; Criscione, S.W.; Caligiana, A.; Brocculi, G.; Adney, E.M.; Boeke, J.D.; et al. L1 drives IFN in senescent cells and promotes age-associated inflammation. Nature 2019, 566, 73–78. [Google Scholar] [CrossRef] [PubMed]
  22. Ogrodnik, M.; Zhu, Y.; Langhi, L.G.P.; Tchkonia, T.; Krüger, P.; Fielder, E.; Victorelli, S.; Ruswhandi, R.A.; Giorgadze, N.; Pirtskhalava, T.; et al. Obesity-Induced Cellular Senescence Drives Anxiety and Impairs Neurogenesis. Cell Metab. 2019, 29, 1061–1077. [Google Scholar] [CrossRef] [PubMed]
  23. Biran, A.; Zada, L.; Abou Karam, P.; Vadai, E.; Roitman, L.; Ovadya, Y.; Porat, Z.; Krizhanovsky, V. Quantitative identification of senescent cells in aging and disease. Aging Cell 2017, 16, 661–671. [Google Scholar] [CrossRef] [PubMed]
  24. Milanovic, M.; Fan, D.N.Y.; Belenki, D.; Däbritz, J.H.M.; Zhao, Z.; Yu, Y.; Dörr, J.R.; Dimitrova, L.; Lenze, D.; Monteiro Barbosa, I.A.; et al. Senescence-associated reprogramming promotes cancer stemness. Nature 2018, 553, 96–100. [Google Scholar] [CrossRef] [PubMed]
  25. Sorokina, A.G.; Orlova, Y.A.; Grigorieva, O.A.; Novoseletskaya, E.S.; Basalova, N.A.; Alexandrushkina, N.A.; Vigovskiy, M.A.; Kirillova, K.I.; Balatsky, A.V.; Samokhodskaya, L.M.; et al. Correlations between biomarkers of senescent cell accumulation at the systemic, tissue and cellular levels in elderly patients. Exp. Gerontol. 2023, 177, 112176. [Google Scholar] [CrossRef] [PubMed]
  26. Ogrodnik, M.; Salmonowicz, H.; Gladyshev, V.N. Integrating cellular senescence with the concept of damage accumulation in aging: Relevance for clearance of senescent cells. Aging Cell 2019, 18, e12841. [Google Scholar] [CrossRef]
  27. Biran, A.; Perelmutter, M.; Gal, H.; Burton, D.G.A.; Ovadya, Y.; Vadai, E.; Geiger, T.; Krizhanovsky, V. Senescent cells communicate via intercellular protein transfer. Genes Dev. 2015, 29, 791–802. [Google Scholar] [CrossRef]
  28. Terlecki-Zaniewicz, L.; Lämmermann, I.; Latreille, J.; Bobbili, M.R.; Pils, V.; Schosserer, M.; Weinmüllner, R.; Dellago, H.; Skalicky, S.; Pum, D.; et al. Small extracellular vesicles and their miRNA cargo are anti-apoptotic members of the senescence-associated secretory phenotype. Aging 2018, 10, 1103–1132. [Google Scholar] [CrossRef]
  29. Mosteiro, L.; Pantoja, C.; Alcazar, N.; Marión, R.M.; Chondronasiou, D.; Rovira, M.; Fernandez-Marcos, P.J.; Muñoz-Martin, M.; Blanco-Aparicio, C.; Pastor, J.; et al. Tissue damage and senescence provide critical signals for cellular reprogramming in vivo. Science 2016, 354, aaf4445. [Google Scholar] [CrossRef] [PubMed]
  30. Baker, D.J.; Wijshake, T.; Tchkonia, T.; LeBrasseur, N.K.; Childs, B.G.; van de Sluis, B.; Kirkland, J.L.; van Deursen, J.M. Clearance of p16Ink4a-positive senescent cells delays ageing-associated disorders. Nature 2011, 479, 232–236. [Google Scholar] [CrossRef] [PubMed]
  31. Childs, B.G.; Durik, M.; Baker, D.J.; van Deursen, J.M. Cellular senescence in aging and age-related disease: From mechanisms to therapy. Nat. Med. 2015, 21, 1424–1435. [Google Scholar] [CrossRef]
  32. He, S.; Sharpless, N.E. Senescence in Health and Disease. Cell 2017, 169, 1000–1011. [Google Scholar] [CrossRef]
  33. Moiseeva, V.; Cisneros, A.; Sica, V.; Deryagin, O.; Lai, Y.; Jung, S.; Andrés, E.; An, J.; Segalés, J.; Ortet, L.; et al. Senescence atlas reveals an aged-like inflamed niche that blunts muscle regeneration. Nature 2023, 613, 169–178. [Google Scholar] [CrossRef]
  34. Xu, M.; Pirtskhalava, T.; Farr, J.N.; Weigand, B.M.; Palmer, A.K.; Weivoda, M.M.; Inman, C.L.; Ogrodnik, M.B.; Hachfeld, C.M.; Fraser, D.G.; et al. Senolytics improve physical function and increase lifespan in old age. Nat. Med. 2018, 24, 1246–1256. [Google Scholar] [CrossRef]
  35. Beckman, J.A.; Duncan, M.S.; Damrauer, S.M.; Wells, Q.S.; Barnett, J.V.; Wasserman, D.H.; Bedimo, R.J.; Butt, A.A.; Marconi, V.C.; Sico, J.J.; et al. Microvascular Disease, Peripheral Artery Disease, and Amputation. Circulation 2019, 140, 449–458. [Google Scholar] [CrossRef]
  36. Guven, G.; Hilty, M.P.; Ince, C. Microcirculation: Physiology, Pathophysiology, and Clinical Application. Blood Purif. 2020, 49, 143–150. [Google Scholar] [CrossRef]
  37. Jonk, A.M.; Houben, A.J.H.M.; Jongh, R.T.d.; Serné, E.H.; Schaper, N.C.; Stehouwer, C.D.A. Microvascular Dysfunction in Obesity: A Potential Mechanism in the Pathogenesis of Obesity-Associated Insulin Resistance and Hypertension. Physiology 2007, 22, 252–260. [Google Scholar] [CrossRef] [PubMed]
  38. Mengozzi, A.; de Ciuceis, C.; Dell’oro, R.; Georgiopoulos, G.; Lazaridis, A.; Nosalski, R.; Pavlidis, G.; Tual-Chalot, S.; Agabiti-Rosei, C.; Anyfanti, P.; et al. The importance of microvascular inflammation in ageing and age-related diseases: A position paper from the ESH working group on small arteries, section of microvascular inflammation. J. Hypertens. 2023, 41, 1521–1543. [Google Scholar] [CrossRef] [PubMed]
  39. Scioli, M.G.; Bielli, A.; Arcuri, G.; Ferlosio, A.; Orlandi, A. Ageing and microvasculature. Vasc. Cell 2014, 6, 19. [Google Scholar] [CrossRef] [PubMed]
  40. Donato, A.J.; Machin, D.R.; Lesniewski, L.A. Mechanisms of Dysfunction in the Aging Vasculature and Role in Age-Related Disease. Circ. Res. 2018, 123, 825–848. [Google Scholar] [CrossRef] [PubMed]
  41. Tomasian, D.; Keaney, J.F.; Vita, J.A. Antioxidants and the bioactivity of endothelium-derived nitric oxide. Cardiovasc. Res. 2000, 47, 426–435. [Google Scholar] [CrossRef]
  42. Edelberg, J.M.; Tang, L.; Hattori, K.; Lyden, D.; Rafii, S. Young adult bone marrow-derived endothelial precursor cells restore aging-impaired cardiac angiogenic function. Circ. Res. 2002, 90, E89–E93. [Google Scholar] [CrossRef]
  43. Weidinger, F.F.; McLenachan, J.M.; Cybulsky, M.I.; Gordon, J.B.; Rennke, H.G.; Hollenberg, N.K.; Fallon, J.T.; Ganz, P.; Cooke, J.P. Persistent dysfunction of regenerated endothelium after balloon angioplasty of rabbit iliac artery. Circulation 1990, 81, 1667–1679. [Google Scholar] [CrossRef]
  44. Kang, D.H.; Anderson, S.; Kim, Y.G.; Mazzalli, M.; Suga, S.; Jefferson, J.A.; Gordon, K.L.; Oyama, T.T.; Hughes, J.; Hugo, C.; et al. Impaired angiogenesis in the aging kidney: Vascular endothelial growth factor and thrombospondin-1 in renal disease. Am. J. Kidney Dis. 2001, 37, 601–611. [Google Scholar] [CrossRef]
  45. Bagi, Z.; Broskova, Z.; Feher, A. Obesity and coronary microvascular disease—implications for adipose tissue-mediated remote inflammatory response. Curr. Vasc. Pharmacol. 2014, 12, 453–461. [Google Scholar] [CrossRef]
