Abstract
Over two-thirds of individuals aged 65 and older are obese or overweight in the United States. Epidemiological data show an association between the degree of adiposity and cognitive dysfunction in the elderly. In this review, the pathophysiological roles of microvascular mechanisms, including impaired endothelial function and neurovascular coupling responses, microvascular rarefaction, and blood-brain barrier disruption in the genesis of cognitive impairment in geriatric obesity are considered. The potential contribution of adipose-derived factors and fundamental cellular and molecular mechanisms of senescence to exacerbated obesity-induced cerebromicrovascular impairment and cognitive decline in aging are discussed.
Keywords: aging, endothelial dysfunction, metabolic syndrome, neurovascular coupling, senescence
INTRODUCTION
Currently, more than 35% of individuals aged 65 and older are obese (over 55% of black women) and if the current trend continues, nearly half of the elderly population in the United States will be obese by 2030 (1). In this age-group, the prevalence of overweight is 78.4% for men and 68.6% for women (2). There is increasing evidence that obesity has deleterious effects on the brain and cognitive function (3–8; Fig. 1). Importantly, several epidemiological studies, including the Framingham Heart Study; the Health, Aging and Body Composition (ABC) study; the Swedish Adoption/Twin Study of Aging; and Baltimore Longitudinal Study on Aging, suggest that aging and obesity exert synergistic negative effects on cognition (9–17). Furthermore, the Whitehall II Study also shows that early midlife obesity is associated with lower executive function and lower Mini Mental State Examination (MMSE) scores and impaired memory, ability, and executive function later in life (18). In the past decade, significant progress has been made in this research field, and many new concepts have emerged that shed light on the cellular and molecular mechanism underlying obesity-induced cognitive impairment in the elderly. The current view is that obesity both promotes the development of vascular cognitive impairment (VCI) (19) [the most important form of Alzheimer’s disease-related dementia (ADRD)] and also increases the incidence of Alzheimer’s disease (AD) (20).
Figure 1.
Obesity in aging promotes cognitive impairment and dementia. A: prevalence of dementia by BMI status, across age categories. Note that obesity in aging is associated with a significant increase in the prevalence of dementia. Figure is reprinted with permission from reference (8). B: obesity is associated with impaired cognitive performance [lower Rapid Visual Information Processing (RVIP) accuracy score] in older participants of the Oklahoma Longitudinal Study on Aging (>60 yr old). The RVIP task [Cambridge Neuropsychological Test Automated Battery (CANTAB) battery of tests] is a sensitive serial discrimination task where task performance reflects visual sustained attention (vigilance) and working memory capabilities. fMRI studies show that frontal, parietal, and cerebellar regions are activated during the task. Older individuals exhibit a decreased performance on the RVIP task (7), which is further exacerbated by obesity. Data are replotted from reference (44). *Significant difference between the two groups. BMI, body mass index; fMRI, functional magnetic resonance imaging.
There is increasing evidence that both aging and obesity cause structural and functional impairment in the cerebral microcirculation, which plays a crucial role in the pathogenesis of both VCI and AD. In this review, potential microvascular contributions to cognitive impairment associated with obesity in the elderly are discussed. Obesity-related alterations in three main regulatory paradigms involved in the regulation of cerebral blood flow (CBF): cerebral autoregulation, endothelium-mediated vasodilation, and neurovascular coupling responses responsible for functional hyperemia. Pathophysiological consequences of cerebromicrovascular dysregulation in obesity are explored, including blood-brain barrier (BBB) disruption, neuroinflammation, exacerbation of neurodegeneration, microvascular rarefaction, and ischemic neuronal dysfunction and damage. In addition, potential obesity-related mechanisms such as adipose tissue dysfunction, hyperinsulinemia, and altered gut-brain axis, which may be causally linked to microvascular dysfunction, are considered. Finally, the evidence for the causal role of cellular senescence in exacerbation of the deleterious effect of obesity on cerebrovascular function and cognition in aging is critically examined. Understanding the cellular mechanisms behind the synergistic interaction of aging and obesity on cognitive decline is important to develop effective interventions for prevention.
LINKS AMONG AGING, OBESITY, AND COGNITIVE DECLINE
Epidemiological Studies
Several large-scale longitudinal and cross-sectional studies have contributed to our understanding on the negative interaction of aging and obesity on cognitive impairment (21). In the Health Aging and Body Composition Study (Health ABC study), more than 3,000 participants between the ages of 70 and 79 yr were followed up for 8 yr, and the associations between baseline measures of overall and regional adiposity and change in cognitive function over time were examined. The results showed that higher measures of radiographically measured total fat mass and subcutaneous fat were associated with worsening cognitive function after 7 yr (16). In the Framingham Heart study with participants of mean age around 66 yr, the obese individuals demonstrated lower cognitive performance after controlling for other risk factor such as hypertension (12). The Baltimore Longitudinal Study on Aging (BLSA) conducted in more than 1,700 participants with a mean age of 55 yr also reported that obesity indices (larger waist circumference and waist-hip ratio) were associated with poorer performance on cognitive tests over time (13). Similarly, the Neurological Diseases in Central Spain (NEDICES), a population-based cross-sectional study with ∼2,000 elderly subjects aged 65 yr or older showed that obese or overweight status was associated with the lowest quartiles of global cognitive functions (22). Studies conducted as part of the Women’s Health Initiative (WHI) in elderly postmenopausal women also reported similar findings (23), suggesting that there are no gender differences in the observed negative interaction of aging and obesity on cognition. In addition, aged individuals with comorbidities associated with obesity such as hypertension, diabetes, hypercholesterolemia, or sedentary life style showed greater decline in memory, dexterity, and executive functions (17, 24–26). In particular, in older adults with central obesity, even modest degrees of hyperglycemia were shown to exacerbate cognitive decline (27). In older patients with heart failure, cerebral hypoperfusion due to a decreased cardiac output and microvascular consequences of obesity interact to adversely influence cognitive function (28). Similar negative interaction has also been reported for patients with obstructive sleep apnea where obesity reduced the capacity for working memory relative to nonobese patients with sleep apnea (29).
It should be noted that although in most clinical studies, a strong association between obesity and cognitive decline is evident in midlife, in late life, there are important confounding factors, which may affect this association. In fact, there are few studies that appear to suggest that obese older individuals may have certain health benefits (30, 31). Several theories have been put forward to explain this “obesity paradox” (32). It is possible that the obesity paradox represents an artifact arising from biases in observational studies (e.g., inadequate adjustment for smoking, which causes weight loss and significantly increases risk for vascular diseases). Another important concern is reverse causation due to illness-induced weight loss. These potential hypotheses were further explored in the British Whitehall II Study where obesity at age 50 was a strong predictor of dementia but not at ages 60 or 70. Furthermore, incident dementia cases had higher body mass indices (BMIs) up to 16 yr before diagnosis but lower BMIs from 8 yr before diagnosis (33). Evidence from longitudinal preclinical studies on aged mice fed a high-fat diet support this concept, suggesting that weight loss due to chronic disease (e.g., cancer) predicts a significant decline in performance in behavioral studies. It is also possible that an inherent selection bias in large-scale clinical studies where the unhealthiest obese patients are naturally excluded by early mortality may also contribute to the obesity paradox (34). Further, analyses based on BMI measurements alone might be inaccurate, as it neglects lean and fat tissue distribution. Central adiposity assessed by waist-to-hip ratio or waist circumference combined with measurements of body composition may be more consistent when determining the effects of obesity on cognition. To overcome the inherent limitations of clinical studies and to provide mechanical insight into the pathogenesis of cognitive decline associated with geriatric obesity, several well-controlled preclinical studies were conducted on lean and obese animal models of aging. These studies provide strong support for the concept that aging exacerbates the deleterious effects of obesity on cognition (see Preclinical Studies).
Preclinical Studies
The deleterious effects of obesity on cognition and cerebral health have been well documented in rodent models (35–39). For example, feeding a high-fat diet (HFD) for 4 to 6 mo to mice results in impaired performance in the T-maze test (40), the Morris water maze test (41), and other behavioral tasks (35–38). There are a number of extant studies that have investigated the interaction of aging and obesity on cognitive decline (37–39). Using mouse models with HFD-induced obesity, several studies have demonstrated that advanced aging and diet-induced obesity exert synergistic deleterious effects on cognitive function and cerebral health (35–37), extending the clinical observations. It is a strength of these studies that similar level of obesity can be induced both in young and aged mice using an identical chronic HFD feeding paradigm. Thus, it is possible to assess the influence of aging per se, independent of the duration or severity of obesity. Using this approach, it was demonstrated that aging exacerbates HFD-induced decline in learning and memory function in mice (36) assessed in the elevated plus maze and Y-maze tests (38). Further, midlife obesity was also associated with compromised visual recognition memory in novel object recognition test in mice (42). Interestingly, there are data suggesting that females may be more at risk for midlife obesity-induced vascular cognitive impairment and dementia (VCID) than males. A recent study reported that feeding a HFD to middle-aged female mice results in greater weight gain and glucose intolerance than in males and that greater visceral fat mass gain and increased systemic TNF-α levels in females correlated with more pronounced spatial memory deficits in females as compared with males (43).
MICROVASCULAR MECHANISMS CONTRIBUTING TO COGNITIVE IMPAIRMENT
The high metabolic demands of the brain are met by a dense microcirculatory network that is estimated to span ∼600 km in total length in humans. The cerebral microcirculation ensures appropriate distribution of oxygen, glucose, and other nutrients to the neural tissue, and it is also responsible for washout of metabolic by-products, maintenance of the ionic milieu, formation of the blood-brain barrier (BBB), and regulation of transport of various substances across it. Thus, microvascular health plays a critical role in the maintenance of normal neuronal and cognitive function (44–58). Cerebromicrovascular dysfunction and microvascular damage has been increasingly recognized as key contributors to age- and obesity-associated cognitive impairment. Clinical studies show that obesity promotes dysregulation of cerebral blood flow (Figs. 2 and 3), which directly relates to cognitive decline (28, 59–64). Experimental studies extend the clinical findings and provide mechanistic insight into the synergistic effects of obesity and aging on cerebromicrovascular function. Here, we provide an overview of the specific pathogenic roles of endothelial dysfunction, neurovascular impairment, microvascular rarefaction, and blood-brain barrier disruption in the pathogenesis of VCI associated with geriatric obesity (Fig. 4).
Figure 2.
Cerebral blood flow is decreased in obese subjects. A shows the relationship between body mass index (BMI) and age-adjusted mean baseline blood flow velocities (BFV) in right and left middle cerebral artery (□MCAR, ■MCAL). B shows that mean BFV in MCAR (P = 0.017) and MCAL (P = 0.0002) are higher for normal weight (BMI < 25 kg/m2) than overweight (BMI 25–30 kg/m2) and obese subjects (BMI > 30 kg/m2). C and D show the average cerebrovascular resistance (CVR in □MCAR and ■MCAL during baseline and head-up tilt (mean ± SE). The figures are reprinted with permission from reference (63).
Figure 3.
Obesity and the metabolic syndrome impair CBF. A: CBF is decreased proportional to the number of metabolic syndrome factors (including abdominal obesity, triglycerides, HDL-cholesterol, blood pressure, and fasting glucose) present in an individual. Lower CBF was reported to most robustly associate with abdominal obesity, and only to a lesser extent with triglycerides and fasting glucose (59). B: participants with metabolic syndrome and obesity show significantly lower CBF in large portions of the cortical surface of the frontal and parietal lobes, and the lateral and superior portions of the temporal and occipital lobes (yellow: voxel-wise results at P < 0.05, FEW corrected, controlling for age, sex, and reference cluster. Resting CBF assessments were made using background-suppressed pseudocontinuous arterial spin labeled (pcASL) MRI. The figures are reprinted with permission from reference (59). CBF, cerebral blood flow.
Figure 4.
Proposed scheme for cerebromicrovascular contributions to obesity-induced cognitive decline in older adults. Excessive accumulation of fat in obesity is associated with adipose tissue dysfunction and low grade inflammation, which results in altered secretion of adipokines and proinflammatory cytokines. These circulating factors mediate the crosstalk between adipose tissue and the brain by impairing the cerebral microcirculation. In aging heightened inflammatory status of the adipose tissue promotes increased systemic inflammation, which—together with age-related impairment of cellular stress resilience pathways—play a key role in the increased vulnerability of obese elderly patients for cognitive impairment. Functional and structural impairment of the cerebral microcirculation results in endothelial dysfunction, neurovascular dysfunction, and microvascular rarefaction, all of which contribute to a significant decline in cerebral blood flow. Microvascular inflammation and disruption of the blood-brain barrier exacerbate neuroinflammation. Obesity is also associated with dysbiosis. Age-related breakdown of the intestinal barrier promotes the leakage of bacterial breakdown products to the circulation, exacerbating microvascular inflammation and blood-brain barrier dysfunction (PAMPs: pathogen-associated molecular patterns). The resulting ischemic and inflammatory foci play a role in the pathogenesis of cognitive impairment. The model predicts that the aforementioned obesity-related structural and functional cerebromicrovascular alterations synergize to promote cognitive impairment in high-risk older adults.
Endothelial Dysfunction and Neurovascular Uncoupling
Microvascular endothelial cells play a critical role in CBF regulation through the production of a variety of vasoactive mediators including the gasotransmitter nitric oxide (NO) (65). Endothelium-dependent, NO-mediated microvascular dilation contributes to the maintenance of resting CBF, as studies show that acute blockade of NO synthase decreases CBF and results in cerebral hypoperfusion (65, 66). Aging and obesity-associated endothelial dysfunction, characterized by decreased NO bioavailability, has been shown to cause cerebral hypoperfusion leading to cognitive decline (67, 68). In addition to NO, endothelial cells also produce other vasoactive mediators including endothelin-1 as well as vasoactive arachidonic acid metabolites including prostacyclin, 20-HETE, and thromboxanes. Age-related impairment in endothelial NO production may also affect prostacyclin-mediated vasodilatory responses in older humans in the peripheral circulation (69). Further, obesity is also associated with diminished synthesis of prostaglandin I2 (PGI2), which contributes to impaired peripheral vasodilatory responses in rodent models (70). There is initial preclinical evidence that interaction of obesity and aging also alters synthesis of vasoactive arachidonic acid metabolites in the brain (39).
One of the important mechanisms that contribute to endothelial dysfunction in aging and obesity is oxidative stress (38, 58, 65, 71–77). Both aging and obesity are associated with increased production of mitochondrial superoxide production mediated in part by increased expression of NADPH oxidases in the brain vasculature and also in the other organs (78–82). Importantly, obesity and aging have synergistic effects on endothelial oxidative stress and upregulation of NADPH oxidase expression (38). Increased levels of superoxide derived from NADPH oxidases and mitochondrial sources react with endothelium-derived NO to form peroxynitrite, thus decreasing the bioavailability of NO in aging and obesity (78, 83, 84).
In addition to increased obesity-related free radical production, decreased antioxidant defense mechanisms also contribute to increased oxidative stress in aging (77, 85–88). Nuclear factor-erythroid 2-related factor 2 (Nrf2) is an evolutionarily conserved transcription factor that regulates the expression of antioxidative and anti-inflammatory genes in the vasculature (77). Previous studies demonstrated that aging is associated with impaired Nrf2 signaling in the vasculature, which in turn increases the sensitivity to oxidative stress-induced vascular damage(87). Accordingly, Nrf2-deficient mice exhibit increased HFD/obesity-related vascular oxidative stress, which exacerbates endothelial dysfunction (86, 89, 90).
Emerging evidence suggests a crucial role for endothelial NO production in neurovascular coupling responses (NVC) (74–76, 86, 91–95). NVC (“functional hyperemia”) is a vital homeostatic mechanism involved in moment-to-moment adjustment of regional blood flow to the energetic demands of neurons during periods of intense neuronal activity (73, 96, 97; Fig. 5). Functional hyperemia not only ensures adequate supply of oxygen and glucose to astrocytes and neurons but also effectively clears the metabolic by-products of neuronal activity. NVC depends on an orchestrated interplay between neurons, astrocytes, endothelial cells, and smooth muscle cells culminating in coupling of increased blood flow to neuronal activity (73). Pharmacological inhibition of NVC significantly impairs learning and memory in mice, highlighting the importance of normal NVC in the maintenance of cognitive functions (94). It is significant that obesity results in neurovascular uncoupling (Fig. 5), which effect is exacerbated in aging, promoting cognitive decline (38, 98). Importantly, treatment with apocynin, a NADPH oxidase inhibitor, improves endothelium-dependent NVC in aged obese mice, suggesting a critical role for increased oxidative stress in neurovascular dysfunction (38). Further evidence for this concept is provided by studies demonstrating that Nrf2 dysfunction also exacerbates obesity-induced neurovascular uncoupling and cognitive impairment, mimicking the aging phenotype (86). In addition to Nrf2, previous studies also provide evidence that insulin-like growth factor-1 (IGF-1)-mediated pathways exert multifaceted cerebromicrovascular protective effects, which act to preserve endothelial vasodilation and NVC (47, 76, 99–104). Aging results in decreased levels of circulating IGF-1 (102, 105–107). Mouse models of genetic IGF-1 deficiency were shown to exhibit accelerated neurovascular aging phenotype, characterized by neurovascular uncoupling, impaired endothelial NO production, and cognitive impairment (76). IGF-1 receptors are abundantly expressed in different cells of the neurovascular unit including endothelial cells, astrocytes, and smooth muscle cells. There is now evidence that cell-type specific depletion of IGF-1 receptors in endothelial cells mimic several aspects of age-related neurovascular uncoupling (Tarantini, Csiszar and Ungvari 2020, manuscript in preparation). Importantly, previous studies also show that genetic IGF-1 deficiency also exacerbates obesity-induced endothelial dysfunction in Lewis dwarf rats (108), mimicking the aging phenotype.
