Skip to main content
The Journals of Gerontology Series A: Biological Sciences and Medical Sciences logoLink to The Journals of Gerontology Series A: Biological Sciences and Medical Sciences
. 2024 Mar 22;79(7):glae083. doi: 10.1093/gerona/glae083

Exercise to Mitigate Cerebrovascular Aging: A Geroscience Perspective

Amani M Norling 1,2,, Lewis A Lipsitz 3,4
Editor: Roger A Fielding5
PMCID: PMC11167493  PMID: 38516994

Abstract

Aging is characterized by a progressive loss of cellular functions that increase the risk of developing chronic diseases, vascular dysfunction, and neurodegenerative conditions. The field of geroscience has identified cellular and molecular hallmarks of aging that may serve as targets for future interventions to reduce the risk of age-related disease and disability. These hallmarks include genomic instability, telomere attrition, epigenetic alterations, loss of proteostasis, deregulated nutrient sensing, mitochondrial dysfunction, cellular senescence, stem cell exhaustion, and altered intercellular communication. Several studies show that exercise may favorably affect these processes and thereby have antiaging properties. The primary mechanisms through which exercise confers protective benefits in the brain are still incompletely understood. To better understand these effects and leverage them to help promote brain health, we present current findings supporting the notion that adaptive responses to exercise play a pivotal role in mitigating the hallmarks of aging and their effects on the aging cerebrovasculature, and ultimately contribute to the maintenance of brain function across the healthspan.

Keywords: Brain, Cardiorespiratory fitness, Cognition, Hallmarks of aging


Aging entails dynamic cellular and molecular changes that result in loss of function across multiple organ systems, including the brain (1). In the brain, aging is associated with an increased risk of neurodegenerative pathologies such as Alzheimer’s disease (AD) (2) and related cognitive disorders (3). To better understand the fundamental mechanisms of aging that may lead to disease, Lopez-Otin and colleagues identified 12 “Hallmarks of Aging” (1) including genomic instability, telomere attrition, epigenetic alterations, loss of proteostasis, disabled macroautophagy, deregulated nutrient sensing, mitochondrial dysfunction, cellular senescence, stem cell exhaustion, altered intercellular communication, chronic inflammation, and dysbiosis.

The introduction of this framework provided a systematic perspective on the biological complexity of aging, which allowed for the identification of key processes and facilitated the development of targeted interventions to reduce or reverse specific effects of cellular aging. But perhaps most importantly, the framework integrated various levels of aging, from molecular to cellular to systemic, offering a comprehensive view of how different aspects of aging interact with each other. Building upon the concept of geroscience, which proposes that these “hallmarks of aging” increase a person’s vulnerability to age-related disease, here we provide a review of how these age-related biological changes interact with each other, disrupt normal cerebrovascular mechanisms, and may ultimately lead to cognitive decline. Moreover, we present a novel interpretation highlighting the role of exercise as a key modulator of biological and cerebrovascular aging, and, in turn, cognitive aging. In this review, we present current knowledge while also highlighting the critical gaps in research regarding the influence of exercise on the intricate processes of cerebrovascular aging. By leveraging the fundamental biological and physiological mechanisms underlying cerebrovascular disease and cognitive decline, our goals are to suggest new opportunities for future translational geroscience research, and pave the way for more efficacious, personalized exercise interventions that effectively preserve and enhance cognitive function with aging.

Lifestyle variables may influence the hallmarks of aging and their effects on brain function. Modifiable cardiovascular risk factors such as obesity, diabetes, and hypertension can exacerbate cellular aging leading to earlier and more pronounced vascular diseases, neuropathologies, and cognitive decline (4). Conversely, countermeasures such as a commitment to a healthful diet, maintaining optimal blood glucose and blood pressure levels, avoidance of excess adiposity, and engagement in regular exercise, are widely recommended approaches to ward off cerebrovascular abnormalities and their harmful consequences on brain function (5). Although adherence to any of the healthful lifestyle behaviors contributes to the maintenance of vascular and cerebrovascular health, exercise and its clinical correlate, cardiorespiratory fitness (CRF), are crucial contributors. However, despite the highly recognized benefits of exercise, the underlying mechanisms through which these benefits translate to cognitive resilience in aging remain elusive. In this review, we propose that exercise plays a major role in the vital connection between the hallmarks of aging, the cerebrovasculature, and maintenance of cognitive function (Table 1; Figure 1). Under this framework, the effects of exercise in promoting CRF and mitigating molecular and cellular aging (87,88) provide a compelling argument for its prevailing role in preserving brain function in aging (89).

Table 1.

The Role of Exercise in Mitigating the Cellular Effects of Aging

Hallmarks of Aging Cellular Hallmarks of Aging Effects Exercise Effects
Genomic instability ↑DNA damage (6)
↑DNA mutations (6,7)
↑Apoptosis (8)
↑Senescence (9,10)
↑P53 (9,10)
↑Failure of BBB (11)
↑Waste product buildup (12–14)
↑Hypoperfusion (15)
↑Neuronal damage (15)
↓ROS (16)
↑VEGF (17,18)
↑DNA repair (19,20)
↓Methylation (21)
↓Genomic instability (22)
Telomeres ↑Shortening (23) ↑Genomic instability (23)
↑Cellular senescence (23)
↑Telomerase (24)
↑Clusterin (25)
↓SASP (26)
↓Telomere Shortening (27)
Epigenetic alterations ↓DNA methylation (28)
↑Dysregulated histone modification (29)
↑Senescence (30)
↑Inflammation (30)
↑Atherosclerosis (28)
↑Vascular diseases (28)
↑Methylation (31)
↑Nrf2 (32)
↑SIRT1 (33)
↓Inflammatory cytokines (34).
↑SOD (35,36)
Loss of proteostasis ↑Misfolded proteins (37) ↓Endothelial function (38)
↓Glymphatic clearance (39)
↓Misfolded proteins (40)
↓Protein aggregation (41)
↑Proteosome activity (42)
↑Glymphatic clearance (41)
Nutrient sensing ↓AMPK (43)
↓Sirtuins (43)
↓IGF-1 (43)
↑Cellular metabolic dysfunction (44)
↑Endothelial dysfunction (45)
↑AMPK (46)
↑IGF-1 (47)
↑Sirtuins (48)
↑NAD+ (49)
↑PGC-1α (50)
↑Vascular function (51,52)
Mitochondrial dysfunction ↓Autophagy/mitophagy (53)
↑ROS (53)
↑Inflammatory cytokines (53,54)
↑Endothelial dysfunction (55,56)
↓Cellular function (54,57)
↔mTOR (58)
↑Autophagy/ Mitophagy (59)
↑p53 (60)
↑Mitochondrial biogenesis (61)
↑Mitochondrial DNA repair (60)
↑Vascular function (59)
↑Capillary density (59)
Cellular senescence ↑SASPs (62)
↓NO (63)
↓Endothelial function (64)
↑Failure of BBB (65)
↑Neuroinflammation (66)
↓Waste product removal (67)
↓Neuronal function (63)
↑P16 (68)
↑NO (69)
↔p53 (68)
↓ROS (68)
Stem cell exhaustion ↓Stem cell activity (70) ↑Vascular disease (71)
↓Cerebral blood flow (72)
↑Endothelial progenitor cells (73)
↑SDF-1 (74)
↑NO (75)
↑P13K/AKT (76,77)
Cellular communication ↓Endocrine signaling (78)
↓Neuronal signaling (78)
↑Immune cell malfunction (70)
↑Inflammation (70)
↑Waste products (79,80)
↓Vascular inflammation (81)
↑Myokine IL-6 (82)
↑IL-10 (83,84)
↑IL-1 (83,85)
↓TNF-α (83,86)

Notes: AMPK = adenosine monophosphate kinase; IGF-1 = insulin like growth factor-1; IL = interleukins 10, 6, 1; mTOR = mammalian target of rapamycin; NAD+ = nicotinamide adenine dicleotide; NO = nitric oxide; Nrf2 = nuclear factor erythroid-2-related factor 2; P13k/AKT = phosphoinositide 3-kinases/protein kinase; p53 = protein 53;p16 = tumor suppressor protein; PGC-1 = peroxisome proliferator-activated receptor gamma coactivator1-α; ROS = reactive oxygen species; SASP = senescence associated secretory phenotype; SDF-1 = stromal cell-derived factor-1; SIRT1 = Sirtuin-1; SOD = superoxide dismutase; TNF-α = tumor necrosis factor; VEGF = vascular endothelial growth factor.

Figure 1.

Figure 1.

Aging and cardiovascular risk factors interact and affect the hallmarks of aging, which have downstream effects on vascular function. The consequential loss of cerebrovascular integrity leads to neurodegenerative changes and cognitive impairment. Exercise inhibits the cellular effects of aging and mitigates cardiovascular risk factors, resulting in maintenance of brain health and cognition across the healthspan.

Importantly, although many of the benefits of exercise can be explained through the lens of molecular and cellular aging, it is important to clarify that it is unlikely that these changes mediate the totality of the exercise–cerebrovascular risk relationship, especially when considering the direct and acute physiological effects of exercise on cardiac output and cerebral blood flow (CBF). Clarifying these intricate relationships may lead to more effective strategies to promote maintenance of brain health and cognition in aging, potentially reducing the prevalence and impact of neurodegenerative diseases.

Although we fully acknowledge that a discussion encompassing all 12 hallmarks offers a more comprehensive perspective on the aging process, the thrust of our discussion focuses on those that affect cerebrovasculature and cognitive health and may be directly influenced by exercise. We begin our narrative review with a discussion of the cerebrovasculature, hemodynamics, and how they are affected in aging, followed by brief discussions of the hallmarks of aging that affect the brain. For each hallmark, we present evidence for the protective role of exercise on the fundamental mechanisms of aging and how these effects may translate into the maintenance of cerebrovascular health and cognitive function in advanced age.

Search Method

We conducted a comprehensive search of relevant literature across various academic databases such as PubMed, Scopus, and Google Scholar using a combination of relevant keywords and advanced search filters (hallmarks of aging and relevant terms; aging; vascular; cerebrovascular; geroscience; exercise; physical activity). These terms were combined using the Boolean operator “AND” with terms specific to hallmarks of aging such as “genomic instability,” “p53,” “autophagy,” AND “vascular,” “brain” and “exercise.” In addition, we scanned reference lists from relevant manuscripts to identify additional studies.

The Cerebrovasculature, Hemodynamics, and Aging

Although the brain constitutes 2% of body weight, this highly metabolic organ (90) receives 15% of the cardiac output and 20% of the available systemic oxygen via an intricate vascular network that spans more than 400 miles (91). Due to the critical link between the brain’s high metabolic demands and its requirement for a continuous blood supply (90), brain metabolism and CBF are tightly linked and can be influenced by fluctuations in arterial blood pressure, intracranial pressure, and systemic CO2 and O2 levels (91).

To satisfy its critical need for CBF, the brain is endowed with an autoregulatory system that maintains a relatively constant blood flow over a wide physiologic range of perfusion pressures. Although the exact determinants of cerebral autoregulation are not yet fully understood, vasoactive agents, such as nitric oxide (NO), H+, K+, and adenosine, are released when CBF falls and elicits dilation of vessels (91). In the absence of neurodegenerative disease, brain energy metabolism and neuronal activity are directly coordinated such that cerebral activation is consistently followed within seconds by dilation of blood vessels and an increase in CBF (92). The relationship between task-dependent neural activation and the corresponding increase in CBF, termed neurovascular coupling (NVC) (93), is a fundamental physiological process that ensures an adequate blood supply to metabolically active regions of the brain.

With aging, however, cellular and molecular alterations in the brain’s blood vessels result in loss of structural integrity and compromised cerebral hemodynamics, leading to ischemic neuronal damage and associated cognitive dysfunction (94). For example, microvascular changes predispose the cerebral circulation to hypoperfusion (95,96), precipitating the breakdown of the blood–brain barrier (BBB), neuroinflammation, cerebral microhemorrhages, impairments in CBF regulation, and ultimately neurodegeneration and cognitive impairments (94). Similarly, disruptions in NVC can lead to a mismatch between neuronal activity and blood supply, triggering ischemic damage, neuronal dysfunction, cognitive decline, and various neurological disorders (93). Other factors such as systemic and cerebral buildup of lipids and atherosclerotic plaques (97), thickening and stiffening of arterial walls, and loss of pericytes that surround the cerebral capillaries (98) precipitate further endothelial dysfunction (95,99). A key feature of the ensuing endothelial dysfunction is the marked decrease in the production of the vasodilator NO and an increase in the vasoconstrictor endothelin-1 (100,101), which can impair CBF regulation and potentially contribute to the development of neurovascular disorders (102).

Moreover, age-related elevations in arterial blood pressure can have direct effects on CBF. In people with impaired cerebral autoregulation, which may occur in the presence of untreated hypertension, as blood pressure increases, blood flow to the brain may also increase (91), raising the risk of cerebral hyperperfusion. Consequently, to protect the brain from the detrimental effects of hyperperfusion, cerebral vessels vasoconstrict, thus initiating compensatory reductions in CBF (91). However, with long-term hypertension management, as shown in the SPRINT MIND trial (103), improvements in cerebral autoregulatory capacity and carotid distensibility were possible, potentially leading to increased CBF (103). Our earlier findings support the notion that the management of blood pressure in older adults with hypertension can have beneficial outcomes on both CBF and carotid distensibility, suggesting a restoration of autoregulatory efficiency with appropriate treatment (104). Conversely, reductions in CBF due to orthostatic hypotension, postprandial hypotension, or other daily hypotensive events can precipitate ischemic damage to watershed regions of the brain, resulting in ischemic injury, and neuroinflammation. In support of this notion, animal and human studies show that a decrease in perfusion is associated with the accumulation of white matter lesions (105) in the deep watershed zones of the brain, which significantly increase the risk of dementia and age-related cognitive impairment (102).

Loss of arterial elasticity is another significant age-related vascular change. Vascular stiffness, particularly in the aorta, increases flow pulsatility to high-perfusion organs, such as the brain. The increase in pulsatility results in cerebral microvascular damage and subsequent cognitive decline in older adults (106). Additionally, arterial stiffness can impair the brain’s autoregulatory mechanism leaving the brain vulnerable to extremes in perfusion pressures, contributing to cognitive decline (107). Furthermore, arterial stiffness increases cardiac left ventricular afterload. This increase causes myocardial stiffening, left ventricular hypertrophy, and decreased diastolic ventricular filling, thereby decreasing cardiac output (Q), particularly during preload reduction (108). Due to the dependence of the brain on cardiac blood supply, reductions in Q can compromise cerebral white matter integrity (109–111) and have been shown to disrupt proteostasis, leading to an abnormal buildup of cellular waste products such as amyloid beta (Aβ), hyperphosphorylated tau (p-tau), and neurofibrillary tangles in the brain (12–14). Although the exact mechanisms underlying neurodegeneration and cognitive decline in AD have yet to be fully defined, recent attempts identified the role of activated microglia (112) and neuroinflammation (113) in the pathogenesis of the disease. Promising evidence indicates that alterations in the cerebrovasculature may be mitigated by physical exercise, which has been associated with enhanced endothelial function and vascular health (88,114,115).

Hallmarks of Aging, Their Impact on the Cerebrovasculature, and the Effects of Exercise

The fundamental mechanisms of biological aging (116) are not distinct processes, but interact to affect cellular function (94). For instance, genomic instability, telomere attrition, epigenetic changes, and loss of proteostasis are incontrovertibly harmful cellular events associated with aging and are therefore considered primary “hallmarks” of aging. Disruptions in nutrient sensing, mitochondrial functions, and cellular senescence are termed antagonistic hallmarks because they initially compensate or counteract the damage induced by the primary hallmarks, but under chronic conditions, become harmful over time. Lastly, integrative hallmarks such as stem cell depletion and disrupted cell-to-cell communication, which manifest after irreparable damage from the primary and antagonistic hallmarks, can impair a cell’s repair capacity and cause further cellular degeneration (1,23,38,62,71,116–120).

Genomic Instability

Genomic instability refers to damage in the DNA sequence of a cell, which may be isolated to certain regions of the genome or may involve the entire genome (6). DNA damage can manifest as a result of mutations, replication errors, deletions or rearrangements (6), telomere shortening (121), and disruptions in tissues and organs including endothelial and vascular functions (122).

DNA damage impairs molecular integrity and alters protein coding, giving rise to mutations that promote aging. For instance, oxidative damage may transform DNA into a DNA adduct (8-OHdG) with a high mutagenic potential, resulting in DNA mutations and instability (7). Similarly, DNA instability occurs as a consequence of defects in DNA repair mechanisms and leads to increased rates of DNA mutations (121). With increased instability, the replicative phenotype shifts to a survival protective mechanism that includes apoptosis and cellular senescence (8). If DNA repair is initiated, tumor protein 53 (p53), a transcription factor that plays critical roles in cell growth, DNA repair, and apoptosis, can become overexpressed and induce cellular senescence to protect against further genomic instability (9,10). In the setting of the brain, senescent cells can lead to dysfunction of the neurovasculature and failure of the BBB (11), potentially inducing hypoperfusion, increased permeability, and subsequent neuronal damage (Table 1; Figure 1) (15).

Effect of Exercise on Genomic Instability

Moderate-intensity exercise serves an important role in the maintenance of cellular health and function, contributing to genomic stability as demonstrated by a study in healthy men between 40 and 74 years of age (19). Compared to the control group, following 16 weeks of combined aerobic and resistance training, DNA strand breaks and oxidative DNA damage decreased, while a nonstatistical increase in DNA repair capacity was observed in plasma lymphocytes in the exercise group (mean age = 58.4 ± 10.2) (19). The beneficial impact of exercise on DNA integrity is reinforced by research indicating that an exercise-induced increase in reactive oxygen species (ROS) elicits an upsurge in the levels of antioxidant enzymes, both in human skeletal muscle (123) and in plasma (124). These findings are supported by basic and human research showing exercise-induced upregulation of antioxidant enzymatic genes associated with defense against ROS (16,124,125) and DNA repair (20). Exercise has also been shown to promote regulation of DNA methylation in human skeletal muscle (21), support endothelial function (114), and mitigate genomic chromosomal damage in older adults (22). However, the effects of exercise on these mechanisms in the human brain have not been thoroughly investigated.

Central to the beneficial effects of exercise on genomic stability is the indirect but nevertheless critical role of vascular endothelial growth factor (VEGF), which is reduced in aging skeletal muscles where VEGF mRNA and protein levels, and capillarization have been shown to decrease in men (126) and women (127). Controlled animal studies (128) and vitro research (129) show that exercise-induced VEGF and angiogenesis improve skeletal muscle (130) and cerebral vascular density (131) and cerebral blood supply (128). In turn, the improvement in blood flow may contribute to genomic stability by reducing DNA damage that occurs from hypoxia and oxidative stress (129).

In humans, exercise training has been shown to increase VEGF in serum (17) and plasma (18), whereas other studies reported no effect of exercise training on VEGF in older adults (132). The discrepancies between positive and negative findings may be attributed to different exercise modalities (aerobic vs resistance), exercise intensity, and the heterogeneity of participant selection (132), highlighting the need for more research.

Although the question of whether systemic VEGF can cross the BBB remains an unresolved area of investigation, evidence from animal research reported exercise-induced elevations in hippocampal VEGF (133,134), suggesting VEGF penetration into the brain. However, evidence in the human brain is still emerging, with limited indications of an increase in VEGF in cerebrospinal fluid after exercise. Yang et al. (135) explored the impact of acute moderate-intensity upper body resistance training on VEGF in the cerebrospinal fluid (CSF) of older patients with normal-pressure hydrocephalus. Lumbar CSF was collected 1 and 2 hours before exercise, and at 3 hourly time points after exercise. Compared to the control group, a significant increase in CSF-VEGF was observed only in the exercise group (135).

Considering the evidence suggesting a possible link between exercise-induced VEGF, angiogenesis, and genomic stability in the human brain, the lack of a definitive link highlights the need for further research to uncover how a stable and responsive peripheral circulatory system, fortified against genomic instability via exercise-induced VEGF, may contribute to the preservation of cerebral vascular health. During aging when VEGF levels are known to decrease (126), increasing VEGF through exercise may be particularly beneficial.

Telomere Shortening

Telomeres are protective chromosomal endcaps whose primary function is to defend chromosomes against genomic instability during cell division. As such, telomeres, which do not fully replicate during mitotic division, gradually shorten with aging. Following repeated cell divisions, telomeres reach a critical length, lose their protective functions, and ultimately initiate cellular senescence (23). Dysfunctional telomeres may reflect overall cellular aging and may serve as biomarkers for an increased risk of developing vascular dementia (VaD). One study reported that older adults diagnosed with VaD exhibited shorter telomeres compared to adults with cerebrovascular or cardiovascular diseases without cognitive decline, adults with AD, and healthy controls. These results suggested a link between telomere length and vascular dementia, independent of other factors (136). A subsequent study, which did not include a control group, reported no differences in telomere length between adults with late-onset AD and VaD (137).

Exercise Effects on Telomere Shortening

Telomerase is an enzymatic protein that resists cellular aging by countering telomeric shortening. However, during embryonic development, the human catalytic subunit of telomerase (hTERT) becomes inactivated in most somatic cells, limiting cell division resulting in undetectable telomerase expression (138,139). Nevertheless, in healthy adults, telomerase may still be active in cells that have a high mitotic potential, such as germline cells, and self-renewing cells, such as peripheral blood mononuclear cells (138).

Evidence shows that chronic exercise can counteract telomere shortening via an increase in telomerase in human mononuclear cells and may thereby have a protective role in systemic vascular aging (24,27,140,141). Other studies reported negative correlations between leukocyte telomere length and cardiovascular and cerebrovascular diseases such as ischemic stroke (142,143), suggesting that maintaining telomere length with physical activity may benefit the vascular system, with possible implications for cerebrovascular health.

Elevations in telomerase activity in trained athletes may impact other systemic cellular aging processes. Protection against telomere shortening preserves genomic stability (23,27), which may moderate endothelial cellular aging and senescence, and reduce concentrations of pro-inflammatory cytokines, collectively referred to as senescent-associated secretory phenotypes (SASPs) (26), that further contribute to telomere shortening (144).

Another factor that may play a role in the relationship between exercise, telomere shortening, and brain health is the exercise-induced extracellular chaperone protein, clusterin (CLU). CLU, which is elevated in older adults with AD and mild cognitive impairment (MCI) (25), is associated with reductions in brain inflammation and is emerging as a critical player in mitigating cellular stress and the inflammatory response (25,145). Although indirect, CLU’s role in protecting cells against inflammation may provide a significant buffer against the cascade of cellular aging and telomere erosion, which may have cerebral vascular benefits. In support of this, researchers examined key plasma proteins before and after 6 months of training in 20y adults (mean age = 69.68 ± 7.95) with amnestic MCI (146). The exercise program consisted of 3 weekly sessions of combined aerobic and resistance training. Although there was no control group, compared to baseline values, CLU increased (146). Given the anti-inflammatory effects of CLU, an exercise-induced increase in CLU may benefit the brain if it prevents the adverse effects of systemic inflammation on BBB dysfunction (147) and neurodegeneration (148). In summary, the presented studies suggest that exercise may protect against neuronal aging via beneficial effects on telomerase activity, telomere length, and inflammation.

Epigenetic Alterations

Epigenetic alterations, including histone modifications and abnormal DNA methylation, alter gene expression without altering the underlying DNA sequence, and can occur following exposure to environmental factors and lifestyle behaviors (149). Disruptions in blood flow within the vascular walls, for instance, have been shown to increase DNA methyltransferase, which initiates broad changes in DNA methylation that can affect gene activity and impair DNA repair capacity (28). These changes, along with histone modifications (29), can lead to DNA double-strand breaks, which can indirectly induce genomic damage (118) and ultimately affect the pathogenesis of AD (150). For instance, DNA methylation may silence genes responsible for the clearance of waste products (150) thereby contributing to disruptions in proteostasis. In turn, a breakdown of proteostasis alters the function of vascular cells, giving rise to endothelial cell senescence, vascular remodeling, atherosclerosis, and inflammation (Table 1; Figure 1) (30,151).

Exercise Effects on Epigenetic Alterations

Exercise results in alterations of DNA methylation profiles and histone modifications (31), which have systemic effects and potential implications for the cerebral vasculature. Albeit indirect, collective evidence from exercise studies suggests a linkage between exercise-induced epigenetic changes, antioxidant expression, and cerebral vasculature.

ROS are recognized stimulants for transcription of genes involved in the adaptive response to exercise-induced oxidative stress (32,152). A key element in this antioxidant response to exercise is the “master regulator,” nuclear factor erythroid-2-related factor (Nrf2) (32). Nrf2 is a transcription factor that is deacetylated by sirtuins (33). Sirtuins (mainly, 1, 6, and 7) are highly conserved proteins of NAD+-dependent histone deacetylases that target histones and transcription factors, contributing to epigenetic regulation of cell phenotypes. The interaction between Nrf2 and SIRT1enhances Nrf2 binding to the antioxidant responsive element (ARE), which regulates transcription of various cytoprotective and antioxidant genes (33) such as superoxide dismutase (SOD) (32,152). Once activated, the Nrf2-ARE mechanism enhances cellular energy and redox balance, protects against neurotoxicity (153), and suppresses inflammation by inhibiting IL-6, IL-1β, and IL-17 gene expression (34).

The downstream activation of Nrf2 and its associated protective effects in animals provides a compelling link between systemic physiological adaptations and prevention or mitigation of cerebral vascular and neurological dysfunctions. For instance, Nrf2 appears to improve systemic vascular endothelial dysfunction by inhibiting oxidative stress (154,155), whereas reduced Nrf2 activity is inversely related to several brain conditions including experimental stroke (156,157), brain injury, cerebrovascular, and neurodegenerative models (158). To determine the impact of Nrf2 upregulation in cerebral vessels following ischemic stroke, Alfieri et al. (159) compared 2 groups of rats where 1 group was pretreated with an Nrf2 stimulator prior to middle cerebral artery occlusion and the other was not. Expression of heme oxygenase, an enzyme involved in catabolism of heme that becomes activated during cellular stress and inflammation, increased in the cerebral microvessels surrounding the infarct in the untreated ischemia group. However, pre-activation with an Nrf2 stimulant (sulforaphane, SFN) increased Nrf2 expression in cerebral microvessels and prevented neurological and BBB dysfunction in the pretreated animals (159). In another study, 2 groups of mice (wild-type and Nrf2 knockout) were pretreated with SFN. In the wild-type group, Nrf2 increased and resulted in the upregulation of antioxidant enzymes in the striatum and cortex, with parallel reductions in inflammatory cytokines and gliosis. These effects were not observed in the Nrf2 knockout mice (160). Collectively, these results provide evidence for the role of pharmacologically activated Nrf2 on antioxidant gene expression, and their potential protective effect on cerebral microvessels in animals.

Evidence from exercise studies in animals support the notion that physical activity may exert comparable stimulatory effects on sirtuins and the Nrf2 signaling cascade (32,35). In humans, activation of the Nrf2 pathway in skeletal muscle and blood leads to enhanced expression of SOD genes, with higher responses following HIIT (35,36). However, this response is diminished in sedentary older participants (161). Compared to their sedentary counterparts, active older adults exhibited a higher Nrf2-mediated antioxidant response, suggesting that chronic exercise may mitigate an age-related decline in Nrf2 signaling (162).

Little is known about exercise-induced Nrf2 expression in the brain. However, evidence for Nrf2 changes in hippocampal, cortical, and hypothalamic structures has been reported following vigorous exercise in adult mice (163). The robust activation of Nrf2 in animal studies, particularly within brain tissues, constitutes a potential neuroprotective mechanism elicited by exercise. Whether comparable exercise-induced Nrf2 effects occur in the human brain is unknown. However, the broader implications of Nrf2-signaling for genomic integrity suggest that mechanisms of antioxidant protection with downstream effects on brain blood vessels may be operational in humans. This proposition is strengthened by evidence linking Nrf2 upregulation with neuroprotection and Nrf2 deficiencies with neurodegenerative diseases which are characterized by increased oxidative stress (159,164). To enhance our understanding and potential therapeutics for neurodegeneration, further research in humans is needed to clarify the precise epigenetic pathways through which exercise might improve Nrf2 signaling and its effects on cerebrovascular function and brain health.

Loss of Proteostasis

Proteostasis entails an intricate balance between synthesis, folding, and degradation of proteins. With aging, the capacity of this system to maintain its coordinated functions becomes disrupted and leads to an accumulation of misfolded proteins. In this misfolded state, proteins lose structural integrity and assume a toxic profile that can initiate cellular and tissue damage (37). Examples of misfolded proteins include Aβ, alpha synuclein, prion proteins, and superoxide dismutase which is implicated in apoptosis (165–167). Protein misfolding can damage neuronal and glial cells, leading to cellular dysfunction, neurodegenerative diseases (AD, Parkinson’s, Creutzfeldt–Jakob, Huntington, amyotrophic lateral sclerosis), and cognitive decline (165). In the vascular system, an accumulation of misfolded proteins, such as members of the apolipoprotein family, α-antitrypsin and medin, contribute to endothelial dysfunction, a key factor in the development of systemic and central vascular diseases (38,168), cerebrovascular dysfunction (169), and cognitive impairment (170).

Lastly, the role of the glymphatic system can be considered in the context of loss of proteostasis. The glymphatic system is a newly discovered cerebral waste drainage system in the brain, similar in function to the systemic lymphatic system (171). The glymphatic system is intricately involved in the regulation of interstitial Aβ deposition (41), whose aggregation is implicated in neurodegenerative processes. With aging, the activity of the glymphatic system is reduced, such that cerebral waste clearance is lower in older compared to younger adults (39). The reductions in glymphatic clearance have been associated with a decrease in cerebral gray matter volume and parallel reductions in cognitive function in older adults (Table 1; Figure 1) (172).

Effects of Exercise on Proteostasis

Exercise enhances proteostasis by promoting autophagy (173). For instance, the proteosome activity improved in rats (174), and in older hypertensive adults following aerobic and resistance exercise training (42), while exercise-induced degradation of misfolded proteins was shown in both animal and human muscle (40). Consequently, by stimulating autophagy and removing toxic protein aggregates and damaged organelles in the brain and in blood vessels, exercise aids in the maintenance of neural function and CBF (175). In turn, adequate CBF, not only safeguards the delivery of nutrients to the brain but may also assist in the removal of waste products, including misfolded proteins (41,79,176).

Additionally, emerging evidence indicates that exercise may enhance glymphatic activity (41,177). To illustrate, glymphatic transport is propelled by the pulsatility of the arterial walls and moves in the same direction as blood flow (14) along astrocyte-supported perivascular channels that drive CSF into the brain and interstitial fluid out of the brain along perivenous spaces (14). Given the dependence of the glymphatic system on arterial pulsatility, the clearance of waste products may be partially driven by changes in physical activity (41,177). Ultimately, the interdependence between cardiac output, arterial pulsatility, and the glymphatic system highlights the role of exercise-induced neuroprotection via cerebral waste product removal (176,177). In this way, the glymphatic system plays a part, albeit indirectly, in autophagy and mitochondrial function and in sustaining cellular homeostasis.

Nutrient Sensing

Nutrient-sensing pathways, including adenosine monophosphate kinase (AMPK), mammalian target of rapamycin (mTOR), sirtuins, and IGF-1, are also dysregulated with aging (43). AMPK maintains mitochondrial homeostasis and mitophagy (mitochondrial autophagy), and is promptly activated following energetic stress induced by nutrient deficiency, hypoxia, ischemia, and exercise (51,178). Once activated, AMPK plays a role in endothelial NO synthase (eNOS) production (179) and angiogenesis (51). By contrast, mTOR stimulates anabolic processes under conditions of high nutrient availability, and plays a central role in energy metabolism by integrating energetic and growth factor signaling with cellular processes such as stress resistance and autophagy (180). Given its function as a signaling target for growth factors, mTOR plays a critical function in regulating angiogenesis in endothelial cells (45). Together, AMPK and mTOR serve as modulators of the signaling cascades of IGF-1 involved in glucose sensing, angiogenesis, and capillarization (181).

Sirtuins, silent information regulators (SIRT 1-7), are conserved histone deacetylases that belong to a class of nicotinamide adenine dinucleotide (NAD+)-dependent enzymes that play an essential role in regulating cellular responses to nutrient availability (44). Sirtuins influence metabolism by regulating gluconeogenesis and fatty acid oxidation by deacetylating transcription factors (44). Once activated, sirutins deacetylate eNOS leading to increased production of NO in endothelial cells. In addition, sirtuins participate in growth factor regulation and can promote angiogenesis. As such, members of the sirtuin family initiate broad effects that result in enhanced vascular health (48), extended lifespans, and protection from neurodegenerative diseases (44). Interestingly, evidence for expression of SIRT1 in the rodent and human central nervous systems has been reported (182). In animals, the presence of SIRT1 in the small resistance vessels of the brain contributed to cerebral blood vessel dilation, whereas its inhibition decreased dilation of blood vessels (183). Similarly, in a model of cerebral ischemia, SIRT1 preserved CBF and restored cerebrovascular reserve in mice with SIRT1 overexpression compared to a group of wild-type mice (184).

Effects of Exercise on Nutrient-Sensing Pathways

Key nutrient-sensing pathways including IGF-1, NAD+, and sirtuins are modulated by resistance (47) and anaerobic exercise (49,185). Exercise-induced release of IGF-1 stimulates ribosomal biogenesis and protein synthesis (50), which have been positively associated with blood vessel density in the brain, CBF, and cognitive function (117,186–189). Similarly, exercise increases NAD+ levels, and counteracts the age-related decline in NAD+ in both animals and humans (190). The increase in NAD+ activates sirtuins in a wide variety of tissues that influence a broad range of cellular and vascular processes described earlier (44,48). Although human studies to examine the direct role of exercise-induced SIRT1 on cerebral circulation are lacking, evidence from animal studies highlights the role of SIRT on cerebral vasodilation (183), and underscores its potential impact on CBF (184).

Lastly, the AMPK pathway is also activated with moderate-to-heavy exercise intensities (46). The upregulation of AMPK conserves adenosine triphosphate leading to activation of PGC-1α and its downstream effects on mitochondrial biogenesis (50), which is critical to cellular energy production. Moreover, activation of AMPK has protective effects. AMPK inhibits inducible transcription factors involved in inflammatory responses in the vasculature (52), improves vascular function and CBF via NO-dependent vasorelaxation (52), and angiogenesis via an increase in VEGF (51). Therefore, by optimizing nutrient-sensing signaling, exercise may improve the responsiveness of the vascular system to cerebral metabolic demands, and may play a pivotal role in the preservation of vascular and cognitive function in aging (191).

Mitochondrial Dysfunction

Mitochondria are responsible for energy production, intracellular calcium regulation, and their own protein synthesis. With age, however, mitochondrial function and mitophagy (mitochondrial autophagy) activity decline, resulting in impairment in cellular energy production (53,192). Mitophagy, which is instrumental in maintaining cellular homeostasis and promoting cell survival, functions as a protective mechanism that selectively degrades damaged or dysfunctional mitochondria (193). However, when mitophagy is disrupted, failing mitochondria produce high levels of ROS and inflammatory cytokines, which can quickly overwhelm the cell’s antioxidant abilities, leading to inflammation (53) and overall loss of cellular function (54). Furthermore, loss of mitophagy results in decreased energy production, generation of pro-apoptotic factors, and onset of endothelial dysfunction (55). Given these effects, deficits in mitochondrial function are frequently identified as contributors to vascular endothelial dysfunction (53) and neurodegenerative diseases (56,194). Mitochondrial dysfunction has also been implicated in vascular cognitive impairment, MCI, and dementia (Table 1; Figure 1) (195–197).

Effects of Exercise on Mitochondrial Dysfunction

Human studies have shown that exercise initiates widespread changes in muscle mitochondria. Specifically, exercise-induced mechanical shear stress mitigates oxidative damage via the release of PGC-1α (59) whose downstream effects on growth factor cascades enhance muscular mitochondrial and capillary density (61,198). Enhanced mitochondrial and capillary density following chronic endurance exercise improves oxygen delivery, enhances oxidative phosphorylation for energy (adenosine triphosphate; ATP) production, and augments the production of antioxidant enzymes (eg, superoxide dismutase) in muscle (58). Together, these beneficial effects lead to a reduction in ROS and their consequent deleterious effects on vascular function (59,179).

Additionally, physiological adaptations to low- or moderate-intensity exercise regulate the expression of membrane receptor adhesion molecules (199), which amplify mitophagy through the inhibition of mTOR and phosphorylation of AMPK. These coordinated events augment the production and availability of NO and ensure both mitochondrial health and optimal vascular tone (59). Lastly, p53, which takes part in mitochondrial biogenesis and DNA repair, increases in muscle following moderate aerobic exercise (60). Considering the fundamental function of mitochondria in cellular energy production in capillary pericytes that regulate vasodilation and vasoconstriction, the effects of exercise on mitochondrial density may also help to ensure precise regulation of cerebral vessels (200), which is vital for maintaining overall brain health and function.

Cellular Senescence

DNA damage, mitochondrial dysfunction, enhanced ROS production, and telomere attrition are some of the age-related changes that trigger cellular senescence (201). Senescent cells lose their ability to divide but remain metabolically active despite undergoing cell-cycle arrest. In this state, they adopt an immunologic phenotype and begin to secrete interleukins and pro-inflammatory SASPs that promote chronic inflammation (62). Once initiated, cellular senescence negatively affects the proliferation of neural stem cells (202) that give rise to neurons and neuroglia, culminating in a reduction in neurons, oligodendroglia, and astrocytes, which can impair neuronal and synaptic function (203) and result in cognitive decline (63). Indeed, a growing body of evidence links the buildup of senescent cells to neurodegenerative pathologies and dysfunction (204). Reducing senescent cells with treatments known as senolytics (eg, Dasatinib, Quercetin, Rapamycin, Fisetin), which are compounds that help induce senescent cell death, have shown promise in mitigating the effects of cellular senescence in animal (205) and human studies of AD (206).

In endothelial cells, cellular senescence results in decreased production of NO and impaired vasodilation (64), which may compromise the blood flow response to neural activity (93). Furthermore, the accumulation of senescent vascular cells can impair BBB integrity thereby allowing systemic pro-inflammatory cytokines to infiltrate the immune-privileged brain (11,63). The breakdown of the BBB is associated with loss of pericytes and transporter functions, and impaired waste product removal resulting in neuroinflammation and neuronal dysfunction (63,66,67).

Effects of Exercise on Cellular Senescence

Exercise is protective against cellular senescence, especially in vascular endothelial cells, which are critical for preserving vascular function during aging (26,68). This protective effect is facilitated by several mechanisms, including activation of sirtuins, upregulation of telomerase activity, and enhanced NO availability (see discussions earlier). Collectively, these exercise-induced effects affect the expression of tumor suppressor p16ink4a (p16), a cyclin-dependent kinase inhibitor that plays a pivotal role in cellular senescence including contributing to vascular endothelial dysfunction in sedentary older adults (68). When stimulated by exercise, SIRT1-mediated histone deacetylation can result in a more closed chromatin state at the p16 gene locus, which reduces its transcription and subsequent expression (207). This effect is evident in trained athletes, where studies showed upregulated telomerase activity and downregulated p16 expression in circulating peripheral blood mononuclear cells compared to untrained controls (140). Additionally, compared to baseline levels, 12 weeks of exercise reduced p16 levels and expression of cellular senescence markers in older adults (26), further supporting the systemic influence of exercise on cellular senescence. Moreover, chronic exercise restores NO availability (69), and modulates age-related imbalances in p53 levels in the endothelial cells of peripheral vessels in humans (68), thereby reducing oxidative stress in older athletes and safeguarding systemic vascular health (68).

In the context of cerebral vessels, mitigating cellular senescence preserves endothelial and vascular function (68), BBB integrity, and neuronal and glial activity (208,209), which are key determinants of cerebral autoregulation, CBF, and cognitive function (108,210). Although direct assessment of exercise effects on cellular senescence in the human cerebral vasculature is lacking, the established effects of exercise in the peripheral vascular system suggest similar benefits for the cerebral vessels. This generalization is supported by evidence from converging lines of research including a study that linked the accumulation of p16 and senescent cells to the breakdown of the BBB and neurovascular dysfunction in mice (11), coupled with evidence for elevated p16 levels in postmortem brain slices of adults with AD, which were not present in non-AD-matched controls (211).

Therefore, by regulating the molecular pathways that affect cellular senescence, chronic exercise may contribute to the preservation of cerebral vascular integrity and cognitive function in aging. Although promising, verification in humans is required to assess the impact of exercise on cerebral senescence markers and cerebrovascular health.

Stem Cell Exhaustion

The accumulation of DNA damage, buildup of SASPs, telomere shortening, mitochondrial dysfunction, and epigenetic changes in turn lead to reductions in stem cell activity (70). Stem cell exhaustion has far-reaching consequences on repair mechanisms of the endothelium, the BBB, neurogenesis, and neurovascular function. For instance, in the vascular system, stem cell exhaustion contributes to the formation of atherosclerosis (212), which may lead to reductions in CBF (72), buildup of waste products, and neurovascular dysfunction (79). Similarly, loss of repair activities in the vasculature (71) can limit the regenerative ability of neuronal and BBB cells, making the brain more permeable to pathogens and neuroinflammation. Therefore, the effects of stem cell exhaustion, alone or in combination with other hallmarks of aging, can play a significant part in age-related cognitive decline (203).

Exercise Effects on Stem Cells

Exercise affects a cascade of molecular processes that stimulate stem-cell-mediated vascular repair. These processes include the migration and proliferation of endothelial progenitor cells (EPC) from bone marrow and other tissue reservoirs into the peripheral blood (73), and the upregulation of growth factors in humans (213). The exercise-induced increase in VEGF enhances the proliferation and migration of EPCs (214). In parallel, stromal cell-derived factor-1 (SDF-1) functions as a chemoattractant that helps guide EPCs to vascular injury sites, as demonstrated in damaged murine carotid arteries (74). Moreover, activation of the PI3K/Akt signaling pathway promotes the survival and function of EPCs (76,77), and the upregulation of eNOS. Once activated, NOS contributes further to the mobilization of EPCs and modulation of vascular tone via an increase in NO (75).

Ultimately, these molecular changes are vital for maintaining the integrity of the BBB and ensuring adequate CBF, which is necessary for the delivery of EPCs to areas within the brain that require repair following intracerebral hemorrhage and ischemic stroke. One study of patients with intracerebral hemorrhage observed that increased levels of circulating EPCs were associated with better clinical outcomes, supporting their role in neurovascular repair processes (215). Another study found that EPCs isolated from blood following ischemic stroke in older adults (mean age = 70) were associated with better BBB permeability, which independently predicted better clinical outcomes (216). A direct link between exercise-induced EPC mobilization and enhanced cerebral vascular function in humans remains to be established.

Altered Intercellular Communication

With aging, endocrine, neuroendocrine, and neuronal signaling begins to fail (78). Loss of inhibitory signaling in these systems can lead to chronic release of stress hormones and cytokines, giving rise to immune system malfunction and inflammation (70). The inflammatory consequence of age-related failure of intercellular communication has been termed “inflammaging.” This phenomenon is characterized by an overproduction of pro-inflammatory cytokines (70), and the breakdown of the BBB (147). This inflammatory response is increasingly identified as a significant risk factor for strokes, and onset of cerebral small vessel disease (217). Furthermore, the breakdown of intercellular communication and the resulting inflammation have been identified as contributors to the buildup of Aβ (80,218), in facilitating the propagation of tau pathology between cells (219), and in cognitive decline (220).

Exercise Effects on Intercellular Communication

Exercise plays a critical role in enhancing intercellular communication, mitigating inflammation, and maintaining the health of blood vessels. Evidence from human studies indicates that aerobic, anaerobic, and resistance training modulate inflammatory biomarkers (81,83). For instance, chronic exercise inhibits pro-inflammatory cytokines (221,222) and protects endothelial (223) and vascular functions (88). One way by which exercise confers these benefits is via the release of interleukin-6 (IL-6) from muscle (82). Contrary to the pro-inflammatory IL-6 released by macrophages, IL-6 released from muscle appears to have anti-inflammatory benefits and is present in significantly higher concentrations than macrophage IL-6 (82). The higher concentrations of myokine-produced IL-6 inhibit systemic inflammation (224) by stimulating the release of other anti-inflammatory interleukins such as IL-10 and IL-1 receptor antagonist, which suppress the pro-inflammatory cytokine TNF-α (86,224). Ultimately, the anti-inflammatory effects of exercise protect the vascular BBB (147), aid in the maintenance of neuronal and glial function, and in the preservation of cognitive function with aging (225).

In summary, the underlying mechanisms of age-related vascular disorders are highly complex, encompassing a wide array of interactions and pathological consequences that give rise to cerebrovascular dysfunction. However, chronic exercise may mitigate the effects of cellular aging (Table 1), and has been shown to be associated with better vascular health, maintenance of brain function in aging (4,81,114,115,226), and decreased mortality risk (227).

Exercise, Cerebral Hemodynamics, and Cognitive Function

As noted earlier, exercise effects on vascular endothelial function underlie some of its benefits on cerebral hemodynamics and cognition (115). This notion is supported by evidence from a large meta-analysis of 51 randomized controlled trials of 2 260 middle-aged and older adults that reported improvements in endothelial function with exercise (228). Moreover, lifelong exercisers who maintained higher CRF into older age had higher CBF and cerebrovascular reactivity (CVR) compared to their sedentary counterparts (96,226). However, even shorter duration exercise training (12 weeks) can improve cerebral hemodynamics (229), and cerebral blood volume (CBV) in the dentate gyrus in healthy older adults (230). Interestingly, a parallel animal study by the same authors showed correlations between elevations in CBV and neurogenesis (230). Although a large number of studies report the beneficial effects of exercise on cerebral hemodynamics, a recent study that examined the acute effects of exercise intensity on peripheral vascular adaptations in healthy young adults reported unchanged CVR across all exercise conditions, including moderate-intensity exercise and sedentary controls (231). Importantly, this study was conducted in a very small and young sample of adults, limiting the translation to older adults. Another study in individuals with stable coronary heart disease reported inconsistent enhancements in cerebrovascular response following acute exercise (232). These findings highlight the complexity of the vascular response to exercise, especially in populations with pre-existing health conditions. Furthermore, although exercise is generally known to improve endothelial dysfunction, a key component in cardiovascular and cerebrovascular disease, whether higher exercise intensities confer greater benefits than moderate-intensity training (MIT) remains a topic of ongoing research (233).

The comprehensive benefits of exercise on various aspects of health and aging, including its impact on endothelial function and cerebral hemodynamics, are associated with maintenance (4,226) and recovery of cognitive function in older age (234–237). In an early exercise study, sedentary adults between 60 and 75 years of age were randomly assigned to a walking or stretching intervention and received neurocognitive assessments before and after 6 months. Compared to the stretching group, adults in the walking group demonstrated improvements in executive function (238). Additionally, age-related cognitive decline was attenuated (239) even when exercise interventions were undertaken in late adulthood (239,240). Furthermore, meta-analytic studies showed that the benefits of exercise on cognition extend to adults with MCI or dementia (237,241). Nevertheless, more recent studies challenge the efficacy of exercise in improving cognitive function in older adults (231,242,243). These incongruities highlight the need for more research into the underlying cellular and molecular mechanisms of exercise-induced benefits to the brain.

Effects of Different Exercise Intensities on the Cerebrovasculature

In the following section, we briefly describe how exercise-induced intensity-dependent metabolic energetic pathways may differentially impact the brain via distinct effects on CBF and cerebral waste product clearance.

To fuel physical activity, the human body relies on 3 energy systems: the phosphagen, anaerobic, and aerobic pathways, whose activations are dependent on the intensity and duration of the activity, and the availability and demand for oxygen (244). Although the 3 systems are simultaneously activated to varying degrees during exercise, one system will dominate depending on the activity type and energy requirements. For instance, short, intense activities such as heavy resistance training or sprinting lasting for 10–15 seconds activate the phosphagen system (244,245), whereas prolonged light-intensity training to MIT like swimming, cycling, or long-distance running is governed by the aerobic system (244). By contrast, anaerobic glycolysis becomes the dominant energetic pathway during high-intensity interval training (HIIT) (244).

Different energetic pathways exert a variety of effects on the hallmarks of aging, which in turn may affect the brain as described in the previous sections. To illustrate, the phosphagen and anaerobic systems, which are activated via resistance training and HIIT, can stimulate mitochondrial biogenesis, protect telomeres against attrition, and increase DNA repair mechanisms and proteostasis activity, while also regulating nutrient sensing via mitigation of insulin insensitivity (246). Activating the aerobic system enhances mitochondrial biogenesis and function and cytokine signaling pathways, leading to improved cell-to-cell communication, thus reducing inflammation and cellular senescence (246).

Additionally, it is well documented that MIT confers significant circulatory benefits including improvements in CBF (88,96,114,115,226,247), due to the more pronounced elevations in exercise-induced vascular shear stress (blood, velocity, and pressure) (248,249), studies show that HIIT results in markedly greater increases in cardiac output and vascular adaptations (250,251) and more substantial cardiovascular benefits (252) with greater reductions in cardiovascular risk factors (253).

Although research on the specific effects of HIIT on cerebral hemodynamics is limited (249), the vascular adaptations to HIIT suggest potential positive implications for CBF and cerebral waste product clearance. For instance, studies show that during incremental exercise, an initial increase in arterial partial pressure of carbon dioxide (PaCO2) serves as a cerebral vasodilator (254). Furthermore, due to the dependence on glucose metabolism, as exercise intensity rises, the rise in PaCO2 is followed by an increase in lactic acid and H+, which is quickly buffered by bicarbonate (HCO3−) and can be co-transported with lactate across the BBB (255). Consequently, the increase in H+ alters the concentration gradient between the brain and the blood, and stimulates cerebral uptake and utilization of lactate (256), which along with increasing levels of CO2, enhances vasodilation and helps augment CBF (257,258). Interestingly, evidence from animal studies suggests that elevated levels of cerebral lactate in the perivascular fluid stimulate the glymphatic system, thereby increasing the clearance of cerebral waste products (259,260).

In summary, synergistic metabolic systems activate different energetic pathways and offer multipronged protection against the hallmarks of aging and associated endothelial, and vascular dysfunction (69,179), resulting in a more resilient vascular system, maintenance of CBF in aging (96,226), and enhanced clearance of cerebral waste products (259,260). This intricate relationship between various exercise intensities and aging emphasizes the significance of a holistic approach to health and well-being, and the need for more research into the differential effects of exercise intensities on the brain.

Conclusion

Aging is a multifactorial cellular and systemic process that is profoundly influenced by modifiable cardiovascular risk factors. The presence or absence of these risk factors exert direct and indirect effects on the cellular and molecular hallmarks of aging, further aggravating or enhancing these processes. In the vascular system, the aging cellular milieu leads to impairments in endothelial repair mechanisms and function, which consequently impair the cerebrovasculature and have downstream effects on cognitive function. Importantly, evolving evidence reveals the pivotal role of exercise in this complex interplay (Table 1; Figure 1). In this role, exercise not only mitigates cellular and molecular hallmarks of aging but also protects the cerebrovasculature and promotes the maintenance of brain health.

The conclusions of our narrative review must be tempered by the methodological limitations of currently published research in this field. First, the preponderance of studies we cited were based on cross-sectional or observational research, much of which was conducted in model animals. Although these studies offer valuable initial insights, they do not enable the causal inference that can only be established through more rigorous clinical trials in humans. Furthermore, studies with positive findings are more likely to be published, overshadowing those with null or negative results. Finally, research involving diverse populations, including varying age groups, ethnicities, and those with disease comorbidities, is lacking.

Despite the current wealth of knowledge, a comprehensive understanding of exercise effects on cerebrovascular pathology and repair mechanisms has remained fragmented, leaving several questions unanswered. Primarily, a relatively limited number of studies examined the effects of lifelong exercise and its effects on cerebrovascular function. The question of whether exercise undertaken after the onset of age-related cellular and cerebrovascular changes can confer similar benefits to lifelong CRF is unknown and requires empirical confirmation. Second, the dose–response relationship between exercise training and cerebrovascular and cognitive functions in older adults is unknown, and a comprehensive understanding of optimal exercise interventions remains elusive. Randomized clinical trials (261,262), which interrogate the effects of different exercise variables (eg, frequency, intensity, duration, modality) on the cerebrovasculature and brain function would provide necessary data to expand current knowledge of the neurobiology of aging and the mechanisms that underlie neuroprotection. Additionally, studies encompassing combined interventions that incorporate exercise with diet, senolytics, and other interventions that target the hallmarks of aging present promising avenues for future preventative or therapeutic interventions. To better understand how exercise affects cerebrovascular health and aging, future research must adopt comprehensive longitudinal studies focused on understanding the long-term implications of exercise interventions and their effects on the biological hallmarks of aging across different demographics.

Irrespective of the limitations of previous research, the current body of knowledge regarding the effects of exercise on cerebrovascular health is nevertheless promising. As a noninvasive, cost-effective, and holistic approach, optimized exercise interventions defend against cerebrovascular pathologies and their effects on cognition in aging. Given the rapidly expanding aging population and its projected societal impact, exercise may be a vital intervention to aid in the prevention or deferment of age-related cognitive impairments.

Contributor Information

Amani M Norling, The Hinda and Arthur Marcus Institute for Aging Research, Hebrew SeniorLife, Boston, Massachusetts, USA; Department of Medicine, Harvard Medical School, Boston, Massachusetts, USA.

Lewis A Lipsitz, The Hinda and Arthur Marcus Institute for Aging Research, Hebrew SeniorLife, Boston, Massachusetts, USA; Department of Medicine, Harvard Medical School, Boston, Massachusetts, USA.

Roger A Fielding, (Medical Sciences Section).

Funding

This work was supported by grants from the National Institute on Aging (T32 AG023480 and R21 AG073886). L.A.L. holds the Irving and Edyth S. Usen Chair in Geriatric Medicine at Hebrew SeniorLife.

Conflict of Interest

None.

Author Contributions

A.M.N. and L.A.L. conceptualized the review and made substantial contributions to this manuscript. A.M.N. was responsible for drafting and revising the manuscript. L.A.L. provided critical revisions and made substantial editorial contributions. A.M.N. and L.A.L. have reviewed and approved the final version of the manuscript.

References

  • 1. López-Otín C, Blasco MA, Partridge L, Serrano M, Kroemer G.. Hallmarks of aging: an expanding universe. Cell. 2023;186:243–278. 10.1016/j.cell.2022.11.001 [DOI] [PubMed] [Google Scholar]
  • 2. Wyss-Coray T. Ageing, neurodegeneration and brain rejuvenation. Nature. 2016;539(7628):180–186. 10.1038/nature20411 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 3. Salthouse TA. Independence of age-related influences on cognitive abilities across the life span. Dev Psychol. 1998;34(5):851–864. 10.1037//0012-1649.34.5.851 [DOI] [PubMed] [Google Scholar]
  • 4. Barnes DE, Yaffe K.. The projected effect of risk factor reduction on Alzheimer’s disease prevalence. Lancet Neurol. 2011;10(9):819–828. 10.1016/S1474-4422(11)70072-2 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 5. Lazar RM, Howard VJ, Kernan WN, et al. ; American Heart Association Stroke Council. A primary care agenda for brain health: a scientific statement from the American Heart Association. Stroke. 2021;52(6):e295–e308. 10.1161/STR.0000000000000367 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 6. Aguilera A, Gómez-González B.. Genome instability: a mechanistic view of its causes and consequences. Nat Rev Genet. 2008;9(3):204–217. 10.1038/nrg2268 [DOI] [PubMed] [Google Scholar]
  • 7. Emam AN, Girgis E, Khalil WK, Mohamed MB, eds. Toxicity of Plasmonic Nanomaterials and Their Hybrid Nanocomposites. Advances in Molecular Toxicology. Elsevier; 2014. [Google Scholar]
  • 8. Clementi E, Inglin L, Beebe E, Gsell C, Garajova Z, Markkanen E.. Persistent DNA damage triggers activation of the integrated stress response to promote cell survival under nutrient restriction. BMC Biol. 2020;18:1–15. 10.1186/s12915-020-00771-x [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 9. Vousden KH, Lane DP.. p53 in health and disease. Nat Rev Mol Cell Biol. 2007;8(4):275–283. 10.1038/nrm2147 [DOI] [PubMed] [Google Scholar]
  • 10. Liu D, Xu Y.. p53, Oxidative stress, and aging. Antioxid Redox Signal. 2011;15(6):1669–1678. 10.1089/ars.2010.3644 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 11. Yamazaki Y, Baker DJ, Tachibana M, et al. Vascular cell senescence contributes to blood–brain barrier breakdown. Stroke. 2016;47(4):1068–1077. 10.1161/STROKEAHA.115.010835 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 12. Nedergaard M. Garbage truck of the brain. Science. 2013;340(6140):1529–1530. 10.1126/science.1240514 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 13. ElAli A, Thériault P, Préfontaine P, Rivest S.. Mild chronic cerebral hypoperfusion induces neurovascular dysfunction, triggering peripheral beta-amyloid brain entry and aggregation. Acta Neuropathol Commun. 2013;1(1):75. 10.1186/2051-5960-1-75 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 14. Mestre H, Tithof J, Du T, et al. Flow of cerebrospinal fluid is driven by arterial pulsations and is reduced in hypertension. Nat Commun. 2018;9(1):1–9. 10.1038/s41467-018-07318-3 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 15. Wong SM, Jansen JF, Zhang CE, et al. Blood-brain barrier impairment and hypoperfusion are linked in cerebral small vessel disease. Neurology. 2019;92(15):e1669–e1677. 10.1212/WNL.0000000000007263 [DOI] [PubMed] [Google Scholar]
  • 16. Gomez-Cabrera M-C, Domenech E, Viña J.. Moderate exercise is an antioxidant: upregulation of antioxidant genes by training. Free Radic Biol Med. 2008;44(2):126–131. 10.1016/j.freeradbiomed.2007.02.001 [DOI] [PubMed] [Google Scholar]
  • 17. Kim H-B, Seo M-W, Jung HC.. Effects of Aerobic vs. Resistance Exercise on Vascular Function and Vascular Endothelial Growth Factor in Older Women. MDPI; 2023:2479. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 18. Schobersberger W, Hobisch-Hagen P, Fries D, et al. Increase in immune activation, vascular endothelial growth factor and erythropoietin after an ultramarathon run at moderate altitude. Immunobiology. 2000;201(5):611–620. 10.1016/S0171-2985(00)80078-9 [DOI] [PubMed] [Google Scholar]
  • 19. Soares JP, Silva AM, Oliveira MM, Peixoto F, Gaivão I, Mota MP.. Effects of combined physical exercise training on DNA damage and repair capacity: role of oxidative stress changes. Age. 2015;37:1–12. 10.1007/s11357-015-9799-4 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 20. Radák Z, Naito H, Kaneko T, et al. Exercise training decreases DNA damage and increases DNA repair and resistance against oxidative stress of proteins in aged rat skeletal muscle. Pflugers Arch. 2002;445:273–278. 10.1007/s00424-002-0918-6 [DOI] [PubMed] [Google Scholar]
  • 21. Sailani MR, Halling JF, Møller HD, et al. Lifelong physical activity is associated with promoter hypomethylation of genes involved in metabolism, myogenesis, contractile properties and oxidative stress resistance in aged human skeletal muscle. Sci Rep. 2019;9(1):3272. 10.1038/s41598-018-37895-8 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 22. Franzke B, Halper B, Hofmann M, et al. ; Vienna Active Ageing Study Group (VAAS). The effect of six months of elastic band resistance training, nutritional supplementation or cognitive training on chromosomal damage in institutionalized elderly. Exp Gerontol. 2015;65:16–22. 10.1016/j.exger.2015.03.001 [DOI] [PubMed] [Google Scholar]
  • 23. O’sullivan RJ, Karlseder J.. Telomeres: protecting chromosomes against genome instability. Nat Rev Mol Cell Biol. 2010;11(3):171–181. 10.1038/nrm2848 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 24. Werner CM, Hecksteden A, Morsch A, et al. Differential effects of endurance, interval, and resistance training on telomerase activity and telomere length in a randomized, controlled study. Eur Heart J. 2019;40(1):34–46. 10.1093/eurheartj/ehy585 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 25. Gupta VB, Doecke JD, Hone E, et al. ; AIBL Research Group. Plasma apolipoprotein J as a potential biomarker for Alzheimer’s disease: Australian Imaging, Biomarkers and Lifestyle study of aging. Alzheimers Dement (Amst). 2016;3(1):18–26. 10.1016/j.dadm.2015.12.001 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 26. Englund DA, Sakamoto AE, Fritsche CM, et al. Exercise reduces circulating biomarkers of cellular senescence in humans. Aging Cell. 2021;20(7):e13415. 10.1111/acel.13415 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 27. Song S, Lee E, Kim H.. Does exercise affect telomere length? A systematic review and meta-analysis of randomized controlled trials. Medicina (Kaunas). 2022;58(2):242. 10.3390/medicina58020242 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 28. Dunn J, Thabet S, Jo H.. Flow-dependent epigenetic DNA methylation in endothelial gene expression and atherosclerosis. Arterioscler Thromb Vasc Biol. 2015;35(7):1562–1569. 10.1161/ATVBAHA.115.305042 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 29. Yi S-J, Kim K.. New insights into the role of histone changes in aging. Int J Mol Sci . 2020;21(21):8241. 10.3390/ijms21218241 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 30. Crouch J, Shvedova M, Thanapaul RJRS, Botchkarev V, Roh D.. Epigenetic regulation of cellular senescence. Cells. 2022;11(4):672. 10.3390/cells11040672 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 31. Sellami M, Bragazzi N, Prince MS, Denham J, Elrayess M.. Regular, intense exercise training as a healthy aging lifestyle strategy: preventing DNA damage, telomere shortening and adverse DNA methylation changes over a lifetime. Front Genet. 2021;12:652497. 10.3389/fgene.2021.652497 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 32. Mallard AR, Spathis JG, Coombes JS.. Nuclear factor (erythroid-derived 2)-like 2 (Nrf2) and exercise. Free Radic Biol Med. 2020;160:471–479. 10.1016/j.freeradbiomed.2020.08.024 [DOI] [PubMed] [Google Scholar]
  • 33. Singh CK, Chhabra G, Ndiaye MA, Garcia-Peterson LM, Mack NJ, Ahmad N.. The role of sirtuins in antioxidant and redox signaling. Antioxid Redox Signal. 2018;28(8):643–661. 10.1089/ars.2017.7290 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 34. Kobayashi E, Suzuki T, Funayama R, et al. Nrf2 suppresses macrophage inflammatory response by blocking proinflammatory cytokine transcription. Nat Commun. 2016;7(11624). 10.1038/ncomms11624 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 35. Done AJ, Newell MJ, Traustadóttir T.. Effect of exercise intensity on Nrf2 signalling in young men. Free Radic Res. 2017;51(6):646–655. 10.1080/10715762.2017.1353689 [DOI] [PubMed] [Google Scholar]
  • 36. Ballmann C, McGinnis G, Peters B, et al. Exercise-induced oxidative stress and hypoxic exercise recovery. Eur J Appl Physiol. 2014;114:725–733. 10.1007/s00421-013-2806-5 [DOI] [PubMed] [Google Scholar]
  • 37. Klaips CL, Jayaraj GG, Hartl FU.. Pathways of cellular proteostasis in aging and disease. J Cell Biol. 2018;217(1):51–63. 10.1083/jcb.201709072 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 38. Ungvari Z, Tarantini S, Donato AJ, Galvan V, Csiszar A.. Mechanisms of vascular aging. Circ Res. 2018;123(7):849–867. 10.1161/CIRCRESAHA.118.311378 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 39. Dai Z, Yang Z, Chen X, et al. The aging of glymphatic system in human brain and its correlation with brain charts and neuropsychological functioning. Cereb Cortex. 2023;33(12):7896–7903. 10.1093/cercor/bhad086 [DOI] [PubMed] [Google Scholar]
  • 40. VerPlank JJ, Lokireddy S, Zhao J, Goldberg AL.. 26S Proteasomes are rapidly activated by diverse hormones and physiological states that raise cAMP and cause Rpn6 phosphorylation. Proc Natl Acad Sci USA. 2019;116(10):4228–4237. 10.1073/pnas.1809254116 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 41. He XF, Liu DX, Zhang Q, et al. Voluntary exercise promotes glymphatic clearance of amyloid beta and reduces the activation of astrocytes and microglia in aged mice. Front Mol Neurosci. 2017;10:144. 10.3389/fnmol.2017.00144 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 42. Teixeira M, Gouveia M, Duarte A, et al. Regular exercise participation contributes to better proteostasis, inflammatory profile, and vasoactive profile in patients with hypertension. Am J Hypertens. 2020;33(2):119–123. 10.1093/ajh/hpz160 [DOI] [PubMed] [Google Scholar]
  • 43. Donato AJ, Walker AE, Magerko KA, et al. Life-long caloric restriction reduces oxidative stress and preserves nitric oxide bioavailability and function in arteries of old mice. Aging Cell. 2013;12(5):772–783. 10.1111/acel.12103 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 44. Duan W. Sirtuins: from metabolic regulation to brain aging. Front Aging Neurosci. 2013;5:36. 10.3389/fnagi.2013.00036 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 45. Wang S, Amato KR, Song W, et al. Regulation of endothelial cell proliferation and vascular assembly through distinct mTORC2 signaling pathways. Mol Cell Biol. 2015;35(7):1299–1313. 10.1128/MCB.00306-14 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 46. Chen Z-P, Stephens TJ, Murthy S, et al. Effect of exercise intensity on skeletal muscle AMPK signaling in humans. Diabetes. 2003;52(9):2205–2212. 10.2337/diabetes.52.9.2205 [DOI] [PubMed] [Google Scholar]
  • 47. Tsai C-L, Wang C-H, Pan C-Y, Chen F-C.. The effects of long-term resistance exercise on the relationship between neurocognitive performance and GH, IGF-1, and homocysteine levels in the elderly. Front Behav Neurosci. 2015;9:23. 10.3389/fnbeh.2015.00023 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 48. Ministrini S, Puspitasari YM, Beer G, Liberale L, Montecucco F, Camici GG.. Sirtuin 1 in endothelial dysfunction and cardiovascular aging. Front Physiol. 2021;12:733696. 10.3389/fphys.2021.733696 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 49. White AT, Schenk S.. NAD+/NADH and skeletal muscle mitochondrial adaptations to exercise. Am J Physiol Endocrinol Metab. 2012;303(3):E308–E321. 10.1152/ajpendo.00054.2012 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 50. Carapeto PV, Aguayo-Mazzucato C.. Effects of exercise on cellular and tissue aging. Aging (Albany NY). 2021;13(10):14522–14543. 10.18632/aging.203051 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 51. Li Y, Sun R, Zou J, Ying Y, Luo Z.. Dual roles of the AMP-activated protein kinase pathway in angiogenesis. Cells. 2019;8(7):752. 10.3390/cells8070752 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 52. Srivastava RAK, Pinkosky SL, Filippov S, Hanselman JC, Cramer CT, Newton RS.. AMP-activated protein kinase: an emerging drug target to regulate imbalances in lipid and carbohydrate metabolism to treat cardio-metabolic diseases: thematic review series: new lipid and lipoprotein targets for the treatment of cardiometabolic diseases. J Lipid Res. 2012;53(12):2490–2514. 10.1194/jlr.R025882 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 53. Amorim JA, Coppotelli G, Rolo AP, Palmeira CM, Ross JM, Sinclair DA.. Mitochondrial and metabolic dysfunction in ageing and age-related diseases. Nat Rev Endocrinol. 2022;18(4):243–258. 10.1038/s41574-021-00626-7 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 54. Grimm A, Friedland K, Eckert A.. Mitochondrial dysfunction: the missing link between aging and sporadic Alzheimer’s disease. Biogerontology. 2016;17:281–296. 10.1007/s10522-015-9618-4 [DOI] [PubMed] [Google Scholar]
  • 55. Li Y, Meng W, Hou Y, et al. Dual role of mitophagy in cardiovascular diseases. J Cardiovasc Pharmacol. 2021;78(1):e30–e39. 10.1097/FJC.0000000000001046 [DOI] [PubMed] [Google Scholar]
  • 56. Kirkman DL, Robinson AT, Rossman MJ, Seals DR, Edwards DG.. Mitochondrial contributions to vascular endothelial dysfunction, arterial stiffness, and cardiovascular diseases. Am J Physiol Heart Circ Physiol. 2021;320(5):H2080–H2100. 10.1152/ajpheart.00917.2020 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 57. Aanerud J, Borghammer P, Chakravarty MM, et al. Brain energy metabolism and blood flow differences in healthy aging. J Cereb Blood Flow Metab. 2012;32(7):1177–1187. 10.1038/jcbfm.2012.18 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 58. Alizadeh Pahlavani H, Laher I, Knechtle B, Zouhal H.. Exercise and mitochondrial mechanisms in patients with sarcopenia. Front Physiol. 2022;13:1040381. 10.3389/fphys.2022.1040381 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 59. Zhang X, Gao F.. Exercise improves vascular health: role of mitochondria. Free Radic Biol Med. 2021;177:347–359. 10.1016/j.freeradbiomed.2021.11.002 [DOI] [PubMed] [Google Scholar]
  • 60. Tachtsis B, Smiles WJ, Lane SC, Hawley JA, Camera DM.. Acute endurance exercise induces nuclear p53 abundance in human skeletal muscle. Front Physiol. 2016;7:144. 10.3389/fphys.2016.00144 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 61. Kim B, Lee H, Kawata K, Park J-Y.. Exercise-mediated wall shear stress increases mitochondrial biogenesis in vascular endothelium. PLoS One. 2014;9(11):e111409. 10.1371/journal.pone.0111409 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 62. Gorgoulis V, Adams PD, Alimonti A, et al. Cellular senescence: defining a path forward. Cell. 2019;179(4):813–827. 10.1016/j.cell.2019.10.005 [DOI] [PubMed] [Google Scholar]
  • 63. Graves SI, Baker DJ.. Implicating endothelial cell senescence to dysfunction in the ageing and diseased brain. Basic Clin Pharmacol Toxicol. 2020;127(2):102–110. 10.1111/bcpt.13403 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 64. 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(13):1541–1544. 10.1161/01.cir.0000013836.85741.17 [DOI] [PubMed] [Google Scholar]
  • 65. Nagata K, Yamazaki T, Takano D, et al. Cerebral circulation in aging. Ageing Res Rev. 2016;30:49–60. 10.1016/j.arr.2016.06.001 [DOI] [PubMed] [Google Scholar]
  • 66. Obermeier B, Daneman R, Ransohoff RM.. Development, maintenance and disruption of the blood-brain barrier. Nat Med. 2013;19(12):1584–1596. 10.1038/nm.3407 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 67. Verheggen I, Van Boxtel M, Verhey F, Jansen J, Backes W.. Interaction between blood-brain barrier and glymphatic system in solute clearance. Neurosci Biobehav Rev. 2018;90:26–33. 10.1016/j.neubiorev.2018.03.028 [DOI] [PubMed] [Google Scholar]
  • 68. Rossman MJ, Kaplon RE, Hill SD, et al. 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(5):H890–H895. 10.1152/ajpheart.00416.2017 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 69. Taddei S, Galetta F, Virdis A, et al. Physical activity prevents age-related impairment in nitric oxide availability in elderly athletes. Circulation. 2000;101(25):2896–2901. 10.1161/01.cir.101.25.2896 [DOI] [PubMed] [Google Scholar]
  • 70. Tenchov R, Sasso JM, Wang X, Zhou QA.. Aging hallmarks and anti-aging strategies: a landscape of research advancement. ACS Chem Neurosci. 2024;15(3):408–446. 10.1021/acschemneuro.3c00532 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 71. Jia G, Aroor AR, Jia C, Sowers JR.. Endothelial cell senescence in aging-related vascular dysfunction. Biochim Biophys Acta Mol Basis Dis. 2019;1865(7):1802–1809. 10.1016/j.bbadis.2018.08.008 [DOI] [PubMed] [Google Scholar]
  • 72. Li B, Lu X, Moeini M, Sakadžić S, Thorin E, Lesage F.. Atherosclerosis is associated with a decrease in cerebral microvascular blood flow and tissue oxygenation. PLoS One. 2019;14(8):e0221547. 10.1371/journal.pone.0221547 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 73. Xia WH, Li J, Su C, et al. Physical exercise attenuates age-associated reduction in endothelium-reparative capacity of endothelial progenitor cells by increasing CXCR4/JAK-2 signaling in healthy men. Aging Cell. 2012;11(1):111–119. 10.1111/j.1474-9726.2011.00758.x [DOI] [PubMed] [Google Scholar]
  • 74. Yin Y, Zhao X, Fang Y, et al. SDF-1α involved in mobilization and recruitment of endothelial progenitor cells after arterial injury in mice. Cardiovasc Pathol. 2010;19(4):218–227. 10.1016/j.carpath.2009.04.002 [DOI] [PubMed] [Google Scholar]
  • 75. Duda DG, Fukumura D, Jain RK.. Role of eNOS in neovascularization: NO for endothelial progenitor cells. Trends Mol Med. 2004;10(4):143–145. 10.1016/j.molmed.2004.02.001 [DOI] [PubMed] [Google Scholar]
  • 76. Chen H, Chen C, Spanos M, et al. Exercise training maintains cardiovascular health: signaling pathways involved and potential therapeutics. Signal Transduct Target Ther. 2022;7(1):306. 10.1038/s41392-022-01153-1 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 77. Yang Z, Xia W-H, Zhang Y-Y, et al. Shear stress-induced activation of Tie2-dependent signaling pathway enhances reendothelialization capacity of early endothelial progenitor cells. J Mol Cell Cardiol. 2012;52(5):1155–1163. 10.1016/j.yjmcc.2012.01.019 [DOI] [PubMed] [Google Scholar]
  • 78. Guo S, Lo EH.. Dysfunctional cell-cell signaling in the neurovascular unit as a paradigm for central nervous system disease. Stroke. 2009;40(3_suppl_1):S4–S7. 10.1161/STROKEAHA.108.534388 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 79. Iadecola C, Smith EE, Anrather J, et al. ; American Heart Association Stroke Council Council on Arteriosclerosis Thrombosis and Vascular Biology Council on Cardiovascular Radiology and Intervention Council on Hypertension Council on Lifestyle and Cardiometabolic Health. The Neurovasculome: key roles in brain health and cognitive impairment: a scientific statement from the American Heart Association/American Stroke Association. Stroke. 2023;54(6):e251–e271. 10.1161/STR.0000000000000431 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 80. Ramanathan A, Nelson AR, Sagare AP, Zlokovic BV.. Impaired vascular-mediated clearance of brain amyloid beta in Alzheimer’s disease: the role, regulation and restoration of LRP1. Front Aging Neurosci. 2015;7:136. 10.3389/fnagi.2015.00136 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 81. Woods JA, Wilund KR, Martin SA, Kistler BM.. Exercise, inflammation and aging. Aging Dis. 2012;3(1):130–140. [PMC free article] [PubMed] [Google Scholar]
  • 82. Fischer CP. Interleukin-6 in acute exercise and training: what is the biological relevance. Exerc Immunol Rev. 2006;12(6-33):41. [PubMed] [Google Scholar]
  • 83. Cerqueira E, Marinho DA, Neiva HP, Lourenço O.. Inflammatory effects of high and moderate intensity exercise—a systematic review. Front Physiol. 2020;10:1550. 10.3389/fphys.2019.01550 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 84. Brandt C, Pedersen BK.. The role of exercise-induced myokines in muscle homeostasis and the defense against chronic diseases. Biomed Res Int. 2010;2010:520258. 10.1155/2010/520258 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 85. Pedersen BK, Febbraio MA.. Muscle as an endocrine organ: focus on muscle-derived interleukin-6. Physiol Rev. 2008;88(4):1379–1406. 10.1152/physrev.90100.2007 [DOI] [PubMed] [Google Scholar]
  • 86. Starkie R, Ostrowski SR, Jauffred S, Febbraio M, Pedersen BK.. Exercise and IL-6 infusion inhibit endotoxin-induced TNF-α production in humans. FASEB J. 2003;17(8):1–10. 10.1096/fj.02-0670fje [DOI] [PubMed] [Google Scholar]
  • 87. Green DJ, Hopman MT, Padilla J, Laughlin MH, Thijssen DH.. Vascular adaptation to exercise in humans: role of hemodynamic stimuli. Physiol Rev. 2017;97(2):495–528. 10.1152/physrev.00014.2016 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 88. Green DJ, Smith KJ.. Effects of exercise on vascular function, structure, and health in humans. Cold Spring Harb Perspect Med. 2018;8(4):a029819. 10.1101/cshperspect.a029819 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 89. Bliss ES, Wong RH, Howe PR, Mills DE.. Benefits of exercise training on cerebrovascular and cognitive function in ageing. J Cereb Blood Flow Metab. 2021;41(3):447–470. 10.1177/0271678X20957807 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 90. Williams L, Leggett R.. Reference values for resting blood flow to organs of man. Clin Phys Physiol Meas. 1989;10(3):187. 10.1088/0143-0815/10/3/001 [DOI] [PubMed] [Google Scholar]
  • 91. Cipolla MJ. The cerebral circulation. In: Integrated Systems Physiology: From Molecule to Function. Vol. 1. Morgan & Claypool Life Sciences; 2009:1–59. 10.4199/c00005ed1v01y200912isp002 [DOI] [PubMed] [Google Scholar]
  • 92. Figley CR, Stroman PW.. The role(s) of astrocytes and astrocyte activity in neurometabolism, neurovascular coupling, and the production of functional neuroimaging signals. Eur J Neurosci. 2011;33(4):577–588. 10.1111/j.1460-9568.2010.07584.x [DOI] [PubMed] [Google Scholar]
  • 93. Filosa JA, Morrison HW, Iddings JA, Du W, Kim KJ.. Beyond neurovascular coupling, role of astrocytes in the regulation of vascular tone. Neuroscience. 2016;323:96–109. 10.1016/j.neuroscience.2015.03.064 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 94. Fulop GA, Tarantini S, Yabluchanskiy A, et al. Role of age-related alterations of the cerebral venous circulation in the pathogenesis of vascular cognitive impairment. Am J Physiol Heart Circ Physiol. 2019;316(5):H1124–H1140. 10.1152/ajpheart.00776.2018 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 95. Kovacic JC, Moreno P, Nabel EG, Hachinski V, Fuster V.. Cellular senescence, vascular disease, and aging: part 2 of a 2-part review: clinical vascular disease in the elderly. Circulation. 2011;123(17):1900–1910. 10.1161/CIRCULATIONAHA.110.009118 [DOI] [PubMed] [Google Scholar]
  • 96. Ainslie PN, Cotter JD, George KP, et al. Elevation in cerebral blood flow velocity with aerobic fitness throughout healthy human ageing. J Physiol. 2008;586(16):4005–4010. 10.1113/jphysiol.2008.158279 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 97. Duong MT, Nasrallah IM, Wolk DA, Chang CC, Chang T-Y.. Cholesterol, atherosclerosis, and APOE in vascular contributions to cognitive impairment and dementia (VCID): potential mechanisms and therapy. Front Aging Neurosci. 2021;13:647990. 10.3389/fnagi.2021.647990 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 98. Hall CN, Reynell C, Gesslein B, et al. Capillary pericytes regulate cerebral blood flow in health and disease. Nature. 2014;508(7494):55–60. 10.1038/nature13165 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 99. Gorelick PB, Scuteri A, Black SE, et al. ; American Heart Association Stroke Council, Council on Epidemiology and Prevention, Council on Cardiovascular Nursing, Council on Cardiovascular Radiology and Intervention, and Council on Cardiovascular Surgery and Anesthesia. Vascular contributions to cognitive impairment and dementia: a statement for healthcare professionals from the American Heart Association/American Stroke Association. Stroke. 2011;42(9):2672–2713. 10.1161/STR.0b013e3182299496 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 100. Forstermann U, Munzel T.. Endothelial nitric oxide synthase in vascular disease: from marvel to menace. Circulation. 2006;113(13):1708–1714. 10.1161/CIRCULATIONAHA.105.602532 [DOI] [PubMed] [Google Scholar]
  • 101. Festoff BW, Sajja RK, van Dreden P, Cucullo L.. HMGB1 and thrombin mediate the blood-brain barrier dysfunction acting as biomarkers of neuroinflammation and progression to neurodegeneration in Alzheimer’s disease. J Neuroinflamm. 2016;13(1):1–12. 10.1186/s12974-016-0670-z [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 102. Cechetti F, Pagnussat AS, Worm PV, et al. Chronic brain hypoperfusion causes early glial activation and neuronal death, and subsequent long-term memory impairment. Brain Res Bull. 2012;87(1):109–116. 10.1016/j.brainresbull.2011.10.006 [DOI] [PubMed] [Google Scholar]
  • 103. Dolui S, Detre JA, Gaussoin SA, et al. Association of intensive vs standard blood pressure control with cerebral blood flow: secondary analysis of the SPRINT MIND randomized clinical trial. JAMA Neurol. 2022;79(4):380–389. 10.1001/jamaneurol.2022.0074 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 104. Lipsitz LA, Gagnon M, Vyas M, et al. Antihypertensive therapy increases cerebral blood flow and carotid distensibility in hypertensive elderly subjects. Hypertension. 2005;45(2):216–221. 10.1161/01.HYP.0000153094.09615.11 [DOI] [PubMed] [Google Scholar]
  • 105. Joutel A, Monet-Leprêtre M, Gosele C, et al. Cerebrovascular dysfunction and microcirculation rarefaction precede white matter lesions in a mouse genetic model of cerebral ischemic small vessel disease. J Clin Invest. 2010;120(2):433–445. 10.1172/JCI39733 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 106. Mitchell GF, Van Buchem MA, Sigurdsson S, et al. Arterial stiffness, pressure and flow pulsatility and brain structure and function: the Age, Gene/Environment Susceptibility–Reykjavik study. Brain. 2011;134(11):3398–3407. 10.1093/brain/awr253 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 107. Meyer ML, Palta P, Tanaka H, et al. Association of central arterial stiffness and pressure pulsatility with mild cognitive impairment and dementia: the Atherosclerosis Risk in Communities Study-Neurocognitive Study (ARIC-NCS). J Alzheimers Dis. 2017;57(1):195–204. 10.3233/JAD-161041 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 108. Tarumi T, Zhang R.. Cerebral blood flow in normal aging adults: cardiovascular determinants, clinical implications, and aerobic fitness. J Neurochem. 2017;144:595–608. 10.1111/jnc.14234 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 109. Suri S, Chiesa ST, Zsoldos E, et al. Associations between arterial stiffening and brain structure, perfusion, and cognition in the Whitehall II Imaging Sub-study: a retrospective cohort study. PLoS Med. 2020;17(12):e1003467. 10.1371/journal.pmed.1003467 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 110. Tsao CW, Himali JJ, Beiser AS, et al. Association of arterial stiffness with progression of subclinical brain and cognitive disease. Neurology. 2016;86(7):619–626. 10.1212/WNL.0000000000002368 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 111. Singer J, Trollor JN, Baune BT, Sachdev PS, Smith E.. Arterial stiffness, the brain and cognition: a systematic review. Ageing Res Rev. 2014;15:16–27. 10.1016/j.arr.2014.02.002 [DOI] [PubMed] [Google Scholar]
  • 112. Kitazaw M, Yamasaki TR, Laferla FM.. Microglia as a potential bridge between the amyloid β-peptide and tau. Ann N Y Acad Sci. 2004;1035(1):85–103. 10.1196/annals.1332.006 [DOI] [PubMed] [Google Scholar]
  • 113. Walters A, Phillips E, Zheng R, Biju M, Kuruvilla T.. Evidence for neuroinflammation in Alzheimer’s disease. Prog Neurol Psychiatry. 2016;20(5):25–31. 10.1002/pnp.444 [DOI] [Google Scholar]
  • 114. Ding YH, Li J, Yao WX, Rafols JA, Clark JC, Ding Y.. Exercise preconditioning upregulates cerebral integrins and enhances cerebrovascular integrity in ischemic rats. Acta Neuropathol. 2006;112(1):74–84. 10.1007/s00401-006-0076-6 [DOI] [PubMed] [Google Scholar]
  • 115. DeSouza CA, Shapiro LF, Clevenger CM, et al. Regular aerobic exercise prevents and restores age-related declines in endothelium-dependent vasodilation in healthy men. Circulation. 2000;102(12):1351–1357. 10.1161/01.cir.102.12.1351 [DOI] [PubMed] [Google Scholar]
  • 116. López-Otín C, Blasco MA, Partridge L, Serrano M, Kroemer G.. The hallmarks of aging. Cell. 2013;153(6):1194–1217. 10.1016/j.cell.2013.05.039 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 117. Ungvari Z, Kaley G, de Cabo R, Sonntag WE, Csiszar A.. Mechanisms of vascular aging: new perspectives . J Gerontol A Biol Sci Med Sci. 2010;65(10):1028–1041. 10.1093/gerona/glq113 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 118. Ding Y-N, Tang X, Chen H-Z, Liu D-P.. Epigenetic regulation of vascular aging and age-related vascular diseases. Adv Exp Med Biol.. 2018;1086:55–75. 10.1007/978-981-13-1117-8_4 [DOI] [PubMed] [Google Scholar]
  • 119. Katsuumi G, Shimizu I, Yoshida Y, Minamino T.. Vascular senescence in cardiovascular and metabolic diseases. Front Cardiovasc Med. 2018;5:18. 10.3389/fcvm.2018.00018 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 120. Ya J, Bayraktutan U.. Vascular ageing: mechanisms, risk factors, and treatment strategies. Int J Mol Sci . 2023;24(14):11538. 10.3390/ijms241411538 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 121. Aubert G, Lansdorp PM.. Telomeres and aging. Physiol Rev. 2008;88(2):557–579. 10.1152/physrev.00026.2007 [DOI] [PubMed] [Google Scholar]
  • 122. Bautista-Niño PK, Portilla-Fernandez E, Vaughan DE, Danser AJ, Roks AJ.. DNA damage: a main determinant of vascular aging. Int J Mol Sci . 2016;17(5):748. 10.3390/ijms17050748 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 123. Samjoo I, Safdar A, Hamadeh M, Raha S, Tarnopolsky M.. The effect of endurance exercise on both skeletal muscle and systemic oxidative stress in previously sedentary obese men. Nutr Diabetes. 2013;3(9):e88–e88. 10.1038/nutd.2013.30 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 124. Leonardo-Mendonça RC, Concepción-Huertas M, Guerra-Hernandez E, Zabala M, Escames G, Acuna-Castroviejo D.. Redox status and antioxidant response in professional cyclists during training. Eur J Sport Sci. 2014;14(8):830–838. 10.1080/17461391.2014.915345 [DOI] [PubMed] [Google Scholar]
  • 125. Rádak Z, Marton O, Nagy E, Koltai E, Goto S.. The complex role of physical exercise and reactive oxygen species on brain. J Sport Health Sci. 2013;2(2):87–93. 10.1016/j.jshs.2013.04.001 [DOI] [Google Scholar]
  • 126. Ryan NA, Zwetsloot KA, Westerkamp LM, Hickner RC, Pofahl WE, Gavin TP.. Lower skeletal muscle capillarization and VEGF expression in aged vs. young men. J Appl Physiol (1985) 2006;100(1):178–185. 10.1152/japplphysiol.00827.2005 [DOI] [PubMed] [Google Scholar]
  • 127. Croley AN, Zwetsloot KA, Westerkamp LM, et al. Lower capillarization, VEGF protein, and VEGF mRNA response to acute exercise in the vastus lateralis muscle of aged vs. young women. J Appl Physiol (1985) 2005;99(5):1872–1879. 10.1152/japplphysiol.00498.2005 [DOI] [PubMed] [Google Scholar]
  • 128. Ding YH, Li J, Zhou Y, Rafols JA, Clark JC, Ding Y.. Cerebral angiogenesis and expression of angiogenic factors in aging rats after exercise. Curr Neurovasc Res. 2006;3(1):15–23. 10.2174/156720206775541787 [DOI] [PubMed] [Google Scholar]
  • 129. Venkatesh GH. Hypoxic stress perturb DNA repair mechanisms leading to genetic instability. In: Chakraborti S, Ray BK, Roychoudhury S, eds. Handbook of Oxidative Stress in Cancer: Mechanistic Aspects. Springer; 2022:859–874. [Google Scholar]
  • 130. Amaral SL, Papanek PE, Greene AS.. Angiotensin II and VEGF are involved in angiogenesis induced by short-term exercise training. Am J Physiol Heart Circ Physiol. 2001;281(3):H1163–H1169. 10.1152/ajpheart.2001.281.3.H1163 [DOI] [PubMed] [Google Scholar]
  • 131. Morland C, Andersson KA, Haugen OP, et al. Exercise induces cerebral VEGF and angiogenesis via the lactate receptor HCAR1. Nat Commun. 2017;8(1):15557. 10.1038/ncomms15557 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 132. Vital TM, Stein AM, de Melo Coelho FG, Arantes FJ, Teodorov E, Santos-Galduróz RF.. Physical exercise and vascular endothelial growth factor (VEGF) in elderly: a systematic review. Arch Gerontol Geriatr. 2014;59(2):234–239. 10.1016/j.archger.2014.04.011 [DOI] [PubMed] [Google Scholar]
  • 133. Karakilic A, Yüksel O, Kızıldağ S, et al. Regular aerobic exercise increased VEGF levels in both soleus and gastrocnemius muscles correlated with hippocampal learning and VEGF levels. Acta Neurobiol Exp. 2021;81(1):1–9. 10.21307/ane-2021-001 [DOI] [PubMed] [Google Scholar]
  • 134. Fabel K, Fabel K, Tam B, et al. VEGF is necessary for exercise-induced adult hippocampalneurogenesis. Eur J Neurosci. 2003 2003;18:2803–2812. 10.1046/j.1460-9568.2003.03041.x [DOI] [PubMed] [Google Scholar]
  • 135. Yang J, Shanahan KJ, Shriver LP, Luciano MG.. Exercise-induced changes of cerebrospinal fluid vascular endothelial growth factor in adult chronic hydrocephalus patients. J Clin Neurosci. 2016;24:52–56. 10.1016/j.jocn.2015.08.019 [DOI] [PubMed] [Google Scholar]
  • 136. Von Zglinicki T, Serra V, Lorenz M, et al. Short telomeres in patients with vascular dementia: an indicator of low antioxidative capacity and a possible risk factor? Lab Invest. 2000;80(11):1739–1747. 10.1038/labinvest.3780184 [DOI] [PubMed] [Google Scholar]
  • 137. Löf-Öhlin ZM, Hagnelius N-O, Nilsson TK.. Relative telomere length in patients with late-onset Alzheimer’s dementia or vascular dementia. Neuroreport. 2008;19(12):1199–1202. 10.1097/WNR.0b013e3283089220 [DOI] [PubMed] [Google Scholar]
  • 138. Wright WE, Piatyszek MA, Rainey WE, Byrd W, Shay JW.. Telomerase activity in human germline and embryonic tissues and cells. Dev Genet. 1996;18(2):173–179. [DOI] [PubMed] [Google Scholar]
  • 139. Cifuentes-Rojas C, Shippen DE.. Telomerase regulation. Mutat Res. 2012;730(1–2):20–27. 10.1016/j.mrfmmm.2011.10.003 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 140. Werner C, Fürster T, Widmann T, et al. Physical exercise prevents cellular senescence in circulating leukocytes and in the vessel wall. Circulation. 2009;120(24):2438–2447. 10.1161/circulationaha.109.861005 [DOI] [PubMed] [Google Scholar]
  • 141. Frej F, Peter MN.. Telomere biology and vascular aging. In: Nilsson PM, Olsen MH, Laurent S, eds. Early Vascular Aging (EVA). Elsevier; 2015:201–211. [Google Scholar]
  • 142. Brouilette SW, Moore JS, McMahon AD, et al. ; West of Scotland Coronary Prevention Study Group. Telomere length, risk of coronary heart disease, and statin treatment in the West of Scotland Primary Prevention Study: a nested case-control study. Lancet (Lond). 2007;369(9556):107–114. 10.1016/S0140-6736(07)60071-3 [DOI] [PubMed] [Google Scholar]
  • 143. Yetim E, Topcuoglu MA, Yurur Kutlay N, Tukun A, Oguz KK, Arsava EM.. The association between telomere length and ischemic stroke risk and phenotype. Sci Rep. 2021;11(1):10967. 10.1038/s41598-021-90435-9 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 144. Baechle JJ, Chen N, Makhijani P, Winer S, Furman D, Winer DA.. Chronic inflammation and the hallmarks of aging. Mol Metab. 2023;74:101755. 10.1016/j.molmet.2023.101755 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 145. Arce Rentería M, Gillett SR, McClure LA, et al. C-reactive protein and risk of cognitive decline: the REGARDS study. PLoS One. 2020;15(12):e0244612. 10.1371/journal.pone.0244612 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 146. De Miguel Z, Khoury N, Betley MJ, et al. Exercise plasma boosts memory and dampens brain inflammation via clusterin. Nature. 2021;600(7889):494–499. 10.1038/s41586-021-04183-x [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 147. Galea I. The blood–brain barrier in systemic infection and inflammation. Cell Mol Immunol. 2021;18(11):2489–2501. 10.1038/s41423-021-00757-x [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 148. Wyss-Coray T. Inflammation in Alzheimer disease: driving force, bystander or beneficial response? Nat Med. 2006;12(9):1005–1015. 10.1038/nm1484 [DOI] [PubMed] [Google Scholar]
  • 149. Harvey ZH, Chen Y, Jarosz DF.. Protein-based inheritance: epigenetics beyond the chromosome. Mol Cell. 2018;69(2):195–202. 10.1016/j.molcel.2017.10.030 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 150. Xiao X, Liu X, Jiao B.. Epigenetics: recent advances and its role in the treatment of Alzheimer’s disease. Front Neurol. 2020;11:538301. 10.3389/fneur.2020.538301 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 151. Keen A, Zhang F, Reader JS, Tzima E.. Proteostasis and resilience in the mechanically-stressed vascular endothelium. Curr Opin Physiol. 2023;34:100673. 10.1016/j.cophys.2023.100673 [DOI] [Google Scholar]
  • 152. Hybertson BM, Gao B, Bose SK, McCord JM.. Oxidative stress in health and disease: the therapeutic potential of Nrf2 activation. Mol Aspects Med. 2011;32(4-6):234–246. 10.1016/j.mam.2011.10.006 [DOI] [PubMed] [Google Scholar]
  • 153. Johnson JA, Johnson DA, Kraft AD, et al. The Nrf2–ARE pathway: an indicator and modulator of oxidative stress in neurodegeneration. Ann N Y Acad Sci. 2008;1147(1):61–69. 10.1196/annals.1427.036 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 154. Karan A, Bhakkiyalakshmi E, Jayasuriya R, Sarada D, Ramkumar KM.. The pivotal role of nuclear factor erythroid 2-related factor 2 in diabetes-induced endothelial dysfunction. Pharmacol Res. 2020;153:104601. 10.1016/j.phrs.2019.104601 [DOI] [PubMed] [Google Scholar]
  • 155. Huang Z, Wu M, Zeng L, Wang D.. The beneficial role of Nrf2 in the endothelial dysfunction of atherosclerosis. Cardiol Res Pract. 2022;2022:1–7. 10.1155/2022/4287711 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 156. Yang C, Zhang X, Fan H, Liu Y.. Curcumin upregulates transcription factor Nrf2, HO-1 expression and protects rat brains against focal ischemia. Brain Res. 2009;1282:133–141. 10.1016/j.brainres.2009.05.009 [DOI] [PubMed] [Google Scholar]
  • 157. Shih AY, Li P, Murphy TH.. A small-molecule-inducible Nrf2-mediated antioxidant response provides effective prophylaxis against cerebral ischemia in vivo. J Neurosci. 2005;25(44):10321–10335. 10.1523/JNEUROSCI.4014-05.2005 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 158. Sivandzade F, Prasad S, Bhalerao A, Cucullo L.. NRF2 and NF-қB interplay in cerebrovascular and neurodegenerative disorders: molecular mechanisms and possible therapeutic approaches. Redox Biol. 2019;21:101059. 10.1016/j.redox.2018.11.017 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 159. Alfieri A, Srivastava S, Siow RC, et al. Sulforaphane preconditioning of the Nrf2/HO-1 defense pathway protects the cerebral vasculature against blood–brain barrier disruption and neurological deficits in stroke. Free Radic Biol Med. 2013;65:1012–1022. 10.1016/j.freeradbiomed.2013.08.190 [DOI] [PubMed] [Google Scholar]
  • 160. Jazwa A, Rojo AI, Innamorato NG, Hesse M, Fernández-Ruiz J, Cuadrado A.. Pharmacological targeting of the transcription factor Nrf2 at the basal ganglia provides disease modifying therapy for experimental parkinsonism. Antioxid Redox Signal. 2011;14(12):2347–2360. 10.1089/ars.2010.3731 [DOI] [PubMed] [Google Scholar]
  • 161. Done AJ, Gage MJ, Nieto NC, Traustadóttir T.. Exercise-induced Nrf2-signaling is impaired in aging. Free Radic Biol Med. 2016;96:130–138. 10.1016/j.freeradbiomed.2016.04.024 [DOI] [PubMed] [Google Scholar]
  • 162. Safdar A, deBeer J, Tarnopolsky MA.. Dysfunctional Nrf2–Keap1 redox signaling in skeletal muscle of the sedentary old. Free Radic Biol Med. 2010;49(10):1487–1493. 10.1016/j.freeradbiomed.2010.08.010 [DOI] [PubMed] [Google Scholar]
  • 163. Tutakhail A, Nazary QA, Lebsir D, Kerdine-Romer S, Coudore F.. Induction of brain Nrf2-HO-1 pathway and antinociception after different physical training paradigms in mice. Life Sci. 2018;209:149–156. 10.1016/j.lfs.2018.08.004 [DOI] [PubMed] [Google Scholar]
  • 164. Sandberg M, Patil J, D’Angelo B, Weber SG, Mallard C.. NRF2-regulation in brain health and disease: implication of cerebral inflammation. Neuropharmacology. 2014;79:298–306. 10.1016/j.neuropharm.2013.11.004 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 165. Ochneva A, Zorkina Y, Abramova O, et al. Protein Misfolding and aggregation in the brain: Common Pathogenetic pathways in neurodegenerative and mental disorders. Int J Mol Sci . 2022;23(22):14498. 10.3390/ijms232214498 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 166. Rozales K, Younis A, Saida N, et al. Differential roles for DNAJ isoforms in HTT-polyQ and FUS aggregation modulation revealed by chaperone screens. Nat Commun. 2022;13(1):516. 10.1038/s41467-022-27982-w [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 167. Tsoi PS, Quan MD, Ferreon JC, Ferreon ACM.. Aggregation of disordered proteins associated with neurodegeneration. Int J Mol Sci. 2023;24(4):3380. 10.3390/ijms24043380 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 168. Miura Y, Tsumoto H, Iwamoto M, et al. Age-associated proteomic alterations in human aortic media. Geriatr Gerontol Int. 2019;19(10):1054–1062. 10.1111/ggi.13757 [DOI] [PubMed] [Google Scholar]
  • 169. Degenhardt K, Wagner J, Skodras A, et al. Medin aggregation causes cerebrovascular dysfunction in aging wild-type mice. Proc Natl Acad Sci USA. 2020;117(38):23925–23931. 10.1073/pnas.2011133117 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 170. Karamanova N, Truran S, Serrano GE, et al. Endothelial immune activation by medin: potential role in cerebrovascular disease and reversal by monosialoganglioside-containing nanoliposomes. J Am Heart Assoc. 2020;9(2):e014810. 10.1161/JAHA.119.014810 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 171. Jessen NA, Munk ASF, Lundgaard I, Nedergaard M.. The Glymphatic System: a beginner’s guide. Neurochem Res. 2015;40(12):2583–2599. 10.1007/s11064-015-1581-6 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 172. Siow TY, Toh CH, Hsu J-L, et al. Association of sleep, neuropsychological performance, and gray matter volume with glymphatic function in community-dwelling older adults. Neurology. 2022;98(8):e829–e838. 10.1212/WNL.0000000000013215 [DOI] [PubMed] [Google Scholar]
  • 173. Campos JC, Queliconi BB, Bozi LH, et al. Exercise reestablishes autophagic flux and mitochondrial quality control in heart failure. Autophagy. 2017;13(8):1304–1317. 10.1080/15548627.2017.1325062 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 174. McMillan EM, Paré M-F, Baechler BL, Graham DA, Rush JW, Quadrilatero J.. Autophagic signaling and proteolytic enzyme activity in cardiac and skeletal muscle of spontaneously hypertensive rats following chronic aerobic exercise. PLoS One. 2015;10(3):e0119382. 10.1371/journal.pone.0119382 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 175. Bonanni R, Cariati I, Tarantino U, D’Arcangelo G, Tancredi V.. Physical exercise and health: a focus on its protective role in neurodegenerative diseases. J Funct Morphol Kinesiol. 2022;7(2):38. 10.3390/jfmk7020038 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 176. Iliff JJ, Wang M, Zeppenfeld DM, et al. Cerebral arterial pulsation drives paravascular CSF–interstitial fluid exchange in the murine brain. J Neurosci. 2013;33(46):18190–18199. 10.1523/jneurosci.1592-13.2013 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 177. von Holstein-Rathlou S, Petersen NC, Nedergaard M.. Voluntary running enhances glymphatic influx in awake behaving, young mice. Neurosci Lett. 2018;662:253–258. 10.1016/j.neulet.2017.10.035 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 178. Herzig S, Shaw RJ.. AMPK: guardian of metabolism and mitochondrial homeostasis. Nat Rev Mol Cell Biol. 2018;19(2):121–135. 10.1038/nrm.2017.95 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 179. Zhang Y, Lee T-S, Kolb EM, et al. AMP-activated protein kinase is involved in endothelial NO synthase activation in response to shear stress. Arterioscler Thromb Vasc Biol. 2006;26(6):1281–1287. 10.1161/01.ATV.0000221230.08596.98 [DOI] [PubMed] [Google Scholar]
  • 180. Caccamo A, Majumder S, Richardson A, Strong R, Oddo S.. Molecular interplay between mammalian target of rapamycin (mTOR), amyloid-β, and Tau: effects on cognitive impairments. J Biol Chem. 2010;285(17):13107–13120. 10.1074/jbc.M110.100420 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 181. Panwar V, Singh A, Bhatt M, et al. Multifaceted role of mTOR (mammalian target of rapamycin) signaling pathway in human health and disease. Signal Transduct Target Ther. 2023;8(1):375. 10.1038/s41392-023-01608-z [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 182. Zakhary SM, Ayubcha D, Dileo JN, et al. Distribution analysis of deacetylase SIRT1 in rodent and human nervous systems. Anat Rec (Hoboken). 2010;293(6):1024–1032. 10.1002/ar.21116 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 183. Tajbakhsh N, Sokoya EM.. Regulation of cerebral vascular function by sirtuin 1. Microcirculation. 2012;19(4):336–342. 10.1111/j.1549-8719.2012.00167.x [DOI] [PubMed] [Google Scholar]
  • 184. Hattori Y, Okamoto Y, Nagatsuka K, et al. SIRT1 attenuates severe ischemic damage by preserving cerebral blood flow. Neuroreport. 2015;26(3):113–117. 10.1097/WNR.0000000000000308 [DOI] [PubMed] [Google Scholar]
  • 185. Ungvari Z, Fazekas-Pongor V, Csiszar A, Kunutsor SK.. The multifaceted benefits of walking for healthy aging: from Blue Zones to molecular mechanisms. GeroScience. 2023;45:3211–3239. 10.1007/s11357-023-00873-8 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 186. Sonntag WE, Eckman DM, Ingraham J, Riddle DR.. Regulation of cerebrovascular aging. In: Riddle DR, ed. Brain Aging: Models, Methods, and Mechanisms. CRC Press; 2007:279. 10.1201/9781420005523-12 [DOI] [PubMed] [Google Scholar]
  • 187. Sonntag WE, Lynch C, Thornton P, Khan A, Bennett S, Ingram R.. The effects of growth hormone and IGF-1 deficiency on cerebrovascular and brain ageing. J Anat. 2000;197 Pt 4(4):575–585. 10.1046/j.1469-7580.2000.19740575.x [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 188. Stein AM, Silva TMV, Coelho FGM, et al. Physical exercise, IGF-1 and cognition A systematic review of experimental studies in the elderly. Dement Neuropsychol. 2018;12(2):114–122. 10.1590/1980-57642018dn12-020003 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 189. Angelini A, Bendini C, Neviani F, et al. Insulin-like growth factor-1 (IGF-1): relation with cognitive functioning and neuroimaging marker of brain damage in a sample of hypertensive elderly subjects. Arch Gerontol Geriatr. 2009;49:5–12. 10.1016/j.archger.2009.09.006 [DOI] [PubMed] [Google Scholar]
  • 190. Fang EF, Lautrup S, Hou Y, et al. NAD+ in aging: molecular mechanisms and translational implications. Trends Mol Med. 2017;23(10):899–916. 10.1016/j.molmed.2017.08.001 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 191. Fagerli E, Escobar I, Ferrier FJ, Jackson CW, Perez-Lao EJ, Perez-Pinzon MA.. Sirtuins and cognition: implications for learning and memory in neurological disorders. Front Physiol. 2022;13:908689. 10.3389/fphys.2022.908689 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 192. Glick D, Barth S, Macleod KF.. Autophagy: cellular and molecular mechanisms. J Pathol. 2010;221(1):3–12. 10.1002/path.2697 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 193. Kim I, Rodriguez-Enriquez S, Lemasters JJ.. Selective degradation of mitochondria by mitophagy. Arch Biochem Biophys. 2007;462(2):245–253. 10.1016/j.abb.2007.03.034 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 194. Sharma C, Kim S, Nam Y, Jung UJ, Kim SR.. Mitochondrial dysfunction as a driver of cognitive impairment in Alzheimer’s disease. Int J Mol Sci . 2021;22(9):4850. 10.3390/ijms22094850 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 195. Schreckenberger ZJ, Wenceslau CF, Joe B, McCarthy CG.. Mitophagy in hypertension-associated premature vascular aging. Am J Hypertens. 2020;33(9):804–812. 10.1093/ajh/hpaa058 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 196. Bhatia S, Rawal R, Sharma P, Singh T, Singh M, Singh V.. Mitochondrial dysfunction in Alzheimer’s disease: opportunities for drug development. Curr Neuropharmacol. 2022;20(4):675–692. 10.2174/1570159X19666210517114016 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 197. Apaijai N, Sriwichaiin S, Phrommintikul A, et al. Cognitive impairment is associated with mitochondrial dysfunction in peripheral blood mononuclear cells of elderly population. Sci Rep. 2020;10(1):21400. 10.1038/s41598-020-78551-4 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 198. Arany Z, Foo S-Y, Ma Y, et al. HIF-independent regulation of VEGF and angiogenesis by the transcriptional coactivator PGC-1α. Nature. 2008;451(7181):1008–1012. 10.1038/nature06613 [DOI] [PubMed] [Google Scholar]
  • 199. Koh Y, Park J.. Cell adhesion molecules and exercise. J Inflamm Res. 2018;11:297–306. 10.2147/jir.s170262 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 200. Sure VN, Sakamuri SS, Sperling JA, et al. A novel high-throughput assay for respiration in isolated brain microvessels reveals impaired mitochondrial function in the aged mice. Geroscience. 2018;40:365–375. 10.1007/s11357-018-0037-8 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 201. Han Y, Kim SY.. Endothelial senescence in vascular diseases: current understanding and future opportunities in senotherapeutics. Exp Mol Med. 2023;55(1):1–12. 10.1038/s12276-022-00906-w [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 202. Daniele S, Da Pozzo E, Iofrida C, Martini C.. Human neural stem cell aging is counteracted by α-glycerylphosphorylethanolamine. ACS Chem Neurosci. 2016;7(7):952–963. 10.1021/acschemneuro.6b00078 [DOI] [PubMed] [Google Scholar]
  • 203. Nicaise AM, Willis CM, Crocker SJ, Pluchino S.. Stem cells of the aging brain. Front Aging Neurosci. 2020;12:247. 10.3389/fnagi.2020.00247 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 204. Dehkordi SK, Walker J, Sah E, et al. Profiling senescent cells in human brains reveals neurons with CDKN2D/p19 and tau neuropathology. Nat Aging. 2021;1(12):1107–1116. 10.1038/s43587-021-00142-3 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 205. Zhang P, Kishimoto Y, Grammatikakis I, et al. Senolytic therapy alleviates Aβ-associated oligodendrocyte progenitor cell senescence and cognitive deficits in an Alzheimer’s disease model. Nat Neurosci. 2019;22(5):719–728. 10.1038/s41593-019-0372-9 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 206. Gonzales MM, Garbarino VR, Kautz TF, et al. Senolytic therapy in mild Alzheimer’s disease: a phase 1 feasibility trial. Nat Med. 2023;29:2481–2488. 10.1038/s41591-023-02543-w [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 207. Li Y, Tollefsbol TO.. p16INK4a suppression by glucose restriction contributes to human cellular lifespan extension through SIRT1-mediated epigenetic and genetic mechanisms. PLoS One. 2011;6(2):e17421. 10.1371/journal.pone.0017421 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 208. Sikora E, Bielak-Zmijewska A, Dudkowska M, et al. Cellular senescence in brain aging. Front Aging Neurosci. 2021;13:646924. 10.3389/fnagi.2021.646924 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 209. Dos Santos LM, Trombetta-Lima M, Eggen B, Demaria M.. Cellular senescence in brain aging and neurodegeneration. Ageing Res Rev. 2023;93:102141. 10.1016/j.arr.2023.102141 [DOI] [PubMed] [Google Scholar]
  • 210. Tarumi T, Gonzales MM, Fallow B, et al. Central artery stiffness, neuropsychological function, and cerebral perfusion in sedentary and endurance-trained middle-aged adults. J Hypertens. 2013;31(12):2400–2409. 10.1097/HJH.0b013e328364decc [DOI] [PubMed] [Google Scholar]
  • 211. McShea A, Harris P, Webster KR, Wahl AF, Smith MA.. Abnormal expression of the cell cycle regulators P16 and CDK4 in Alzheimer’s disease. Am J Pathol. 1997;150(6):1933. [PMC free article] [PubMed] [Google Scholar]
  • 212. Williamson K, Stringer S, Alexander M.. Endothelial progenitor cells enter the aging arena. Front Physiol. 2012;3:30. 10.3389/fphys.2012.00030 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 213. Mitsiou G, Tokmakidis SP, Dinas PC, Smilios I, Nanas S.. Endothelial progenitor cell mobilization based on exercise volume in patients with cardiovascular disease and healthy individuals: a systematic review and meta-analysis. Eur Heart J Open. 2022;2(6):oeac078. 10.1093/ehjopen/oeac078 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 214. Moore M, Hattori K, Heissig B, et al. Mobilization of endothelial and hematopoietic stem and progenitor cells by adenovector-mediated elevation of serum levels of SDF-1, VEGF, and angiopoietin-1. Ann N Y Acad Sci. 2001;938(1):36–47. 10.1111/j.1749-6632.2001.tb03572.x [DOI] [PubMed] [Google Scholar]
  • 215. Pías-Peleteiro J, Pérez-Mato M, López-Arias E, et al. Increased endothelial progenitor cell levels are associated with good outcome in intracerebral hemorrhage. Sci Rep. 2016;6(1):28724. 10.1038/srep28724 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 216. Sargento-Freitas J, Aday S, Nunes C, et al. Endothelial progenitor cells enhance blood–brain barrier permeability in subacute stroke. Neurology. 2018;90(2):e127–e134. 10.1212/WNL.0000000000004801 [DOI] [PubMed] [Google Scholar]
  • 217. Low A, Mak E, Rowe JB, Markus HS, O’Brien JT.. Inflammation and cerebral small vessel disease: a systematic review. Ageing Res Rev. 2019;53:100916. 10.1016/j.arr.2019.100916 [DOI] [PubMed] [Google Scholar]
  • 218. Hansen DV, Hanson JE, Sheng M.. Microglia in Alzheimer’s disease. J Cell Biol. 2018;217(2):459–472. 10.1083/jcb.201709069 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 219. Lewis J, Dickson DW.. Propagation of tau pathology: hypotheses, discoveries, and yet unresolved questions from experimental and human brain studies. Acta Neuropathol. 2015;131:27–48. 10.1007/s00401-015-1507-z [DOI] [PubMed] [Google Scholar]
  • 220. Olabarria M, Noristani HN, Verkhratsky A, Rodríguez JJ.. Concomitant astroglial atrophy and astrogliosis in a triple transgenic animal model of Alzheimer’s disease. Glia. 2010;58(7):831–838. 10.1002/glia.20967 [DOI] [PubMed] [Google Scholar]
  • 221. Cronin O, Keohane DM, Molloy MG, Shanahan F.. The effect of exercise interventions on inflammatory biomarkers in healthy, physically inactive subjects: a systematic review. QJM. 2017;110(10):629–637. 10.1093/qjmed/hcx091 [DOI] [PubMed] [Google Scholar]
  • 222. Gleeson M, Bishop NC, Stensel DJ, Lindley MR, Mastana SS, Nimmo MA.. The anti-inflammatory effects of exercise: mechanisms and implications for the prevention and treatment of disease. Nat Rev Immunol. 2011;11(9):607–615. 10.1038/nri3041 [DOI] [PubMed] [Google Scholar]
  • 223. Green DJ, Walsh JH, Maiorana A, Best MJ, Taylor RR, O’Driscoll JG.. Exercise-induced improvement in endothelial dysfunction is not mediated by changes in CV risk factors: pooled analysis of diverse patient populations . Am J Physiol Heart Circ Physiol. 2003;285(6):H2679–H2687. 10.1152/ajpheart.00519.2003 [DOI] [PubMed] [Google Scholar]
  • 224. Pedersen BK. Muscular interleukin-6 and its role as an energy sensor. Med Sci Sports Exerc. 2012;44(3):392–396. 10.1249/MSS.0b013e31822f94ac [DOI] [PubMed] [Google Scholar]
  • 225. Wang M, Zhang H, Liang J, Huang J, Chen N.. Exercise suppresses neuroinflammation for alleviating Alzheimer’s disease. J Neuroinflammation. 2023;20(1):76. 10.1186/s12974-023-02753-6 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 226. Bailey DM, Marley CJ, Brugniaux JV, et al. Elevated aerobic fitness sustained throughout the adult lifespan is associated with improved cerebral hemodynamics. Stroke. 2013;44(11):3235–3238. 10.1161/STROKEAHA.113.002589 [DOI] [PubMed] [Google Scholar]
  • 227. Kokkinos P, Faselis C, Samuel IBH, et al. Cardiorespiratory fitness and mortality risk across the spectra of age, race, and sex. J Am Coll Cardiol. 2022;80(6):598–609. 10.1016/j.jacc.2022.05.031 [DOI] [PubMed] [Google Scholar]
  • 228. Ashor AW, Lara J, Siervo M, et al. Exercise modalities and endothelial function: a systematic review and dose–response meta-analysis of randomized controlled trials. Sports Med. 2015;45:279–296. 10.1007/s40279-014-0272-9 [DOI] [PubMed] [Google Scholar]
  • 229. Fu TC, Wang CH, Lin PS, et al. Aerobic interval training improves oxygen uptake efficiency by enhancing cerebral and muscular hemodynamics in patients with heart failure. Int J Cardiol. 2013;167(1):41–50. 10.1016/j.ijcard.2011.11.086 [DOI] [PubMed] [Google Scholar]
  • 230. Pereira AC, Huddleston DE, Brickman AM, et al. An in vivo correlate of exercise-induced neurogenesis in the adult dentate gyrus. Proc Natl Acad Sci USA. 2007;104(13):5638–5643. 10.1073/pnas.0611721104 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 231. Weston ME, Koep JL, Lester AB, Barker AR, Bond B.. The acute effect of exercise intensity on peripheral and cerebral vascular function in healthy adults. J Appl Physiol (1985) 2022;133(2):461–470. 10.1152/japplphysiol.00772.2021 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 232. Bérubé B, Boidin M, Gayda M, et al. Acute effects of exercise on cerebrovascular response and cognitive performance in individuals with stable coronary heart disease. Brain Res. 2021;1772:147671. 10.1016/j.brainres.2021.147671 [DOI] [PubMed] [Google Scholar]
  • 233. Kolmos M, Krawcyk RS, Kruuse C.. Effect of high-intensity training on endothelial function in patients with cardiovascular and cerebrovascular disease: a systematic review. SAGE Open Med. 2016;4:2050312116682253. 10.1177/2050312116682253 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 234. Baker LD, Frank LL, Foster-Schubert K, et al. Aerobic exercise improves cognition for older adults with glucose intolerance, a risk factor for Alzheimer’s disease. J Alzheimers Dis. 2010;22(2):569–579. 10.3233/JAD-2010-100768 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 235. Becker L, Kutz DF, Voelcker-Rehage C.. Exercise-induced changes in basal ganglia volume and their relation to cognitive performance. J Neurol Neuromed. 2016;1(5):19–24. 10.29245/2572.942X/2016/5.1044 [DOI] [Google Scholar]
  • 236. Cassilhas RC, Lee K, Fernandes J, et al. Spatial memory is improved by aerobic and resistance exercise through divergent molecular mechanisms. Neuroscience. 2012;202:309–317. 10.1016/j.neuroscience.2011.11.029 [DOI] [PubMed] [Google Scholar]
  • 237. Heyn P, Abreu BC, Ottenbacher KJ.. The effects of exercise training on elderly persons with cognitive impairment and dementia: a meta-analysis. Arch Phys Med Rehabil. 2004;85(10):1694–1704. 10.1016/j.apmr.2004.03.019 [DOI] [PubMed] [Google Scholar]
  • 238. Kramer AF, Hahn S, Cohen NJ, et al. Ageing, fitness and neurocognitive function. Nature. 1999;400:418–419. 10.1038/22682 [DOI] [PubMed] [Google Scholar]
  • 239. Bherer L, Erickson KI, Liu-Ambrose T.. A review of the effects of physical activity and exercise on cognitive and brain functions in older adults. J Aging Res. 2013;2013:657508. 10.1155/2013/657508 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 240. Erickson KI, Banducci SE, Weinstein AM, et al. The brain-derived neurotrophic factor Val66Met polymorphism moderates an effect of physical activity on working memory performance. Psychol Sci. 2013;24(9):1770–1779. 10.1177/0956797613480367 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 241. Gates N, Singh MAF, Sachdev PS, Valenzuela M.. The effect of exercise training on cognitive function in older adults with mild cognitive impairment: a meta-analysis of randomized controlled trials. Am J Geriatr Psychiatry. 2013;21(11):1086–1097. 10.1016/j.jagp.2013.02.018 [DOI] [PubMed] [Google Scholar]
  • 242. Lenze EJ, Voegtle M, Miller JP, et al. Effects of mindfulness training and exercise on cognitive function in older adults: a randomized clinical trial. JAMA. 2022;328(22):2218–2229. 10.1001/jama.2022.21680 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 243. Li H, Su W, Dang H, et al. Exercise training for mild cognitive impairment adults older than 60: a systematic review and meta-analysis. J Alzheimers Dis. 2022;88(Preprint):1263–1278. 10.3233/jad-220243 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 244. Barclay C. Energy demand and supply in human skeletal muscle. J Muscle Res Cell Motil. 2017;38(2):143–155. 10.1007/s10974-017-9467-7 [DOI] [PubMed] [Google Scholar]
  • 245. Kemp GJ, Meyerspeer M, Moser E.. Absolute quantification of phosphorus metabolite concentrations in human muscle in vivo by 31P MRS: a quantitative review . NMR Biomed. 2007;20(6):555–565. 10.1002/nbm.1192 [DOI] [PubMed] [Google Scholar]
  • 246. Rebelo-Marques A, De Sousa Lages A, Andrade R, et al. Aging hallmarks: the benefits of physical exercise. Front Endocrinol. 2018;9:258. 10.3389/fendo.2018.00258 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 247. DuBose LE, Weng TB, Pierce GL, et al. Association between cardiorespiratory fitness and cerebrovascular reactivity to a breath-hold stimulus in older adults: influence of aerobic exercise training. J Appl Physiol (1985) 2022;132(6):1468–1479. 10.1152/japplphysiol.00599.2021 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 248. Willie CK, Tzeng YC, Fisher JA, Ainslie PN.. Integrative regulation of human brain blood flow. J Physiol. 2014;592(5):841–859. 10.1113/jphysiol.2013.268953 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 249. Calverley TA, Ogoh S, Marley CJ, et al. HIITing the brain with exercise; mechanisms, consequences and practical recommendations. J Physiol. 2020;598:2513–2530. 10.1113/jp275021 [DOI] [PubMed] [Google Scholar]
  • 250. Wisløff U, Støylen A, Loennechen JP, et al. Superior cardiovascular effect of aerobic interval training versus moderate continuous training in heart failure patients: a randomized study. Circulation. 2007;115(24):3086–3094. 10.1161/CIRCULATIONAHA.106.675041 [DOI] [PubMed] [Google Scholar]
  • 251. Ramos JS, Dalleck LC, Tjonna AE, Beetham KS, Coombes JS.. The impact of high-intensity interval training versus moderate-intensity continuous training on vascular function: a systematic review and meta-analysis. Sports Med. 2015;45(5):679–692. 10.1007/s40279-015-0321-z [DOI] [PubMed] [Google Scholar]
  • 252. Weston KS, Wisloff U, Coombes JS.. High-intensity interval training in patients with lifestyle-induced cardiometabolic disease: a systematic review and meta-analysis. Br J Sports Med. 2014;48(16):1227–1234. 10.1136/bjsports-2013-092576 [DOI] [PubMed] [Google Scholar]
  • 253. Ross LM, Porter RR, Durstine JL.. High-intensity interval training (HIIT) for patients with chronic diseases. J Sport Health Sci. 2016;5(2):139–144. 10.1016/j.jshs.2016.04.005 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 254. Querido JS, Sheel AW.. Regulation of cerebral blood flow during exercise. Sports Med. 2007;37:765–782. 10.2165/00007256-200737090-00002 [DOI] [PubMed] [Google Scholar]
  • 255. Becker HM, Bröer S, Deitmer JW.. Facilitated lactate transport by MCT1 when coexpressed with the sodium bicarbonate cotransporter (NBC) in Xenopus oocytes. Biophys J. 2004;86(1):235–247. 10.1016/S0006-3495(04)74099-0 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 256. Ide K, Horn A, Secher NH.. Cerebral metabolic response to submaximal exercise. J Appl Physiol (1985) 1999;87(5):1604–1608. 10.1152/jappl.1999.87.5.1604 [DOI] [PubMed] [Google Scholar]
  • 257. Rasmussen P, Madsen CA, Nielsen HB, et al. Coupling between the blood lactate-to-pyruvate ratio and MCA V mean at the onset of exercise in humans. J Appl Physiol. 2009;107(6):1799–1805. 10.1152/japplphysiol.00468.2009 [DOI] [PubMed] [Google Scholar]
  • 258. Rasmussen P, Plomgaard P, Krogh-Madsen R, et al. MCA V mean and the arterial lactate-to-pyruvate ratio correlate during rhythmic handgrip. J Appl Physiol. 2006;101(5):1406–1411. 10.1152/japplphysiol.00423.2006 [DOI] [PubMed] [Google Scholar]
  • 259. Ball KK, Cruz NF, Mrak RE, Dienel GA.. Trafficking of glucose, lactate, and amyloid-β from the inferior colliculus through perivascular routes. J Cereb Blood Flow Metab. 2010;30(1):162–176. 10.1038/jcbfm.2009.206 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 260. Lundgaard I, Lu ML, Yang E, et al. Glymphatic clearance controls state-dependent changes in brain lactate concentration. J Cereb Blood Flow Metab. 2017;37(6):2112–2124. 10.1177/0271678X16661202 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 261. Erickson KI, Grove GA, Burns JM, et al. Investigating gains in neurocognition in an intervention trial of exercise (IGNITE): protocol. Contemp Clin Trials. 2019;85:105832. 10.1016/j.cct.2019.105832 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 262. Broatch JR, Zarekookandeh N, Glarin R, et al. Protocol: Train Smart Study: protocol for a randomised trial investigating the role of exercise training dose on markers of brain health in sedentary middle-aged adults. BMJ Open. 2023;13(5):e069413. 10.1136/bmjopen-2022-069413 [DOI] [PMC free article] [PubMed] [Google Scholar]

Articles from The Journals of Gerontology Series A: Biological Sciences and Medical Sciences are provided here courtesy of Oxford University Press

RESOURCES