  46. Verma, S.; Li, S.H.; Wang, C.H.; Fedak, P.W.; Li, R.K.; Weisel, R.D.; Mickle, D.A. Resistin promotes endothelial cell activation: Further evidence of adipokine-endothelial interaction. Circulation 2003, 108, 736–740. [Google Scholar] [CrossRef]
  47. Wassmann, S.; Stumpf, M.; Strehlow, K.; Schmid, A.; Schieffer, B.; Böhm, M.; Nickenig, G. Interleukin-6 induces oxidative stress and endothelial dysfunction by overexpression of the angiotensin II type 1 receptor. Circ. Res. 2004, 94, 534–541. [Google Scholar] [CrossRef]
  48. Chen, C.; Jiang, J.; Lü, J.M.; Chai, H.; Wang, X.; Lin, P.H.; Yao, Q. Resistin decreases expression of endothelial nitric oxide synthase through oxidative stress in human coronary artery endothelial cells. Am. J. Physiol. Heart Circ. Physiol. 2010, 299, H193–H201. [Google Scholar] [CrossRef]
  49. Dick, G.M.; Katz, P.S.; Farias, M., 3rd; Morris, M.; James, J.; Knudson, J.D.; Tune, J.D. Resistin impairs endothelium-dependent dilation to bradykinin, but not acetylcholine, in the coronary circulation. Am. J. Physiol. Heart Circ. Physiol. 2006, 291, H2997–H3002. [Google Scholar] [CrossRef]
  50. Kibel, A.; Selthofer-Relatic, K.; Drenjancevic, I.; Bacun, T.; Bosnjak, I.; Kibel, D.; Gros, M. Coronary microvascular dysfunction in diabetes mellitus. J. Int. Med. Res. 2017, 45, 1901–1929. [Google Scholar] [CrossRef]
  51. Haffner, S.J.; Cassells, H. Hyperglycemia as a cardiovascular risk factor. Am. J. Med. 2003, 115 (Suppl. 8A), 6s–11s. [Google Scholar] [CrossRef]
  52. Brownlee, M. Biochemistry and molecular cell biology of diabetic complications. Nature 2001, 414, 813–820. [Google Scholar] [CrossRef]
  53. Koller, A.; Balasko, M.; Bagi, Z. Endothelial regulation of coronary microcirculation in health and cardiometabolic diseases. Intern. Emerg. Med. 2013, 8 (Suppl. 1), S51–S54. [Google Scholar] [CrossRef]
  54. Okon, E.B.; Chung, A.W.; Rauniyar, P.; Padilla, E.; Tejerina, T.; McManus, B.M.; Luo, H.; van Breemen, C. Compromised arterial function in human type 2 diabetic patients. Diabetes 2005, 54, 2415–2423. [Google Scholar] [CrossRef]
  55. Horton, W.B.; Barrett, E.J. Microvascular Dysfunction in Diabetes Mellitus and Cardiometabolic Disease. Endocr. Rev. 2021, 42, 29–55. [Google Scholar] [CrossRef]
  56. Shah, M.S.; Brownlee, M. Molecular and Cellular Mechanisms of Cardiovascular Disorders in Diabetes. Circ. Res. 2016, 118, 1808–1829. [Google Scholar] [CrossRef]
  57. Yin, J.; Wang, S.; Liu, Y.; Chen, J.; Li, D.; Xu, T. Coronary microvascular dysfunction pathophysiology in COVID-19. Microcirculation 2021, 28, e12718. [Google Scholar] [CrossRef]
  58. Carnevale, S.; Beretta, P.; Morbini, P. Direct endothelial damage and vasculitis due to SARS-CoV-2 in small bowel submucosa of COVID-19 patient with diarrhea. J. Med. Virol. 2021, 93, 61–63. [Google Scholar] [CrossRef]
  59. Varga, Z.; Flammer, A.J.; Steiger, P.; Haberecker, M.; Andermatt, R.; Zinkernagel, A.S.; Mehra, M.R.; Schuepbach, R.A.; Ruschitzka, F.; Moch, H. Endothelial cell infection and endotheliitis in COVID-19. Lancet 2020, 395, 1417–1418. [Google Scholar] [CrossRef]
  60. Guo, J.; Wei, X.; Li, Q.; Li, L.; Yang, Z.; Shi, Y.; Qin, Y.; Zhang, X.; Wang, X.; Zhi, X.; et al. Single-cell RNA analysis on ACE2 expression provides insights into SARS-CoV-2 potential entry into the bloodstream and heart injury. J. Cell Physiol. 2020, 235, 9884–9894. [Google Scholar] [CrossRef]
  61. Sławiński, G.; Lewicka, E. What should a cardiologist know about coronavirus disease 2019? Kardiol. Pol. 2020, 78, 278–283. [Google Scholar] [CrossRef]
  62. Long, M.; Huang, Z.; Zhuang, X.; Huang, Z.; Guo, Y.; Liao, X.; Luo, C. Association of Inflammation and Endothelial Dysfunction with Coronary Microvascular Resistance in Patients with Cardiac Syndrome X. Arq. Bras. Cardiol. 2017, 109, 397–403. [Google Scholar] [CrossRef]
  63. Deanfield, J.E.; Halcox, J.P.; Rabelink, T.J. Endothelial function and dysfunction: Testing and clinical relevance. Circulation 2007, 115, 1285–1295. [Google Scholar] [CrossRef]
  64. Alexander, Y.; Osto, E.; Schmidt-Trucksäss, A.; Shechter, M.; Trifunovic, D.; Duncker, D.J.; Aboyans, V.; Bäck, M.; Badimon, L.; Cosentino, F.; et al. Endothelial function in cardiovascular medicine: A consensus paper of the European Society of Cardiology Working Groups on Atherosclerosis and Vascular Biology, Aorta and Peripheral Vascular Diseases, Coronary Pathophysiology and Microcirculation, and Thrombosis. Cardiovasc. Res. 2021, 117, 29–42. [Google Scholar] [CrossRef]
  65. Erusalimsky, J.D.; Kurz, D.J. Cellular senescence in vivo: Its relevance in ageing and cardiovascular disease. Exp. Gerontol. 2005, 40, 634–642. [Google Scholar] [CrossRef]
  66. Itoh, Y.; Toriumi, H.; Yamada, S.; Hoshino, H.; Suzuki, N. Resident endothelial cells surrounding damaged arterial endothelium reendothelialize the lesion. Arterioscler. Thromb. Vasc. Biol. 2010, 30, 1725–1732. [Google Scholar] [CrossRef]
  67. McDonald, A.I.; Shirali, A.S.; Aragón, R.; Ma, F.; Hernandez, G.; Vaughn, D.A.; Mack, J.J.; Lim, T.Y.; Sunshine, H.; Zhao, P.; et al. Endothelial Regeneration of Large Vessels Is a Biphasic Process Driven by Local Cells with Distinct Proliferative Capacities. Cell Stem Cell 2018, 23, 210–225. [Google Scholar] [CrossRef]
  68. Lee, M.Y.K.; Cai, Y.; Wang, Y.; Liao, S.-Y.; Liu, Y.; Zhang, Y.; Bai, B.; Tse, H.-F.; Vanhoutte, P.M. Differential genomic changes caused by cholesterol- and PUFA-rich diets in regenerated porcine coronary endothelial cells. Physiol. Genom. 2012, 44, 551–561. [Google Scholar] [CrossRef]
  69. Kunz, J.; Keim, U. On the regeneration of aortic endothelium at different ages. Mech. Ageing Dev. 1975, 4, 361–369. [Google Scholar] [CrossRef]
  70. Yan, F.; Liu, X.; Ding, H.; Zhang, W. Paracrine mechanisms of endothelial progenitor cells in vascular repair. Acta Histochem. 2022, 124, 151833. [Google Scholar] [CrossRef]
  71. Tao, J.; Wang, Y.; Yang, Z.; Tu, C.; Xu, M.G.; Wang, J.M.; Wang, Q.; Zeng, Q.Y.; Chen, G.W.; Ma, H. A study of association between age-related circulating endothelial progenitor cells and arterial elasticity. Zhonghua Xin Xue Guan Bing Za Zhi 2005, 33, 347–350. [Google Scholar]
  72. Williamson, K.; Stringer, S.E.; Alexander, M.Y. Endothelial progenitor cells enter the aging arena. Front. Physiol. 2012, 3, 30. [Google Scholar] [CrossRef] [PubMed]
  73. Felice, F.; Barsotti, M.C.; Poredos, P.; Balbarini, A.; Di Stefano, R. Effect of aging on metabolic pathways in endothelial progenitor cells. Curr. Pharm. Des. 2013, 19, 2351–2365. [Google Scholar] [CrossRef] [PubMed]
  74. Van Craenenbroeck, A.H.; Van Craenenbroeck, E.M. Endothelial progenitor cells and cardiovascular risk: Does ageing trump all other factors? Ann. Transl. Med. 2016, 4, 553. [Google Scholar] [CrossRef] [PubMed]
  75. Erusalimsky, J.D.; Skene, C. Mechanisms of endothelial senescence. Exp. Physiol. 2009, 94, 299–304. [Google Scholar] [CrossRef] [PubMed]
  76. Erusalimsky, J.D. Vascular endothelial senescence: From mechanisms to pathophysiology. J. Appl. Physiol. 2009, 106, 326–332. [Google Scholar] [CrossRef] [PubMed]
  77. Aviv, H.; Khan, M.Y.; Skurnick, J.; Okuda, K.; Kimura, M.; Gardner, J.; Priolo, L.; Aviv, A. Age dependent aneuploidy and telomere length of the human vascular endothelium. Atherosclerosis 2001, 159, 281–287. [Google Scholar] [CrossRef] [PubMed]
  78. Fenton, M.; Barker, S.; Kurz, D.J.; Erusalimsky, J.D. Cellular senescence after single and repeated balloon catheter denudations of rabbit carotid arteries. Arterioscler. Thromb. Vasc. Biol. 2001, 21, 220–226. [Google Scholar] [CrossRef]
  79. van der Loo, B.; Fenton, M.J.; Erusalimsky, J.D. Cytochemical detection of a senescence-associated beta-galactosidase in endothelial and smooth muscle cells from human and rabbit blood vessels. Exp. Cell Res. 1998, 241, 309–315. [Google Scholar] [CrossRef]
  80. Gimbrone, M.A. Culture of vascular endothelium. Prog. Hemost. Thromb. 1976, 3, 1–28. [Google Scholar]
  81. Levine, E.M.; Mueller, S.N. Cultured vascular endothelial cells as a model system for the study of cellular senescence. Int. Rev. Cytol. Suppl. 1979, 10, 67–76. [Google Scholar]
  82. Thorgeirsson, G.; Robertson, A.L. The vascular endothelium-pathobiologic significance. Am. J. Pathol. 1978, 93, 803–848. [Google Scholar] [PubMed]
  83. Pawlowski, N.A.; Kaplan, G.; Abraham, E.; Cohn, Z.A. The selective binding and transmigration of monocytes through the junctional complexes of human endothelium. J. Exp. Med. 1988, 168, 1865–1882. [Google Scholar] [CrossRef]
  84. Maciag, T.; Hoover, G.A.; Stemerman, M.B.; Weinstein, R. Serial propagation of human endothelial cells in vitro. J. Cell Biol. 1981, 91, 420–426. [Google Scholar] [CrossRef] [PubMed]
  85. Tokunaga, O.; Fan, J.L.; Watanabe, T. Atherosclerosis- and age-related multinucleated variant endothelial cells in primary culture from human aorta. Am. J. Pathol. 1989, 135, 967–976. [Google Scholar] [PubMed]
  86. Mueller, S.N.; Rosen, E.M.; Levine, E.M. Cellular senescence in a cloned strain of bovine fetal aortic endothelial cells. Science 1980, 207, 889–891. [Google Scholar] [CrossRef] [PubMed]
  87. Duthu, G.S.; Smith, J.R. In vitro proliferation and lifespan of bovine aorta endothelial cells: Effect of culture conditions and fibroblast growth factor. J. Cell. Physiol. 1980, 103, 385–392. [Google Scholar] [CrossRef] [PubMed]
  88. Rosen, E.M.; Mueller, S.N.; Noveral, J.P.; Levine, E.M. Proliferative characteristics of clonal endothelial cell strains. J. Cell. Physiol. 1981, 107, 123–137. [Google Scholar] [CrossRef] [PubMed]
  89. Swanson, E.C.; Manning, B.; Zhang, H.; Lawrence, J.B. Higher-order unfolding of satellite heterochromatin is a consistent and early event in cell senescence. J. Cell Biol. 2013, 203, 929–942. [Google Scholar] [CrossRef]
  90. Johnson, T.E.; Umbenhauer, D.R.; Hill, R.; Bradt, C.; Mueller, S.N.; Levine, E.M.; Nichols, W.W. Karyotypic and phenotypic changes during in vitro aging of human endothelial cells. J. Cell. Physiol. 1992, 150, 17–27. [Google Scholar] [CrossRef]
  91. Boumendil, C.; Hari, P.; Olsen, K.C.F.; Acosta, J.C.; Bickmore, W.A. Nuclear pore density controls heterochromatin reorganization during senescence. Genes. Dev. 2019, 33, 144–149. [Google Scholar] [CrossRef] [PubMed]
  92. Katsuumi, G.; Shimizu, I.; Yoshida, Y.; Minamino, T. Vascular Senescence in Cardiovascular and Metabolic Diseases. Front. Cardiovasc. Med. 2018, 5, 18. [Google Scholar] [CrossRef] [PubMed]
  93. Minamino, T.; Miyauchi, H.; Yoshida, T.; Ishida, Y.; Yoshida, H.; Komuro, I. Endothelial cell senescence in human atherosclerosis: Role of telomere in endothelial dysfunction. Circulation 2002, 105, 1541–1544. [Google Scholar] [CrossRef] [PubMed]
  94. Chala, N.; Moimas, S.; Giampietro, C.; Zhang, X.; Zambelli, T.; Exarchos, V.; Nazari-Shafti, T.Z.; Poulikakos, D.; Ferrari, A. Mechanical Fingerprint of Senescence in Endothelial Cells. Nano Lett. 2021, 21, 4911–4920. [Google Scholar] [CrossRef] [PubMed]
  95. Bloom, S.I.; Islam, M.T.; Lesniewski, L.A.; Donato, A.J. Mechanisms and consequences of endothelial cell senescence. Nat. Rev. Cardiol. 2023, 20, 38–51. [Google Scholar] [CrossRef] [PubMed]
  96. Repin, V.S.; Dolgov, V.V.; Zaikina, O.E.; Novikov, I.D.; Antonov, A.S.; Nikolaeva, M.A.; Smirnov, V.N. Heterogeneity of endothelium in human aorta. A quantitative analysis by scanning electron microscopy. Atherosclerosis 1984, 50, 35–52. [Google Scholar] [CrossRef]
  97. Bürrig, K.F. The endothelium of advanced arteriosclerotic plaques in humans. Arter. Thromb. 1991, 11, 1678–1689. [Google Scholar] [CrossRef]
  98. Morgan, R.G.; Ives, S.J.; Lesniewski, L.A.; Cawthon, R.M.; Andtbacka, R.H.I.; Noyes, R.D.; Richardson, R.S.; Donato, A.J. Age-related telomere uncapping is associated with cellular senescence and inflammation independent of telomere shortening in human arteries. Am. J. Physiol. Heart Circ. Physiol. 2013, 305, H251–H258. [Google Scholar] [CrossRef]
  99. Bautista-Niño, P.K.; Portilla-Fernandez, E.; Rubio-Beltrán, E.; van der Linden, J.J.; de Vries, R.; van Veghel, R.; de Boer, M.; Durik, M.; Ridwan, Y.; Brandt, R.; et al. Local endothelial DNA repair deficiency causes aging-resembling endothelial-specific dysfunction. Clin. Sci. 2020, 134, 727–746. [Google Scholar] [CrossRef]
  100. Evans, C.E.; Iruela-Arispe, M.L.; Zhao, Y.-Y. Mechanisms of Endothelial Regeneration and Vascular Repair and Their Application to Regenerative Medicine. Am. J. Pathol. 2021, 191, 52–65. [Google Scholar] [CrossRef]
  101. Durik, M.; Kavousi, M.; van der Pluijm, I.; Isaacs, A.; Cheng, C.; Verdonk, K.; Loot, A.E.; Oeseburg, H.; Bhaggoe, U.M.; Leijten, F.; et al. Nucleotide excision DNA repair is associated with age-related vascular dysfunction. Circulation 2012, 126, 468–478. [Google Scholar] [CrossRef]
  102. Bai, B.; Wang, Y. Methods to investigate the role of SIRT1 in endothelial senescence. Methods Mol. Biol. 2013, 965, 327–339. [Google Scholar] [CrossRef]