Figure 5.
Obesity impairs neurovascular coupling responses. A: obesity impairs neurovascular coupling in mice. Representative pseudocolour laser speckle flowmetry maps of baseline CBF (top) and CBF changes in the whisker barrel field relative to baseline during contralateral whisker stimulation (bottom, right oval, 30 s, 5 Hz) in standard diet-fed lean and high-fat diet-fed obese mice. Color bar represents CBF as percent change from baseline. B shows the time-course of CBF changes after the start of contralateral whisker stimulation (horizontal bars). Summary data are shown in C. Data are mean ± S.E. (n = 6–8 in each group), *P < 0.05 vs. lean control; #P < 0.05 vs. untreated (one-way ANOVA with post hoc Tukey’s tests). D and E: obesity impairs neurovascular coupling in older humans. Neurovascular coupling responses were assessed by functional near-infrared spectroscopy (fNIRS) during a finger-tapping task in normal weight (BMI 18–25, n = 10) and obese (BMI > 30, n = 10) older adults (>65 years of age). Data were analyzed using the Brain AnalyzIR toolbox (97) based on a general linear model (GLM) approach. Task-related changes in oxygenated hemoglobin (HbO) concentration [calculated using the Beer–Lambert law (96)] was used as an index of functional hyperemia. The design matrix included boxcar regressors for each stimulation, and a canonical hemodynamic response function was used to identify activated cortical regions. β-Weights, scaling the predictors, were then used for group-level statistics, where a t contrast of [BMI 18–25] – [BMI >30] was applied (*P < 0.05). In D solid lines represent statistically significant difference between groups in task-evoked neurovascular coupling responses in the area and vicinity of the left primary motor cortex, evidenced by the increased HbO concentration observed in the normal weight older adult group when compared with their obese counterparts. Bar graphs (E) represent calculated changes in HbO. Note that neurovascular responses, that show an age-related decline even in older adults, are inverted in obese older adults. Position of fNIRS light sources (s14 and s15) and light detectors (d13, d15, and d16) are shown in D. Data are replotted from previously published studies (45, 86). BMI, body mass index; CBF, cerebral blood flow.
Microvascular Rarefaction
Microvascular rarefaction, manifested by a decline in capillary density, contributes to cognitive impairment through a decline in CBF, reducing metabolic support for neurons (65, 109). Previous studies demonstrate that obesity results in decreased capillary density in the cortex and hippocampus, and this effect is exacerbated in aging (38, 109–111). Importantly, the extent of obesity-induced capillary rarefaction in the hippocampus is directly correlated to the extent of cognitive impairment (38), providing additional evidence for the close association between dysregulation of CBF and neuronal dysfunction. It is also possible that comorbidities associated with obesity, such as hypertension, play also a pathogenic role in worsening capillary rarefaction observed with aging (102). The mechanisms underlying cerebromicrovascular rarefaction in aging and obesity may include impaired endothelial NO bioavailability (109, 112–114), loss of pericytes (38), increased endothelial apoptosis (115, 116), decreased levels of proangiogenic factors [e.g., VEGF (117), IGF-1 (102, 105–107, 118)], and impaired endothelial angiogenic processes (38, 102, 119–123). Overexpression of VEGF in vivo in the aged rodent brain or in vitro VEGF treatment of cultured primary microvascular endothelial cells derived from aged rats results in impaired angiogenic responses, consistent with the concept that aging results in endothelial resistance to angiogenic stimuli (121). Aging-induced impairment of endothelial angiogenic processes and resistance to family-wise error (FWE). VEGF have been attributed to decreased expression of VEGF receptors (124), dysregulation of angiogenic miRNA expression (122), impaired sirtuin 1 (SIRT1) activation (119, 125), and impaired Nrf2 signaling (123). Further studies are warranted to determine how diet-induced obesity impacts these synergistic mechanisms in the cerebral microcirculation.
Blood-Brain Barrier Damage and Neuroinflammation
Blood-brain barrier (BBB) is a specialized structure formed by endothelial cells of cerebral microvessels, pericyte, astrocyte end-feet, and basal membrane in the central nervous system. This heavily restricted barrier maintains CNS homeostasis by facilitating transport of essential nutrient molecules, regulating ion balance and preventing the influx of serum-derived factors into the brain parenchyma (55,56, 126). The integrity of BBB is critical for the maintenance of proper neuronal function (127). BBB leakage or increased permeability is commonly associated with cognitive impairment under various pathological conditions including but not limited to AD, diabetes, stroke, and traumatic brain injury (55, 56, 126, 128). In fact, a recent study reported increased BBB permeability as an early biomarker for cognitive dysfunction in humans independent of the presence of AD-related biomarkers like Aβ and/or tau in the hippocampus (129).
Both aging and obesity promote BBB disruption (128), and our studies demonstrate that their effects are synergistic (37, 39, 86). The mechanisms underlying exacerbated obesity-induced BBB damage in aging are likely multifaceted. First, alterations in the expression of tight junction and adherens junction proteins including occludin, claudins, and cadherins might impair BBB integrity (38). Additionally, both aging and obesity are likely to result in posttranslational modifications, including phosphorylation, palmitoylation, glycosylation, acetylation, and methylation of tight junction proteins, which may affect their stability and proper cellular localization (130). Pericytes are also critical structural component of BBB and pericyte-deficient Pdgfrβ−/− mice have increased BBB permeability (131). In that regard, it is significant that aged obese mice have less pericyte coverage in the cerebral microvessels than younger ones (37). Lastly, cells forming the BBB have a high metabolic rate, consistent with the high energy demands for active ATP-dependent transporters. Proteomic analysis from freshly isolated cerebral microvessels indicates that several proteins important for cellular energy metabolism are downregulated in diet-induced obesity (132), suggesting that impaired energy metabolism in the endothelial cells could also potentially contribute to BBB disruption. There is strong evidence that age-related decline in cellular NAD+ levels and uncoupling of the mitochondrial electron transport chain contribute importantly to impaired energy metabolism of cerebromicrovascular endothelial cells (51–53, 57, 71, 74, 75). Although the precise mechanisms that contribute to the hypometabolic state of microvascular endothelial cells observed in obesity are not known, decreases in circulating levels of adiponectin (high molecular weight form), a hormone known to stimulate energy metabolism through AMPK pathway, could potentially play a role (133, 134).
One of the major consequences of BBB breakdown is leakage of plasma constituents including IgG, thrombin, and fibrinogen into the brain parenchyma (37). Increased infiltration of plasma proteins through the BBB promotes neuroinflammation mediated through activation of resident immune cells, especially microglia (37). For example, interaction of IgG with Fc gamma receptors (FcγR) results in microglia activation (135), leading to secretion of proinflammatory cytokines, chemokines, and reactive oxygen species. There is evidence demonstrating synergistic interaction of aging and HFD-induced obesity to exacerbate leakage of IgG and promote microglia activation in the mouse hippocampus (37, 39). Activated microglia may also cause further BBB damage, thus driving a vicious cycle of neuroinflammation (136). Chronic unresolved inflammation in obesity adversely affects neuronal function related to cognition (137–141). Increased presence of activated microglia in the hippocampi of obese aged mice is associated with exacerbated impairment of long-term potentiation (LTP) of excitatory synaptic transmission, an important cellular correlate for learning and memory (39). It is significant that Nrf2-deficient mice exhibit exacerbated HFD/obesity-related BBB disruption, neuroinflammation, and LTP impairment in the hippocampi, mimicking the aging phenotype (86).
OBESITY-RELATED FACTORS THAT CONTRIBUTE TO CEREBROMICROVASCULAR IMPAIRMENT
The cellular mechanisms underlying the increased susceptibility of the elderly to obesity-induced cerebromicrovascular impairment and cognitive decline are likely multifaceted. Here, we discuss the potential role of adipose tissue inflammation, altered adipokine secretion, insulin resistance, and alterations of the gut-brain axis.
Adipose Tissue Dysfunction
Once considered an inert fat storage organ, adipose tissue is now recognized as an active endocrine organ that secretes a variety of adipokines, which can act both at peripheral and central sites. Excessive accumulation of fat in obesity is associated with adipose tissue dysfunction. This results in dysregulated secretion of adipokines including proinflammatory cytokines and chemokines, rendering the adipose tissue as a major contributor to systemic inflammation. Emerging studies suggest that the crosstalk between adipose tissue and the brain plays a key role in the increased vulnerability of obese elderly patients for cognitive impairment. In this section, we discuss potential adipose tissue-related mechanisms that can affect cerebral microcirculation and cognition.
Heightened inflammatory status of the adipose tissue promotes systemic inflammation.
Obesity is associated with low-grade inflammation within the adipose tissue (including increased infiltration and activation of macrophages, proinflammatory changes in the cellular secretome), which results in elevated levels of circulating proinflammatory mediators (142–146). Based on the observations from clinical studies investigating the effects of weight loss strategies on systemic inflammation (147, 148), it can be inferred that adipose tissue dysfunction and its heightened inflammatory status contribute significantly to systemic inflammation in obesity. In particular, inflammatory cytokines and neuroinflammation (36, 37, 39, 86, 137, 149–163) have an important role in impaired neuronal function and the pathogenesis of both VCI and AD (164–169).
Adipose tissue is capable of handling excess energy intake by expansion of existing adipocytes (hypertrophy) and also through adipogenesis where the progenitor cells proliferate and differentiate to generate new adipocytes (hyperplasia). Inadequate expansion of adipocytes results in hypertrophied adipocytes, which tilts the secretory profile of adipocytes favoring inflammation (170). With long-term obesity, this is followed by infiltration of immune cells in the adipose tissue, most notably macrophages, CD8+ T cells, mast cells, and B cells. Obesity is also known to alter the polarization of adipose tissue macrophages from anti-inflammatory M2 to proinflammatory M1 phenotype, leading to persistent unresolved inflammation (171). Activated macrophages and inflamed adipocytes secrete a variety of cytokines and chemokines such as IL-6 and TNF-α, which enter the circulation and lead to systemic inflammation. Additionally, toll-like receptors (e.g., TLR4) are abundantly expressed both on adipocytes and macrophages. When stimulated by circulating bacterial breakdown products (see Altered Gut-Brain Axis (Dysbiosis) in these cells, multiple inflammatory signal transduction cascades are activated, promoting the secretion of a range of inflammatory cytokines and acute-phase proteins. There is strong evidence that aging exacerbates obesity-induced inflammation in the adipose tissue (37–39, 172–174), which contributes to the development of several secondary diseases such as the metabolic syndrome, insulin resistance, type 2 diabetes mellitus, and hypertension. The heightened inflammatory status of the adipose tissue and the consequential increases in circulating cytokines are also thought to play a critical role in exacerbation of VCI and AD in older obese individuals.
Studies have shown a causal link between systemic inflammation and cognitive impairment (175). Circulating inflammatory mediators can affect cerebromicrovascular function and cognition through several mechanisms. First, they promote microvascular oxidative stress and endothelial dysfunction, induce endothelial activation, and impair cellular energy metabolism. Further, circulating cytokines have also been demonstrated to disrupt BBB function by modifying tight junction structures (176), inducing endothelial apoptosis (177) and glycocalyx degradation on the apical endothelium (178). Cytokines like IL6, TNFα, IL-1β, and IL-1α can selectively cross BBB using active transport systems (179–181) and activate resident glial cells to foster neuroinflammation and cognitive decline.
Altered adipokine secretion.
In addition to cytokines, dysregulation in the secretion and signaling of other adipokines (leptin, adiponectin, and resistin) has also been implicated in the pathogenesis of neurovascular diseases (182).
Leptin is a peptide hormone secreted in proportion to white adipose tissue mass. Originally the effect of leptin was only considered in the hypothalamus where it is involved in the regulation of central control of food intake and energy homeostasis. However, identification of the leptin receptor (LepR) on endothelial cells and LepR-mediated transport mechanisms at the BBB (183, 184) suggests that leptin can also affect the microcirculation and thereby potentially modulate microvascular contributions to cognitive decline. However, the vascular (and cognitive) effects of leptin signaling are likely complex. On endothelial cells, leptin has been shown to upregulate endothelin-1, as well as to stimulate the expression of adhesion molecules and induce oxidative stress (185). There are also studies showing that leptin induces hypertension and/or endothelial dysfunction (186–190). Leptin-deficient and whole-body leptin receptor-deficient mice are protected from neointimal hyperplasia in response to arterial wall injury (191). Clinical studies show that high leptin levels predict acute cardiovascular events, coronary restenosis, and stroke (191). Yet, LepR deficiency causes cognitive impairment in Zucker rats and db/db mice (192), and endothelial-specific LepR deficiency was reported to associate with poor vascular outcomes (193). Studies show that leptin responsiveness decreases with aging and obesity, which may be related to defective leptin transport across BBB, downregulation of LepRs, and/or impaired leptin signaling downstream of LepRs (194, 195). Leptin resistance is associated with high circulating levels of leptin both in aging and obesity (196). Studies investigating the direct effects of leptin resistance on the cerebral microvessels are warranted.
Resistin is a proinflammatory adipokine, which promotes insulin resistance (197) and atherosclerosis (182, 198, 199). Elevated resistin level is associated with an increased risk of ischemic stroke (199–204). Resistin was shown to increase permeability in a cell culture-based blood-brain barrier model (205). Resistin has also been causally linked to endothelial dysfunction (206, 207) Yet, its role in dysregulation of CBF and NVC responses, BBB disruption, and cognitive decline (208) remains elusive.
Adiponectin is an adipokine produced primarily in adipose tissue, which circulates at high concentrations and modulates metabolic processes, including glucose regulation and fatty acid oxidation, and confers potent anti-inflammatory effects (209–213). It acts as an insulin-sensitizing hormone in muscle and liver (209). Through these actions, it ameliorates diabetes and prolongs life span in mouse models of type 2 diabetes (e.g., db/db mice on high-fat diet) (214). Adiponectin activates the AMPK (AMP-activated protein kinase)–PGC1α (peroxisome proliferator-activated receptor γ coactivator 1 α) axis in cells (211). Importantly, aging and obesity associate with decreased adiponectin levels (134, 215). Decreased adiponectin levels have also been observed in elderly patients with neurocognitive disorders (216). In contrast, the antiaging dietary regimen caloric restriction increases circulating adiponectin levels in experimental animals (215, 217–222). Adiponectin was shown to confer multifaceted neuroprotective and vasoprotective effects (212, 218, 220, 223). Adiponectin receptors (AdipoR1 and AdipoR2) are expressed in the hippocampus and other brain regions, and adiponectin was shown to promote synaptic transmission and memory function (224, 225). Accordingly, AdipoRon, a small molecule pan-adiponectin receptor agonist has been also shown to modulate hippocampal synaptic transmission (226) and attenuate neuroinflammation (227).
Adiponectin also exerts diverse endothelial protective effects. It was shown to protect endothelial cells against high glucose and oxidized LDL-induced oxidative stress (228, 229), increase the production of NO1179 (230), and maintain capillarity and microvascular blood flow (231). The pan-adiponectin receptor agonist AdipoRon was shown to improve endothelial function (232). Adiponectin was also reported to inhibit atherogenesis (212) and to modulate inflammatory processes in cerebromicrovascular endothelial cells (233). Further, several studies established a critical role of adiponectin in antiaging vascular effects of caloric restriction (218, 220). Exercise training and weight loss were also shown to increase adiponectin levels, which associate with improvement of microvascular endothelial function (234, 235). Whether therapies targeting adiponectin signaling can exert similar improvements in brain microvascular function in obese elderly patients remains to be determined.
Insulin Resistance
Obesity is commonly associated with hyperinsulinemia and insulin resistance, a prerequisite for prediabetes and type 2 diabetes (236). Clinical studies have shown that diabetes or prediabetes accelerates the progression from mild cognitive impairment to dementia (12, 237–239), with age and the duration of diabetes being the major risk factors (240).
Intact insulin signaling in the brain is important for normal cognitive functions. High-fat diet-induced obesity has been shown to induce insulin resistance in the hippocampus (241, 242), a region known to regulate learning and memory. Preclinical studies have shown that hippocampal-specific insulin resistance impairs spatial learning and neuroplasticity without affecting peripheral glucose homeostasis (243), suggesting insulin resistance in the brain could contribute to obesity-induced cognitive dysfunction. Although the exact mechanisms underlying obesity-induced insulin resistance in the hippocampus are not known, reduced receptor-mediated transport of insulin across BBB or reduced expression of insulin receptors in the hippocampus could play a role (244, 245). In addition to its direct actions on neurons, insulin signaling can also modulate cognitive functions through its actions on the brain microvasculature. Under insulin-sensitive states, insulin activates eNOS to produce NO through the phosphatidyl inositol (PI)-3-kinase-Akt signaling pathway, resulting in increased tissue perfusion and subsequent augmentation of glucose disposal (246, 247). Obesity-induced insulin resistance in the hippocampal microvessels led to decreased insulin-mediated microvascular perfusion and eNOS expression in the hippocampus (241). In insulin-resistant obese Zucker rats, treatment with insulin sensitizing agents like metformin and rosiglitazone was reported to improve endothelial NO mediation (248) and partially rescue cerebral microvascular rarefaction (109). Considering that BBB damage precedes cognitive dysfunction in obesity (249, 250), insulin resistance in the cerebromicrovascular endothelial cells as a causative factor for BBB damage and cognitive decline in obesity needs to be investigated.
Altered Gut-Brain Axis (Dysbiosis)
The gut microbiome, with an estimated 100 trillion microrganisms, has emerged as an important contributor to cognitive health. A change in the composition of the gut microbiome due to loss of beneficial bacteria or overgrowth of harmful bacteria leading to an overall decrease in microbial diversity is called dysbiosis. Both aging and obesity are associated with a dysbiotic microbiome (251–253). Specifically, increased levels of Firmicutes (F) and decreased levels of Bacteroides (B) phylum bacteria have been reported both in obesity and aging (254–256). More importantly, these changes in the microbiome are linked with impaired CBF, BBB impairment, and cognitive dysfunction (254, 257). Clinical studies show that patients with dementia have a higher F/B ratio (258) and elderly patients with similar dysbiotic microbiome perform poor in cognitive tests (259). Similarly in preclinical studies, obese mice with poor microbial diversity exhibited impaired spatial memory (260), and fecal/cecal transplantation from high-fat diet-fed mice to germ-free mice resulted in selective disruptions in exploratory, cognitive, and stereotypical behavior in the absence of obesity (261). These studies suggest that dysbiosis could contribute to obesity- and/or aging-induced cognitive dysfunction.
One of the major mechanisms by which dysbiotic gut microbiota may impact cognition is through promoting BBB impairment. Brainste et al. (262) showed that germ-free mice (both during the intrauterine and the postnatal period) had increased BBB permeability with reduced expression of the tight junction proteins, occludin, and claudin-5. Exposure of germ-free mice to normal microbiota reversed the above mentioned adverse effects on BBB (262), suggesting gut microbiota-brain communication is essential for normal development and maintenance of BBB function. Although there are correlational studies connecting gut microbiome perturbations and obesity and aging-induced BBB dysfunction and cognitive decline (254, 257), the direct cause-effect relationship needs further investigation. Dysbiosis can also indirectly affect cognition through promoting systemic inflammation. Rodent studies have shown that intake of western diet compromises the gut barrier by decreasing the level of tight junction protein ZO-1 and transepithelial resistance in the colon (263). The resulting leaky gut makes it easier for the entry of bacteria-derived lipopolysaccharide (LPS) in to the circulation, leading to endotoxemia and systemic inflammation (257). In addition, dysbiosis also results in decreased production of beneficial short chain fatty acids (SCFAs) such as acetate, propionate, and butyrate by microbial fermentation of indigestible carbohydrates. Obesity is associated with decreased plasma levels of SCFAs (264), which are known to have anti-inflammatory and immune-modulatory effects. Especially, sodium butyrate has been shown to improve cognitive function by increasing brain-derived neurotrophic factor (BDNF) levels in the brain (265). It is also highly possible that butyrate can modulate the aging process due to its epigenetic actions by inhibition of histone deacetylase activity (266).
CELLULAR SENESCENCE: A POTENTIAL MECHANISM FOR ACCELERATED VASCULAR AGING IN OBESITY
Cellular senescence is a cell-autonomous aging process characterized by irreversible cell cycle arrest, expression of a senescence-associated secretory phenotype (SASP), heterochromatin foci, and increased expression of cell cycle inhibitors like p16. Senescent cells accumulate in various tissues of the body including the brain during aging and have been implicated in the pathogenesis of age-related diseases (77, 85, 149, 267–275). One of the major mechanisms through which senescent cells contribute to aging and age-related diseases is through SASP where the secretome containing proinflammatory mediators and matrix-degrading proteases detrimentally affect the tissue microenvironment, impairing normal tissue function and rejuvenation. Elimination of senescent cells that expresses p16 protein has been recently reported to improve life span and health span in rodents (276–280), consistent with the notion that senescent cells drive organismal aging.
Emerging evidence suggest that cellular senescence in the vascular cells could mediate aging and obesity-induced vascular pathologies. Primary cerebrovascular endothelial cells and pericytes isolated from aged mice had higher SA-β gal activity and increased expression of cell cycle inhibitors, p16 and p21, when compared with young mice (281). BulbR1 (H/H) mice, which exhibit an increased number of senescent endothelial cells and pericytes demonstrated less coverage of tight junction proteins in the cortical microvessels and a compromised BBB integrity (281). Metabolic factors that have relevance for obesity and the metabolic syndrome, including high glucose levels, oxidized low-density lipoproteins, and advanced glycation end products, have been reported to induce premature senescence in endothelial cells (282–284). We have recently demonstrated that obesity increases expression of senescence markers in the mouse cerebral circulation, and this effect is exacerbated by genetic depletion of Nrf2 (86). Further, Nrf2 deficiency accelerates age-associated induction of senescence and inflammation in the hippocampus (85). These studies point to a potential role for accelerated vascular senescence in the brain contributing to the adverse interaction of aging and obesity in the pathogenesis of VCI. It is important to better understand the mechanisms by which metabolic factors in obesity might induce premature senescence in the vasculature. Further studies elucidating the cell types that become senescent in aging and obesity in the cerebral vasculature will provide crucial details on the cellular mechanisms involved in senescence-mediated cognitive aging. Identification of senescent cells by assessing their transcriptomic profile [single cell RNA sequencing (273)], by flow cytometry (285), or by immunohistology should be attempted in obese aged animals. The effects of senolytic treatments in these models should also be tested (285).
INTERVENTION STRATEGIES
Exercise
Several studies have documented the beneficial effects of exercise on age- and obesity-dependent neurovascular dysfunction, cerebral blood flow, and cognition. In older obese/overweight individuals, a morning bout of moderate-intensity exercise, with subsequent light-intensity walking breaks from sitting, improved cerebral blood flow measured by transcranial Doppler (286). In another study, 4-mo high-intensity interval training improved cerebral oxygen extraction along with positive cognitive outcomes including improved short-term and verbal memory, attention, and processing speed in middle-aged obese patients (287). In addition, three separate meta-analyses of longitudinal studies have reported that physical activity delays or prevents late-life cognitive decline and dementia (288–290). Some studies have also compared the effects of different types of exercise on microvascular and cognitive outcomes in aging. Acute aerobic, but not resistance, training was shown to improve attention and working memory in aged individuals (291). Similarly, moderate aerobic exercise for 24 wk improved vasomotor organization, attention, and concentration in healthy aged subjects (292). In another study, a supervised aerobic intervention for 6 mo also improved fluency and resting cerebral blood flow in healthy low-active middle-aged and older adults in the Brain in Motion (BIM) study (293). Several other studies also overwhelmingly support the positive effects of aerobic exercise on cerebral blood flow and cognitive outcomes in older individuals (294–296). Interestingly, exercise was able to confer similar cognitive benefits either alone or in combination with dietary intervention in obese elderly patients (297). Although the majority of studies suggest that exercise benefits obese older adults, some studies did not find any association of physical activity and the prevalence of cognitive impairment in the elderly (298, 299). The presence of comorbidities like diabetes may likely contribute to the observed inconsistency in the positive effect of exercise in obese elderly individuals (300).
Preclinical studies have provided additional evidence elucidating microvascular mechanisms contributing to exercise-mediated beneficial cognitive outcomes in aging and obesity. Voluntary wheel running for 6 mo in midlife reduced BBB permeability, increased microvessel pericyte coverage, reduced microglial activation, and preserved basement membrane in the microvasculature of APOE-deficient mice (301). Six weeks of voluntary wheel running also appears to increase capillarization and VEGF levels in the hippocampus of middle-aged mice (302). Chronic physical activity after the onset of obesity also improved insulin-mediated vasodilation in the cerebral vessels in middle-aged rats (303). These aforementioned exercise-induced microvascular protective effects likely can be attributed, at least in part, to reduced systemic inflammatory status. Results from the Health ABC and NHANESIII studies show that self-reported physical activity is associated with reduced levels of circulating IL6, TNF-α, and C-reactive protein (CRP) levels, and this association is independent of both BMI and waist-to-hip ratio in older adults (304, 305). Although the existing evidence supports the concept that exercise improves cognition via exerting microvascular protective effects, additional studies are needed to completely understand the circulating mediators and the exact cellular and molecular mechanisms involved in its effects on neurovascular coupling and brain capillarization, especially in obese elderly individuals.
Dietary Interventions
Weight loss mediated through various forms of dietary interventions including calorie restriction (CR), intermittent fasting, and consumption of a Mediterranean diet have inconsistent cognitive outcomes in the obese elderly population. Three months of 30% CR increased verbal memory scores, which correlated with reduced body weight, fasting insulin, and CRP levels in overweight aged subjects (306), and the same is true for patients with mild cognitive impairment (MCI) (307). Importantly, improved cognition was observed only during the negative energy phase of CR, which is no longer sustained during the subsequent weight maintenance phase (308). However, some studies report that weight loss by CR alone was not sufficient to improve cognition, unless combined with exercise (309, 310). This could be due to the adverse side effects of CR including decrease in muscle mass, which adversely affects the overall glucose metabolism and negates the positive effects of weight loss on cognition. Hence, intermittent fasting (various dietary regimen with alternating fasting and nonfasting cycles) has emerged as a better alternative to CR, as it has been shown to improve cognition in the obese elderly (311) without adverse side effects (312). Previous studies demonstrated that CR in aged rodents increases Nrf2 activity, increases the angiogenic potential, and reduces the cellular and mitochondrial oxidative stress in cerebromicrovascular endothelial cells (119), and these changes at the level of microvasculature are at least in part mediated through circulating factors (120). Additional studies are needed to understand the source and the microvascular impact of these circulating factors in the context of VCI.
Changes in diet composition including Mediterranean diet rich in olive oil or the ketogenic diet low in carbohydrate and high in fat have also been shown to affect cognition positively in the elderly population (313, 314). Especially, adherence to the Mediterranean diet improved endothelial function marked by increases in flow-mediated dilation (314), increases in serum NO, decline in ROS and endothelin-1 production (315) and improves the regenerative capacity of endothelial progenitor cells (316). However, most of the aforementioned studies focused on the peripheral vasculature, and the effects of diet composition on cerebral microvasculature are far from clear.
Other Nonlifestyle Interventions
Although diet and exercise seem effective in overweight or moderately obese individuals, lifestyle interventions are not amenable for severely obese patients. Bariatric surgery is a popular nonlifestyle intervention for obese subjects with a BMI ≥ 40 to yield sustained weight reductions. Results from the Longitudinal Assessment of Bariatric Surgery project demonstrated improved executive and memory performance and was maintained 2–3 yr after surgery-induced weight loss, whereas this effect was lost in the subset of participants who regained weight (317, 318). As seen with other weight loss strategies, bariatric surgery-mediated cognitive improvements are associated with improved metabolic outcomes and reduced systemic inflammation (319), which could affect brain microvasculature to impact cognition.
In women, the role of estrogen in modulation of vascular function and cognition should not be overlooked (320). Surgical menopause in women ≤45 yr of age through bilateral oophorectomy significantly affects cognitive performance (321, 322). In contrast, estrogen replacement through hormone replacement therapy in older women was shown to improve cognitive test scores, especially when started early during the postmenopausal period (323).The protective role of estrogen on endothelial function has been extensively studied and reviewed elsewhere (324).
PERSPECTIVES
It is becoming increasingly accepted that microvascular mechanisms could play a critical role in aging-induced and obesity-related cognitive impairment. Rescuing microvascular function for treatment and prevention of cognitive decline is a promising approach, as the cerebral vasculature and the neurovascular unit are more accessible targets for pharmacological and nonpharmacological (e.g., dietary, exercise) interventions than nonvascular cells in the brain. Further translational studies are warranted to test the cerebromicrovascular and cognitive protective effects of combinations of various exercise protocols, dietary regimens, and antiaging pharmacological interventions in obese older adults at risk for VCI.
GRANTS
This work was supported by grants from the Oklahoma Center for the Advancement of Science and Technology (to A. Csiszar, A. Yabluchanskiy, and Z. Ungvari), the National Institute on Aging (NIA; R01-AG047879; R01-AG038747; R01-AG055395; R01-AG068295), the National Institute of Neurological Disorders and Stroke (R01-NS056218, R01-NS100782), the National Institute of General Medical Sciences Oklahoma Shared Clinical and Translational Resources (GM104938, to A. Yabluchanskiy), the Presbyterian Health Foundation, the NIA-supported Geroscience Training Program in Oklahoma (T32AG052363), the Oklahoma Nathan Shock Center (P30AG050911), the Cellular and Molecular GeroScience CoBRE (1P20GM125528, sub#5337).
DISCLOSURES
No conflicts of interest, financial or otherwise, are declared by the authors.
AUTHOR CONTRIBUTIONS
S.T., A.Y., and T.C. prepared figures; P.B., A.C., T.K., S.T., A.N-T., C.A., A.Y., T.C., A.L., and A.T. drafted manuscript; P.B., A.C., T.K., S.T., A.N-T., C.A., A.Y., T.C., A.L., and A.T. edited and revised manuscript; P.B., A.C., T.K., S.T., A.N-T., C.A., A.Y., T.C., A.L., and A.T. approved final version of manuscript.
REFERENCES
- 1.Wang YC, Colditz GA, Kuntz KM. Forecasting the obesity epidemic in the aging U.S. population. Obesity (Silver Spring) 15: 2855–2865, 2007. doi: 10.1038/oby.2007.339. [DOI] [PubMed] [Google Scholar]
- 2.Flegal KM, Carroll MD, Kit BK, Ogden CL. Prevalence of obesity and trends in the distribution of body mass index among US adults, 1999-2010. JAMA 307: 491–497, 2012. doi: 10.1001/jama.2012.39. [DOI] [PubMed] [Google Scholar]
- 3.Beydoun MA, Beydoun HA, Wang Y. Obesity and central obesity as risk factors for incident dementia and its subtypes: a systematic review and meta-analysis. Obes Rev 9: 204–218, 2008. doi: 10.1111/j.1467-789X.2008.00473.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 4.Gustafson DR, Karlsson C, Skoog I, Rosengren L, Lissner L, Blennow K. Mid-life adiposity factors relate to blood-brain barrier integrity in late life. J Intern Med 262: 643–650, 2007. doi: 10.1111/j.1365-2796.2007.01869.x. [DOI] [PubMed] [Google Scholar]
- 5.Prickett C, Brennan L, Stolwyk R. Examining the relationship between obesity and cognitive function: a systematic literature review. Obes Res Clin Pract 9: 93–113, 2015. doi: 10.1016/j.orcp.2014.05.001. [DOI] [PubMed] [Google Scholar]
- 6.Prickett C, Stolwyk R, O’Brien P, Brennan L. Neuropsychological functioning in mid-life treatment-seeking adults with obesity: a cross-sectional study. Obes Surg 28: 532–540, 2018. doi: 10.1007/s11695-017-2894-0. [DOI] [PubMed] [Google Scholar]
- 7.Neale C, Johnston P, Hughes M, Scholey A. Functional activation during the rapid visual information processing task in a middle aged cohort: an fMRI study. PLoS One 10: e0138994, 2015. doi: 10.1371/journal.pone.0138994. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 8.Nepal B, Brown L, Ranmuthugala G. Modelling the impact of modifying lifestyle risk factors on dementia prevalence in Australian population aged 45 years and over, 2006-2051. Australas J Ageing 29: 111–116, 2010. doi: 10.1111/j.1741-6612.2010.00392.x. [DOI] [PubMed] [Google Scholar]
- 9.Dahl A, Hassing LB, Fransson E, Berg S, Gatz M, Reynolds CA, Pedersen NL. Being overweight in midlife is associated with lower cognitive ability and steeper cognitive decline in late life. J Gerontol A Biol Sci Med Sci 65A: 57–62, 2010. doi: 10.1093/gerona/glp035. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 10.Dahl AK, Hassing LB. Obesity and cognitive aging. Epidemiol Rev 35: 22–32, 2013. doi: 10.1093/epirev/mxs002. [DOI] [PubMed] [Google Scholar]
- 11.Dahl AK, Hassing LB, Fransson EI, Gatz M, Reynolds CA, Pedersen NL. Body mass index across midlife and cognitive change in late life. Int J Obes 37: 296–302, 2013. doi: 10.1038/ijo.2012.37. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 12.Elias MF, Elias PK, Sullivan LM, Wolf PA, D’Agostino RB. Obesity, diabetes and cognitive deficit: The Framingham Heart Study. Neurobiol Aging 26: 11–16, 2005. doi: 10.1016/j.neurobiolaging.2005.08.019. [DOI] [PubMed] [Google Scholar]
- 13.Gunstad J, Lhotsky A, Wendell CR, Ferrucci L, Zonderman AB. Longitudinal examination of obesity and cognitive function: results from the Baltimore longitudinal study of aging. Neuroepidemiology 34: 222–229, 2010. doi: 10.1159/000297742. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 14.Hassing LB, Dahl AK, Pedersen NL, Johansson B. Overweight in midlife is related to lower cognitive function 30 years later: a prospective study with longitudinal assessments. Dement Geriatr Cogn Disord 29: 543–552, 2010. doi: 10.1159/000314874. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 15.Hassing LB, Dahl AK, Thorvaldsson V, Berg S, Gatz M, Pedersen NL, Johansson B. Overweight in midlife and risk of dementia: a 40-year follow-up study. Int J Obes 33: 893–898, 2009. doi: 10.1038/ijo.2009.104. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 16.Kanaya AM, Lindquist K, Harris TB, Launer L, Rosano C, Satterfield S, Yaffe K, Study HA. Total and regional adiposity and cognitive change in older adults: The Health, Aging and Body Composition (ABC) study. Arch Neurol 66: 329–335, 2009. doi: 10.1001/archneurol.2008.570. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 17.Wolf PA, Beiser A, Elias MF, Au R, Vasan RS, Seshadri S. Relation of obesity to cognitive function: importance of central obesity and synergistic influence of concomitant hypertension. The Framingham Heart Study. Curr Alzheimer Res 4: 111–116, 2007. doi: 10.2174/156720507780362263. [DOI] [PubMed] [Google Scholar]
- 18.Sabia S, Kivimaki M, Shipley MJ, Marmot MG, Singh-Manoux A. Body mass index over the adult life course and cognition in late midlife: the Whitehall II Cohort Study. Am J Clin Nutr 89: 601–607, 2009. doi: 10.3945/ajcn.2008.26482. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 19.Gorelick PB, Scuteri A, Black SE, Decarli C, Greenberg SM, Iadecola C, Launer LJ, Laurent S, Lopez OL, Nyenhuis D, Petersen RC, Schneider JA, Tzourio C, Arnett DK, Bennett DA, Chui HC, Higashida RT, Lindquist R, Nilsson PM, Roman GC, Sellke FW, Seshadri S; 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 42: 2672–2713, 2011. doi: 10.1161/STR.0b013e3182299496. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 20.Luchsinger JA. Adiposity, hyperinsulinemia, diabetes and Alzheimer's disease: an epidemiological perspective. Eur J Pharmacol 585: 119–129, 2008. doi: 10.1016/j.ejphar.2008.02.048. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 21.Kivimaki M, Kuosma E, Ferrie JE, Luukkonen R, Nyberg ST, Alfredsson L, Batty GD, Brunner EJ, Fransson E, Goldberg M, Knutsson A, Koskenvuo M, Nordin M, Oksanen T, Pentti J, Rugulies R, Shipley MJ, Singh-Manoux A, Steptoe A, Suominen SB, Theorell T, Vahtera J, Virtanen M, Westerholm P, Westerlund H, Zins M, Hamer M, Bell JA, Tabak AG, Jokela M. Overweight, obesity, and risk of cardiometabolic multimorbidity: pooled analysis of individual-level data for 120 813 adults from 16 cohort studies from the USA and Europe. Lancet Public Health 2: e277–e285, 2017. doi: 10.1016/S2468-2667(17)30074-9. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 22.Benito-León J, Mitchell AJ, Hernández-Gallego J, Bermejo-Pareja F. Obesity and impaired cognitive functioning in the elderly: a population-based cross-sectional study (NEDICES). Eur J Neurol 20: 899–906, 2013. doi: 10.1111/ene.12083. [DOI] [PubMed] [Google Scholar]
- 23.Kerwin DR, Zhang Y, Kotchen JM, Espeland MA, Van Horn L, McTigue KM, Robinson JG, Powell L, Kooperberg C, Coker LH, Hoffmann R. The cross-sectional relationship between body mass index, waist-hip ratio, and cognitive performance in postmenopausal women enrolled in the Women's Health Initiative. J Am Geriatr Soc 58: 1427–1432, 2010. doi: 10.1111/j.1532-5415.2010.02969.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 24.Elias MF, Elias PK, Sullivan LM, Wolf PA, D'Agostino RB. Lower cognitive function in the presence of obesity and hypertension: the Framingham heart study. Int J Obes 27: 260–268, 2003. doi: 10.1038/sj.ijo.802225. [DOI] [PubMed] [Google Scholar]
- 25.Waldstein SR, Katzel LI. Interactive relations of central versus total obesity and blood pressure to cognitive function. Int J Obes (Lond) 30: 201–207, 2006. doi: 10.1038/sj.ijo.0803114. [DOI] [PubMed] [Google Scholar]
- 26.Zaninotto P, Batty GD, Allerhand M, Deary IJ. Cognitive function trajectories and their determinants in older people: 8 years of follow-up in the English Longitudinal Study of Ageing. J Epidemiol Community Health 72: 685–694, 2018. doi: 10.1136/jech-2017-210116. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 27.Ganguli M, Beer JC, Zmuda JM, Ryan CM, Sullivan KJ, Chang CH, Rao RH. Aging, diabetes, obesity, and cognitive decline: a population-based study. J Am Geriatr Soc 68: 991–998, 2020. doi: 10.1111/jgs.16321. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 28.Alosco ML, Spitznagel MB, Raz N, Cohen R, Sweet LH, Colbert LH, Josephson R, van Dulmen M, Hughes J, Rosneck J, Gunstad J. Obesity interacts with cerebral hypoperfusion to exacerbate cognitive impairment in older adults with heart failure. Cerebrovasc Dis Extra 2: 88–98, 2012. doi: 10.1159/000343222. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 29.Shen YC, Kung SC, Chang ET, Hong YL, Wang LY. The impact of obesity in cognitive and memory dysfunction in obstructive sleep apnea syndrome. Int J Obes 43: 355–361, 2019. doi: 10.1038/s41366-018-0138-6. [DOI] [PubMed] [Google Scholar]
- 30.Bagger YZ, Tankó LB, Alexandersen P, Qin G, Christiansen C. The implications of body fat mass and fat distribution for cognitive function in elderly women. Obes Res 12: 1519–1526, 2004. doi: 10.1038/oby.2004.189. [DOI] [PubMed] [Google Scholar]
- 31.Kuo HK, Jones RN, Milberg WP, Tennstedt S, Talbot L, Morris JN, Lipsitz LA. Cognitive function in normal-weight, overweight, and obese older adults: an analysis of the Advanced Cognitive Training for Independent and Vital Elderly cohort. J Am Geriatr Soc 54: 97–103, 2006. doi: 10.1111/j.1532-5415.2005.00522.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 32.Hainer V, Aldhoon-Hainerová I. Obesity paradox does exist. Diabetes Care 36 Suppl 2: S276–S281, 2013. doi: 10.2337/dcS13-2023. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 33.Singh-Manoux A, Dugravot A, Shipley M, Brunner EJ, Elbaz A, Sabia S, Kivimaki M. Obesity trajectories and risk of dementia: 28 years of follow-up in the Whitehall II Study. Alzheimers Dement 14: 178–186, 2018. doi: 10.1016/j.jalz.2017.06.2637. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 34.Banack HR, Kaufman JS. The "obesity paradox" explained. Epidemiology 24: 461–462, 2013. doi: 10.1097/EDE.0b013e31828c776c. [DOI] [PubMed] [Google Scholar]
- 35.Bruce-Keller AJ, White CL, Gupta S, Knight AG, Pistell PJ, Ingram DK, Morrison CD, Keller JN. NOX activity in brain aging: exacerbation by high fat diet. Free Radic Biol Med 49: 22–30, 2010. doi: 10.1016/j.freeradbiomed.2010.03.006. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 36.Spencer SJ, D'Angelo H, Soch A, Watkins LR, Maier SF, Barrientos RM. High-fat diet and aging interact to produce neuroinflammation and impair hippocampal- and amygdalar-dependent memory. Neurobiol Aging 58: 88–101, 2017. doi: 10.1016/j.neurobiolaging.2017.06.014. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 37.Tucsek Z, Toth P, Sosnowsk D, Gautam T, Mitschelen M, Koller A, Szalai G, Sonntag WE, Ungvari Z, Csiszar A. Obesity in aging exacerbates blood brain barrier disruption, neuroinflammation and oxidative stress in the mouse hippocampus: effects on expression of genes involved in beta-amyloid generation and Alzheimer's disease. J Gerontol A Biol Sci Med Sci 69: 1212–1226, 2014. doi: 10.1093/gerona/glt177. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 38.Tucsek Z, Toth P, Tarantini S, Sosnowska D, Gautam T, Warrington JP, Giles CB, Wren JD, Koller A, Ballabh P, Sonntag WE, Ungvari Z, Csiszar A. Aging exacerbates obesity-induced cerebromicrovascular rarefaction, neurovascular uncoupling, and cognitive decline in mice. J Gerontol A Biol Sci Med Sci 69: 1339–1352, 2014. doi: 10.1093/gerona/glu080. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 39.Valcarcel-Ares MN, Tucsek Z, Kiss T, Giles CB, Tarantini S, Yabluchanskiy A, Balasubramanian P, Gautam T, Galvan V, Ballabh P, Richardson A, Freeman WM, Wren JD, Deak F, Ungvari Z, Csiszar A. Obesity in aging exacerbates neuroinflammation, dysregulating synaptic function-related genes and altering eicosanoid synthesis in the mouse hippocampus: potential role in impaired synaptic plasticity and cognitive decline. J Gerontol A Biol Sci Med Sci 74: 290–298, 2018. doi: 10.1093/gerona/gly127. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 40.Morrison CD, Pistell PJ, Ingram DK, Johnson WD, Liu Y, Fernandez-Kim SO, White CL, Purpera MN, Uranga RM, Bruce-Keller AJ, Keller JN. High fat diet increases hippocampal oxidative stress and cognitive impairment in aged mice: implications for decreased Nrf2 signaling. J Neurochem 114: 1581–1589, 2010. doi: 10.1111/j.1471-4159.2010.06865.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 41.Lin B, Hasegawa Y, Takane K, Koibuchi N, Cao C, Kim-Mitsuyama S. High-fat-diet intake enhances cerebral amyloid angiopathy and cognitive impairment in a mouse model of alzheimer's disease, independently of metabolic disorders. J Am Heart Assoc 5: e003154, 2016. doi: 10.1161/JAHA.115.003154. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 42.Pétrault O, Pétrault M, Ouk T, Bordet R, Bérézowski V, Bastide M. Visceral adiposity links cerebrovascular dysfunction to cognitive impairment in middle-aged mice. Neurobiol Dis 130: 104536, 2019. doi: 10.1016/j.nbd.2019.104536. [DOI] [PubMed] [Google Scholar]
- 43.Salinero AE, Robison LS, Gannon OJ, Riccio D, Mansour F, Abi‐Ghanem C, Zuloaga KL. Sex‐specific effects of high‐fat diet on cognitive impairment in a mouse model of VCID. FASEB J 34: 15108–15122, 2020. (ed.)doi: 10.1096/fj.202000085R. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 44.Csipo T, Lipecz A, Fulop GA, Hand RA, Ngo BN, Dzialendzik M, Tarantini S, Balasubramanian P, Kiss T, Yabluchanska V, Silva-Palacios F, Courtney DL, Dasari TW, Sorond F, Sonntag WE, Csiszar A, Ungvari Z, Yabluchanskiy A. Age-related decline in peripheral vascular health predicts cognitive impairment. Geroscience 41: 125–136, 2019. doi: 10.1007/s11357-019-00063-5. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 45.Csipo T, Mukli P, Lipecz A, Tarantini S, Bahadli D, Abdulhussein O, Owens C, Kiss T, Balasubramanian P, Nyul-Toth A, Hand RA, Yabluchanska V, Sorond FA, Csiszar A, Ungvari Z, Yabluchanskiy A. Assessment of age-related decline of neurovascular coupling responses by functional near-infrared spectroscopy (fNIRS) in humans. Geroscience 41: 495–509, 2019. doi: 10.1007/s11357-019-00122-x. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 46.Csiszar A, Yabluchanskiy A, Ungvari A, Ungvari Z, Tarantini S. Overexpression of catalase targeted to mitochondria improves neurovascular coupling responses in aged mice. Geroscience 41: 609–617, 2019. doi: 10.1007/s11357-019-00111-0. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 47.Farias Quipildor GE, Mao K, Hu Z, Novaj A, Cui MH, Gulinello M, Branch CA, Gubbi S, Patel K, Moellering DR, Tarantini S, Kiss T, Yabluchanskiy A, Ungvari Z, Sonntag WE, Huffman DM. Central IGF-1 protects against features of cognitive and sensorimotor decline with aging in male mice. Geroscience 41: 185–208, 2019. doi: 10.1007/s11357-019-00065-3. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 48.Fulop GA, Ahire C, Csipo T, Tarantini S, Kiss T, Balasubramanian P, Yabluchanskiy A, Farkas E, Toth A, Nyul-Toth A, Toth P, Csiszar A, Ungvari Z. Cerebral venous congestion promotes blood-brain barrier disruption and neuroinflammation, impairing cognitive function in mice. Geroscience 41: 575–589, 2019. doi: 10.1007/s11357-019-00110-1. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 49.Iadecola C. The neurovascular unit coming of age: a journey through neurovascular coupling in health and disease. Neuron 96: 17–42, 2017. doi: 10.1016/j.neuron.2017.07.030. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 50.Iadecola C, Gottesman RF. Cerebrovascular alterations in Alzheimer disease. Circ Res 123: 406–408, 2018. doi: 10.1161/CIRCRESAHA.118.313400. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 51.Kiss T, Balasubramanian P, Valcarcel-Ares MN, Tarantini S, Yabluchanskiy A, Csipo T, Lipecz A, Reglodi D, Zhang XA, Bari F, Farkas E, Csiszar A, Ungvari Z. Nicotinamide mononucleotide (NMN) treatment attenuates oxidative stress and rescues angiogenic capacity in aged cerebromicrovascular endothelial cells: a potential mechanism for prevention of vascular cognitive impairment. GeroScience 41: 619–630, 2019. doi: 10.1007/s11357-019-00074-2. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 52.Kiss T, Giles CB, Tarantini S, Yabluchanskiy A, Balasubramanian P, Gautam T, Csipo T, Nyul-Toth A, Lipecz A, Szabo C, Farkas E, Wren JD, Csiszar A, Ungvari Z. Nicotinamide mononucleotide (NMN) supplementation promotes anti-aging miRNA expression profile in the aorta of aged mice, predicting epigenetic rejuvenation and anti-atherogenic effects. Geroscience 41: 419–439, 2019. doi: 10.1007/s11357-019-00095-x. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 53.Kiss T, Nyul-Toth A, Balasubramanian P, Tarantini S, Ahire C, Yabluchanskiy A, Csipo T, Farkas E, Wren JD, Garman L, Csiszar A, Ungvari Z. Nicotinamide mononucleotide (NMN) supplementation promotes neurovascular rejuvenation in aged mice: transcriptional footprint of SIRT1 activation, mitochondrial protection, anti-inflammatory, and anti-apoptotic effects. Geroscience 42: 527–546, 2020. doi: 10.1007/s11357-020-00165-5. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 54.Lipecz A, Csipo T, Tarantini S, Hand RA, Ngo BN, Conley S, Nemeth G, Tsorbatzoglou A, Courtney DL, Yabluchanska V, Csiszar A, Ungvari ZI, Yabluchanskiy A. Age-related impairment of neurovascular coupling responses: a dynamic vessel analysis (DVA)-based approach to measure decreased flicker light stimulus-induced retinal arteriolar dilation in healthy older adults. Geroscience 41: 341–349, 2019. doi: 10.1007/s11357-019-00078-y. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 55.Sweeney MD, Kisler K, Montagne A, Toga AW, Zlokovic BV. The role of brain vasculature in neurodegenerative disorders. Nat Neurosci 21: 1318–1331, 2018. doi: 10.1038/s41593-018-0234-x. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 56.Sweeney MD, Sagare AP, Zlokovic BV. Blood-brain barrier breakdown in Alzheimer disease and other neurodegenerative disorders. Nat Rev Neurol 14: 133–150, 2018. doi: 10.1038/nrrheum.2018.1, doi:. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 57.Tarantini S, Yabluchanskiy A, Csipo T, Fulop G, Kiss T, Balasubramanian P, DelFavero J, Ahire C, Ungvari A, Nyul-Toth A, Farkas E, Benyo Z, Toth A, Csiszar A, Ungvari Z. Treatment with the poly(ADP-ribose) polymerase inhibitor PJ-34 improves cerebromicrovascular endothelial function, neurovascular coupling responses and cognitive performance in aged mice, supporting the NAD+ depletion hypothesis of neurovascular aging. Geroscience 41: 533–542, 2019. doi: 10.1007/s11357-019-00101-2. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 58.Wiedenhoeft T, Tarantini S, Nyul-Toth A, Yabluchanskiy A, Csipo T, Balasubramanian P, Lipecz A, Kiss T, Csiszar A, Csiszar A, Ungvari Z. Fusogenic liposomes effectively deliver resveratrol to the cerebral microcirculation and improve endothelium-dependent neurovascular coupling responses in aged mice. Geroscience 41: 711–725, 2019. doi: 10.1007/s11357-019-00102-1. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 59.Birdsill AC, Carlsson CM, Willette AA, Okonkwo OC, Johnson SC, Xu G, Oh JM, Gallagher CL, Koscik RL, Jonaitis EM, Hermann BP, LaRue A, Rowley HA, Asthana S, Sager MA, Bendlin BB. Low cerebral blood flow is associated with lower memory function in metabolic syndrome. Obesity (Silver Spring ) 21: 1313–1320, 2013. doi: 10.1002/oby.20170. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 60.Espeland MA, Luchsinger JA, Neiberg RH, Carmichael O, Laurienti PJ, Pi‐Sunyer X, Wing RR, Cook D, Horton E, Casanova R, Erickson K, Nick Bryan R; the Action for Health in Diabetes Brain Magnetic Resonance Imaging Research Group. Long term effect of intensive lifestyle intervention on cerebral blood flow. J Am Geriatr Soc 66: 120–126, 2018. doi: 10.1111/jgs.15159. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 61.Hurr C, Patik JC, Kim K, Brothers RM. Blunted cerebral vascular responsiveness to hypercapnia in obese individuals. Exp Physiol 102: 1300–1308, 2017. doi: 10.1113/EP086446. [DOI] [PubMed] [Google Scholar]
- 62.MacIntosh BJ, Shirzadi Z, Atwi S, Detre JA, Dolui S, Bryan RN, Launer LJ, Swardfager W. Metabolic and vascular risk factors are associated with reduced cerebral blood flow and poorer midlife memory performance. Hum Brain Mapp 41: 855–864, 2020. doi: 10.1002/hbm.24844. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 63.Selim M, Jones R, Novak P, Zhao P, Novak V. The effects of body mass index on cerebral blood flow velocity. Clin Auton Res 18: 331–338, 2008. doi: 10.1007/s10286-008-0490-z. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 64.Willeumier KC, Taylor DV, Amen DG. Elevated BMI is associated with decreased blood flow in the prefrontal cortex using SPECT imaging in healthy adults. Obesity (Silver Spring ) 19: 1095–1097, 2011. doi: 10.1038/oby.2011.16. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 65.Toth P, Tarantini S, Csiszar A, Ungvari Z. Functional vascular contributions to cognitive impairment and dementia: mechanisms and consequences of cerebral autoregulatory dysfunction, endothelial impairment, and neurovascular uncoupling in aging. Am J Physiol Heart Circ Physiol 312: H1–H20, 2017. doi: 10.1152/ajpheart.00581.2016. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 66.Demchenko IT, Luchakov YI, Moskvin AN, Gutsaeva DR, Allen BW, Thalmann ED, Piantadosi CA. Cerebral blood flow and brain oxygenation in rats breathing oxygen under pressure. J Cereb Blood Flow Metab 25: 1288–1300, 2005. doi: 10.1038/sj.jcbfm.9600110. [DOI] [PubMed] [Google Scholar]
- 67.Sabayan B, Westendorp RG, Grond J, Stott DJ, Sattar N, van Osch MJ, van Buchem MA, de Craen AJ. Markers of endothelial dysfunction and cerebral blood flow in older adults. Neurobiol Aging 35: 373–377, 2014. doi: 10.1016/j.neurobiolaging.2013.08.020. [DOI] [PubMed] [Google Scholar]
- 68.Toda N, Ayajiki K, Okamura T. Obesity-induced cerebral hypoperfusion derived from endothelial dysfunction: one of the risk factors for Alzheimer's disease. Curr Alzheimer Res 11: 733–744, 2014. doi: 10.2174/156720501108140910120456. [DOI] [PubMed] [Google Scholar]
- 69.Nicholson WT, Vaa B, Hesse C, Eisenach JH, Joyner MJ. Aging is associated with reduced prostacyclin-mediated dilation in the human forearm. Hypertension 53: 973–978, 2009. doi: 10.1161/HYPERTENSIONAHA.108.121483. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 70.Hodnett BL, Dearman JA, Carter CB, Hester RL. Attenuated PGI2 synthesis in obese Zucker rats. Am J Physiol Regul Integr Comp Physiol 296: R715–R721, 2009. doi: 10.1152/ajpregu.90330.2008. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 71.Csiszar A, Tarantini S, Yabluchanskiy A, Balasubramanian P, Kiss T, Farkas E, Baur JA, Ungvari ZI. Role of endothelial NAD+ deficiency in age-related vascular dysfunction. Am J Physiol Heart Circ Physiol 316: H1253–H1266. doi: 10.1152/ajpheart.00039.2019. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 72.Phillips SA, Sylvester FA, Frisbee JC. Oxidant stress and constrictor reactivity impair cerebral artery dilation in obese Zucker rats. Am J Physiol Regul Integr Comp Physiol 288: R522–R530, 2005. doi: 10.1152/ajpregu.00655.2004. [DOI] [PubMed] [Google Scholar]
- 73.Tarantini S, Tran CHT, Gordon GR, Ungvari Z, Csiszar A. Impaired neurovascular coupling in aging and Alzheimer's disease: Contribution of astrocyte dysfunction and endothelial impairment to cognitive decline. Exp Gerontol 94: 52–58, 2017. doi: 10.1016/j.exger.2016.11.004. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 74.Tarantini S, Valcarcel-Ares MN, Toth P, Yabluchanskiy A, Tucsek Z, Kiss T, Hertelendy P, Kinter M, Ballabh P, Sule Z, Farkas E, Baur JA, Sinclair DA, Csiszar A, Ungvari Z. Nicotinamide mononucleotide (NMN) supplementation rescues cerebromicrovascular endothelial function and neurovascular coupling responses and improves cognitive function in aged mice. Redox Biol 24: 101192, 2019. doi: 10.1016/j.redox.2019.101192. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 75.Tarantini S, Valcarcel-Ares NM, Yabluchanskiy A, Fulop GA, Hertelendy P, Gautam T, Farkas E, Perz A, Rabinovitch PS, Sonntag WE, Csiszar A, Ungvari Z. Treatment with the mitochondrial-targeted antioxidant peptide SS-31 rescues neurovascular coupling responses and cerebrovascular endothelial function and improves cognition in aged mice. Aging Cell 17: e12731, 2018. doi: 10.1111/acel.12731. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 76.Toth P, Tarantini S, Ashpole NM, Tucsek Z, Milne GL, Valcarcel-Ares NM, Menyhart A, Farkas E, Sonntag WE, Csiszar A, Ungvari Z. IGF-1 deficiency impairs neurovascular coupling in mice: implications for cerebromicrovascular aging. Aging Cell 14: 1034–1044, 2015. doi: 10.1111/acel.12372. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 77.Ungvari Z, Tarantini S, Nyul-Toth A, Kiss T, Yabluchanskiy A, Csipo T, Balasubramanian P, Lipecz A, Benyo Z, Csiszar A. Nrf2 dysfunction and impaired cellular resilience to oxidative stressors in the aged vasculature: from increased cellular senescence to the pathogenesis of age-related vascular diseases. Geroscience 41: 727–738, 2019. doi: 10.1007/s11357-019-00107-w. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 78.Csiszar A, Ungvari Z, Edwards JG, Kaminski P, Wolin MS, Koller A, Kaley G. Aging-induced phenotypic changes and oxidative stress impair coronary arteriolar function. Circ Res 90: 1159–1166, 2002. doi: 10.1161/01.RES.0000020401.61826.EA. [DOI] [PubMed] [Google Scholar]
- 79.Lynch CM, Kinzenbaw DA, Chen X, Zhan S, Mezzetti E, Filosa J, Ergul A, Faulkner JL, Faraci FM, Didion SP. Nox2-derived superoxide contributes to cerebral vascular dysfunction in diet-induced obesity. Stroke 44: 3195–3201, 2013. doi: 10.1161/STROKEAHA.113.001366. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 80.Park L, Anrather J, Girouard H, Zhou P, Iadecola C. Nox2-derived reactive oxygen species mediate neurovascular dysregulation in the aging mouse brain. J Cereb Blood Flow Metab 27: 1908–1918, 2007. doi: 10.1038/sj.jcbfm.9600491. [DOI] [PubMed] [Google Scholar]
- 81.Parker-Duffen JL, Nakamura K, Silver M, Kikuchi R, Tigges U, Yoshida S, Denzel MS, Ranscht B, Walsh K. T-cadherin is essential for adiponectin-mediated revascularization. J Biol Chem 288: 24886–24897, 2013. doi: 10.1074/jbc.M113.454835. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 82.Toth P, Tarantini S, Tucsek Z, Ashpole NM, Sosnowska D, Gautam T, Ballabh P, Koller A, Sonntag WE, Csiszar A, Ungvari Z. Resveratrol treatment rescues neurovascular coupling in aged mice: role of improved cerebromicrovascular endothelial function and downregulation of NADPH oxidase. Am J Physiol Heart Circ Physiol 306: H299–H308, 2014. doi: 10.1152/ajpheart.00744.2013. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 83.Bourgoin F, Bachelard H, Badeau M, Mélançon S, Pitre M, Larivière R, Nadeau A. Endothelial and vascular dysfunctions and insulin resistance in rats fed a high-fat, high-sucrose diet. Am J Physiol Heart Circ Physiol 295: H1044–H1055, 2008. doi: 10.1152/ajpheart.00516.2008. [DOI] [PubMed] [Google Scholar]
- 84.Galili O, Versari D, Sattler KJ, Olson ML, Mannheim D, McConnell JP, Chade AR, Lerman LO, Lerman A. Early experimental obesity is associated with coronary endothelial dysfunction and oxidative stress. Am J Physiol Heart Circ Physiol 292: H904–H911, 2007. doi: 10.1152/ajpheart.00628.2006. [DOI] [PubMed] [Google Scholar]
- 85.Fulop GA, Kiss T, Tarantini S, Balasubramanian P, Yabluchanskiy A, Farkas E, Bari F, Ungvari Z, Csiszar A. Nrf2 deficiency in aged mice exacerbates cellular senescence promoting cerebrovascular inflammation. Geroscience 40: 513–521, 2018. doi: 10.1007/s11357-018-0047-6. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 86.Tarantini S, Valcarcel-Ares MN, Yabluchanskiy A, Tucsek Z, Hertelendy P, Kiss T, Gautam T, Zhang XA, Sonntag WE, de Cabo R, Farkas E, Elliott ME, Kinter MT, Deak F, Ungvari Z, Csiszar A. Nrf2 deficiency exacerbates obesity-induced oxidative stress, neurovascular dysfunction, blood brain barrier disruption, neuroinflammation, amyloidogenic gene expression and cognitive decline in mice, mimicking the aging phenotype. J Gerontol A Biol Sci Med Sci 73: 853–863, 2018. doi: 10.1093/gerona/glx177. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 87.Ungvari Z, Bailey-Downs L, Gautam T, Sosnowska D, Wang M, Monticone RE, Telljohann R, Pinto JT, de Cabo R, Sonntag WE, Lakatta E, Csiszar A. Age-associated vascular oxidative stress, Nrf2 dysfunction and NF-kB activation in the non-human primate Macaca mulatta. J Gerontol A Biol Sci Med Sci 66A: 866–875, 2011. doi: 10.1093/gerona/glr092. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 88.Ungvari Z, Bailey-Downs L, Sosnowska D, Gautam T, Koncz P, Losonczy G, Ballabh P, de Cabo R, Sonntag WE, Csiszar A. Vascular oxidative stress in aging: a homeostatic failure due to dysregulation of Nrf2-mediated antioxidant response. Am J Physiol Heart Circ Physiol 301: H363–H372, 2011. doi: 10.1152/ajpheart.01134.2010. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 89.Ungvari Z, Bagi Z, Feher A, Recchia FA, Sonntag WE, Pearson K, de Cabo R, Csiszar A. Resveratrol confers endothelial protection via activation of the antioxidant transcription factor Nrf2. Am J Physiol Heart Circ Physiol 299: H18–H24, 2010. doi: 10.1152/ajpheart.00260.2010. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 90.Ungvari ZI, Bailey-Downs L, Gautam T, Jimenez R, Losonczy G, Zhang C, Ballabh P, Recchia FA, Wilkerson DC, Sonntag WE, Pearson KJ, de Cabo R, Csiszar A. Adaptive induction of NF-E2-Related Factor-2-driven antioxidant genes in endothelial cells in response to hyperglycemia. Am J Physiol Heart Circ Physiol 300: H1133–H1140, 2011. doi: 10.1152/ajpheart.00402.2010. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 91.Chen BR, Kozberg MG, Bouchard MB, Shaik MA, Hillman EA. Critical role for the vascular endothelium in functional neurovascular coupling in the brain. J Am Heart Assoc 3: e000787, 2014. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 92.Di Marco LY, Venneri A, Farkas E, Evans PC, Marzo A, Frangi AF. Vascular dysfunction in the pathogenesis of Alzheimer's disease–A review of endothelium-mediated mechanisms and ensuing vicious circles. Neurobiol Dis 82: 593–606, 2015. doi: 10.1016/j.nbd.2015.08.014. [DOI] [PubMed] [Google Scholar]
- 93.Tarantini S, Hertelendy P, Tucsek Z, Valcarcel-Ares MN, Smith N, Menyhart A, Farkas E, Hodges E, Towner R, Deak F, Sonntag WE, Csiszar A, Ungvari Z, Toth P. Pharmacologically-induced neurovascular uncoupling is associated with cognitive impairment in mice. J Cereb Blood Flow Metab 35: 1871–1881, 2015. doi: 10.1038/jcbfm.2015.162. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 94.Tarantini S, Yabluchanksiy A, Fulop GA, Hertelendy P, Valcarcel-Ares MN, Kiss T, Bagwell JM, O'Connor D, Farkas E, Sorond F, Csiszar A, Ungvari Z. Pharmacologically induced impairment of neurovascular coupling responses alters gait coordination in mice. Geroscience 39: 601–614, 2017. [Erratum in Geroscience 2018]. doi: 10.1007/s11357-017-0003-x. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 95.Toth P, Tarantini S, Davila A, Valcarcel-Ares MN, Tucsek Z, Varamini B, Ballabh P, Sonntag WE, Baur JA, Csiszar A, Ungvari Z. Purinergic glio-endothelial coupling during neuronal activity: role of P2Y1 receptors and eNOS in functional hyperemia in the mouse somatosensory cortex. Am J Physiol Heart Circ Physiol 309: H1837–H1845, 2015. doi: 10.1152/ajpheart.00463.2015. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 96.Jacques SL. Optical properties of biological tissues: a review. Phys Med Biol 58: R37–R61, 2013. doi: 10.1088/0031-9155/58/11/R37. [DOI] [PubMed] [Google Scholar]
- 97.Santosa H, Zhai XT, Fishburn F, Huppert T. The NIRS Brain AnalyzIR Toolbox. Algorithms 11: 73, 2018. doi: 10.3390/a11050073. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 98.Li W, Prakash R, Chawla D, Du W, Didion SP, Filosa JA, Zhang Q, Brann DW, Lima VV, Tostes RC, Ergul A. Early effects of high-fat diet on neurovascular function and focal ischemic brain injury. Am J Physiol Regul Integr Comp Physiol 304: R1001–R1008, 2013. doi: 10.1152/ajpregu.00523.2012. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 99.Ashpole NM, Logan S, Yabluchanskiy A, Mitschelen MC, Yan H, Farley JA, Hodges EL, Ungvari Z, Csiszar A, Chen S, Georgescu C, Hubbard GB, Ikeno Y, Sonntag WE. IGF-1 has sexually dimorphic, pleiotropic, and time-dependent effects on healthspan, pathology, and lifespan. Geroscience 39: 129–145, 2017. doi: 10.1007/s11357-017-9971-0. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 100.Fulop GA, Ramirez-Perez FI, Kiss T, Tarantini SV, Ares MN, Toth P, Yabluchanskiy A, Conley SM, Ballabh P, Martinez-Lemus LA, Ungvari Z, Csiszar A. IGF-1 deficiency promotes pathological remodeling of cerebral arteries: a potential mechanism contributing to the pathogenesis of intracerebral hemorrhages in aging. J Gerontol A Biol Sci Med Sci 74: 446–454, 2019. doi: 10.1093/gerona/gly144. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 101.Sonntag WE, Deak F, Ashpole N, Toth P, Csiszar A, Freeman W, Ungvari Z. Insulin-like growth factor-1 in CNS and cerebrovascular aging. Front Aging Neurosci 5: 27, 2013. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 102.Tarantini S, Tucsek Z, Valcarcel-Ares MN, Toth P, Gautam T, Giles CB, Ballabh P, Wei JY, Wren JD, Ashpole NM, Sonntag WE, Ungvari Z, Csiszar A. Circulating IGF-1 deficiency exacerbates hypertension-induced microvascular rarefaction in the mouse hippocampus and retrosplenial cortex: implications for cerebromicrovascular and brain aging. Age (Dordr) 38: 273–289, 2016. doi: 10.1007/s11357-016-9931-0. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 103.Tarantini S, Valcarcel-Ares NM, Yabluchanskiy A, Springo Z, Fulop GA, Ashpole N, Gautam T, Giles CB, Wren JD, Sonntag WE, Csiszar A, Ungvari Z. Insulin-like growth factor 1 deficiency exacerbates hypertension-induced cerebral microhemorrhages in mice, mimicking the aging phenotype. Aging Cell 16: 469–479, 2017. doi: 10.1111/acel.12583. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 104.Toth P, Tucsek Z, Tarantini S, Sosnowska D, Gautam T, Mitschelen M, Koller A, Sonntag WE, Csiszar A, Ungvari Z. IGF-1 deficiency impairs cerebral myogenic autoregulation in hypertensive mice. J Cereb Blood Flow Metab 34: 1887–1897, 2014. doi: 10.1038/jcbfm.2014.156. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 105.Bancu I, Navarro Díaz M, Serra A, Granada M, Lopez D, Romero R, Bonet J. Low insulin-like growth factor-1 level in obesity nephropathy: a new risk factor? PLoS One 11: e0154451, 2016. doi: 10.1371/journal.pone.0154451. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 106.Breese CR, Ingram RL, Sonntag WE. Influence of age and long-term dietary restriction on plasma insulin-like growth factor-1 (IGF-1), IGF-1 gene expression, and IGF-1 binding proteins. J Gerontol 46: B180–B187, 1991. doi: 10.1093/geronj/46.5.B180. [DOI] [PubMed] [Google Scholar]
- 107.Galli G, Pinchera A, Piaggi P, Fierabracci P, Giannetti M, Querci G, Scartabelli G, Manetti L, Ceccarini G, Martinelli S, Di Salvo C, Anselmino M, Bogazzi F, Landi A, Vitti P, Maffei M, Santini F. Serum insulin-like growth factor-1 concentrations are reduced in severely obese women and raise after weight loss induced by laparoscopic adjustable gastric banding. Obes Surg 22: 1276–1280, 2012. doi: 10.1007/s11695-012-0669-1. [DOI] [PubMed] [Google Scholar]
- 108.Bailey-Downs LC, Sosnowska D, Toth P, Mitschelen M, Gautam T, Henthorn JC, Ballabh P, Koller A, Farley JA, Sonntag WE, Csiszar A, Ungvari Z. Growth hormone and IGF-1 deficiency exacerbate high-fat diet-induced endothelial impairment in obese lewis dwarf rats: implications for vascular aging. J Gerontol A Biol Sci Med Sci 67: 553–564, 2012. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 109.Chantler PD, Shrader CD, Tabone LE, d'Audiffret AC, Huseynova K, Brooks SD, Branyan KW, Grogg KA, Frisbee JC. Cerebral cortical microvascular rarefaction in metabolic syndrome is dependent on insulin resistance and loss of nitric oxide bioavailability. Microcirculation 22: 435–445, 2015. [Erratum in Microcirculation 23: 272, 2016]. doi: 10.1111/micc.12209. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 110.Murugesan N, Demarest TG, Madri JA, Pachter JS. Brain regional angiogenic potential at the neurovascular unit during normal aging. Neurobiol Aging 33: 1004.e1001–e1016, 2012. doi: 10.1016/j.neurobiolaging.2011.09.022. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 111.Sonntag WE, Lynch CD, Cooney PT, Hutchins PM. Decreases in cerebral microvasculature with age are associated with the decline in growth hormone and insulin-like growth factor 1. Endocrinology 138: 3515–3520, 1997. doi: 10.1210/endo.138.8.5330. [DOI] [PubMed] [Google Scholar]
- 112.Frisbee JC. Hypertension-independent microvascular rarefaction in the obese Zucker rat model of the metabolic syndrome. Microcirculation 12: 383–392, 2005. doi: 10.1080/10739680590960241. [DOI] [PubMed] [Google Scholar]
- 113.Frisbee JC. Reduced nitric oxide bioavailability contributes to skeletal muscle microvessel rarefaction in the metabolic syndrome. Am J Physiol Regul Integr Comp Physiol 289: R307–R316, 2005. doi: 10.1152/ajpregu.00114.2005. [DOI] [PubMed] [Google Scholar]
- 114.Frisbee JC. Remodeling of the skeletal muscle microcirculation increases resistance to perfusion in obese Zucker rats. Am J Physiol Heart Circ Physiol 285: H104–H111, 2003. doi: 10.1152/ajpheart.00118.2003. [DOI] [PubMed] [Google Scholar]
- 115.Hoffmann JRG, Haendeler J, Aicher A, RöSsig L, Vasa M, Zeiher AM, Dimmeler S. Aging enhances the sensitivity of endothelial cells toward apoptotic stimuli: important role of nitric oxide. Circ Res 89: 709–715, 2001. doi: 10.1161/hh2001.097796. [DOI] [PubMed] [Google Scholar]
- 116.Kobayashi NOBUHIKO, DeLano FrankA, Schmid-SchöNbein GeertW. Oxidative stress promotes endothelial cell apoptosis and loss of microvessels in the spontaneously hypertensive rats. ATVB 25: 2114–2121, 2005. doi: 10.1161/01.ATV.0000178993.13222.f2. [DOI] [PubMed] [Google Scholar]
- 117.Villar-Cheda B, Sousa-Ribeiro D, Rodriguez-Pallares J, Rodriguez-Perez AI, Guerra MJ, Labandeira-Garcia JL. Aging and sedentarism decrease vascularization and VEGF levels in the rat substantia nigra. Implications for Parkinson's disease. J Cereb Blood Flow Metab 29: 230–234, 2009. doi: 10.1038/jcbfm.2008.127. [DOI] [PubMed] [Google Scholar]
- 118.Kiss T, Tarantini S, Csipo T, Balasubramanian P, Nyul-Toth A, Yabluchanskiy A, Wren JD, Garman L, Huffman DM, Csiszar A, Ungvari Z. Circulating anti-geronic factors from heterochonic parabionts promote vascular rejuvenation in aged mice: transcriptional footprint of mitochondrial protection, attenuation of oxidative stress, and rescue of endothelial function by young blood. Geroscience 42: 727–748, 2020. doi: 10.1007/s11357-020-00180-6. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 119.Csiszar A, Gautam T, Sosnowska D, Tarantini S, Banki E, Tucsek Z, Toth P, Losonczy G, Koller A, Reglodi D, Giles CB, Wren JD, Sonntag WE, Ungvari Z. Caloric restriction confers persistent anti-oxidative, pro-angiogenic, and anti-inflammatory effects and promotes anti-aging miRNA expression profile in cerebromicrovascular endothelial cells of aged rats. Am J Physiol Heart Circ Physiol 307: H292–H306, 2014. doi: 10.1152/ajpheart.00307.2014. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 120.Csiszar A, Sosnowska D, Tucsek Z, Gautam T, Toth P, Losonczy G, Colman RJ, Weindruch R, Anderson RM, Sonntag WE, Ungvari Z. Circulating factors induced by caloric restriction in the nonhuman primate Macaca mulatta activate angiogenic processes in endothelial cells. J Gerontol A Biol Sci Med Sci 68: 235–249, 2013. doi: 10.1093/gerona/gls158. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 121.Ungvari Z, Tarantini S, Kiss T, Wren JD, Giles CB, Griffin CT, Murfee WL, Pacher P, Csiszar A. Endothelial dysfunction and angiogenesis impairment in the ageing vasculature. Nat Rev Cardiol 15: 555–565, 2018. doi: 10.1038/s41569-018-0030-z. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 122.Ungvari Z, Tucsek Z, Sosnowska D, Toth P, Gautam T, Podlutsky A, Csiszar A, Losonczy G, Valcarcel-Ares MN, Sonntag WE, Csiszar A. Aging-induced dysregulation of Dicer1-dependent microRNA expression impairs angiogenic capacity of rat cerebromicrovascular endothelial cells. J Gerontol A Biol Sci Med Sci 68: 877–891, 2013. doi: 10.1093/gerona/gls242. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 123.Valcarcel-Ares MN, Gautam T, Warrington JP, Bailey-Downs L, Sosnowska D, de Cabo R, Losonczy G, Sonntag WE, Ungvari Z, Csiszar A. Disruption of Nrf2 signaling impairs angiogenic capacity of endothelial cells: implications for microvascular aging. J Gerontol A Biol Sci Med Sci 67: 821–829, 2012. doi: 10.1093/gerona/glr229. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 124.Baffert F, Thurston G, Rochon-Duck M, Le T, Brekken R, McDonald DM. Age-related changes in vascular endothelial growth factor dependency and angiopoietin-1-induced plasticity of adult blood vessels. Circ Res 94: 984–992, 2004. doi: 10.1161/01.RES.0000125295.43813.1F. [DOI] [PubMed] [Google Scholar]
- 125.Kiss T, Balasubramanian P, Valcarcel-Ares MN, Tarantini S, Yabluchanskiy A, Csipo T, Lipecz A, Reglodi D, Zhang XA, Bari F, Farkas E, Csiszar A, Ungvari Z. Nicotinamide mononucleotide (NMN) treatment attenuates oxidative stress and rescues angiogenic capacity in aged cerebromicrovascular endothelial cells: a potential mechanism for the prevention of vascular cognitive impairment. Geroscience 41: 619–630, 2019. doi: 10.1007/s11357-019-00074-2. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 126.Sweeney MD, Zhao Z, Montagne A, Nelson AR, Zlokovic BV. Blood-brain barrier: from physiology to disease and back. Physiol Rev 99: 21–78, 2019. doi: 10.1152/physrev.00050.2017. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 127.Daneman R, Prat A. The blood-brain barrier. Cold Spring Harb Perspect Biol 7: a020412, 2015. doi: 10.1101/cshperspect.a020412. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 128.Montagne A, Barnes SR, Sweeney MD, Halliday MR, Sagare AP, Zhao Z, Toga AW, Jacobs RE, Liu CY, Amezcua L, Harrington MG, Chui HC, Law M, Zlokovic BV. Blood-brain barrier breakdown in the aging human hippocampus. Neuron 85: 296–302, 2015. doi: 10.1016/j.neuron.2014.12.032. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 129.Nation DA, Sweeney MD, Montagne A, Sagare AP, D'Orazio LM, Pachicano M, Sepehrband F, Nelson AR, Buennagel DP, Harrington MG, Benzinger TLS, Fagan AM, Ringman JM, Schneider LS, Morris JC, Chui HC, Law M, Toga AW, Zlokovic BV. Blood-brain barrier breakdown is an early biomarker of human cognitive dysfunction. Nat Med 25: 270–276, 2019. doi: 10.1038/s41591-018-0297-y. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 130.Stamatovic SM, Johnson AM, Keep RF, Andjelkovic AV. Junctional proteins of the blood-brain barrier: New insights into function and dysfunction. Tissue Barriers 4: e1154641, 2016. doi: 10.1080/21688370.2016.1154641. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 131.Armulik A, Genove G, Mae M, Nisancioglu MH, Wallgard E, Niaudet C, He L, Norlin J, Lindblom P, Strittmatter K, Johansson BR, Betsholtz C. Pericytes regulate the blood-brain barrier. Nature 468: 557–561, 2010. doi: 10.1038/nature09522. [DOI] [PubMed] [Google Scholar]
- 132.Ouyang S, Hsuchou H, Kastin AJ, Wang Y, Yu C, Pan W. Diet-induced obesity suppresses expression of many proteins at the blood-brain barrier. J Cereb Blood Flow Metab 34: 43–51, 2014. doi: 10.1038/jcbfm.2013.166. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 133.Otsuka H, Yanai M, Kobayashi H, Haketa A, Hara M, Sugama K, Kato K, Soma M. High-molecular-weight adiponectin levels in healthy, community-dwelling, elderly Japanese volunteers: a 5-year prospective observational study. Aging Clin Exp Res 30: 791–798, 2018. doi: 10.1007/s40520-017-0840-6. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 134.Tabara Y, Osawa H, Kawamoto R, Tachibana-Iimori R, Yamamoto M, Nakura J, Miki T, Makino H, Kohara K. Reduced high-molecular-weight adiponectin and elevated high-sensitivity C-reactive protein are synergistic risk factors for metabolic syndrome in a large-scale middle-aged to elderly population: the Shimanami Health Promoting Program Study. J Clin Endocrinol Metab 93: 715–722, 2008. doi: 10.1210/jc.2007-0397. [DOI] [PubMed] [Google Scholar]
- 135.Fuller JP, Stavenhagen JB, Teeling JL. New roles for Fc receptors in neurodegeneration-the impact on immunotherapy for Alzheimer's disease. Front Neurosci 8: 235, 2014. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 136.Shigemoto-Mogami Y, Hoshikawa K, Sato K. Activated microglia disrupt the blood-brain barrier and induce chemokines and cytokines in a rat. Front Cell Neurosci 12: 494, 2018. doi: 10.3389/fnhum.2018.00494. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 137.Cope EC, LaMarca EA, Monari PK, Olson LB, Martinez S, Zych AD, Katchur NJ, Gould E. Microglia play an active role in obesity-associated cognitive decline. J Neurosci 38: 8889–8904, 2018. doi: 10.1523/JNEUROSCI.0789-18.2018. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 138.Gerges NZ, Aleisa AM, Alkadhi KA. Impaired long-term potentiation in obese zucker rats: possible involvement of presynaptic mechanism. Neuroscience 120: 535–539, 2003. doi: 10.1016/S0306-4522(03)00297-5. [DOI] [PubMed] [Google Scholar]
- 139.Grillo CA, Piroli GG, Evans AN, Macht VA, Wilson SP, Scott KA, Sakai RR, Mott DD, Lp R. Obesity/hyperleptinemic phenotype adversely affects hippocampal plasticity: effects of dietary restriction. Physiol Behav 104: 235–241, 2011. doi: 10.1016/j.physbeh.2010.10.020. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 140.Hao S, Dey A, Yu X, Stranahan AM. Dietary obesity reversibly induces synaptic stripping by microglia and impairs hippocampal plasticity. Brain Behav Immun 51: 230–239, 2016. doi: 10.1016/j.bbi.2015.08.023. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 141.Hwang LL, Wang CH, Li TL, Chang SD, Lin LC, Chen CP, Chen CT, Liang KC, Ho IK, Yang WS, Chiou LC. Sex differences in high-fat diet-induced obesity, metabolic alterations and learning, and synaptic plasticity deficits in mice. Obesity (Silver Spring) 18: 463–469, 2010. doi: 10.1038/oby.2009.273. [DOI] [PubMed] [Google Scholar]
- 142.Fain JN. Release of interleukins and other inflammatory cytokines by human adipose tissue is enhanced in obesity and primarily due to the nonfat cells. Vitam Horm 74: 443–477, 2006. doi: 10.1016/S0083-6729(06)74018-3. [DOI] [PubMed] [Google Scholar]
- 143.Hocking SL, Wu LE, Guilhaus M, Chisholm DJ, James DE. Intrinsic depot-specific differences in the secretome of adipose tissue, preadipocytes, and adipose tissue-derived microvascular endothelial cells. Diabetes 59: 3008–3016, 2010. doi: 10.2337/db10-0483. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 144.Nishimura S, Manabe I, Nagasaki M, Seo K, Yamashita H, Hosoya Y, Ohsugi MTK, Kadowaki T, Nagai R, Sugiura S. In vivo imaging in mice reveals local cell dynamics and inflammation in obese adipose tissue. J Clin Invest 118: 710–721, 2008. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 145.Schmidt FM, Weschenfelder J, Sander C, Minkwitz J, Thormann J, Chittka T, Mergl R, Kirkby KC, Fasshauer M, Stumvoll M, Holdt LM, Teupser D, Hegerl U, Himmerich H. Inflammatory cytokines in general and central obesity and modulating effects of physical activity. PLoS One 10: e0121971, 2015. doi: 10.1371/journal.pone.0121971. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 146.Surmi BK, Hasty AH. Macrophage infiltration into adipose tissue: initiation, propagation and remodeling. Future Lipidol 3: 545–556, 2008. doi: 10.2217/17460875.3.5.545. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 147.Freitas WR, Oliveira LVF, Perez EA, Ilias EJ, Lottenberg CP, Silva AS, Urbano JJ, Oliveira MC, Vieira RP, Ribeiro-Alves M, Alves VLS, Kassab P, Thuler FR, Malheiros CA. Systemic inflammation in severe obese patients undergoing surgery for obesity and weight-related diseases. Obes Surg 28: 1931–1942, 2018. doi: 10.1007/s11695-017-3104-9. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 148.Niemiro GM, Allen JM, Mailing LJ, Khan NA, Holscher HD, Woods JA, De Lisio M. Effects of endurance exercise training on inflammatory circulating progenitor cell content in lean and obese adults. J Physiol 596: 2811–2822, 2018. doi: 10.1113/JP276023. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 149.Csipo T, Lipecz A, Ashpole NM, Balasubramanian P, Tarantini S. Astrocyte senescence contributes to cognitive decline. Geroscience, 42: 51–55, 2020. doi: 10.1007/s11357-019-00140-9. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 150.Erion JR, Wosiski-Kuhn M, Dey A, Hao S, Davis CL, Pollock NK, Stranahan AM. Obesity elicits interleukin 1-mediated deficits in hippocampal synaptic plasticity. J Neurosci 34: 2618–2631, 2014. doi: 10.1523/JNEUROSCI.4200-13.2014. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 151.Freeman LR, Small BJ, Bickford PC, Umphlet C, Granholm AC. A high-fat/high-cholesterol diet inhibits growth of fetal hippocampal transplants via increased inflammation. Cell Transplant 20: 1499–1514, 2011. doi: 10.3727/096368910X557281. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 152.Kim JD, Yoon NA, Jin S, Diano S. Microglial UCP2 mediates inflammation and obesity induced by high-fat feeding. Cell Metab 30: 952–962, 2019. doi: 10.1016/j.cmet.2019.08.010. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 153.Kirk RA, Kesner RP, Wang LM, Wu Q, Towner RA, Hoffman JM, Morton KA. Lipopolysaccharide exposure in a rat sepsis model results in hippocampal amyloid-beta plaque and phosphorylated tau deposition and corresponding behavioral deficits. Geroscience 41: 467–481, 2019. doi: 10.1007/s11357-019-00089-9. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 154.Knight EM, Martins IV, Gumusgoz S, Allan SM, Lawrence CB. High-fat diet-induced memory impairment in triple-transgenic Alzheimer's disease (3xTgAD) mice is independent of changes in amyloid and tau pathology. Neurobiol Aging 35: 1821–1832, 2014. doi: 10.1016/j.neurobiolaging.2014.02.010. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 155.Lye JJ, Latorre E, Lee BP, Bandinelli S, Holley JE, Gutowski NJ, Ferrucci L, Harries LW. Astrocyte senescence may drive alterations in GFAPalpha, CDKN2A p14(ARF), and TAU3 transcript expression and contribute to cognitive decline. Geroscience 41: 561–573, 2019. doi: 10.1007/s11357-019-00100-3. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 156.McFadden T, Musaus M, Nelsen JL, Martin K, Jones N, Smith P, Kugler H, Jarome TJ. Dysregulation of protein degradation in the hippocampus is associated with impaired spatial memory during the development of obesity. Behav Brain Res 393: 112787, 2020. doi: 10.1016/j.bbr.2020.112787. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 157.Moreno-Navarrete JM, Blasco G, Puig J, Biarnes C, Rivero M, Gich J, Fernandez-Aranda F, Garre-Olmo J, Ramio-Torrenta L, Alberich-Bayarri A, Garcia-Castro F, Pedraza S, Ricart W, Fernandez-Real JM. Neuroinflammation in obesity: circulating lipopolysaccharide-binding protein associates with brain structure and cognitive performance. Int J Obes 41: 1627–1635, 2017. doi: 10.1038/ijo.2017.162. [DOI] [PubMed] [Google Scholar]
- 158.Nerurkar PV, Johns LM, Buesa LM, Kipyakwai G, Volper E, Sato R, Shah P, Feher D, Williams PG, Nerurkar VR. Momordica charantia (bitter melon) attenuates high-fat diet-associated oxidative stress and neuroinflammation. J Neuroinflammation 8: 64, 2011. doi: 10.1186/1742-2094-8-64. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 159.Nuzzo D, Baldassano S, Amato A, Picone P, Galizzi G, Caldara GF, Di Carlo M, Mulè F. Glucagon-like peptide-2 reduces the obesity-associated inflammation in the brain. Neurobiol Dis 121: 296–304, 2019. doi: 10.1016/j.nbd.2018.10.012. [DOI] [PubMed] [Google Scholar]
- 160.Samara A, Murphy T, Strain J, Rutlin J, Sun P, Neyman O, Sreevalsan N, Shimony JS, Ances BM, Song SK, Hershey T, Eisenstein SA. Neuroinflammation and white matter alterations in obesity assessed by diffusion basis spectrum imaging. Front Hum Neurosci 13: 464, 2019. doi: 10.3389/fnins.2019.00464. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 161.Sobesky JL, Barrientos RM, De May HS, Thompson BM, Weber MD, Watkins LR, Maier SF. High-fat diet consumption disrupts memory and primes elevations in hippocampal IL-1beta, an effect that can be prevented with dietary reversal or IL-1 receptor antagonism. Brain Behav Immun 42: 22–32, 2014. doi: 10.1016/j.bbi.2014.06.017. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 162.Toedebusch CM, Garcia VB, Snyder JC, Jones MR, Schulz DJ, Johnson GC, Villalon E, Coates JR, Garcia ML. Lumbar spinal cord microglia exhibited increased activation in aging dogs compared with young adult dogs. Geroscience 42: 169–182, 2020. doi: 10.1007/s11357-019-00133-8. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 163.Towner RA, Saunders D, Smith N, Gulej R, McKenzie T, Lawrence B, Morton KA. Anti-inflammatory agent, OKN-007, reverses long-term neuroinflammatory responses in a rat encephalopathy model as assessed by multi-parametric MRI: implications for aging-associated neuroinflammation. Geroscience 41: 483–494, 2019. doi: 10.1007/s11357-019-00094-y. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 164.Casaletto KB, Elahi FM, Staffaroni AM, Walters S, Contreras WR, Wolf A, Dubal D, Miller B, Yaffe K, Kramer JH. Cognitive aging is not created equally: differentiating unique cognitive phenotypes in "normal" adults. Neurobiol Aging 77: 13–19, 2019. doi: 10.1016/j.neurobiolaging.2019.01.007. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 165.Chi GC, Fitzpatrick AL, Sharma M, Jenny NS, Lopez OL, St D. Inflammatory biomarkers predict domain-specific cognitive decline in older adults. J Gerontol A Biol Sci Med Sci 72: 796–803, 2017. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 166.Harrison SL, de Craen AJ, Kerse N, Teh R, Granic A, Davies K, Wesnes KA, den Elzen WP, Gussekloo J, Kirkwood TB, Robinson L, Jagger C, Siervo M, Bc S. Predicting risk of cognitive decline in very old adults using three models: the Framingham stroke risk profile; the cardiovascular risk factors, aging, and dementia model; and oxi-inflammatory biomarkers. J Am Geriatr Soc 65: 381–389, 2017. doi: 10.1111/jgs.14532. [DOI] [PubMed] [Google Scholar]
- 167.Lai KSP, Liu CS, Rau A, Lanctot KL, Kohler CA, Pakosh M, Carvalho AF, Herrmann N. Peripheral inflammatory markers in Alzheimer's disease: a systematic review and meta-analysis of 175 studies. J Neurol Neurosurg Psychiatry 88: 876–882, 2017. doi: 10.1136/jnnp-2017-316201. [DOI] [PubMed] [Google Scholar]
- 168.Ng A, Tam WW, Zhang MW, Ho CS, Husain SF, McIntyre RS, Ho RC. IL-1beta, IL-6, TNF- alpha and CRP in elderly patients with depression or Alzheimer's disease: Systematic review and meta. Sci Rep 8: 12050, 2018. doi: 10.1038/s41598-018-30487-6. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 169.Shen XN, Niu LD, Wang YJ, Cao XP, Liu Q, Tan L, Zhang C, Jt Y. Inflammatory markers in Alzheimer's disease and mild cognitive impairment: a meta-analysis and systematic review of 170 studies. J Neurol Neurosurg Psychiatry 90: 590–598, 2019. doi: 10.1136/jnnp-2018-319148. [DOI] [PubMed] [Google Scholar]
- 170.Crewe C, An YA, Scherer PE. The ominous triad of adipose tissue dysfunction: inflammation, fibrosis, and impaired angiogenesis. J Clin Invest 127: 74–82, 2017. doi: 10.1172/JCI88883. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 171.Castoldi A, Naffah de Souza C, Câmara NO, Moraes-Vieira PM. The macrophage switch in obesity development. Front Immunol 6: 637, 2015. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 172.Bailey-Downs LC, Tucsek Z, Toth P, Sosnowska D, Gautam T, Sonntag WE, Csiszar A, Ungvari Z. Aging exacerbates obesity-induced oxidative stress and inflammation in perivascular adipose tissue in mice: a paracrine mechanism contributing to vascular redox dysregulation and inflammation. J Gerontol A Biol Sci Med Sci 68: 780–792, 2013. doi: 10.1093/gerona/gls238. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 173.Starr ME, Evers BM, Saito H. Age-associated increase in cytokine production during systemic inflammation: adipose tissue as a major source of IL-6. J Gerontol A Biol Sci Med Sci 64: 723–730, 2009. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 174.Wu D, Ren Z, Pae M, Guo W, Cui X, Merrill AH, Meydani SN. Aging up-regulates expression of inflammatory mediators in mouse adipose tissue. J Immunol 179: 4829–4839, 2007. doi: 10.4049/jimmunol.179.7.4829. [DOI] [PubMed] [Google Scholar]
- 175.Lin T, Liu GA, Perez E, Rainer RD, Febo M, Cruz-Almeida Y, Ebner NC. Systemic inflammation mediates age-related cognitive deficits. Front Aging Neurosci 10: 236, 2018. doi: 10.3389/fnagi.2018.00236. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 176.Zhou T, Zhao L, Zhan R, He Q, Tong Y, Tian X, Wang H, Zhang T, Fu Y, Sun Y, Xu F, Guo X, Fan D, Han H, Chui D. Blood-brain barrier dysfunction in mice induced by lipopolysaccharide is attenuated by dapsone. Biochem Biophys Res Commun 453: 419–424, 2014. doi: 10.1016/j.bbrc.2014.09.093. [DOI] [PubMed] [Google Scholar]
- 177.Cardoso FL, Kittel A, Veszelka S, Palmela I, Toth A, Brites D, Deli MA, Brito MA. Exposure to lipopolysaccharide and/or unconjugated bilirubin impair the integrity and function of brain microvascular endothelial cells. PLoS One 7: e35919, 2012. doi: 10.1371/journal.pone.0035919. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 178.Wiesinger A, Peters W, Chappell D, Kentrup D, Reuter S, Pavenstädt H, Oberleithner H, Kümpers P. Nanomechanics of the endothelial glycocalyx in experimental sepsis. PLoS One 8: e80905, 2013. doi: 10.1371/journal.pone.0080905. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 179.Banks WA, Kastin AJ, Gutierrez EG. Penetration of interleukin-6 across the murine blood-brain barrier. Neurosci Lett 179: 53–56, 1994. doi: 10.1016/0304-3940(94)90933-4. [DOI] [PubMed] [Google Scholar]
- 180.Banks WA, Ortiz L, Plotkin SR, Kastin AJ. Human interleukin (IL) 1 alpha, murine IL-1 alpha and murine IL-1 beta are transported from blood to brain in the mouse by a shared saturable mechanism. J Pharmacol Exp Ther 259: 988–996, 1991. [PubMed] [Google Scholar]
- 181.Gutierrez EG, Banks WA, Kastin AJ. Murine tumor necrosis factor alpha is transported from blood to brain in the mouse. J Neuroimmunol 47: 169–176, 1993. doi: 10.1016/0165-5728(93)90027-V. [DOI] [PubMed] [Google Scholar]
- 182.Opatrilova R, Caprnda M, Kubatka P, Valentova V, Uramova S, Nosal V, Gaspar L, Zachar L, Mozos I, Petrovic D, Dragasek J, Filipova S, Busselberg D, Zulli A, Rodrigo L, Kruzliak P, Krasnik V. Adipokines in neurovascular diseases. Biomed Pharmacother 98: 424–432, 2018. doi: 10.1016/j.biopha.2017.12.074. [DOI] [PubMed] [Google Scholar]
- 183.Banks WA, Kastin AJ, Huang W, Jaspan JB, Maness LM. Leptin enters the brain by a saturable system independent of insulin. Peptides 17: 305–311, 1996. doi: 10.1016/0196-9781(96)00025-3. [DOI] [PubMed] [Google Scholar]
- 184.Di Spiezio A, Sandin ES, Dore R, Müller-Fielitz H, Storck SE, Bernau M, Mier W, Oster H, Jöhren O, Pietrzik CU, Lehnert H, Schwaninger M. The LepR-mediated leptin transport across brain barriers controls food reward. Mol Metab 8: 13–22, 2018. doi: 10.1016/j.molmet.2017.12.001. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 185.Teixeira TM, da Costa DC, Resende AC, Soulage CO, Bezerra FF, Daleprane JB. Activation of Nrf2-antioxidant signaling by 1,25-dihydroxycholecalciferol prevents leptin-induced oxidative stress and inflammation in human endothelial cells. J Nutr 147: 506–513, 2017. doi: 10.3945/jn.116.239475. [DOI] [PubMed] [Google Scholar]
- 186.Faulkner JL, Kennard S, Huby AC, Antonova G, Lu Q, Jaffe IZ, Patel VS, Fulton DJR, Belin de Chantemele EJ. Progesterone predisposes females to obesity-associated leptin-mediated endothelial dysfunction via upregulating endothelial MR (mineralocorticoid receptor) expression. Hypertension 74: 678–686, 2019. doi: 10.1161/HYPERTENSIONAHA.119.12802. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 187.Huby AC, Antonova G, Groenendyk J, Gomez-Sanchez CE, Bollag WB, Filosa JA, de Chantemele BELIN, Adipocyte EJ. Derived hormone leptin is a direct regulator of aldosterone secretion, which promotes endothelial dysfunction and cardiac fibrosis. Circulation 132: 2134–2145, 2015. doi: 10.1161/CIRCULATIONAHA.115.018226. [DOI] [PubMed] [Google Scholar]
- 188.Huby AC, Otvos L, Jr, Belin de Chantemele , EJ. Leptin induces hypertension and endothelial dysfunction via aldosterone-dependent mechanisms in obese female mice. Hypertension 67: 1020–1028, 2016. doi: 10.1161/HYPERTENSIONAHA.115.06642. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 189.Knudson JD, Dincer UD, Zhang C, Swafford AN, Jr,Koshida R, Picchi A, Focardi M, Dick GM, Tune JD. Leptin receptors are expressed in coronary arteries, and hyperleptinemia causes significant coronary endothelial dysfunction. Am J Physiol Heart Circ Physiol 289: H48–H56, 2005. doi: 10.1152/ajpheart.01159.2004. [DOI] [PubMed] [Google Scholar]
- 190.Korda M, Kubant R, Patton S, Malinski T. Leptin-induced endothelial dysfunction in obesity. Am J Physiol Heart Circ Physiol 295: H1514–H1521, 2008. doi: 10.1152/ajpheart.00479.2008. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 191.Beltowski J. Leptin and atherosclerosis. Atherosclerosis 189: 47–60, 2006. doi: 10.1016/j.atherosclerosis.2006.03.003. [DOI] [PubMed] [Google Scholar]
- 192.Li XL, Aou S, Oomura Y, Hori N, Fukunaga K, Hori T. Impairment of long-term potentiation and spatial memory in leptin receptor-deficient rodents. Neuroscience 113: 607–615, 2002. doi: 10.1016/S0306-4522(02)00162-8. [DOI] [PubMed] [Google Scholar]
- 193.Hubert A, Bochenek ML, Schütz E, Gogiraju R, Münzel T, Schäfer K. Selective deletion of leptin signaling in endothelial cells enhances neointima formation and phenocopies the vascular effects of diet-induced obesity in mice. Arterioscler Thromb Vasc Biol 37: 1683–1697, 2017. doi: 10.1161/ATVBAHA.117.309798. [DOI] [PubMed] [Google Scholar]
- 194.de Git KCG, Adan RAH. Leptin resistance in diet-induced obesity: the role of hypothalamic inflammation. Obes Rev 16: 207–224, 2015. . doi: 10.1111/obr.12243. [DOI] [PubMed] [Google Scholar]
- 195.Myers MG, Heymsfield SB, Haft C, Kahn BB, Laughlin M, Leibel RL, Tschöp MH, Yanovski JA. Challenges and opportunities of defining clinical leptin resistance. Cell Metab 15: 150–156, 2012. doi: 10.1016/j.cmet.2012.01.002. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 196.Sasaki T. Age-associated weight gain, leptin, and SIRT1: a possible role for hypothalamic SIRT1 in the prevention of weight gain and aging through modulation of leptin sensitivity. Front Endocrinol (Lausanne) 6: 109, 2015. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 197.Steppan CM, Bailey ST, Bhat S, Brown EJ, Banerjee RR, Wright CM, Patel HR, Ahima RS, Lazar MA. The hormone resistin links obesity to diabetes. Nature 409: 307–312, 2001. doi: 10.1038/35053000. [DOI] [PubMed] [Google Scholar]
- 198.Rubio-Guerra AF, Cabrera-Miranda LJ, Vargas-Robles H, Maceda-Serrano A, Lozano-Nuevo JJ, Escalante-Acosta BA. Correlation between levels of circulating adipokines and adiponectin/resistin index with carotid intima-media thickness in hypertensive type 2 diabetic patients. Cardiology 125: 150–153, 2013. doi: 10.1159/000348651. [DOI] [PubMed] [Google Scholar]
- 199.Weikert C, Westphal S, Berger K, Dierkes J, MöHlig M, Spranger J, Rimm EB, Willich SN, Boeing H, Pischon T. Plasma resistin levels and risk of myocardial infarction and ischemic stroke. J Clin Endocrinol Metab 93: 2647–2653, 2008. doi: 10.1210/jc.2007-2735. [DOI] [PubMed] [Google Scholar]
- 200.Bouziana S, Tziomalos K, Goulas A, Vyzantiadis TA, Papadopoulou M, Panderi A, Etaatzitolios A. Effects of major adipokines and the -420 C > G resistin gene polymorphism on the long-term outcome of patients with acute ischemic stroke. Int J Neurosci 129: 978–985, 2019. doi: 10.1080/00207454.2019.1596906. [DOI] [PubMed] [Google Scholar]
- 201.Dong XL, Xu SJ, Zhang L, Zhang XQ, Liu T, Gao QY, Qian QQ, Sun BL, Yang MF. Serum resistin levels may contribute to an increased risk of acute cerebral infarction. Mol Neurobiol 54: 1919–1926, 2017. doi: 10.1007/s12035-016-9751-3. [DOI] [PubMed] [Google Scholar]
- 202.Kapłon‐Cieślicka A., Tymińska A., Rosiak M., Ozierański K., Peller M., Eyileten C., Kondracka A., Pordzik J., Mirowska‐Guzel D., Opolski G., Postuła M., Filipiak K. Resistin is a prognostic factor for death in type 2 diabetes. Diabetes Metab Res Rev 35: e3098, 2018. doi: 10.1002/dmrr.3098. [DOI] [PubMed] [Google Scholar]
- 203.Kochanowski J, Grudniak M, Baranowska-Bik A, Wolinska-Witort E, Kalisz M, Baranowska B, Bik W. Resistin levels in women with ischemic stroke. Neuro Endocrinol Lett 33: 603–607, 2012. [PubMed] [Google Scholar]
- 204.Rajpathak SN, Kaplan RC, Wassertheil-Smoller S, Cushman M, Rohan TE, McGinn AP, Wang T, Strickler HD, Scherer PE, Mackey R, Curb D, Ho GY. Resistin, but not adiponectin and leptin, is associated with the risk of ischemic stroke among postmenopausal women: results from the Women's Health Initiative. Stroke 42: 1813–1820, 2011. doi: 10.1161/STROKEAHA.110.607853. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 205.Xiaoying L, Li T, Yu S, Jiusheng J, Jilin Z, Jiayi W, Dongxin L, Wengang F, Xinyue Z, Hao Y, Yuhua C, Deshu S. Resistin-inhibited neural stem cell-derived astrocyte differentiation contributes to permeability destruction of the blood-brain barrier. Neurochem Res 44: 905–916, 2019. doi: 10.1007/s11064-019-02726-3. [DOI] [PubMed] [Google Scholar]
- 206.Ramirez JL, Khetani SA, Zahner GJ, Spaulding KA, Schaller MS, Gasper WJ, Hills NK, Schafer AL, Grenon SM. Serum resistin is associated with impaired endothelial function and a higher rate of adverse cardiac events in patients with peripheral artery disease. J Vasc Surg 69: 497–506, 2019. doi: 10.1016/j.jvs.2018.05.251. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 207.Samsamshariat SZA, Sakhaei F, Salehizadeh L, Keshvari M, Asgary S. Relationship between resistin, endothelin-1, and flow-mediated dilation in patient with and without metabolic syndrome. Adv Biomed Res 8: 16, 2019. doi: 10.4103/abr.abr_126_18. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 208.Miralbell J, Lopez-Cancio E, Lopez-Oloriz J, Arenillas JF, Barrios M, Soriano-Raya JJ, Galan A, Caceres C, Alzamora M, Pera G, Toran P, Davalos A, Mataro M. Cognitive patterns in relation to biomarkers of cerebrovascular disease and vascular risk factors. Cerebrovasc Dis 36: 98–105, 2013. doi: 10.1159/000352059. [DOI] [PubMed] [Google Scholar]
- 209.Hotta K, Funahashi T, Bodkin NL, Ortmeyer HK, Arita Y, Hansen BC, Matsuzawa Y. Circulating concentrations of the adipocyte protein adiponectin are decreased in parallel with reduced insulin sensitivity during the progression to type 2 diabetes in rhesus monkeys. Diabetes 50: 1126–1133, 2001. doi: 10.2337/diabetes.50.5.1126. [DOI] [PubMed] [Google Scholar]
- 210.Kubota N, Terauchi Y, Yamauchi T, Kubota T, Moroi M, Matsui J, Eto K, Yamashita T, Kamon J, Satoh H, Yano W, Froguel P, Nagai R, Kimura S, Kadowaki T, Noda T. Disruption of adiponectin causes insulin resistance and neointimal formation. J Biol Chem 277: 25863–25866, 2002. doi: 10.1074/jbc.C200251200. [DOI] [PubMed] [Google Scholar]
- 211.Yamauchi T, Kamon J, Minokoshi Y, Ito Y, Waki H, Uchida S, Yamashita S, Noda M, Kita S, Ueki K, Eto K, Akanuma Y, Froguel P, Foufelle F, Ferre P, Carling D, Kimura S, Nagai R, Kahn BB, Kadowaki T. Adiponectin stimulates glucose utilization and fatty-acid oxidation by activating AMP-activated protein kinase. Nat Med 8: 1288–1295, 2002. doi: 10.1038/nm788. [DOI] [PubMed] [Google Scholar]
- 212.Yamauchi T, Kamon J, Waki H, Imai Y, Shimozawa N, Hioki K, Uchida S, Ito Y, Takakuwa K, Matsui J, Takata M, Eto K, Terauchi Y, Komeda K, Tsunoda M, Murakami K, Ohnishi Y, Naitoh T, Yamamura K, Ueyama Y, Froguel P, Kimura S, Nagai R, Kadowaki T. Globular adiponectin protected ob/ob mice from diabetes and ApoE-deficient mice from atherosclerosis. J Biol Chem 278: 2461–2468, 2003. doi: 10.1074/jbc.M209033200. [DOI] [PubMed] [Google Scholar]
- 213.Yamauchi T, Kamon J, Waki H, Terauchi Y, Kubota N, Hara K, Mori Y, Ide T, Murakami K, Tsuboyama-Kasaoka N, Ezaki O, Akanuma Y, Gavrilova O, Vinson C, Reitman ML, Kagechika H, Shudo K, Yoda M, Nakano Y, Tobe K, Nagai R, Kimura S, Tomita M, Froguel P, Kadowaki T. The fat-derived hormone adiponectin reverses insulin resistance associated with both lipoatrophy and obesity. Nat Med 7: 941–946, 2001. doi: 10.1038/90984. [DOI] [PubMed] [Google Scholar]
- 214.Okada-Iwabu M, Yamauchi T, Iwabu M, Honma T, Hamagami K, Matsuda K, Yamaguchi M, Tanabe H, Kimura-Someya T, Shirouzu M, Ogata H, Tokuyama K, Ueki K, Nagano T, Tanaka A, Yokoyama S, Kadowaki T. A small-molecule AdipoR agonist for type 2 diabetes and short life in obesity. Nature 503: 493–499, 2013. doi: 10.1038/nature12656. [DOI] [PubMed] [Google Scholar]
- 215.Miller KN, Burhans MS, Clark JP, Howell PR, Polewski MA, DeMuth TM, Eliceiri KW, Lindstrom MJ, Ntambi JM, Anderson RM. Aging and caloric restriction impact adipose tissue, adiponectin, and circulating lipids. Aging Cell 16: 497–507, 2017. doi: 10.1111/acel.12575. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 216.Gilbert T, Roche S, Blond E, Bar JY, Drai J, Cuerq C, Haution-Bitker M, Ecochard R, Bonnefoy M. Association between peripheral leptin and adiponectin levels and cognitive decline in patients with neurocognitive disorders ≥65 years. JAD 66: 1255–1264, 2018. doi: 10.3233/JAD-180533. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 217.Ding Q, Ash C, Mracek T, Merry B, Bing C. Caloric restriction increases adiponectin expression by adipose tissue and prevents the inhibitory effect of insulin on circulating adiponectin in rats. J Nutr Biochem 23: 867–874, 2012. doi: 10.1016/j.jnutbio.2011.04.011. [DOI] [PubMed] [Google Scholar]
- 218.Kondo M, Shibata R, Miura R, Shimano M, Kondo K, Li P, Ohashi T, Kihara S, Maeda N, Walsh K, Ouchi N, Murohara T. Caloric restriction stimulates revascularization in response to ischemia via adiponectin-mediated activation of endothelial nitric-oxide synthase. J Biol Chem 284: 1718–1724, 2009. doi: 10.1074/jbc.M805301200. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 219.Niemann B, Silber RE, Rohrbach S. Age-specific effects of short- and long-term caloric restriction on the expression of adiponectin and adiponectin receptors: influence of intensity of food restriction. Exp Gerontol 43: 706–713, 2008. doi: 10.1016/j.exger.2008.02.008. [DOI] [PubMed] [Google Scholar]
- 220.Shinmura K, Tamaki K, Saito K, Nakano Y, Tobe T, Bolli R. Cardioprotective effects of short-term caloric restriction are mediated by adiponectin via activation of AMP-activated protein kinase. Circulation 116: 2809–2817, 2007. doi: 10.1161/CIRCULATIONAHA.107.725697. [DOI] [PubMed] [Google Scholar]
- 221.Xu XJ, Babo E, Qin F, Croteau D, Colucci WS. Short-term caloric restriction in db/db mice improves myocardial function and increases high molecular weight (HMW) adiponectin. IJC Metab Endocr 13: 28–34, 2016. doi: 10.1016/j.ijcme.2016.10.002. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 222.Zhu M, Miura J, Lu LX, Bernier M, DeCabo R, Lane MA, Roth GS, Dk I. Circulating adiponectin levels increase in rats on caloric restriction: the potential for insulin sensitization. Exp Gerontol 39: 1049–1059, 2004. doi: 10.1016/j.exger.2004.03.024. [DOI] [PubMed] [Google Scholar]
- 223.Antoniades C, Antonopoulos AS, Tousoulis D, Stefanadis C. Adiponectin: from obesity to cardiovascular disease. Obes Rev 10: 269–279, 2009. doi: 10.1111/j.1467-789X.2009.00571.x. [DOI] [PubMed] [Google Scholar]
- 224.Bloemer J, Pinky PD, Smith WD, Bhattacharya D, Chauhan A, Govindarajulu M, Hong H, Dhanasekaran M, Judd R, Amin RH, Reed MN, Suppiramaniam V. Adiponectin knockout mice display cognitive and synaptic deficits. Front Endocrinol (Lausanne) 10: 819, 2019. doi: 10.3389/fendo.2019.00819. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 225.Weisz F, Piccinin S, Mango D, Ngomba RT, Mercuri NB, Nicoletti F, Nistico R. The role of adiponectin receptors in the regulation of synaptic transmission in the hippocampus. Synapse 71: e21964, 2017. doi: 10.1002/syn.21964. [DOI] [PubMed] [Google Scholar]
- 226.Zhang D, Wang X, Wang B, Garza JC, Fang X, Wang J, Scherer PE, Brenner R, Zhang W, Lu XY. Adiponectin regulates contextual fear extinction and intrinsic excitability of dentate gyrus granule neurons through AdipoR2 receptors. Mol Psychiatry 22: 1044–1055, 2017. doi: 10.1038/mp.2016.58. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 227.Zheng J, Sun Z, Liang F, Xu W, Lu J, Shi L, Shao A, Yu J, Zhang J. AdipoRon attenuates neuroinflammation after intracerebral hemorrhage through AdipoR1-AMPK pathway. Neuroscience 412: 116–130, 2019. doi: 10.1016/j.neuroscience.2019.05.060. [DOI] [PubMed] [Google Scholar]
- 228.Motoshima H, Wu X, Mahadev K, Goldstein BJ. Adiponectin suppresses proliferation and superoxide generation and enhances eNOS activity in endothelial cells treated with oxidized LDL. Biochem Biophys Res Commun 315: 264–271, 2004. doi: 10.1016/j.bbrc.2004.01.049. [DOI] [PubMed] [Google Scholar]
- 229.Ouedraogo R, Wu X, Xu SQ, Fuchsel L, Motoshima H, Mahadev K, Hough K, Scalia R, Bj G. Adiponectin suppression of high-glucose-induced reactive oxygen species in vascular endothelial cells: evidence for involvement of a cAMP signaling pathway. Diabetes 55: 1840–1846, 2006. doi: 10.2337/db05-1174. [DOI] [PubMed] [Google Scholar]
- 230.Chen H, Montagnani M, Funahashi T, Shimomura I, Quon MJ. Adiponectin stimulates production of nitric oxide in vascular endothelial cells. J Biol Chem 278: 45021–45026, 2003. doi: 10.1074/jbc.M307878200. [DOI] [PubMed] [Google Scholar]
- 231.Tanigawa T, Shibata R, Ouchi N, Kondo K, Ishii M, Katahira N, Kambara T, Inoue Y, Takahashi R, Ikeda N, Kihara S, Ueda H, Murohara T. Adiponectin deficiency exacerbates age-related hearing impairment. Cell Death Dis 5: e1189–e1189, 2014. doi: 10.1038/cddis.2014.140. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 232.Choi SR, Lim JH, Kim MY, Kim EN, Kim Y, Choi BS, Kim YS, Kim HW, Lim KM, Kim MJ, Park CW. Adiponectin receptor agonist AdipoRon decreased ceramide, and lipotoxicity, and ameliorated diabetic nephropathy. Metabolism 85: 348–360, 2018. doi: 10.1016/j.metabol.2018.02.004. [DOI] [PubMed] [Google Scholar]
- 233.Spranger J, Verma S, Gohring I, Bobbert T, Seifert J, Sindler AL, Pfeiffer A, Hileman SM, Tschop M, Banks WA. Adiponectin does not cross the blood-brain barrier but modifies cytokine expression of brain endothelial cells. Diabetes 55: 141–147, 2006. doi: 10.2337/diabetes.55.01.06.db05-1077. [DOI] [PubMed] [Google Scholar]
- 234.Csipo T, Fulop GA, Lipecz A, