Abstract
The blood–brain barrier (BBB) is a critical physiochemical interface that regulates communication between the brain and blood. It is comprised of brain endothelial cells which regulate the BBB’s barrier and interface properties and is surrounded by supportive brain cell types including pericytes and astrocytes. Recent reports have suggested that the BBB undergoes dysfunction during normative aging and in disease. In this review, we consider the effect of cellular senescence, one of the nine hallmarks of aging, on the BBB. We first characterize known normative age–related changes at the BBB, and then evaluate changes in neurodegenerative diseases, with an emphasis on if/how cellular senescence is influencing these changes. We then discuss what insight has been gained from in vitro and in vivo studies of cellular senescence at the BBB. Finally, we evaluate mechanisms by which cellular senescence in peripheral pathologies can indirectly or directly affect BBB function.
Keywords: Blood–brain barrier, cellular senescence, brain endothelial cells, pericytes, astrocytes, aging
Impact Statement
There have been significant advancements in understanding and characterizing the phenotype of cellular senescence over the last decade, and novel therapeutic approaches that eliminate senescent cells have advanced to clinical trials for a range of age-associated diseases, including those of the central nervous system (CNS). However, much remains to be learned about the predominant cell types that become senescent in the CNS and their functional effects. This review summarizes the known age-associated changes of the blood–brain barrier (BBB) induced by senescence, senescent changes in diseases affecting the CNS, and considers how senolytics may impact important BBB functions.
Introduction
Cellular senescence
Cellular senescence is a complex phenomenon first identified by Hayflick and Moorhead 1 in 1961. In their seminal publication, which has now been cited over 1800 times, they described isolated human fibroblasts having a “finite limit of cultivation.” That is, after about 50 subcultures, these cells “degenerated” and stopped proliferating. Further research determined that senescent cells accumulate with age and established cellular senescence as a hallmark of aging.2,3 Today, cellular senescence is generally defined by an irreversible arrest of the cell cycle with accompanying stereotyped phenotypic changes.
In the past 20 years, there has been significant insight into the phenotype of senescent cells; thorough phenotypic characterization is crucial as there is yet to be any universal and specific markers to determine whether a cell is senescent. Beyond irreversible cell-cycle arrest (regulated by p16INK4a and/or p53/p21),4,5 features of senescent cells include, but are not limited to, secretion of inflammatory proteins (e.g. pro-inflammatory cytokines and chemokines, angiogenic factors),6,7 high lysosomal activity (e.g. increased senescence-associated β-galactosidase, SA-β-gal), 21 morphological differences (e.g. cell blebbing), 8 and nuclear changes (e.g. downregulated lamin B1, Figure 1(A)). 9 Due to the heterogeneity of cellular senescence, the International Cell Senescence Association published their consensus on how to identify cellular senescence, outlining detailed cellular and molecular changes, and we refer the reader to these excellent analyses.10,11
Figure 1.
Cellular senescence and the BBB. (A) A variety of different stressors can induce cell-cycle arrest including telomere attrition, oxidative stress, DNA damage, and epigenetic stress. From here, a cell can mend itself via internal mechanisms and re-enter the cell cycle or, if the damage is overtly destructive, a cell can turn on apoptotic programs to prevent damaged machinery from impacting the health of the tissue. However, in some cases (the mechanisms by which are still under investigation), a cell can stay in cell-cycle arrest and, over an undetermined period of time (anywhere between 10 days to 6 weeks in vivo), enters a state of cellular senescence where it can no longer re-enter the cell cycle. Besides irreversible cell-cycle arrest, the phenotype of cellular senescence consists of (but is not limited to) increased p16 and p21/p53, decreased nuclear lamin B1, senescence-associated heterochromatin foci (SAHF), increased senescence-associated β-galactosidase, increased reactive oxygen species, and a senescence-associated secretory phenotype (SASP) which is a pattern of secreting cytokines and proteases. (B) The BBB primarily consists of brain endothelial cells (BECs, pink) which are sealed together by tight junction proteins. BECs are embedded in a basement membrane and are physically surrounded by pericytes (yellow) and astrocytes (purple) in the brain which support BBB induction and maintenance as important components of the neurovascular unit (NVU). The schematic is of a capillary cross-section in the brain. (C) BECs predominantly exist in a quiescent state (G0 resting phase of the cell cycle). This process is regulated by Wnt/β-catenin as well as sonic hedgehog signaling. Quiescent BECs can re-enter the cell cycle with appropriate stimuli. This figure was made with BioRender. BBB: blood–brain barrier.
Cellular senescence does have beneficial features in that it prevents proliferation of damaged cells and triggers their breakdown by the immune system. However, the onset and prolonged duration of cellular senescence can lead to dysfunctional stem cells and widespread inflammation attributed to the senescence-associated secretory phenotype (SASP). This chronic senescent phenotype has been identified as a contributor to the aging brain and age-related brain diseases including Alzheimer’s disease (AD) and frontotemporal dementia.12,13 In 2015, the first senolytic therapeutic strategy, a combination of dasatinib and quercetin, was reported, 14 which boosted enthusiasm and heightened interest in the geroscience community. At the time of submission of this review, there are 16 active clinical trials that look at the effect of senolytics in a variety of age-associated diseases including chronic kidney disease, osteoarthritis, and AD.
The discovery of senolytics has strengthened the research into cellular senescence and its influence in disease. Recent studies have explored evidence for senescence in brain cells including astrocytes, 15 microglia, 16 and neurons. 17 In addition, there is significant evidence for endothelial cell senescence in age-related peripheral diseases as well as vascular-associated disease.18,19 Central nervous system (CNS) vascular dysfunction, including impairment of the microvascular blood–brain barrier (BBB), can be a major contributor to age-associated CNS diseases. However, relatively little is known about the extent that cellular senescence contributes to age- and disease-associated changes of the BBB. In this review, we will discuss the current literature that has elucidated the relations of cellular senescence to BBB dysfunction.
The vascular BBB
The vascular BBB is an essential brain interface primarily comprised of brain endothelial cells (BECs) that protect the brain from exposure to potentially harmful substances in the circulation and regulate the transport of essential nutrients and signaling molecules between brain and blood compartments (Figure 1(B)). The BBB’s interface and barrier properties are conferred, in part, by unique BEC features that include low levels of pinocytosis, tight junction proteins that limit paracellular leakage, and transporters that regulate which substances can enter or leave the brain. 20 BECs are surrounded by supportive brain cell types including astrocytes and pericytes which make up the neurovascular unit (NVU). Many excellent reviews highlight the multifaceted features of the BBB in health and development, as well as in aging and disease.20–22 Recently, multiple modes of BBB dysfunction have been associated with aging, including increased leakiness, impaired transport of molecules such as glucose, 23 amyloid-beta peptide,24,25 and xenobiotics.26,27 It is theorized that these changes at the BBB may be drivers of cognitive dysfunction in aging and may be further impaired in neurodegenerative diseases like AD.28–34 Several reports have sought to understand how epigenetic stress and mitochondrial dysfunction act; two of the nine “hallmarks of aging” contribute to BBB dysfunction.35,36 In this review, we focus on a third: cellular senescence.
Quiescent resting state of BECs
As background, it is important to distinguish between quiescence and senescence. In contrast to senescence, which is generally irreversible, 10 quiescence is a generally reversible state of cell-cycle arrest that is essential for certain types of cells with strict proliferative guidelines such as lymphocytes and stem cells. 37 Quiescent cells can share some features of senescent cells, such as increased lysosome content/SA-β-gal activity, although the molecular underpinnings can differ. 38 Notably, the BBB, and in particular BECs, primarily exist in a quiescent state (Figure 1(C)). 39 Quiescence is regulated by nutritive and growth factors and/or contact inhibition, and maintains cells in a stable state where they are non-proliferative, yet able to re-enter the cell cycle with appropriate stimuli. In the mature, healthy BBB, the quiescent endothelium exhibits very little proliferation/migration, vascular leakage, and expression of leukocyte adhesion molecules. 39 Wnt/β-catenin and sonic hedgehog signaling regulate vascular quiescence and are crucial to the integrity of the BBB.39,40 Conversely, cellular senescence can occur in response to a variety of cell stressors (including DNA damage, oncogenic mutations, telomere attrition, and oxidative stress).41–44 This review will specifically examine cellular senescence and its involvement at the BBB.
Aging
First, we will review changes at the aging BBB such as permeability, morphology, and transport kinetics. We include core features of the BBB including the basement membranes (BMs) and glycocalyx, as well as consider the effect of advanced age and relative systemic changes such as blood pressure and pulse flow.
Normative age–related changes at the vascular BBB
The vascular BBB has long been known to undergo morphological and functional changes with normative aging. 22 Studies have shown that brain capillaries have a decreased number of BECs that become more elongated with age. 45 Furthermore, there is an overall decrease in capillary diameter, particularly BECs in white matter, due to loss of cytoplasm. 46 This thinning is thought to be related to a loss of pericytes. 46 Mitochondrial number and gap junctions in BECs do not vary, which proposes that some facets of the BBB remain intact with healthy aging. More recent studies have found decreased tight junction expression 47 and evidence for a shift toward increased transcytosis 48 at the BBB with age, indicative of changes in BBB structure and function.
Whereas morphological studies found little or no evidence for BBB disruption, immunohistochemical studies have been mixed. Cuffing studies of albumin, fibrinogen, or immunoglobulin G (IgG) occurs in young as well as aged brains, with a variance too large to allow conclusions. 49 Cerebrospinal fluid (CSF)/serum ratios of albumin do increase with healthy aging, 50 but may reflect decreased CSF reabsorption rather than or in addition to BBB disruption. 51
Imaging studies using dynamic contrast-enhanced magnetic resonance imaging (MRI) have found that in healthy humans, there is a modest increase in permeability of a low-molecular weight gadolinium-based tracer in the hippocampus that occurs with age. 32 This compares to early tracer studies, which found little evidence for disruption of the BBB to sucrose, 52 albumin, 53 or horseradish peroxidase. 54 Another imaging study found increased leakage in the BBB of white matter, gray matter, and the hippocampus, where the white and gray matter leakage correlated with cognitive decline. 55 It should be noted that the degree of increased leakage that occurs with healthy aging is small and highly variable among individuals. In fact, one study using dynamic contrast-enhanced MRI found that a 90+ individual had BBB integrity matching the 20-year-old group. 55 In retrospect, the small increase and large variation explains why it has been so difficult for morphometric and immunohistochemical studies to detect disruption. Therefore, while there is a trend toward increased BBB disruption with age, this is not a universal feature of aging.
Aging is associated with changes in the transport kinetics of several substances. Peptide transport system-1 56 and the brain-to-blood transport of amyloid-beta peptide 57 decrease with aging. A reduced transport of interleukin-1 beta across the BBB has been postulated to underlie the diminished fever seen with aging. 58 Other transporters that are altered with normative aging are the large neutral amino acid transporter, the efflux transporter P-glycoprotein, and the transporters for choline, triiodothyronine, tumor necrosis factor-alpha, and glucose. 22 It is unclear the degree to which the decrease in BBB transport systems arises from a defect in BBB function versus an adaptive response of the BBB to a decreased demand for transporter substrates by the brain. The composition of the lipid membrane dictates the degree to which small, lipid soluble substances can cross the BBB by way of the non-saturable mechanism of transcellular diffusion. Fluorescence polarization studies suggest that membrane composition of BECs changes little with aging. 59
As the above illustrates, the vascular BBB undergoes structural and functional changes with normative aging. This raises the question as to whether the age-related changes in BBB are accompanied by cellular senescence of BECs of which we detail in a later section.
Normative age–related changes at the BBB extracellular matrix
Senescent cells and their associated secretory phenotype promote dysfunctional tissue turnover, which results in depletion of extracellular matrix (ECM) in some tissues and abnormal matrix deposition in others.60,61 For the brain in general, and the microvascular ECM in particular, there is an overall increase in degradative processes that is often attributed to matrix metalloproteases-2 and -9 (MMPs).62,63 The higher protease activity found in aged tissues is usually not accompanied by similar increases in inhibitors of activity, such as the tissue inhibitors of MMPs.64,65 The resulting milieu can affect the ECM on both the luminal and abluminal side of the BBB.
The luminal ECM is known as the glycocalyx, a dynamic thin gel-like layer present along the endothelium throughout the macro- and micro-vasculature. The glycocalyx is comprised primarily of heparan sulfate and chondroitin sulfate proteoglycans, non-sulfated glycosaminoglycans, and their associated binding proteins, which are constantly modified by interactions with the circulation.66,67 Content and synthesis of hyaluronic acid, a component of the glycocalyx, increases with aging. 68 Furthermore, aging is associated with a thinner glycocalyx and lower barrier function in the peripheral vasculature. 69 Thinning of the glycocalyx is multifactorial and reflects increases in protease activity and oxidative processes; at the same time, there are decreases in protective factors such as heparan sulfates and sirtuins. 70
On the abluminal side of the microvasculature is the BM, a thin and intricate layer of ECM that supports the BECs and invests the pericytes. The BM is composed of an intercalating network of laminin, collagen IV, and fibronectin interspersed with proteoglycans and glycoproteins. 71 In contrast to the dynamic turnover of the glycocalyx, the BM is relatively stable with sheets of collagen IV fibers and laminin chains that are interwoven with a repository of deposition products and post-translational modifications. The BM can thicken during normative aging, a process that is accelerated by disease processes.72,73 For example, advanced glycation end products are non-enzymatic, post-translational modifications of proteins such as type IV collagen that are increased in aging, hypertension, diabetes, and inflammatory processes that increase carbonyl stress. Several studies have demonstrated that these alterations interfere with BBB function.74,75
Advanced age and relevant systemic age-related changes
During normative aging, there are increases in systolic blood pressure and pulse pressure, with changes in diastolic blood pressure being more variable. As a result, there is an increase in the prevalence of the syndrome of isolated systolic hypertension in older adults. 76 The effect of widened pulse pressure on brain microvasculature is typically implied rather than directly studied, but there is increasing evidence that higher intravascular stress has a detrimental effect on BBB function and negatively impacts glymphatic clearance.77,78 The beneficial effect of treating hypertension on cognitive outcomes may reflect a dampening of subsequent damage to the brain microvasculature that could include stabilization of BBB ECM.79,80 Studies that administered chronic senolytics to mice suggest benefits include reducing calcifications in the vascular ECM and improving nitric oxide bioavailability to the general vasculature. 81
The off-target effects of the numerous medications that are taken by an aging population could affect cellular senescence. The range of medications that potentially influence the microvasculature is beyond the scope of this review, but certain classes of commonly used medications likely improve BBB function and mitigate senescence changes. Drugs affecting the latter would include medications that lower systolic blood pressure or glucose, such as angiotensin inhibitors, selective sodium-glucose cotransporter 2 inhibitors, and glucagon-like peptide-1 receptor agonists.82–84 The increasing use of biologics that can alter the entire immune system might dampen inflammation seen with aging (known as inflammaging) and subsequent senescence, but could also detrimentally affect compensatory processes that ward off infections. 85 Future studies will establish how senolytics that mitigate the senescent changes in the BBB influence the effect of medications on the aged brain.70,86
Brain diseases
Neurodegenerative diseases
Neurodegenerative diseases are associated with both BBB dysfunctions and with aging.87,88 This raises the possibilities that accelerated senescence of BECs could be the driver for BBB dysfunction in neurodegenerative diseases or that neurodegenerative diseases can accelerate the aging of the BBB with the accumulation of senescent BECs. BBB dysfunctions, cellular senescence, and neurodegenerative diseases all have common factors in their pathology including the mechanisms of oxidative stress, mitochondrial dysfunction, and inflammation. Therefore, one would predict that many neurodegenerative diseases or age-associated diseases with altered CNS function are associated with both BBB dysfunctions and cellular senescence. It is clear, given that senescent cells can induce other cells to become senescent in a paracrine-like fashion, that cellular senescence in other cells of the NVU is important in BBB dysfunction. It is also supported that BBB dysfunction can lead to cellular senescence in other, adjacent cells. For example, Preininger and Kaufer have reviewed how an aging, leaky BBB can induce TGFβ signaling, leading to senescence in astrocytes. 89 However, there are relatively few studies examining BEC senescence in neurodegenerative diseases to date. Brain microvessels from subjects with higher levels of tauopathy have upregulation of senescence-related genes. 90 This suggests that tau pathology can contribute to cellular senescence in BECs. More studies are necessary to study effects of other neurodegenerative proteinopathies like Parkinson’s disease (alpha synuclein) and AD (amyloid beta).
Contribution of apolipoprotein E, a common genetic risk factor for AD
Apolipoprotein E (apoE) is a lipoprotein component that helps shuttle lipids and cholesterol around the body. 91 In humans, apoE is primarily produced by the liver in the periphery, and by astrocytes and microglia in the CNS. Because plasma apoE does not enter the brain, it is possible that peripheral and CNS sources of apoE differentially influence disease processes. There are three isoforms of human apoE: E2, E3, and E4. In AD, apoE4 is the greatest known genetic risk factor and thus heavily studied in neurodegenerative contexts. 92 To date, there have been very few studies investigating the impact of apoE genotype on cellular senescence, particularly at the BBB. In vitro studies have shown quercetin, one component of the senolytic cocktail of dasatinib plus quercetin, can prevent the enhanced redox state of apoE4-treated neuroblastoma cells (SK-N-SH cells) potentially by modifying the structure of apoE4. 93 As oxidative stress can lead to senescence, preventing this E4 modification could slow this process. Other accumulating evidence suggests that apoE4 accelerates cellular senescence of microglia. 94 Cellular apoE levels can accelerate senescence by destabilizing heterochromatin, while loss of apoE resists senescence; 95 therefore, a change in the cellular apoE level could trigger senescence of a given cell, but these studies remain to be performed in BECs. However, while apoE genotype can regulate apoE protein level, apoE4 produces less apoE protein compared to apoE3 or apoE2, 96 suggesting an apoE4 genotype is not likely to lead to senescence via heterochromatin destabilization.
Importantly, human clinical trials are just beginning to investigate the impact of senolytics in AD and whether there are apoE genotype effects of cellular senescence. There is currently a National Institutes of Health (NIH) grant committed to investigating the impact of the apoE genotype on senescence in AD and vascular cognitive impairment and dementia. 97 In a preclinical study, mice overexpressing AD genes (5XFAD) crossed with apoE3/E4 transgenic mice (E3FAD, E4FAD) were fed rapamycin for 16 weeks, beginning before the onset of amyloid-beta peptide deposition, which is a distinct pathological feature in AD. 98 E4FAD mice had decreased levels of p-glycoprotein (Pgp), an efflux transporter at the BBB, and increased accumulation of amyloid-beta peptide. When fed rapamycin, a strategy that reduces the SASP phenotype (also known as a senomorphic),99,100 Pgp protein and amyloid-beta peptide levels were restored to the E3FAD level. Cerebral blood flow was also improved in E4FAD rapamycin treated animals indicating a link between cellular senescence and amyloid beta transport at the BBB. Since a senescent phenotype was not assessed in this study, there is no way to identify whether the improvements from rapamycin were due to an impact on senescence or related to its inflammation-suppression role.
apoE genotype may also influence the biological activities of senolytics. While senolytic combinations have not yet been tested, an apoE genotype effect was shown to occur in response to dietary quercetin. Following six weeks of quercetin intake, apoE3 mice had a significant reduction in blood tumor necrosis factor α (TNF-α) levels (44%) while apoE4 mice did not have a significant reduction. 101 TNF-α can directly impact the BBB 102 or can cross the BBB to act within the CNS 103 and can induce senescence in many cell types, including endothelial cells.104,105 The limited data on apoE genotype in BBB senescence clearly warrant further investigations based on the intersecting signaling pathways involved and data reported in peripheral cells.
In vitro/in vivo studies inducing cellular senescence at the BBB
As previously described, cellular senescence is one of the nine hallmarks of aging, 2 and BBB dysfunction can also occur with normative aging. 106 Both processes may be exacerbated by underlying diseases (which will be discussed). However, the extent to which brain microvascular senescence contributes to BBB dysfunction is less clear. This section reviews our current understanding of the relation of brain microvascular senescence to BBB dysfunction based on findings in cell models, rodents, and from single-cell RNA sequencing studies of human BECs.
Brain microvascular/endothelial senescence and BBB dysfunction in rodents
As discussed in previous sections, BBB dysfunction appears to be a feature of normative aging that is exacerbated in neurodegenerative disease and with associated risk factors. Aged mice have been reported to have a leaky BBB,34,107 which can be mediated in part through the decreased expression of endothelial sirtuin-1, 34 an inhibitor of endothelial cell senescence.108,109 Recent analysis of single-cell RNAseq data sets from young versus aged mouse brains found that the percentage of senescent BECs increased from 5.23% to 10.06%. Notably, the method used to calculate this increase relied on enrichment scores for a panel of senescent gene markers, taking into account both the expression levels and numbers of markers expressed in each cell. 110 The authors of this study reported unpublished findings that a similar increase in BEC senescence was found in the p16-3MR senescence reporter mice. Senescence-associated BBB dysfunction has also been demonstrated in rodent models of accelerated senescence. BubR1 hypomorphic mice (BubR1H/H) have a mutation in the BubR1 gene, resulting in reduced levels of the protein. BubR1 is a mitotic checkpoint activator protein that regulates proper chromosomal segregation and attachment to the mitotic spindle. 111 This protein is reduced with aging, and BubR1H/H mice are prone to aneuploidy and develop a progeroid phenotype which includes growth retardation, short lifespan, impaired wound healing, cardiac arrhythmias, and arterial wall stiffening.111,112 Cellular senescence is widespread in many tissues in this model. The introduction of the INK-ATTAC transgene, which expresses a drug-activatable apoptosis-inducing caspase-8 and fluorescent reporter driven by the promoter for p16Ink4a, in BubR1H/H mice results in selective elimination of senescent cells with transgenic caspase-8 activating AP20187 treatment, and improvement of aspects of the aging phenotype. 112 Increased SA-β-gal staining and p16Ink4a mRNA were also noted in cultured BECs and pericytes of BubR1H/H mice versus wild-type (WT) controls. 113 In vivo, it was shown that five-month-old BubR1H/H mice have increased BBB disruption, as measured by greater leakage of the tracers Evans blue and sodium fluorescein into the cerebral cortex in comparison to the WT controls. Reduced levels of tight junction proteins claudin-5, ZO-1, and occludin were also measured in the cerebral cortex of BubR1H/H mice versus WT controls 113 Moreover, a recent study that the stabilization of G-quadruplexes upregulates cysteine rich angiogenic inducer 61 (CCN1) and induces cellular senescence in BECs. 114 CCN1 is a matricellular protein secreted by BECs which binds to heparin sulfate proteoglycans, activating DNA damage factors, inducing expression of MMPs that are part of the SASP, and triggering cellular senescence. There was an age-associated increase in CCN1 from cerebral microvessels isolated from young (4 months) versus old (12 months) mice that was concomitant with increased senescence features. Together, these data suggest an association of brain microvascular senescence with BBB disruption; however, more mechanistic studies are needed for definitive evidence that cellular senescence in BECs or other cells of the NVU is driving the BBB disruption phenotype.
Induction of senescence in BBB cells in vitro
In vitro models of the BBB/primary cultures of BECs and other cells of the NVU have also been used to study aspects of cellular senescence. As initially described by Hayflick and Moorehead 1 in the 1960s, replicative senescence occurs when cells have surpassed their proliferative lifespan, which can be achieved by serial passaging. Stamatovic et al. 34 used two immortalized models of BECs to study effects of passage and hydrogen peroxide-induced senescence on BBB permeability. They found that both models of senescence were associated with increased levels of senescence markers and increased BBB permeability. Moreover, Stamatovic et al. found that primary mouse BECs underwent nearly complete senescence (as defined by SA-β-gal staining, upregulation of p21, DNA damage marked by γH2A.X, and heterochromatin foci) after six complete population doublings. Senescent mouse BECs showed increased BBB leakage as evidenced by reduced transendothelial electrical resistance, and increased permeability to albumin in comparison to non-senescent/low passage mouse BECs.115–117
Senescence can also be induced in vitro through physical or chemical stressors, such as ionizing radiation, chemotherapeutics, or oxidative stress and specific protocols have been described to do so.115–117 Importantly, senescence is a slow process, taking about three to seven days to develop in response to stressors. 118 Ionizing radiation of rat BECs elicited DNA damage, which resulted in greater cell death, while repair of DNA damage was slower than in neurons, astrocytes, and microglia. While ionizing radiation caused apoptosis in about 10% of cells, about 30% of surviving cells showed hallmarks of senescence that included SA-β-gal staining, increased p16Ink4a and p53, an SASP, and reduced proliferative capacity. Oxidative stress was also elevated. 119 Phenotypic characterization of cultured BECs and pericytes from young (5–8 weeks old) versus middle-aged (40–50 weeks old) mice showed that both cell types exhibited increases in SA-β-gal staining and reduced proliferation with age. Interestingly, whereas endothelial cells from older mice had significant elevations in p21 expression but not p16INK4a, pericytes had significant elevations in p16INK4a but not p21. 113 In a triculture model of primary BECs, astrocytes, and pericytes, it was shown that BECs from BubR1H/H mice were more leaky as measured by TEER and leakage to sodium fluorescein and albumin, and had abnormal expression and mislocalization of the tight junction proteins claudin-5, ZO-1, and occludin. 113 BECs and pericytes from BubR1H/H mice had increased SA-β-gal staining and increased expression of p16INK4a versus WT mice. 113 Similar to results in vivo, these studies support that BBB dysfunction is associated with cellular senescence, and that endothelial cells with a senescent phenotype form a less robust BBB in vitro.
Mechanisms by which cellular senescence could influence BBB functions
In the previous section, an overview was provided on current knowledge of how senescent changes particularly in BECs are associated with BBB dysfunction. However, theoretically senescent cells in the brain or even in the periphery could contribute to aspects of BBB dysfunction through their direct and indirect interactions with BEC (Figure 2). Cells of the NVU such as astrocytes and pericytes have established roles in supporting the development and maintenance of the BBB.120,121 Currently, there is minimal evidence informing to what extent these cell types become senescent in vivo; analysis of an existing single-cell RNAseq data set did not detect senescent pericytes, smooth muscle cells, or astrocytes in aged mice. 122 Astrocyte senescence has been observed in a number of in vitro models of disease-associated stimuli, and it is known that astrocytes in vivo adopt a more pro-inflammatory phenotype with aging and in CNS diseases. 15 In vivo, p16INK4a positive astrocytes were found to be increased in the frontal cortex of human brains with aging and AD. 63 Microglia can also have physical associations with the BBB in health and disease, resulting in beneficial or harmful effects on BBB functions that are context-dependent. 123 Increased microglia senescence occurs in the aging brain,110,122 and microglia with disease-associated transcriptomic signatures also have a senescent transcriptomic phenotype. 124 Whole body elimination of senescent cells in aged mice using a genetic/pharmacologic INK-ATTAC strategy or a senolytic drug treatment regimen resulted in a selective decrease of p16INK4a + microglia, and improved cognitive function of aged mice, further supporting that senescent microglia can drive age-associated cognitive decline. How senescence of NVU cells influences BBB functions has not yet been formally addressed but could plausibly contribute to aspects of BBB dysfunction through at least two mechanisms: (1) the loss of protective interactions with BECs and (2) the toxic contribution of the SASP (Figure 2).
Figure 2.
Cellular senescence and its possible contributions to BBB dysfunction. (A) Depiction of a healthy BBB with intact tight junctions and physical associations of BECs with astrocyte endfeet, pericytes, and microglia. (B) Changes of BBB function as a result of cellular senescence. Cellular senescence may occur in endothelial cells (green), resulting in cell-autonomous loss of BBB properties. Senescence of BBB-associated astrocytes, pericytes, and microglia (green cells) could contribute to loss of BBB maintenance/preservation following inflammatory insults. Senescent leukocytes, and particularly monocytes/macrophages (green), may also interact with the BBB or traffic across into brain parenchyma to induce neuroinflammation. The SASP from senescent cells in the brain, circulating leukocytes, or peripheral organs could lead to increases in SASP proteins (green dots) in brain ISF or blood that directly alters non-senescent brain endothelial cell functions, resulting in increased leakiness and leukocyte trafficking (purple cell). SASP proteins could also interact with BBB-associated cell types, altering their BBB-maintenance functions, or modulating their secretory phenotype. An increase in the secretion of inflammatory proteins by non-senescent pericytes (orange dots), astrocytes (purple dots, brain side), endothelial cells (pink dots), microglia (red dots), or monocytes/macrophages (purple dots, blood side) may contribute to the senescence-associated inflammatory milieu/localized reactive gliosis, and propagate BBB dysfunction. This figure was made with BioRender. BBB: blood–brain barrier; BEC: brain endothelial cell; SASP: senescence-associated secretory phenotype; ISF: interstitial fluid.
In addition to CNS cells of the NVU that may contribute to BBB dysfunction, senescence of cells that lie outside of the brain could also impact various functions of the BBB. For example, SASP factors can enter the blood stream as secreted proteins or packaged in exosomes. 125 Thus, an SASP from peripheral cells that are distant from the brain could interact with the luminal surface of BECs, affecting BEC function. A subset of p16INK4a positive infiltrating brain myeloid cells, together with senescent microglia, contributes to age-associated leukocyte trafficking across the BBB into the brain. 126 Systemic elimination of these senescent myeloid cells was associated with improved nest building in aged mice. 126 In summary, endothelial cells of the BBB and associated cells of the NVU can adopt senescent phenotypes with aging and age-associated diseases. However, to what extent cellular senescence within the BBB/NVU/CNS contributes to BBB dysfunction versus cellular senescence at distal sites in blood or peripheral organs remains unclear.
Cellular senescence as it relates to BBB dysfunction in peripheral/systemic diseases
Diabetes/obesity
Diabetes and obesity can elicit many stress pathways causing premature cellular senescence. As recently reviewed, there are multiple metabolic drivers of senescence that fit in with aging including mitochondrial dysfunction, redox imbalances, and hyperglycemia. 127 Exposure of endothelial cells to high glucose alone can trigger a senescent phenotype. 128 In addition, retinal endothelial cells undergo premature senescence as measured by SA-β-gal positive staining following exposure to high glucose. 129 Since senescence of cells can elicit a secretory phenotype, senescence of any cell within the NVU could contribute to the neurovascular inflammation associated with AD.
We have previously shown in a diabetic mouse model that treatment with a mitochondrial carbonic anhydrase inhibitor decreases oxidative stress and prevents BBB disruption and diabetes-induced pericyte loss at the NVU.130,131 The impact of this inhibitor on cellular senescence remains to be investigated. However, as carbonic anhydrases are increased with aging in brain 132 and with diabetes in heart, 133 targeting this enzyme would decrease oxidative stress and mitochondrial dysfunction, which could slow senescence.
While it is known that metabolic disease can induce many of the same stress pathways at the BBB that often lead to senescence of cells,134–136 whether senescence of BBB cells occurs in metabolic diseases has not been largely investigated. The blood–retina barrier (BRB) is a window into the BBB. Studying the BRB, it was found that arginase 1 is a contributor to diabetes-induced senescence of endothelial cells. 137 Arginase is an enzyme that uses l-arginine as a substrate. When the l-arginine supply is insufficient, nitric oxide synthase generates superoxide, limiting synthesis of nitric oxide, and contributing not only to oxidative stress but also to vascular dysfunction.
Peripheral senescent cells can promote insulin resistance, suggesting senescent cells can drive metabolic disease. 138 In either high-fat diet-induced obesity or in a genetic obese model (db/db mice), levels of senescent cells predominantly in the visceral adipose tissue were increased compared to controls. Senescent cell clearance through dasatinib and quercetin treatment improved glucose tolerance and insulin sensitivity but had no impact on body weight. As stated previously, due to the SASP, senescence in any peripheral organ could affect the BBB due to contact through the circulation.
Regular exercise can slow endothelial cell senescence in humans. 139 In endothelial cells collected from the forearm in young sedentary, aged sedentary, and aged individuals who exercised, protein levels of p21 and p16 were increased with age, with no age-related increases in the exercised individuals. Levels of these proteins were also inversely correlated with vascular endothelial function. These data suggest aerobic exercise may be able to protect against endothelial dysfunction that occurs with age by preventing endothelial cell senescence.
Inflammatory diseases
Chronic inflammation slows tissue remodeling and is often accompanied by cellular senescence. Inflammatory diseases that are organ-specific (e.g. originating in the lung like asthma, pneumonia, and COVID-19) as well as systemic diseases (e.g. arthritis) can contribute to vascular senescence. Because all organs are connected by the circulatory system, changes in one organ or cell type could impact other organs or cell types through endocrine signaling.
SARS-CoV-2 is an exemplary viral infection of the respiratory tract that has affected most of the world’s population. While SARS-CoV-2 primarily targets the lungs, a cytokine storm accompanying infection dramatically elevates many cytokines in blood, thus affecting every organ within the body.140,141 We have shown that a fragment of the viral attachment protein of the SARS-CoV-2 virus, S1, can cross the BBB. 142 There are also multiple different ways SARS-CoV-2 can have a detrimental impact on the BBB. 143 Increased oxidative stress and release of cytokines are two ways that the virus could directly impact the BBB. Whether infection increases senescence in BECs is still unknown. However, in a Nature Aging publication earlier this year, it was shown that SARS-CoV-2 infection can induce senescence of neighboring, uninfected cells in a paracrine manner. 144 Administration of dasatinib plus quercetin to rodents infected with SARS-CoV-2 decreased levels of inflammatory factors classified as the SASP.144,145 Therefore, it is plausible that SARS-CoV-2 could induce BBB dysfunction directly by inducing senescence of BECs or indirectly through senescence of adjacent cells or through systemic effects of the SASP.
During systemic inflammation, multiple studies have shown disruption of BBB integrity or enhanced BBB transport of inflammatory factors.146–148 Beyond the vagal afferents and choroid plexus, BECs are the first CNS cell type exposed to the damaging effects of peripheral inflammation. Inflammatory factors can trigger MMP secretion, endoplasmic reticulum stress, and mitochondrial damage. Gastrointestinal disorders not only release inflammatory cytokines but also alter the gut microbiota which can impact the BBB. 149 Thus, various peripheral inflammatory diseases can impact cellular senescence at the BBB.
Cancer
While this review focuses on the unfavorable aspects of cellular senescence, we must acknowledge its biological advantage as a tumor suppressant. Cellular acquisition of a fully senescent phenotype prevents premalignant and malignant tumorous cells from undergoing cell-cycle division. Moreover, the adoption of an SASP phenotype can act on neighboring cancer cells, improving the vasculature for drug delivery and recruiting immune cells to the cancerous region. 150 Many chemotherapeutics have been shown to stimulate therapy-induced senescence (TIS) such as doxorubicin, etoposide, and cisplatin.118,151 TIS cells have been identified in tumors following radiation or chemotherapy, 152 and are known to secrete the inflammatory cytokine interleukin 1α (IL-1α), a crucial SASP initiator and regulator. 152
However, it is also known that 17–75% of cancer victims treated with various chemotherapeutics, including doxorubicin, acquire chemotherapy-induced cognitive dysfunction (CICD) or “chemobrain.” 153 CICD is a syndrome of cognitive impairment that can persist long after treatment, and likely has BBB involvement. 154 TIS cells could be contributing to this debilitating phenotype. Adding to the complexity, it is known that TIS cells can induce ECM cleavage and growth factor release, which can promote tumorigenesis and eventual metastasis. 150 Thus, it is difficult to identify definitively whether senescent cells are pro-tumorigenic or antitumorigenic. Current work is investigating treatment paradigms of TIS coupled with senolytics which could halt tumor progression and provide an opportunity for senescent cell clearance. 150
Systemic cancer cells release IL-6, a key component of the SASP. This can not only disrupt the BBB but also crosses the intact BBB,155,156 thus participating in a neuro-immune axis. 20 If this disruption of the BBB is prevented by eliminating IL-6, the host can live longer with the tumor. 157 Tumor-bearing mouse hosts have increased systemic IL-6. When given antibodies directed at the IL-6 receptor, BBB disruption is prevented and lethality is decreased, despite no change in tumor burden. Similar results were obtained in a Drosophila tumor-bearing model. 157 These data suggest that morbidity in cancer can be improved by targeting downstream effects of inflammatory cytokines like IL-6, which is a predominant SASP factor.
Conclusions
The field of cellular senescence is rapidly expanding. Preclinical studies have demonstrated that senolytics have efficacy in the brain, but the specific cellular targets are unknown.158–160 In this review, we have presented evidence of normative age–associated changes to the BBB and carefully analyzed how some of these changes are reminiscent of cellular senescence. We have also considered age-associated disease in the brain and the periphery, and considered how these changes could induce cellular senescence at the BBB and in particular BECs. Finally, we have examined multiple in vitro and in vivo studies that induce cellular senescence in BBB model systems. Importantly, the hallmarks of aging are intertwined; for example, cellular senescence can be induced by telomere shortening and DNA damage. 2 Moreover, cellular senescence can cause stem cell exhaustion and altered intracellular communication. We have concentrated on cellular senescence, but recognize that some of the age-associated changes in BBB functions that we highlighted could be due, in part, to other hallmarks of aging.
While we have presented multiple studies evaluating BECs and BBB-associated cells like pericytes and astrocytes, more work is needed. Cellular senescence can take anywhere between three and seven days in vitro or ten days to six weeks in vivo to establish. However, it is unknown if the development and progression of the phenotype over time cause different changes to BBB function (e.g. the acute vs chronic phenotype). Furthermore, BECs can induce senescence in nearby cells and adjacent cells can induce senescence in BECs, but it is still not clear what cells or induction mechanism impacts normative age–related BBB changes. With the emergence of clinical trials investigating senolytics, more work is urgently needed to understand the importance and roles of cellular senescence at the BBB. It is still not known if cellular senescence is maladaptive or adaptive, and if it varies by tissue or induction mechanism. The review presented here seeks to highlight the complexities of cellular senescence on the BBB and encourage more studies on this intriguing topic.
Footnotes
Authors’ Contributions: All authors participated in the conceptualization, writing, and revision of this publication.
The author(s) declared no potential conflicts of interest with respect to the research, authorship, and/or publication of this article.
Funding: The author(s) disclosed receipt of the following financial support for the research, authorship, and/or publication of this article: This work was supported by the Biological Mechanisms of Healthy Aging Training Program NIH T32AG066574 (R.C.K.), the Joe W. & Dorthy Dorsett Brown Foundation (M.A.E), P30 DK017047-44 (E.M.R.), P30 AG066509 (E.M.R.), the Veterans Administration (W.A.B.), and R03AG051071 (M.J.R.) and R21AG073676 (M.J.R.).
ORCID iDs: Rachel C Knopp
https://orcid.org/0000-0002-1091-3829
William A Banks
https://orcid.org/0000-0002-0496-3619
References
- 1.Hayflick L, Moorhead PS. The serial cultivation of human diploid cell strains. Exp Cell Res 1961;25:585–621 [DOI] [PubMed] [Google Scholar]
- 2.Lopez-Otin C, Blasco MA, Partridge L, Serrano M, Kroemer G. The hallmarks of aging. Cell 2013;153:1194–217 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 3.Schneider EL. Aging and cultured human skin fibroblasts. J Invest Dermatol 1979;73:15–8 [DOI] [PubMed] [Google Scholar]
- 4.Krishnamurthy J, Torrice C, Ramsey MR, Kovalev GI, Al-Regaiey K, Su L, Sharpless NE. Ink4a/Arf expression is a biomarker of aging. J Clin Invest 2004;114:1299–307 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 5.Mijit M, Caracciolo V, Melillo A, Amicarelli F, Giordano A. Role of p53 in the regulation of cellular senescence. Biomolecules 2020;10:420. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 6.Coppe JP, Patil CK, Rodier F, Sun Y, Munoz DP, Goldstein J, Nelson PS, Desprez PY, Campisi J. Senescence-associated secretory phenotypes reveal cell-nonautonomous functions of oncogenic RAS and the p53 tumor suppressor. Plos Biol 2008;6:2853–68 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 7.Dimri GP, Lee X, Basile G, Acosta M, Scott G, Roskelley C, Medrano EE, 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 U S A 1995;92:9363–7 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 8.Herranz N, Gil J. Mechanisms and functions of cellular senescence. J Clin Invest 2018;128:1238–46 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 9.Lukasova E, Kovarik A, Kozubek S. Consequences of lamin B1 and lamin B receptor downregulation in senescence. Cells 2018;7:11. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 10.Gorgoulis V, Adams PD, Alimonti A, Bennett DC, Bischof O, Bishop C, Campisi J, Collado M, Evangelou K, Ferbeyre G, Gil J, Hara E, Krizhanovsky V, Jurk D, Maier AB, Narita M, Niedernhofer L, Passos JF, Robbins PD, Schmitt CA, Sedivy J, Vougas K, von Zglinicki T, Zhou D, Serrano M, Demaria M. Cellular senescence: defining a path forward. Cell 2019;179:813–27 [DOI] [PubMed] [Google Scholar]
- 11.Kohli J, Wang B, Brandenburg SM, 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;16:2471–98 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 12.Salech F, SanMartin CD, Concha-Cerda J, Romero-Hernandez E, Ponce DP, Liabeuf G, Rogers NK, Murgas P, Bruna B, More J, Behrens MI. Senescence markers in peripheral blood mononuclear cells in amnestic mild cognitive impairment and Alzheimer’s disease. Int J Mol Sci 2022;23:9387. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 13.Gaikwad S, Puangmalai N, Bittar A, Montalbano M, Garcia S, McAllen S, Bhatt N, Sonawane M, Sengupta U, Kayed R. Tau oligomer induced HMGB1 release contributes to cellular senescence and neuropathology linked to Alzheimer’s disease and frontotemporal dementia. Cell Rep 2021;36:109419. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 14.Zhu Y, Tchkonia T, Pirtskhalava T, Gower AC, Ding H, Giorgadze N, Palmer AK, Ikeno Y, Hubbard GB, Lenburg M, O’Hara SP, LaRusso NF, Miller JD, Roos CM, Verzosa GC, LeBrasseur NK, Wren JD, Farr JN, Khosla S, Stout MB, McGowan SJ, Fuhrmann-Stroissnigg H, Gurkar AU, Zhao J, Colangelo D, Dorronsoro A, Ling YY, Barghouthy AS, Navarro DC, Sano T, Robbins PD, Niedernhofer LJ, Kirkland JL. The Achilles’ heel of senescent cells: from transcriptome to senolytic drugs. Aging Cell 2015;14:644–58 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 15.Cohen J, Torres C. Astrocyte senescence: evidence and significance. Aging Cell 2019;18:e12937 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 16.Greenwood EK, Brown DR. Senescent microglia: the key to the ageing brain? Int J Mol Sci 2021;22:4402. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 17.Sikora E, Bielak-Zmijewska A, Dudkowska M, Krzystyniak A, Mosieniak G, Wesierska M, Wlodarczyk J. Cellular senescence in brain aging. Front Aging Neurosci 2021;13:646924. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 18.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;105:1541–4 [DOI] [PubMed] [Google Scholar]
- 19.Jia G, Aroor AR, Jia C, Sowers JR. Endothelial cell senescence in aging-related vascular dysfunction. Biochim Biophys Acta Mol Basis Dis 2019;1865:1802–9 [DOI] [PubMed] [Google Scholar]
- 20.Erickson MA, Banks WA. Neuroimmune axes of the blood-brain barriers and blood-brain interfaces: bases for physiological regulation, disease states, and pharmacological interventions. Pharmacol Rev 2018;70:278–314 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 21.Daneman R, Prat A. The blood-brain barrier. Cold Spring Harb Perspect Biol 2015;7:a020412 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 22.Banks WA, Reed MJ, Logsdon AF, Rhea EM, Erickson MA. Healthy aging and the blood-brain barrier. Nature Aging 2021;1:243–54 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 23.Winkler EA, Nishida Y, Sagare AP, Rege SV, Bell RD, Perlmutter D, Sengillo JD, Hillman S, Kong P, Nelson AR, Sullivan JS, Zhao Z, Meiselman HJ, Wendy RB, Soto J, Abel ED, Makshanoff J, Zuniga E, De Vivo DC, Zlokovic BV. GLUT1 reductions exacerbate Alzheimer’s disease vasculo-neuronal dysfunction and degeneration. Nat Neurosci 2015;18:521–30 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 24.Sagare AP, Bell RD, Zlokovic BV. Neurovascular dysfunction and faulty amyloid beta-peptide clearance in Alzheimer disease. Cold Spring Harb Perspect Med 2012;2:a011452 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 25.Banks WA, Kumar VB, Farr SA, Nakaoke R, Robinson SM, Morley JE. Impairments in brain-to-blood transport of amyloid-beta and reabsorption of cerebrospinal fluid in an animal model of Alzheimer’s disease are reversed by antisense directed against amyloid-beta protein precursor. J Alzheimers Dis 2011;23:599–605 [DOI] [PubMed] [Google Scholar]
- 26.van Assema DM, Lubberink M, Boellaard R, Schuit RC, Windhorst AD, Scheltens P, Lammertsma AA, van Berckel BN. P-glycoprotein function at the blood-brain barrier: effects of age and gender. Mol Imaging Biol 2012;14:771–6 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 27.Chiu C, Miller MC, Monahan R, Osgood DP, Stopa EG, Silverberg GD. P-glycoprotein expression and amyloid accumulation in human aging and Alzheimer’s disease: preliminary observations. Neurobiol Aging 2015;36:2475–82 [DOI] [PubMed] [Google Scholar]
- 28.Sweeney MD, Zhao Z, Montagne A, Nelson AR, Zlokovic BV. Blood-brain barrier: from physiology to disease and back. Physiol Rev 2019;99:21–78 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 29.Nation DA, Sweeney MD, Montagne A, Sagare AP, D’Orazio LM, Pachicano M, Sepehrband F, Nelson AR, Buennagel DP, Harrington MG, Benzinger TLS, Fagan AM, Ringman JM, Schneider LS, Morris JC, Chui HC, Law M, Toga AW, Zlokovic BV. Blood-brain barrier breakdown is an early biomarker of human cognitive dysfunction. Nat Med 2019;25:270–6 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 30.Arvanitakis Z, Capuano AW, Leurgans SE, Bennett DA, Schneider JA. Relation of cerebral vessel disease to Alzheimer’s disease dementia and cognitive function in elderly people: a cross-sectional study. Lancet Neurol 2016;15:934–43 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 31.Iturria-Medina Y, Sotero RC, Toussaint PJ, Mateos-Perez JM, Evans AC, Alzheimer’s Disease Neuroimaging Initiative. Early role of vascular dysregulation on late-onset Alzheimer’s disease based on multifactorial data-driven analysis. Nat Commun 2016;7:11934. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 32.Montagne A, Barnes SR, Sweeney MD, Halliday MR, Sagare AP, Zhao Z, Toga AW, Jacobs RE, Liu CY, Amezcua L, Harrington MG, Chui HC, Law M, Zlokovic BV. Blood-brain barrier breakdown in the aging human hippocampus. Neuron 2015;85:296–302 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 33.Elahy M, Jackaman C, Mamo JC, Lam V, Dhaliwal SS, Giles C, Nelson D, Takechi R. Blood-brain barrier dysfunction developed during normal aging is associated with inflammation and loss of tight junctions but not with leukocyte recruitment. Immun Ageing 2015;12:2. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 34.Stamatovic SM, Martinez-Revollar G, Hu A, Choi J, Keep RF, Andjelkovic AV. Decline in Sirtuin-1 expression and activity plays a critical role in blood-brain barrier permeability in aging. Neurobiol Dis 2019;126:105–16 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 35.Herrera EA, González-Candia A. Gestational hypoxia and blood-brain barrier permeability: early origins of cerebrovascular dysfunction induced by epigenetic mechanisms. Front Physiol 2021;12:717550. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 36.Salmina AB, Kharitonova EV, Gorina YV, Teplyashina EA, Malinovskaya NA, Khilazheva ED, Mosyagina AI, Morgun AV, Shuvaev AN, Salmin VV, Lopatina OL, Komleva YK. Blood-brain barrier and neurovascular unit in vitro models for studying mitochondria-driven molecular mechanisms of neurodegeneration. Int J Mol Sci 2021;22:4661. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 37.Terzi MY, Izmirli M, Gogebakan B. The cell fate: senescence or quiescence. Mol Biol Rep 2016;43:1213–20 [DOI] [PubMed] [Google Scholar]
- 38.Cho S, Hwang ES. Status of mTOR activity may phenotypically differentiate senescence and quiescence. Mol Cells 2012;33:597–604 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 39.Ricard N, Bailly S, Guignabert C, Simons M. The quiescent endothelium: signalling pathways regulating organ-specific endothelial normalcy. Nat Rev Cardiol 2021;18:565–80 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 40.Alvarez JI, Dodelet-Devillers A, Kebir H, Ifergan I, Fabre PJ, Terouz S, Sabbagh M, Wosik K, Bourbonniere L, Bernard M, van Horssen J, de Vries HE, Charron F, Prat A. The Hedgehog pathway promotes blood-brain barrier integrity and CNS immune quiescence. Science 2011;334:1727–31 [DOI] [PubMed] [Google Scholar]
- 41.Kang C, Xu Q, Martin TD, Li MZ, Demaria M, Aron L, Lu T, Yankner BA, Campisi J, Elledge SJ. The DNA damage response induces inflammation and senescence by inhibiting autophagy of GATA4. Science 2015;349:aaa5612 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 42.Baker DJ, Alimirah F, van Deursen JM, Campisi J, Hildesheim J. Oncogenic senescence: a multi-functional perspective. Oncotarget 2017;8:27661–72 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 43.Zhu Y, Liu X, Ding X, Wang F, Geng X. Telomere and its role in the aging pathways: telomere shortening, cell senescence and mitochondria dysfunction. Biogerontology 2019;20:1–16 [DOI] [PubMed] [Google Scholar]
- 44.Kudryavtseva AV, Krasnov GS, Dmitriev AA, Alekseev BY, Kardymon OL, Sadritdinova AF, Fedorova MS, Pokrovsky AV, Melnikova NV, Kaprin AD, Moskalev AA, Snezhkina AV. Mitochondrial dysfunction and oxidative stress in aging and cancer. Oncotarget 2016;7:44879–905 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 45.Bär T. Morphometric evaluation of capillaries in different laminae of rat cerebral cortex by automatic image analysis. Adv Neurol 1978;20:1–9 [PubMed] [Google Scholar]
- 46.Stewart PA, Magliocco M, Hayakawa K, Farrell CL, Del Maestro RF, Girvin J, Kaufmann JC, Vinters HV, Gilbert J. A quantitative analysis of blood-brain barrier ultrastructure in the aging human. Microvasc Res 1987;33:270–82 [DOI] [PubMed] [Google Scholar]
- 47.Bors L, Tóth K, Tóth EZ, Bajza Á, Csorba A, Szigeti K, Máthé D, Perlaki G, Orsi G, Tóth GK, Erdő F. Age-dependent changes at the blood-brain barrier. A comparative structural and functional study in young adult and middle aged rats. Brain Res Bull 2018;139:269–77 [DOI] [PubMed] [Google Scholar]
- 48.Yang AC, Stevens MY, Chen MB, Lee DP, Stähli D, Gate D, Contrepois K, Chen W, Iram T, Zhang L, Vest RT, Chaney A, Lehallier B, Olsson N, du Bois H, Hsieh R, Cropper HC, Berdnik D, Li L, Wang EY, Traber GM, Bertozzi CR, Luo J, Snyder MP, Elias JE, Quake SR, James ML, Wyss-Coray T. Physiological blood-brain transport is impaired with age by a shift in transcytosis. Nature 2020;583:425–30 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 49.Vorbrodt AW, Dobrogowska DH. Immunocytochemical evaluation of blood-brain barrier to endogenous albumin in adult, newborn and aged mice. Folia Histochem Cytobiol 1994;32:63–70 [PubMed] [Google Scholar]
- 50.Farrall AJ, Wardlaw JM. Blood-brain barrier: ageing and microvascular disease – systematic review and meta-analysis. Neurobiol Aging 2009;30:337–52 [DOI] [PubMed] [Google Scholar]
- 51.Chen RL. Is it appropriate to use albumin CSF/plasma ratio to assess blood brain barrier permeability? Neurobiol Aging 2011;32:1338–9 [DOI] [PubMed] [Google Scholar]
- 52.Rapoport SI, Ohno K, Pettigrew KD. Blood-brain barrier permeability in senescent rats. J Gerontol 1979;34:162–9 [DOI] [PubMed] [Google Scholar]
- 53.Sankar R, Blossom E, Clemons K, Charles P. Age-associated changes in the effects of amphetamine on the blood-brain barrier of rats. Neurobiol Aging 1983;4:65–8 [DOI] [PubMed] [Google Scholar]
- 54.Rudick RA, Buell SJ. Integrity of blood-brain barrier to peroxidase in senescent mice. Neurobiol Aging 1983;4:283–7 [DOI] [PubMed] [Google Scholar]
- 55.Verheggen ICM, de Jong JJA, van Boxtel MPJ, Postma AA, Jansen JFA, Verhey FRJ, Backes WH. Imaging the role of blood-brain barrier disruption in normal cognitive ageing. Geroscience 2020;42:1751–64 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 56.Banks WA, Kastin AJ. Aging and the blood-brain barrier: changes in the carrier-mediated transport of peptides in rats. Neuroscience Letters 1985;61:171–5 [DOI] [PubMed] [Google Scholar]
- 57.Silverberg GD, Messier AA, Miller MC, Machan JT, Majmudar SS, Stopa EG, Donahue JE, Johanson CE. Amyloid efflux transporter expression at the blood-brain barrier declines in normal aging. J Neuropathol Exp Neurol 2010;69:1034–43 [DOI] [PubMed] [Google Scholar]
- 58.McLay RN, Kastin AJ, Zadina JE. Passage of interleukin-1-beta across the blood-brain barrier is reduced in aged mice: a possible mechanism for diminished fever in aging. Neuroimmunomodulation 2000;8:148–53 [DOI] [PubMed] [Google Scholar]
- 59.Mooradian AD, Smith TL. The effect of age on lipid composition and order of rat cerebral microvessels. Neurochem Res 1992;17:233–7 [DOI] [PubMed] [Google Scholar]
- 60.Vannier DR, Shapeti A, Chuffart F, Planus E, Manet S, Rivier P, Destaing O, Albiges-Rizo C, Van Oosterwyck H, Faurobert E. CCM2-deficient endothelial cells undergo a ROCK-dependent reprogramming into senescence-associated secretory phenotype. Angiogenesis 2021;24:843–60 [DOI] [PubMed] [Google Scholar]
- 61.Vasko R, Xavier S, Chen J, Lin CH, Ratliff B, Rabadi M, Maizel J, Tanokuchi R, Zhang F, Cao J, Goligorsky MS. Endothelial sirtuin 1 deficiency perpetrates nephrosclerosis through downregulation of matrix metalloproteinase-14: relevance to fibrosis of vascular senescence. J Am Soc Nephrol 2014;25:276–91 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 62.Ritzel RM, Lai YJ, Crapser JD, Patel AR, Schrecengost A, Grenier JM, Mancini NS, Patrizz A, Jellison ER, Morales-Scheihing D, Venna VR, Kofler JK, Liu F, Verma R, McCullough LD. Aging alters the immunological response to ischemic stroke. Acta Neuropathol 2018;136: 89–110 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 63.Bhat R, Crowe EP, Bitto A, Moh M, Katsetos CD, Garcia FU, Johnson FB, Trojanowski JQ, Sell C, Torres C. Astrocyte senescence as a component of Alzheimer’s disease. Plos One 2012;7:e45069 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 64.Fujimoto M, Takagi Y, Aoki T, Hayase M, Marumo T, Gomi M, Nishimura M, Kataoka H, Hashimoto N, Nozaki K. Tissue inhibitor of metalloproteinases protect blood-brain barrier disruption in focal cerebral ischemia. J Cereb Blood Flow Metab 2008;28:1674–85 [DOI] [PubMed] [Google Scholar]
- 65.West MD, Pereira-Smith OM, Smith JR. Replicative senescence of human skin fibroblasts correlates with a loss of regulation and overexpression of collagenase activity. Exp Cell Res 1989;184:138–47 [DOI] [PubMed] [Google Scholar]
- 66.Kutuzov N, Flyvbjerg H, Lauritzen M. Contributions of the glycocalyx, endothelium, and extravascular compartment to the blood-brain barrier. Proc Natl Acad Sci U S A 2018;115:E9429–38 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 67.Logsdon AF, Rhea EM, Reed M, Banks WA, Erickson MA. The neurovascular extracellular matrix in health and disease. Exp Biol Med (Maywood) 2021;246:835–44. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 68.Reed MJ, Damodarasamy M, Banks WA. The extracellular matrix of the blood-brain barrier: structural and functional roles in health, aging, and Alzheimer’s disease. Tissue Barriers 2019;7:1651157. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 69.Machin DR, Bloom SI, Campbell RA, Phuong TTT, Gates PE, Lesniewski LA, Rondina MT, Donato AJ. Advanced age results in a diminished endothelial glycocalyx. Am J Physiol Heart Circ Physiol 2018;315:H531–9 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 70.Cheung TM, Yan JB, Fu JJ, Huang J, Yuan F, Truskey GA. Endothelial cell senescence increases traction forces due to age-associated changes in the Glycocalyx and SIRT1. Cell Mol Bioeng 2015;8:63–75 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 71.Leclech C, Natale CF, Barakat AI. The basement membrane as a structured surface – role in vascular health and disease. J Cell Sci 2020;133:jcs239889 [DOI] [PubMed] [Google Scholar]
- 72.Ceafalan LC, Fertig TE, Gheorghe TC, Hinescu ME, Popescu BO, Pahnke J, Gherghiceanu M. Age-related ultrastructural changes of the basement membrane in the mouse blood-brain barrier. J Cell Mol Med 2019;23:819–27 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 73.Thomsen MS, Routhe LJ, Moos T. The vascular basement membrane in the healthy and pathological brain. J Cereb Blood Flow Metab 2017;37:3300–17 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 74.Shimizu F, Sano Y, Tominaga O, Maeda T, Abe MA, Kanda T. Advanced glycation end-products disrupt the blood-brain barrier by stimulating the release of transforming growth factor-beta by pericytes and vascular endothelial growth factor and matrix metalloproteinase-2 by endothelial cells in vitro. Neurobiol Aging 2013;34:1902–12 [DOI] [PubMed] [Google Scholar]
- 75.Hussain M, Bork K, Gnanapragassam VS, Bennmann D, Jacobs K, Navarette-Santos A, Hofmann B, Simm A, Danker K, Horstkorte R. Novel insights in the dysfunction of human blood-brain barrier after glycation. Mech Ageing Dev 2016;155:48–54 [DOI] [PubMed] [Google Scholar]
- 76.Mitchell GF, Parise H, Benjamin EJ, Larson MG, Keyes MJ, Vita JA, Vasan RS, Levy D. Changes in arterial stiffness and wave reflection with advancing age in healthy men and women: the Framingham Heart Study. Hypertension 2004;43:1239–45 [DOI] [PubMed] [Google Scholar]
- 77.Tsao CW, Seshadri S, Beiser AS, Westwood AJ, Decarli C, Au R, Himali JJ, Hamburg NM, Vita JA, Levy D, Larson MG, Benjamin EJ, Wolf PA, Vasan RS, Mitchell GF. Relations of arterial stiffness and endothelial function to brain aging in the community. Neurology 2013;81:984–91 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 78.Hansen TW, Li Y, Staessen JA, Jeppesen J, Rasmussen S, Wang JG, Thijs L, Ibsen H, Safar ME, Torp-Pedersen C. Independent prognostic value of the ambulatory arterial stiffness index and aortic pulse wave velocity in a general population. J Hum Hypertens 2008;22:214–6 [DOI] [PubMed] [Google Scholar]
- 79.Elias MF, Elias PK, Sullivan LM, Wolf PA, D’Agostino RB. Lower cognitive function in the presence of obesity and hypertension: the Framingham heart study. Int J Obes Relat Metab Disord 2003;27:260–8 [DOI] [PubMed] [Google Scholar]
- 80.Bálint AR, Puskás T, Menyhárt Á, Kozák G, Szenti I, Kónya Z, Marek T, Bari F, Farkas E. Aging Impairs cerebrovascular reactivity at preserved resting cerebral arteriolar tone and vascular density in the laboratory rat. Front Aging Neurosci 2019;11:301. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 81.Roos CM, Zhang B, Palmer AK, Ogrodnik MB, Pirtskhalava T, Thalji NM, Hagler M, Jurk D, Smith LA, Casaclang-Verzosa G, Zhu Y, Schafer MJ, Tchkonia T, Kirkland JL, Miller JD. Chronic senolytic treatment alleviates established vasomotor dysfunction in aged or atherosclerotic mice. Aging Cell 2016;15:973–7 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 82.Sueta D, Koibuchi N, Hasegawa Y, Toyama K, Uekawa K, Katayama T, Ma M, Nakagawa T, Waki H, Maeda M, Ogawa H, Kim-Mitsuyama S. Blood pressure variability, impaired autonomic function and vascular senescence in aged spontaneously hypertensive rats are ameliorated by angiotensin blockade. Atherosclerosis 2014;236:101–7 [DOI] [PubMed] [Google Scholar]
- 83.Park SH, Farooq MA, Gaertner S, Bruckert C, Qureshi AW, Lee HH, Benrahla D, Pollet B, Stephan D, Ohlmann P, Lessinger JM, Mayoux E, Auger C, Morel O, Schini-Kerth VB. Empagliflozin improved systolic blood pressure, endothelial dysfunction and heart remodeling in the metabolic syndrome ZSF1 rat. Cardiovasc Diabetol 2020;19:19. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 84.Almutairi M, Al Batran R, Ussher JR. Glucagon-like peptide-1 receptor action in the vasculature. Peptides 2019;111:26–32 [DOI] [PubMed] [Google Scholar]
- 85.Xu S, Ilyas I, Little PJ, Li H, Kamato D, Zheng X, Luo S, Li Z, Liu P, Han J, Harding IC, Ebong EE, Cameron SJ, Stewart AG, Weng J. Endothelial dysfunction in atherosclerotic cardiovascular diseases and beyond: from mechanism to pharmacotherapies. Pharmacol Rev 2021;73:924–67 [DOI] [PubMed] [Google Scholar]
- 86.Ya J, Kadir RRA, Bayraktutan U. Delay of endothelial cell senescence protects cerebral barrier against age-related dysfunction: role of senolytics and senomorphics. Tissue Barriers. Epub ahead of print 26 July 2022. DOI: 10.1080/21688370.2022.2103353 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 87.Sweeney MD, Sagare AP, Zlokovic BV. Blood-brain barrier breakdown in Alzheimer disease and other neurodegenerative disorders. Nat Rev Neurol 2018;14:133–50 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 88.Hou Y, Dan X, Babbar M, Wei Y, Hasselbalch SG, Croteau DL, Bohr VA. Ageing as a risk factor for neurodegenerative disease. Nat Rev Neurol 2019;15:565–81 [DOI] [PubMed] [Google Scholar]
- 89.Preininger MK, Kaufer D. Blood-brain barrier dysfunction and astrocyte senescence as reciprocal drivers of neuropathology in aging. Int J Mol Sci 2022;23:6217. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 90.Bryant AG, Hu M, Carlyle BC, Arnold SE, Frosch MP, Das S, Hyman BT, Bennett RE. Cerebrovascular senescence is associated with tau pathology in Alzheimer’s disease. Front Neurol 2020;11:575953. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 91.Rebeck GW. The role of APOE on lipid homeostasis and inflammation in normal brains. J Lipid Res 2017;58:1493–9 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 92.Strittmatter WJ, Roses AD. Apolipoprotein E and Alzheimer’s disease. Annu Rev Neurosci 1996;19:53–77 [DOI] [PubMed] [Google Scholar]
- 93.Mountaki C, Dafnis I, Panagopoulou EA, Vasilakopoulou PB, Karvelas M, Chiou A, Karathanos VT, Chroni A. Mechanistic insight into the capacity of natural polar phenolic compounds to abolish Alzheimer’s disease-associated pathogenic effects of apoE4 forms. Free Radic Biol Med 2021;171:284–301 [DOI] [PubMed] [Google Scholar]
- 94.Fernandez CG, Hamby ME, McReynolds ML, Ray WJ. The role of APOE4 in disrupting the homeostatic functions of astrocytes and microglia in aging and Alzheimer’s disease. Front Aging Neurosci 2019;11:14. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 95.Zhao H, Ji Q, Wu Z, Wang S, Ren J, Yan K, Wang Z, Hu J, Chu Q, Hu H, Cai Y, Want Q, Huang D, Ji Z, Li J, Carlos Izpisua Belmonte J, Song M, Zhang W, Qu J, Liu G-H. Destabilizing heterochromatin by APOE mediates senescence. Nature Aging 2022;2:303–16 [DOI] [PubMed] [Google Scholar]
- 96.Riddell DR, Zhou H, Atchison K, Warwick HK, Atkinson PJ, Jefferson J, Xu L, Aschmies S, Kirksey Y, Hu Y, Wagner E, Parratt A, Xu J, Li Z, Zaleska MM, Jacobsen JS, Pangalos MN, Reinhart PH. Impact of apolipoprotein E (ApoE) polymorphism on brain ApoE levels. J Neurosci 2008;28:11445–53 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 97.Kanekiyo T. Endothelial cell senescence and APOE4 in vascular cognitive impairment and dementia, 2022, https://grantome.com/grant/NIH/RF1-AG071226-01
- 98.Lin AL, Parikh I, Yanckello LM, White RS, Hartz AMS, Taylor CE, McCulloch SD, Thalman SW, Xia M, McCarty K, Ubele M, Head E, Hyder F, Sanganahalli BG. APOE genotype-dependent pharmacogenetic responses to rapamycin for preventing Alzheimer’s disease. Neurobiol Dis 2020;139:104834. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 99.Wang R, Yu Z, Sunchu B, Shoaf J, Dang I, Zhao S, Caples K, Bradley L, Beaver LM, Ho E, Löhr CV, Perez VI. Rapamycin inhibits the secretory phenotype of senescent cells by a Nrf2-independent mechanism. Aging Cell 2017;16:564–74 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 100.Niedernhofer LJ, Robbins PD. Senotherapeutics for healthy ageing. Nat Rev Drug Discov 2018;17:377. [DOI] [PubMed] [Google Scholar]
- 101.Boesch-Saadatmandi C, Wolffram S, Minihane AM, Rimbach G. Effect of apoE genotype and dietary quercetin on blood lipids and TNF-alpha levels in apoE3 and apoE4 targeted gene replacement mice. Br J Nutr 2009;101:1440–3 [DOI] [PubMed] [Google Scholar]
- 102.Williams LM, Fujimoto T, Weaver RR, Logsdon AF, Evitts KM, Young JE, Banks WA, Erickson MA. Prolonged culturing of iPSC-derived brain endothelial-like cells is associated with quiescence, downregulation of glycolysis, and resistance to disruption by an Alzheimer’s brain milieu. Fluids Barriers CNS 2022;19:10. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 103.Gutierrez EG, Banks WA, Kastin AJ. Murine tumor necrosis factor alpha is transported from blood to brain in the mouse. J Neuroimmunol 1993;47:169–76 [DOI] [PubMed] [Google Scholar]
- 104.Kandhaya-Pillai R, Miro-Mur F, Alijotas-Reig J, Tchkonia T, Kirkland JL, Schwartz S. TNFα-senescence initiates a STAT-dependent positive feedback loop, leading to a sustained interferon signature, DNA damage, and cytokine secretion. Aging (Albany NY) 2017;9:2411–35 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 105.Khan SY, Awad EM, Oszwald A, Mayr M, Yin X, Waltenberger B, Stuppner H, Lipovac M, Uhrin P, Breuss JM. Premature senescence of endothelial cells upon chronic exposure to TNFα can be prevented by N-acetyl cysteine and plumericin. Sci Rep 2017;7:39501. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 106.Banks WA, Reed MJ, Logsdon AF, Rhea EM, Erickson MA. Healthy aging and the blood-brain barrier. Nat Aging 2021;1:243–54 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 107.Nyul-Toth A, Tarantini S, DelFavero J, Yan F, Balasubramanian P, Yabluchanskiy A, Ahire C, Kiss T, Csipo T, Lipecz A, Farkas AE, Wilhelm I, Krizbai IA, Tang Q, Csiszar A, Ungvari Z. Demonstration of age-related blood-brain barrier disruption and cerebromicrovascular rarefaction in mice by longitudinal intravital two-photon microscopy and optical coherence tomography. Am J Physiol Heart Circ Physiol 2021;320:H1370–92 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 108.Xu C, Wang L, Fozouni P, Evjen G, Chandra V, Jiang J, Lu C, Nicastri M, Bretz C, Winkler JD, Amaravadi R, Garcia BA, Adams PD, Ott M, Tong W, Johansen T, Dou Z, Berger SL. SIRT1 is downregulated by autophagy in senescence and ageing. Nat Cell Biol 2020;22:1170–9 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 109.Ota H, Akishita M, Eto M, Iijima K, Kaneki M, Ouchi Y. Sirt1 modulates premature senescence-like phenotype in human endothelial cells. J Mol Cell Cardiol 2007;43:571–9 [DOI] [PubMed] [Google Scholar]
- 110.Kiss T, Nyúl-Tóth Á, Balasubramanian P, Tarantini S, Ahire C, DelFavero J, Yabluchanskiy A, Csipo T, Farkas E, Wiley G, Garman L, Csiszar A, Ungvari Z. Single-cell RNA sequencing identifies senescent cerebromicrovascular endothelial cells in the aged mouse brain. Geroscience 2020;42:429–44 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 111.Baker DJ, Dawlaty MM, Wijshake T, Jeganathan KB, Malureanu L, van Ree JH, Crespo-Diaz R, Reyes S, Seaburg L, Shapiro V, Behfar A, Terzic A, van de Sluis B, van Deursen JM. Increased expression of BubR1 protects against aneuploidy and cancer and extends healthy lifespan. Nat Cell Biol 2013;15:96–102 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 112.Baker DJ, Wijshake T, Tchkonia T, LeBrasseur NK, Childs BG, van de Sluis B, Kirkland JL, van Deursen JM. Clearance of p16Ink4a-positive senescent cells delays ageing-associated disorders. Nature 2011;479:232–6 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 113.Yamazaki Y, Baker DJ, Tachibana M, Liu CC, van Deursen JM, Brott TG, Bu G, Kanekiyo T. Vascular cell senescence contributes to blood-brain barrier breakdown. Stroke 2016;47:1068–77 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 114.Noh B, Blasco-Conesa MP, Lai YJ, Ganesh BP, Urayama A, Moreno-Gonzalez I, Marrelli SP, McCullough LD, Moruno-Manchon JF. G-quadruplexes stabilization upregulates CCN1 and accelerates aging in cultured cerebral endothelial cells. Front Aging 2021;2:797562. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 115.Noren Hooten N, Evans MK. Techniques to induce and quantify cellular senescence. J Vis Exp 2017;123:55533. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 116.Warnon C, Bouhjar K, Ninane N, Verhoyen M, Fattaccioli A, Fransolet M, Lambert de, Rouvroit C, Poumay Y, Piel G, Mottet D, Debacq-Chainiaux F. HDAC2 and 7 down-regulation induces senescence in dermal fibroblasts. Aging (Albany NY) 2021;13:17978–8005 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 117.Hu X, Zhang H. Doxorubicin-induced cancer cell senescence shows a time delay effect and is inhibited by epithelial-mesenchymal transition (EMT). Med Sci Monit 2019;25:3617–23 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 118.Ewald JA, Desotelle JA, Wilding G, Jarrard DF. Therapy-induced senescence in cancer. J Natl Cancer Inst 2010;102:1536–46 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 119.Ungvari Z, Podlutsky A, Sosnowska D, Tucsek Z, Toth P, Deak F, Gautam T, Csiszar A, Sonntag WE. Ionizing radiation promotes the acquisition of a senescence-associated secretory phenotype and impairs angiogenic capacity in cerebromicrovascular endothelial cells: role of increased DNA damage and decreased DNA repair capacity in microvascular radiosensitivity. J Gerontol A Biol Sci Med Sci 2013;68:1443–57 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 120.Daneman R, Zhou L, Kebede AA, Barres BA. Pericytes are required for blood-brain barrier integrity during embryogenesis. Nature 2010;468:562–6 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 121.Heithoff BP, George KK, Phares AN, Zuidhoek IA, Munoz-Ballester C, Robel S. Astrocytes are necessary for blood-brain barrier maintenance in the adult mouse brain. Glia 2021;69:436–72 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 122.Kiss T, Nyúl-Tóth Á, DelFavero J, Balasubramanian P, Tarantini S, Faakye J, Gulej R, Ahire C, Ungvari A, Yabluchanskiy A, Wiley G, Garman L, Ungvari Z, Csiszar A. Spatial transcriptomic analysis reveals inflammatory foci defined by senescent cells in the white matter, hippocampi and cortical grey matter in the aged mouse brain. Geroscience 2022;44:661–81 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 123.Knopp RC, Banks WA, Erickson MA. Physical associations of microglia and the vascular blood-brain barrier and their importance in development, health, and disease. Curr Opin Neurobiol 2022;77:102648. [DOI] [PubMed] [Google Scholar]
- 124.Hu Y, Fryatt GL, Ghorbani M, Obst J, Menassa DA, Martin-Estebane M, Muntslag TAO, Olmos-Alonso A, Guerrero-Carrasco M, Thomas D, Cragg MS, Gomez-Nicola D. Replicative senescence dictates the emergence of disease-associated microglia and contributes to Abeta pathology. Cell Rep 2021;35:109228. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 125.Basisty N, Kale A, Jeon OH, Kuehnemann C, Payne T, Rao C, Holtz A, Shah S, Sharma V, Ferrucci L, Campisi J, Schilling B. A proteomic atlas of senescence-associated secretomes for aging biomarker development. Plos Biol 2020;18:e3000599 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 126.Zhang X, Pearsall VM, Carver CM, Atkinson EJ, Clarkson BDS, Grund EM, Baez-Faria M, Pavelko KD, Kachergus JM, White TA, Johnson RK, Malo CS, Gonzalez-Suarez AM, Ayasoufi K, Johnson KO, Tritz ZP, Fain CE, Khadka RH, Ogrodnik M, Jurk D, Zhu Y, Tchkonia T, Revzin A, Kirkland JL, Johnson AJ, Howe CL, Thompson EA, LeBrasseur NK, Schafer MJ. Rejuvenation of the aged brain immune cell landscape in mice through p16-positive senescent cell clearance. Nat Commun 2022;13:5671. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 127.Wiley CD, Campisi J. The metabolic roots of senescence: mechanisms and opportunities for intervention. Nat Metab 2021;3:1290–301 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 128.Phoenix A, Chandran R, Ergul A. Cerebral microvascular senescence and inflammation in diabetes. Front Physiol 2022;13:864758. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 129.Bertelli PM, Pedrini E, Hughes D, McDonnell S, Pathak V, Peixoto E, Guduric-Fuchs J, Stitt AW, Medina RJ. Long term high glucose exposure induces premature senescence in retinal endothelial cells. Front Physiol 2022;13:929118. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 130.Price TO, Eranki V, Banks WA, Ercal N, Shah GN. Topiramate treatment protects blood-brain barrier pericytes from hyperglycemia-induced oxidative damage in diabetic mice. Endocrinology 2012;153: 362–72 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 131.Salameh TS, Shah GN, Price TO, Hayden MR, Banks WA. Blood-brain barrier disruption and neurovascular unit dysfunction in diabetic mice: protection with the mitochondrial carbonic anhydrase inhibitor topiramate. J Pharmacol Exp Ther 2016;359:452–9 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 132.Pollard A, Shephard F, Freed J, Liddell S, Chakrabarti L. Mitochondrial proteomic profiling reveals increased carbonic anhydrase II in aging and neurodegeneration. Aging (Albany NY) 2016;8:2425–36 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 133.Torella D, Ellison GM, Torella M, Vicinanza C, Aquila I, Iaconetti C, Scalise M, Marino F, Henning BJ, Lewis FC, Gareri C, Lascar N, Cuda G, Salvatore T, Nappi G, Indolfi C, Torella R, Cozzolino D, Sasso FC. Carbonic anhydrase activation is associated with worsened pathological remodeling in human ischemic diabetic cardiomyopathy. J Am Heart Assoc 2014;3:e000434 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 134.Bogush M, Heldt NA, Persidsky Y. Blood brain barrier injury in diabetes: unrecognized effects on brain and cognition. J Neuroimmune Pharmacol 2017;12:593–601 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 135.Banks WA, Rhea EM. The blood-brain barrier, oxidative stress, and insulin resistance. Antioxidants (Basel) 2021;10:1695. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 136.Rhea EM, Salameh TS, Logsdon AF, Hanson AJ, Erickson MA, Banks WA. Blood-brain barriers in obesity. AAPS J 2017;19:921–30 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 137.Shosha E, Xu Z, Narayanan SP, Lemtalsi T, Fouda AY, Rojas M, Xing J, Fulton D, Caldwell RW, Caldwell RB. Mechanisms of diabetes-induced endothelial cell senescence: role of arginase 1. Int J Mol Sci 2018;19:1215. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 138.Palmer AK, Xu M, Zhu Y, Pirtskhalava T, Weivoda MM, Hachfeld CM, Prata LG, van Dijk TH, Verkade E, Casaclang-Verzosa G, Johnson KO, Cubro H, Doornebal EJ, Ogrodnik M, Jurk D, Jensen MD, Chini EN, Miller JD, Matveyenko A, Stout MB, Schafer MJ, White TA, Hickson LJ, Demaria M, Garovic V, Grande J, Arriaga EA, Kuipers F, von Zglinicki T, LeBrasseur NK, Campisi J, Tchkonia T, Kirkland JL. Targeting senescent cells alleviates obesity-induced metabolic dysfunction. Aging Cell 2019;18:e12950 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 139.Rossman MJ, Kaplon RE, Hill SD, McNamara MN, Santos-Parker JR, Pierce GL, Seals DR, Donato AJ. Endothelial cell senescence with aging in healthy humans: prevention by habitual exercise and relation to vascular endothelial function. Am J Physiol Heart Circ Physiol 2017;313:H890–5 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 140.Gavriatopoulou M, Korompoki E, Fotiou D, Ntanasis-Stathopoulos I, Psaltopoulou T, Kastritis E, Terpos E, Dimopoulos MA. Organ-specific manifestations of COVID-19 infection. Clin Exp Med 2020;20:493–506 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 141.Cron RQ, Caricchio R, Chatham WW. Calming the cytokine storm in COVID-19. Nat Med 2021;27:1674–5 [DOI] [PubMed] [Google Scholar]
- 142.Rhea EM, Logsdon AF, Hansen KM, Williams LM, Reed MJ, Baumann KK, Holden SJ, Raber J, Banks WA, Erickson MA. The S1 protein of SARS-CoV-2 crosses the blood-brain barrier in mice. Nat Neurosci 2021;24:368–78 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 143.Erickson MA, Rhea EM, Knopp RC, Banks WA. Interactions of SARS-CoV-2 with the blood-brain barrier. Int J Mol Sci 2021;22:2681. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 144.Tsuji S, Minami S, Hashimoto R, Konishi Y, Suzuki T, Kondo T, Sasai M, Torii S, Ono C, Shichinohe S, Sato S, Wakita M, Okumura S, Nakano S, Matsudaira T, Matsumoto T, Kawamoto S, Yamamoto M, Watanabe T, Matsuura Y, Takayama K, Kobayashi T, Okamoto T, Hara E. SARS-CoV-2 infection triggers paracrine senescence and leads to a sustained senescence-associated inflammatory response. Nature Aging 2022;2:115–24 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 145.Lee S, Yu Y, Trimpert J, Benthani F, Mairhofer M, Richter-Pechanska P, Wyler E, Belenki D, Kaltenbrunner S, Pammer M, Kausche L, Firsching TC, Dietert K, Schotsaert M, Martínez-Romero C, Singh G, Kunz S, Niemeyer D, Ghanem R, Salzer HJF, Paar C, Mülleder M, Uccellini M, Michaelis EG, Khan A, Lau A, Schönlein M, Habringer A, Tomasits J, Adler JM, Kimeswenger S, Gruber AD, Hoetzenecker W, Steinkellner H, Purfürst B, Motz R, Di Pierro F, Lamprecht B, Osterrieder N, Landthaler M, Drosten C, García-Sastre A, Langer R, Ralser M, Eils R, Reimann M, Fan DNY, Schmitt CA. Virus-induced senescence is a driver and therapeutic target in COVID-19. Nature 2021;599:283–9 [DOI] [PubMed] [Google Scholar]
- 146.Varatharaj A, Galea I. The blood-brain barrier in systemic inflammation. Brain Behav Immun 2017;60:1–12 [DOI] [PubMed] [Google Scholar]
- 147.Galea I. The blood-brain barrier in systemic infection and inflammation. Cell Mol Immunol 2021;18:2489–501 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 148.Erickson MA, Liang WS, Fernandez EG, Bullock KM, Thysell JA, Banks WA. Genetics and sex influence peripheral and central innate immune responses and blood-brain barrier integrity. Plos One 2018; 13:e0205769 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 149.Logsdon AF, Erickson MA, Rhea EM, Salameh TS, Banks WA. Gut reactions: how the blood-brain barrier connects the microbiome and the brain. Exp Biol Med (Maywood) 2018;243:159–65 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 150.Wang L, Lankhorst L, Bernards R. Exploiting senescence for the treatment of cancer. Nat Rev Cancer 2022;22:340–55 [DOI] [PubMed] [Google Scholar]
- 151.Carpenter VJ, Saleh T, Gewirtz DA. Senolytics for cancer therapy: is all that glitters really gold? Cancers (Basel) 2021;13:723. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 152.Orjalo AV, Bhaumik D, Gengler BK, Scott GK, Campisi J. Cell surface-bound IL-1alpha is an upstream regulator of the senescence-associated IL-6/IL-8 cytokine network. Proc Natl Acad Sci U S A 2009;106:17031–6 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 153.Wefel JS, Schagen SB. Chemotherapy-related cognitive dysfunction. Curr Neurol Neurosci Rep 2012;12:267–75 [DOI] [PubMed] [Google Scholar]
- 154.Nguyen LD, Ehrlich BE. Cellular mechanisms and treatments for chemobrain: insight from aging and neurodegenerative diseases. EMBO Mol Med 2020;12:e12075 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 155.Threlkeld SW, Lynch JL, Lynch KM, Sadowska GB, Banks WA, Stonestreet BS. Ovine proinflammatory cytokines cross the murine blood-brain barrier by a common saturable transport mechanism. Neuroimmunomodulation 2010;17:405–10 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 156.Luheshi G, Gay J, Rothwell N. Circulating IL-6 is transported into the brain via a saturable transport mechanism in the rat. Br J Pharmacol 1994;112:637P [Google Scholar]
- 157.Kim J, Chuang HC, Wolf NK, Nicolai CJ, Raulet DH, Saijo K, Bilder D. Tumor-induced disruption of the blood-brain barrier promotes host death. Dev Cell 2021;56:2712–21.e4 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 158.Ogrodnik M, Evans SA, Fielder E, Victorelli S, Kruger P, Salmonowicz H, Weigand BM, Patel AD, Pirtskhalava T, Inman CL, Johnson KO, Dickinson SL, Rocha A, Schafer MJ, Zhu Y, Allison DB, von Zglinicki T, LeBrasseur NK, Tchkonia T, Neretti N, Passos JF, Kirkland JL, Jurk D. Whole-body senescent cell clearance alleviates age-related brain inflammation and cognitive impairment in mice. Aging Cell 2021;20:e13296 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 159.Zhang P, Kishimoto Y, Grammatikakis I, Gottimukkala K, Cutler RG, Zhang S, Abdelmohsen K, Bohr VA, Misra Sen J, Gorospe M, Mattson MP. Senolytic therapy alleviates Aβ-associated oligodendrocyte progenitor cell senescence and cognitive deficits in an Alzheimer’s disease model. Nat Neurosci 2019;22:719–28 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 160.Lim S, Kim TJ, Kim YJ, Kim C, Ko SB, Kim BS. Senolytic therapy for cerebral ischemia-reperfusion injury. Int J Mol Sci 2021;22:11967. [DOI] [PMC free article] [PubMed] [Google Scholar]