  103. Asahara, T.; Murohara, T.; Sullivan, A.; Silver, M.; van der Zee, R.; Li, T.; Witzenbichler, B.; Schatteman, G.; Isner, J.M. Isolation of putative progenitor endothelial cells for angiogenesis. Science 1997, 275, 964–967. [Google Scholar] [CrossRef]
  104. Marcu, R.; Choi, Y.J.; Xue, J.; Fortin, C.L.; Wang, Y.; Nagao, R.J.; Xu, J.; MacDonald, J.W.; Bammler, T.K.; Murry, C.E.; et al. Human Organ-Specific Endothelial Cell Heterogeneity. iScience 2018, 4, 20–35. [Google Scholar] [CrossRef]
  105. Nolan, D.J.; Ginsberg, M.; Israely, E.; Palikuqi, B.; Poulos, M.G.; James, D.; Ding, B.-S.; Schachterle, W.; Liu, Y.; Rosenwaks, Z.; et al. Molecular signatures of tissue-specific microvascular endothelial cell heterogeneity in organ maintenance and regeneration. Dev. Cell 2013, 26, 204–219. [Google Scholar] [CrossRef]
  106. Gebrane-Younès, J.; Drouet, L.; Caen, J.P.; Orcel, L. Heterogeneous distribution of Weibel-Palade bodies and von Willebrand factor along the porcine vascular tree. Am. J. Pathol. 1991, 139, 1471–1484. [Google Scholar]
  107. Matsuoka, R.L.; Buck, L.D.; Vajrala, K.P.; Quick, R.E.; Card, O.A. Historical and current perspectives on blood endothelial cell heterogeneity in the brain. Cell Mol. Life Sci. 2022, 79, 372. [Google Scholar] [CrossRef]
  108. Yuan, S.Y.; Rigor, R.R. Integrated Systems Physiology: From Molecule to Function to Disease. In Regulation of Endothelial Barrier Function; Morgan & Claypool Life Sciences: San Rafael, CA, USA, 2010. [Google Scholar] [CrossRef]
  109. Secomb, T.W.; Pries, A.R. The microcirculation: Physiology at the mesoscale. J. Physiol. 2011, 589, 1047–1052. [Google Scholar] [CrossRef] [PubMed]
  110. Vancheri, F.; Longo, G.; Vancheri, S.; Henein, M. Coronary Microvascular Dysfunction. J. Clin. Med. 2020, 9, 2880. [Google Scholar] [CrossRef] [PubMed]
  111. Kalucka, J.; de Rooij, L.P.M.H.; Goveia, J.; Rohlenova, K.; Dumas, S.J.; Meta, E.; Conchinha, N.V.; Taverna, F.; Teuwen, L.-A.; Veys, K.; et al. Single-Cell Transcriptome Atlas of Murine Endothelial Cells. Cell 2020, 180, 764–779.e20. [Google Scholar] [CrossRef] [PubMed]
  112. Risau, W. Differentiation of endothelium. FASEB J. 1995, 9, 926–933. [Google Scholar] [CrossRef] [PubMed]
  113. Garlanda, C.; Dejana, E. Heterogeneity of endothelial cells. Specific markers. Arterioscler. Thromb. Vasc. Biol. 1997, 17, 1193–1202. [Google Scholar] [CrossRef]
  114. Pulgar, V.M. Transcytosis to Cross the Blood Brain Barrier, New Advancements and Challenges. Front. Neurosci. 2018, 12, 1019. [Google Scholar] [CrossRef] [PubMed]
  115. Pollay, M.; Roberts, P.A. Blood-brain barrier: A definition of normal and altered function. Neurosurgery 1980, 6, 675–685. [Google Scholar] [CrossRef] [PubMed]
  116. Yang, A.C.; Stevens, M.Y.; Chen, M.B.; Lee, D.P.; Stähli, D.; Gate, D.; Contrepois, K.; Chen, W.; Iram, T.; Zhang, L.; et al. Physiological blood-brain transport is impaired with age by a shift in transcytosis. Nature 2020, 583, 425–430. [Google Scholar] [CrossRef] [PubMed]
  117. Kiss, T.; Nyúl-Tóth, Á.; Balasubramanian, P.; Tarantini, S.; Ahire, C.; DelFavero, J.; Yabluchanskiy, A.; Csipo, T.; Farkas, E.; Wiley, G.; et al. Single-cell RNA sequencing identifies senescent cerebromicrovascular endothelial cells in the aged mouse brain. Geroscience 2020, 42, 429–444. [Google Scholar] [CrossRef] [PubMed]
  118. Knopp, R.C.; Erickson, M.A.; Rhea, E.M.; Reed, M.J.; Banks, W.A. Cellular senescence and the blood-brain barrier: Implications for aging and age-related diseases. Exp. Biol. Med. 2023, 248, 399–411. [Google Scholar] [CrossRef] [PubMed]
  119. Vorbrodt, A.W.; Lossinsky, A.S.; Wisniewski, H.M. Localization of alkaline phosphatase activity in endothelia of developing and mature mouse blood-brain barrier. Dev. Neurosci. 1986, 8, 1–13. [Google Scholar] [CrossRef]
  120. Nwafor, D.C.; Brichacek, A.L.; Ali, A.; Brown, C.M. Tissue-Nonspecific Alkaline Phosphatase in Central Nervous System Health and Disease: A Focus on Brain Microvascular Endothelial Cells. Int. J. Mol. Sci. 2021, 22, 5257. [Google Scholar] [CrossRef]
  121. Raub, T.J.; Newton, C.R. Recycling kinetics and transcytosis of transferrin in primary cultures of bovine brain microvessel endothelial cells. J. Cell. Physiol. 1991, 149, 141–151. [Google Scholar] [CrossRef]
  122. DeBault, L.E.; Henriquez, E.; Hart, M.N.; Cancilla, P.A. Cerebral microvessels and derived cells in tissue culture: II. Establishment, identification, and preliminary characterization of an endothelial cell line. In Vitro 1981, 17, 480–494. [Google Scholar] [CrossRef]
  123. Sapatino, B.V.; Welsh, C.J.; Smith, C.A.; Bebo, B.F.; Linthicum, D.S. Cloned mouse cerebrovascular endothelial cells that maintain their differentiation markers for factor VIII, low density lipoprotein, and angiotensin-converting enzyme. Vitr. Cell Dev. Biol. Anim. 1993, 29A, 923–928. [Google Scholar] [CrossRef] [PubMed]
  124. Shi, F.; Audus, K.L. Biochemical characteristics of primary and passaged cultures of primate brain microvessel endothelial cells. Neurochem. Res. 1994, 19, 427–433. [Google Scholar] [CrossRef] [PubMed]
  125. Noren Hooten, N.; Evans, M.K. Techniques to Induce and Quantify Cellular Senescence. J. Vis. Exp. 2017, 123, 55533. [Google Scholar] [CrossRef]
  126. Ungvari, Z.; Podlutsky, A.; Sosnowska, D.; Tucsek, Z.; Toth, P.; Deak, F.; Gautam, T.; Csiszar, A.; Sonntag, W.E. 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, 1443–1457. [Google Scholar] [CrossRef] [PubMed]
  127. Ungvari, Z.; Tarantini, S.; Kiss, T.; Wren, J.D.; Giles, C.B.; Griffin, C.T.; Murfee, W.L.; Pacher, P.; Csiszar, A. Endothelial dysfunction and angiogenesis impairment in the ageing vasculature. Nat. Rev. Cardiol. 2018, 15, 555–565. [Google Scholar] [CrossRef] [PubMed]
  128. Mooradian, A.D.; Smith, T.L. The effect of age on lipid composition and order of rat cerebral microvessels. Neurochem. Res. 1992, 17, 233–237. [Google Scholar] [CrossRef] [PubMed]
  129. Krouwer, V.J.D.; Hekking, L.H.P.; Langelaar-Makkinje, M.; Regan-Klapisz, E.; Post, J.A. Endothelial cell senescence is associated with disrupted cell-cell junctions and increased monolayer permeability. Vascular Cell 2012, 4, 12. [Google Scholar] [CrossRef]
  130. Pelegrí, C.; Canudas, A.M.; del Valle, J.; Casadesus, G.; Smith, M.A.; Camins, A.; Pallàs, M.; Vilaplana, J. Increased permeability of blood-brain barrier on the hippocampus of a murine model of senescence. Mech. Ageing Dev. 2007, 128, 522–528. [Google Scholar] [CrossRef]
  131. Del Valle, J.; Duran-Vilaregut, J.; Manich, G.; Camins, A.; Pallàs, M.; Vilaplana, J.; Pelegrí, C. Time-course of blood-brain barrier disruption in senescence-accelerated mouse prone 8 (SAMP8) mice. Int. J. Dev. Neurosci. 2009, 27, 47–52. [Google Scholar] [CrossRef]
  132. Kiss, T.; Nyúl-Tóth, Á.; DelFavero, J.; Balasubramanian, P.; Tarantini, S.; Faakye, J.; Gulej, R.; Ahire, C.; Ungvari, A.; Yabluchanskiy, A.; et al. 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, 661–681. [Google Scholar] [CrossRef] [PubMed]
  133. Marescal, O.; Cheeseman, I.M. Cellular Mechanisms and Regulation of Quiescence. Dev. Cell 2020, 55, 259–271. [Google Scholar] [CrossRef] [PubMed]
  134. Cho, S.; Hwang, E.S. Status of mTOR activity may phenotypically differentiate senescence and quiescence. Mol. Cells 2012, 33, 597–604. [Google Scholar] [CrossRef] [PubMed]
  135. Dunlay, S.M.; Roger, V.L.; Redfield, M.M. Epidemiology of heart failure with preserved ejection fraction. Nat. Rev. Cardiol. 2017, 14, 591–602. [Google Scholar] [CrossRef] [PubMed]
  136. van Heerebeek, L.; Borbély, A.; Niessen, H.W.M.; Bronzwaer, J.G.F.; van der Velden, J.; Stienen, G.J.M.; Linke, W.A.; Laarman, G.J.; Paulus, W.J. Myocardial structure and function differ in systolic and diastolic heart failure. Circulation 2006, 113, 1966–1973. [Google Scholar] [CrossRef] [PubMed]
  137. Borlaug, B.A. The pathophysiology of heart failure with preserved ejection fraction. Nat. Rev. Cardiol. 2014, 11, 507–515. [Google Scholar] [CrossRef] [PubMed]
  138. Larson, K.F.; Malik, A.; Brozovich, F.V. Aging and Heart Failure with Preserved Ejection Fraction. Compr. Physiol. 2022, 12, 3813–3822. [Google Scholar] [CrossRef]
  139. Del Buono, M.G.; Iannaccone, G.; Scacciavillani, R.; Carbone, S.; Camilli, M.; Niccoli, G.; Borlaug, B.A.; Lavie, C.J.; Arena, R.; Crea, F.; et al. Heart failure with preserved ejection fraction diagnosis and treatment: An updated review of the evidence. Prog. Cardiovasc. Dis. 2020, 63, 570–584. [Google Scholar] [CrossRef]
  140. Pinto, A.R.; Ilinykh, A.; Ivey, M.J.; Kuwabara, J.T.; D’Antoni, M.L.; Debuque, R.; Chandran, A.; Wang, L.; Arora, K.; Rosenthal, N.A.; et al. Revisiting Cardiac Cellular Composition. Circ. Res. 2016, 118, 400–409. [Google Scholar] [CrossRef]
  141. Litviňuková, M.; Talavera-López, C.; Maatz, H.; Reichart, D.; Worth, C.L.; Lindberg, E.L.; Kanda, M.; Polanski, K.; Heinig, M.; Lee, M.; et al. Cells of the adult human heart. Nature 2020, 588, 466–472. [Google Scholar] [CrossRef]
  142. A single-cell transcriptomic atlas characterizes ageing tissues in the mouse. Nature 2020, 583, 590–595. [CrossRef]
  143. Koenig, A.L.; Shchukina, I.; Amrute, J.; Andhey, P.S.; Zaitsev, K.; Lai, L.; Bajpai, G.; Bredemeyer, A.; Smith, G.; Jones, C.; et al. Single-cell transcriptomics reveals cell-type-specific diversification in human heart failure. Nat. Cardiovasc. Res. 2022, 1, 263–280. [Google Scholar] [CrossRef]
  144. Cuijpers, I.; Simmonds, S.J.; van Bilsen, M.; Czarnowska, E.; González Miqueo, A.; Heymans, S.; Kuhn, A.R.; Mulder, P.; Ratajska, A.; Jones, E.A.V.; et al. Microvascular and lymphatic dysfunction in HFpEF and its associated comorbidities. Basic. Res. Cardiol. 2020, 115, 39. [Google Scholar] [CrossRef]
  145. Paulus, W.J.; Tschöpe, C. A novel paradigm for heart failure with preserved ejection fraction: Comorbidities drive myocardial dysfunction and remodeling through coronary microvascular endothelial inflammation. J. Am. Coll. Cardiol. 2013, 62, 263–271. [Google Scholar] [CrossRef]
  146. Mohammed, S.F.; Redfield, M.M. Response to Letters Regarding Article, “Coronary Microvascular Rarefaction and Myocardial Fibrosis in Heart Failure With Preserved Ejection Fraction”. Circulation 2015, 132, e206. [Google Scholar] [CrossRef]
  147. Shah, S.J.; Kitzman, D.W.; Borlaug, B.A.; van Heerebeek, L.; Zile, M.R.; Kass, D.A.; Paulus, W.J. Phenotype-Specific Treatment of Heart Failure With Preserved Ejection Fraction: A Multiorgan Roadmap. Circulation 2016, 134, 73–90. [Google Scholar] [CrossRef] [PubMed]
  148. Yang, J.H.; Obokata, M.; Reddy, Y.N.V.; Redfield, M.M.; Lerman, A.; Borlaug, B.A. Endothelium-dependent and independent coronary microvascular dysfunction in patients with heart failure with preserved ejection fraction. Eur. J. Heart Fail. 2020, 22, 432–441. [Google Scholar] [CrossRef] [PubMed]
  149. Taqueti, V.R.; Solomon, S.D.; Shah, A.M.; Desai, A.S.; Groarke, J.D.; Osborne, M.T.; Hainer, J.; Bibbo, C.F.; Dorbala, S.; Blankstein, R.; et al. Coronary microvascular dysfunction and future risk of heart failure with preserved ejection fraction. Eur. Heart J. 2018, 39, 840–849. [Google Scholar] [CrossRef] [PubMed]
  150. Lam, C.S.P.; Voors, A.A.; de Boer, R.A.; Solomon, S.D.; van Veldhuisen, D.J. Heart failure with preserved ejection fraction: From mechanisms to therapies. Eur. Heart J. 2018, 39, 2780–2792. [Google Scholar] [CrossRef]
  151. Jesel, L.; Abbas, M.; Park, S.-H.; Matsushita, K.; Kindo, M.; Hasan, H.; Auger, C.; Sato, C.; Ohlmann, P.; Mazzucotelli, J.-P.; et al. Atrial Fibrillation Progression Is Associated with Cell Senescence Burden as Determined by p53 and p16 Expression. J. Clin. Med. 2019, 9, 36. [Google Scholar] [CrossRef] [PubMed]
  152. Sano, M.; Minamino, T.; Toko, H.; Miyauchi, H.; Orimo, M.; Qin, Y.; Akazawa, H.; Tateno, K.; Kayama, Y.; Harada, M.; et al. p53-induced inhibition of Hif-1 causes cardiac dysfunction during pressure overload. Nature 2007, 446, 444–448. [Google Scholar] [CrossRef]
  153. Chen, M.S.; Lee, R.T.; Garbern, J.C. Senescence mechanisms and targets in the heart. Cardiovasc. Res. 2022, 118, 1173–1187. [Google Scholar] [CrossRef]
  154. Mojiri, A.; Walther, B.K.; Jiang, C.; Matrone, G.; Holgate, R.; Xu, Q.; Morales, E.; Wang, G.; Gu, J.; Wang, R.; et al. Telomerase therapy reverses vascular senescence and extends lifespan in progeria mice. Eur. Heart J. 2021, 42, 4352–4369. [Google Scholar] [CrossRef]
  155. Yoshida, Y.; Shimizu, I.; Katsuumi, G.; Jiao, S.; Suda, M.; Hayashi, Y.; Minamino, T. p53-Induced inflammation exacerbates cardiac dysfunction during pressure overload. J. Mol. Cell Cardiol. 2015, 85, 183–198. [Google Scholar] [CrossRef]
  156. Gogiraju, R.; Xu, X.; Bochenek, M.L.; Steinbrecher, J.H.; Lehnart, S.E.; Wenzel, P.; Kessel, M.; Zeisberg, E.M.; Dobbelstein, M.; Schäfer, K. Endothelial p53 deletion improves angiogenesis and prevents cardiac fibrosis and heart failure induced by pressure overload in mice. J. Am. Heart Assoc. 2015, 4, e001770. [Google Scholar] [CrossRef]
  157. Gevaert, A.B.; Shakeri, H.; Leloup, A.J.; Van Hove, C.E.; De Meyer, G.R.Y.; Vrints, C.J.; Lemmens, K.; Van Craenenbroeck, E.M. Endothelial Senescence Contributes to Heart Failure With Preserved Ejection Fraction in an Aging Mouse Model. Circ. Heart Fail. 2017, 10, e003806. [Google Scholar] [CrossRef]
  158. Bloom, S.I.; Tucker, J.R.; Lim, J.; Thomas, T.G.; Stoddard, G.J.; Lesniewski, L.A.; Donato, A.J. Aging results in DNA damage and telomere dysfunction that is greater in endothelial versus vascular smooth muscle cells and is exacerbated in atheroprone regions. Geroscience 2022, 44, 2741–2755. [Google Scholar] [CrossRef]
  159. Vermeij, W.P.; Hoeijmakers, J.H.J.; Pothof, J. Genome Integrity in Aging: Human Syndromes, Mouse Models, and Therapeutic Options. Annu. Rev. Pharmacol. Toxicol. 2016, 56, 427–445. [Google Scholar] [CrossRef] [PubMed]
  160. Lim, S.L.; Lam, C.S.P.; Segers, V.F.M.; Brutsaert, D.L.; De Keulenaer, G.W. Cardiac endothelium-myocyte interaction: Clinical opportunities for new heart failure therapies regardless of ejection fraction. Eur. Heart J. 2015, 36, 2050–2060. [Google Scholar] [CrossRef] [PubMed]
  161. Ferrucci, L.; Kuchel, G.A. Heterogeneity of Aging: Individual Risk Factors, Mechanisms, Patient Priorities, and Outcomes. J. Am. Geriatr. Soc. 2021, 69, 610–612. [Google Scholar] [CrossRef] [PubMed]
  162. Xiao, X.; Jiang, H.; Wei, H.; Zhou, Y.; Ji, X.; Zhou, C. Endothelial Senescence in Neurological Diseases. Aging Dis. 2023, 14, 2153–2166. [Google Scholar] [CrossRef]
  163. Wu, C.-M.; Zheng, L.; Wang, Q.; Hu, Y.-W. The emerging role of cell senescence in atherosclerosis. Clin. Chem. Lab. Med. 2021, 59, 27–38. [Google Scholar] [CrossRef]
  164. Sun, Y.; Wang, X.; Liu, T.; Zhu, X.; Pan, X. The multifaceted role of the SASP in atherosclerosis: From mechanisms to therapeutic opportunities. Cell Biosci. 2022, 12, 74. [Google Scholar] [CrossRef]
  165. Bu, L.-L.; Yuan, H.-H.; Xie, L.-L.; Guo, M.-H.; Liao, D.-F.; Zheng, X.-L. New Dawn for Atherosclerosis: Vascular Endothelial Cell Senescence and Death. Int. J. Mol. Sci. 2023, 24, 15160. [Google Scholar] [CrossRef]
  166. Higashi, Y.; Kihara, Y.; Noma, K. Endothelial dysfunction and hypertension in aging. Hypertens. Res. 2012, 35, 1039–1047. [Google Scholar] [CrossRef]
  167. Graves, S.I.; Baker, D.J. Implicating endothelial cell senescence to dysfunction in the ageing and diseased brain. Basic. Clin. Pharmacol. Toxicol. 2020, 127, 102–110. [Google Scholar] [CrossRef] [PubMed]
  168. López-Otín, C.; Blasco, M.A.; Partridge, L.; Serrano, M.; Kroemer, G. The hallmarks of aging. Cell 2013, 153, 1194–1217. [Google Scholar] [CrossRef] [PubMed]
  169. López-Otín, C.; Blasco, M.A.; Partridge, L.; Serrano, M.; Kroemer, G. Hallmarks of aging: An expanding universe. Cell 2023, 186, 243–278. [Google Scholar] [CrossRef] [PubMed]
  170. Abdellatif, M.; Rainer, P.P.; Sedej, S.; Kroemer, G. Hallmarks of cardiovascular ageing. Nat. Rev. Cardiol. 2023, 20, 754–777. [Google Scholar] [CrossRef] [PubMed]
  171. Folgueras, A.R.; Freitas-Rodríguez, S.; Velasco, G.; López-Otín, C. Mouse Models to Disentangle the Hallmarks of Human Aging. Circ. Res. 2018, 123, 905–924. [Google Scholar] [CrossRef]
  172. Leri, A.; Franco, S.; Zacheo, A.; Barlucchi, L.; Chimenti, S.; Limana, F.; Nadal-Ginard, B.; Kajstura, J.; Anversa, P.; Blasco, M.A. Ablation of telomerase and telomere loss leads to cardiac dilatation and heart failure associated with p53 upregulation. EMBO J. 2003, 22, 131–139. [Google Scholar] [CrossRef] [PubMed]
  173. Anderson, R.; Lagnado, A.; Maggiorani, D.; Walaszczyk, A.; Dookun, E.; Chapman, J.; Birch, J.; Salmonowicz, H.; Ogrodnik, M.; Jurk, D.; et al. Length-independent telomere damage drives post-mitotic cardiomyocyte senescence. EMBO J. 2019, 38, e100492. [Google Scholar] [CrossRef] [PubMed]
  174. Madonna, R.; De Caterina, R.; Willerson, J.T.; Geng, Y.-J. Biologic function and clinical potential of telomerase and associated proteins in cardiovascular tissue repair and regeneration. Eur. Heart J. 2011, 32, 1190–1196. [Google Scholar] [CrossRef] [PubMed]
  175. Madonna, R. Vascular rejuvenation: A new therapeutic target? Eur. Heart J. 2021, 42, 4370–4372. [Google Scholar] [CrossRef] [PubMed]
  176. Grunewald, M.; Kumar, S.; Sharife, H.; Volinsky, E.; Gileles-Hillel, A.; Licht, T.; Permyakova, A.; Hinden, L.; Azar, S.; Friedmann, Y.; et al. Counteracting age-related VEGF signaling insufficiency promotes healthy aging and extends life span. Science 2021, 373, eabc8479. [Google Scholar] [CrossRef] [PubMed]
  177. Bai, B.; Liang, Y.; Xu, C.; Lee, M.Y.K.; Xu, A.; Wu, D.; Vanhoutte, P.M.; Wang, Y. Cyclin-dependent kinase 5-mediated hyperphosphorylation of sirtuin-1 contributes to the development of endothelial senescence and atherosclerosis. Circulation 2012, 126, 729–740. [Google Scholar] [CrossRef]
  178. Bai, B.; Man, A.W.C.; Yang, K.; Guo, Y.; Xu, C.; Tse, H.-F.; Han, W.; Bloksgaard, M.; De Mey, J.G.R.; Vanhoutte, P.M.; et al. Endothelial SIRT1 prevents adverse arterial remodeling by facilitating HERC2-mediated degradation of acetylated LKB1. Oncotarget 2016, 7, 39065–39081. [Google Scholar] [CrossRef]
  179. Guo, Y.; Xu, C.; Man, A.W.C.; Bai, B.; Luo, C.; Huang, Y.; Xu, A.; Vanhoutte, P.M.; Wang, Y. Endothelial SIRT1 prevents age-induced impairment of vasodilator responses by enhancing the expression and activity of soluble guanylyl cyclase in smooth muscle cells. Cardiovasc. Res. 2019, 115, 678–690. [Google Scholar] [CrossRef]
  180. Zu, Y.; Liu, L.; Lee, M.Y.K.; Xu, C.; Liang, Y.; Man, R.Y.; Vanhoutte, P.M.; Wang, Y. SIRT1 promotes proliferation and prevents senescence through targeting LKB1 in primary porcine aortic endothelial cells. Circ. Res. 2010, 106, 1384–1393. [Google Scholar] [CrossRef] [PubMed]
  181. Roos, C.M.; Zhang, B.; Palmer, A.K.; Ogrodnik, M.B.; Pirtskhalava, T.; Thalji, N.M.; Hagler, M.; Jurk, D.; Smith, L.A.; Casaclang-Verzosa, G.; et al. Chronic senolytic treatment alleviates established vasomotor dysfunction in aged or atherosclerotic mice. Aging Cell 2016, 15, 973–977. [Google Scholar] [CrossRef] [PubMed]
  182. Zhu, Y.; Tchkonia, T.; Pirtskhalava, T.; Gower, A.C.; Ding, H.; Giorgadze, N.; Palmer, A.K.; Ikeno, Y.; Hubbard, G.B.; Lenburg, M.; et al. The Achilles’ heel of senescent cells: From transcriptome to senolytic drugs. Aging Cell 2015, 14, 644–658. [Google Scholar] [CrossRef] [PubMed]
  183. Zhu, Y.; Tchkonia, T.; Fuhrmann-Stroissnigg, H.; Dai, H.M.; Ling, Y.Y.; Stout, M.B.; Pirtskhalava, T.; Giorgadze, N.; Johnson, K.O.; Giles, C.B.; et al. Identification of a novel senolytic agent, navitoclax, targeting the Bcl-2 family of anti-apoptotic factors. Aging Cell 2016, 15, 428–435. [Google Scholar] [CrossRef] [PubMed]
  184. Tarantini, S.; Balasubramanian, P.; Delfavero, J.; Csipo, T.; Yabluchanskiy, A.; Kiss, T.; Nyúl-Tóth, Á.; Mukli, P.; Toth, P.; Ahire, C.; et al. Treatment with the BCL-2/BCL-xL inhibitor senolytic drug ABT263/Navitoclax improves functional hyperemia in aged mice. Geroscience 2021, 43, 2427–2440. [Google Scholar] [CrossRef] [PubMed]
  185. Ogrodnik, M.; Evans, S.A.; Fielder, E.; Victorelli, S.; Kruger, P.; Salmonowicz, H.; Weigand, B.M.; Patel, A.D.; Pirtskhalava, T.; Inman, C.L.; et al. Whole-body senescent cell clearance alleviates age-related brain inflammation and cognitive impairment in mice. Aging Cell 2021, 20, e13296. [Google Scholar] [CrossRef] [PubMed]
  186. Hickson, L.J.; Langhi Prata, L.G.P.; Bobart, S.A.; Evans, T.K.; Giorgadze, N.; Hashmi, S.K.; Herrmann, S.M.; Jensen, M.D.; Jia, Q.; Jordan, K.L.; et al. Senolytics decrease senescent cells in humans: Preliminary report from a clinical trial of Dasatinib plus Quercetin in individuals with diabetic kidney disease. eBioMedicine 2019, 47, 446–456. [Google Scholar] [CrossRef]
  187. Rossman, M.J.; Kaplon, R.E.; Hill, S.D.; McNamara, M.N.; Santos-Parker, J.R.; Pierce, G.L.; Seals, D.R.; Donato, A.J. 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–H895. [Google Scholar] [CrossRef]
  188. Guerrero, A.; Herranz, N.; Sun, B.; Wagner, V.; Gallage, S.; Guiho, R.; Wolter, K.; Pombo, J.; Irvine, E.E.; Innes, A.J.; et al. Cardiac glycosides are broad-spectrum senolytics. Nat. Metab. 2019, 1, 1074–1088. [Google Scholar] [CrossRef]
  189. Ziff, O.J.; Kotecha, D. Digoxin: The good and the bad. Trends Cardiovasc. Med. 2016, 26, 585–595. [Google Scholar] [CrossRef]
  190. Triana-Martínez, F.; Picallos-Rabina, P.; Da Silva-Álvarez, S.; Pietrocola, F.; Llanos, S.; Rodilla, V.; Soprano, E.; Pedrosa, P.; Ferreirós, A.; Barradas, M.; et al. Identification and characterization of Cardiac Glycosides as senolytic compounds. Nat. Commun. 2019, 10, 4731. [Google Scholar] [CrossRef]
  191. Wang, C.; Qin, L.; Manes, T.D.; Kirkiles-Smith, N.C.; Tellides, G.; Pober, J.S. Rapamycin antagonizes TNF induction of VCAM-1 on endothelial cells by inhibiting mTORC2. J. Exp. Med. 2014, 211, 395–404. [Google Scholar] [CrossRef]
  192. Xu, M.; Tchkonia, T.; Ding, H.; Ogrodnik, M.; Lubbers, E.R.; Pirtskhalava, T.; White, T.A.; Johnson, K.O.; Stout, M.B.; Mezera, V.; et al. JAK inhibition alleviates the cellular senescence-associated secretory phenotype and frailty in old age. Proc. Natl. Acad. Sci. USA 2015, 112, E6301–E6310. [Google Scholar] [CrossRef] [PubMed]
  193. Chen, C.; Liu, Y.; Liu, Y.; Zheng, P. mTOR regulation and therapeutic rejuvenation of aging hematopoietic stem cells. Sci. Signal 2009, 2, ra75. [Google Scholar] [CrossRef] [PubMed]
  194. Bitto, A.; Ito, T.K.; Pineda, V.V.; LeTexier, N.J.; Huang, H.Z.; Sutlief, E.; Tung, H.; Vizzini, N.; Chen, B.; Smith, K.; et al. Transient rapamycin treatment can increase lifespan and healthspan in middle-aged mice. eLife 2016, 5, e16351. [Google Scholar] [CrossRef] [PubMed]
  195. Laberge, R.M.; Sun, Y.; Orjalo, A.V.; Patil, C.K.; Freund, A.; Zhou, L.; Curran, S.C.; Davalos, A.R.; Wilson-Edell, K.A.; Liu, S.; et al. MTOR regulates the pro-tumorigenic senescence-associated secretory phenotype by promoting IL1A translation. Nat. Cell Biol. 2015, 17, 1049–1061. [Google Scholar] [CrossRef]
  196. Chang, J.; Wang, Y.; Shao, L.; Laberge, R.-M.; Demaria, M.; Campisi, J.; Janakiraman, K.; Sharpless, N.E.; Ding, S.; Feng, W.; et al. Clearance of senescent cells by ABT263 rejuvenates aged hematopoietic stem cells in mice. Nat. Med. 2016, 22, 78–83. [Google Scholar] [CrossRef] [PubMed]
  197. Yousefzadeh, M.J.; Zhu, Y.; McGowan, S.J.; Angelini, L.; Fuhrmann-Stroissnigg, H.; Xu, M.; Ling, Y.Y.; Melos, K.I.; Pirtskhalava, T.; Inman, C.L.; et al. Fisetin is a senotherapeutic that extends health and lifespan. eBioMedicine 2018, 36, 18–28. [Google Scholar] [CrossRef]
  198. Johmura, Y.; Yamanaka, T.; Omori, S.; Wang, T.-W.; Sugiura, Y.; Matsumoto, M.; Suzuki, N.; Kumamoto, S.; Yamaguchi, K.; Hatakeyama, S.; et al. Senolysis by glutaminolysis inhibition ameliorates various age-associated disorders. Science 2021, 371, 265–270. [Google Scholar] [CrossRef]
  199. Kirkland, J.L.; Tchkonia, T. Senolytic drugs: From discovery to translation. J. Intern. Med. 2020, 288, 518–536. [Google Scholar] [CrossRef]
  200. Wilson, W.H.; O’Connor, O.A.; Czuczman, M.S.; LaCasce, A.S.; Gerecitano, J.F.; Leonard, J.P.; Tulpule, A.; Dunleavy, K.; Xiong, H.; Chiu, Y.-L.; et al. Navitoclax, a targeted high-affinity inhibitor of BCL-2, in lymphoid malignancies: A phase 1 dose-escalation study of safety, pharmacokinetics, pharmacodynamics, and antitumour activity. Lancet Oncol. 2010, 11, 1149–1159. [Google Scholar] [CrossRef]
  201. de Vos, S.; Leonard, J.P.; Friedberg, J.W.; Zain, J.; Dunleavy, K.; Humerickhouse, R.; Hayslip, J.; Pesko, J.; Wilson, W.H. Safety and efficacy of navitoclax, a BCL-2 and BCL-XL inhibitor, in patients with relapsed or refractory lymphoid malignancies: Results from a phase 2a study. Leuk. Lymphoma 2021, 62, 810–818. [Google Scholar] [CrossRef]
  202. Roberts, A.W.; Davids, M.S.; Pagel, J.M.; Kahl, B.S.; Puvvada, S.D.; Gerecitano, J.F.; Kipps, T.J.; Anderson, M.A.; Brown, J.R.; Gressick, L.; et al. Targeting BCL2 with Venetoclax in Relapsed Chronic Lymphocytic Leukemia. N. Engl. J. Med. 2016, 374, 311–322. [Google Scholar] [CrossRef]
  203. Tuttle, C.S.L.; Waaijer, M.E.C.; Slee-Valentijn, M.S.; Stijnen, T.; Westendorp, R.; Maier, A.B. Cellular senescence and chronological age in various human tissues: A systematic review and meta-analysis. Aging Cell 2020, 19, e13083. [Google Scholar] [CrossRef] [PubMed]
  204. Gasek, N.S.; Kuchel, G.A.; Kirkland, J.L.; Xu, M. Strategies for Targeting Senescent Cells in Human Disease. Nat. Aging 2021, 1, 870–879. [Google Scholar] [CrossRef] [PubMed]
  205. Hernandez-Segura, A.; de Jong, T.V.; Melov, S.; Guryev, V.; Campisi, J.; Demaria, M. Unmasking Transcriptional Heterogeneity in Senescent Cells. Curr. Biol. 2017, 27, 2652–2660.e4. [Google Scholar] [CrossRef] [PubMed]
  206. Rhinn, M.; Ritschka, B.; Keyes, W.M. Cellular senescence in development, regeneration and disease. Development 2019, 146, dev151837. [Google Scholar] [CrossRef] [PubMed]
  207. Moiseeva, V.; Cisneros, A.; Cobos, A.C.; Tarrega, A.B.; Oñate, C.S.; Perdiguero, E.; Serrano, A.L.; Muñoz-Cánoves, P. Context-dependent roles of cellular senescence in normal, aged, and disease states. FEBS J. 2023, 290, 1161–1185. [Google Scholar] [CrossRef] [PubMed]
  208. Kohli, J.; Wang, B.; Brandenburg, S.M.; Basisty, N.; Evangelou, K.; Varela-Eirin, M.; Campisi, J.; Schilling, B.; Gorgoulis, V.; Demaria, M. Algorithmic assessment of cellular senescence in experimental and clinical specimens. Nat. Protoc. 2021, 16, 2471–2498. [Google Scholar] [CrossRef]
  209. Garrido, A.M.; Kaistha, A.; Uryga, A.K.; Oc, S.; Foote, K.; Shah, A.; Finigan, A.; Figg, N.; Dobnikar, L.; Jørgensen, H.; et al. Efficacy and limitations of senolysis in atherosclerosis. Cardiovasc. Res. 2022, 118, 1713–1727. [Google Scholar] [CrossRef]
Figure 1. Mechanism of endothelial cell senescence. Senescent endothelial cells develop a senescence-associated secretory phenotype (SASP) and an increase of SA-β-Gal activity. Senescence results in decrease of endothelial NO synthase expression and reduction of NO production, and the inducible NO synthase (iNOS) and ROS levels increase significantly, leading to DNA damage and promotion of senescence and apoptosis. Senescent cells also exhibit upregulation of p53/p21 or p16/Rb pathways and markers associated with activation of the DNA damage response (p38MAPK and γH2AX). In addition, the VEGF signaling pathway, along with AMPK, mTOR, and SIRT1, is also involved in the endothelial cell senescence.
Figure 1. Mechanism of endothelial cell senescence. Senescent endothelial cells develop a senescence-associated secretory phenotype (SASP) and an increase of SA-β-Gal activity. Senescence results in decrease of endothelial NO synthase expression and reduction of NO production, and the inducible NO synthase (iNOS) and ROS levels increase significantly, leading to DNA damage and promotion of senescence and apoptosis. Senescent cells also exhibit upregulation of p53/p21 or p16/Rb pathways and markers associated with activation of the DNA damage response (p38MAPK and γH2AX). In addition, the VEGF signaling pathway, along with AMPK, mTOR, and SIRT1, is also involved in the endothelial cell senescence.
Ijms 25 01978 g001
Figure 2. Comparison of young and senescent endothelial cells isolated from porcine aorta (Magnification: 4×). Arteries were separated from the heart and immediately placed in freshly prepared cold Earle’s balanced salt solution (EBSS). After clearing all the peripheral connective tissues, aortas were cut longitudinally and pinned down for en face cell isolation in a glass dissecting dish. A sufficient cold EBSS was added to rinse the aortic fragments by gentle shaking. The EBSS was discarded and replaced rapidly by a fresh EBSS to reduce the exposure time to air in order to enhance cell viability. After thorough washing for five times, the EBSS was removed completely before collecting the endothelial cells. Endothelial cells were collected gently from the luminal surface of aorta by scraping with a scalpel blade and collected in pre-warmed DMEM containing 15% fetal bovine serum (FBS) and 1% penicillin-streptomycin-fungizone (PSF). The cell pellets were washed by centrifugation (200× g, 5 min) and re-suspension for two times. Finally, the PAECs were resuspended by the pre-warmed DMEM containing 15% FBS and 1% PSF and transferred into a 60 mm 1% gelatin-coated Petri dish and cultured in DMEM with 15% FBS and 1% PSF in a humidified incubator containing 5% CO2/95% O2 at 37 °C. The medium was replaced every two days. The young cells (left) show a typical “cobblestone” monolayer, whereas after 4-week culture, the senescent (right) endothelial cells exhibit an enlarged, multinucleated and flattened morphology [102].
Figure 2. Comparison of young and senescent endothelial cells isolated from porcine aorta (Magnification: 4×). Arteries were separated from the heart and immediately placed in freshly prepared cold Earle’s balanced salt solution (EBSS). After clearing all the peripheral connective tissues, aortas were cut longitudinally and pinned down for en face cell isolation in a glass dissecting dish. A sufficient cold EBSS was added to rinse the aortic fragments by gentle shaking. The EBSS was discarded and replaced rapidly by a fresh EBSS to reduce the exposure time to air in order to enhance cell viability. After thorough washing for five times, the EBSS was removed completely before collecting the endothelial cells. Endothelial cells were collected gently from the luminal surface of aorta by scraping with a scalpel blade and collected in pre-warmed DMEM containing 15% fetal bovine serum (FBS) and 1% penicillin-streptomycin-fungizone (PSF). The cell pellets were washed by centrifugation (200× g, 5 min) and re-suspension for two times. Finally, the PAECs were resuspended by the pre-warmed DMEM containing 15% FBS and 1% PSF and transferred into a 60 mm 1% gelatin-coated Petri dish and cultured in DMEM with 15% FBS and 1% PSF in a humidified incubator containing 5% CO2/95% O2 at 37 °C. The medium was replaced every two days. The young cells (left) show a typical “cobblestone” monolayer, whereas after 4-week culture, the senescent (right) endothelial cells exhibit an enlarged, multinucleated and flattened morphology [102].
Ijms 25 01978 g002
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Wang, P.; Konja, D.; Singh, S.; Zhang, B.; Wang, Y. Endothelial Senescence: From Macro- to Micro-Vasculature and Its Implications on Cardiovascular Health. Int. J. Mol. Sci. 2024, 25, 1978. https://doi.org/10.3390/ijms25041978

AMA Style

Wang P, Konja D, Singh S, Zhang B, Wang Y. Endothelial Senescence: From Macro- to Micro-Vasculature and Its Implications on Cardiovascular Health. International Journal of Molecular Sciences. 2024; 25(4):1978. https://doi.org/10.3390/ijms25041978

Chicago/Turabian Style

Wang, Peichun, Daniels Konja, Sandeep Singh, Beijia Zhang, and Yu Wang. 2024. "Endothelial Senescence: From Macro- to Micro-Vasculature and Its Implications on Cardiovascular Health" International Journal of Molecular Sciences 25, no. 4: 1978. https://doi.org/10.3390/ijms25041978

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop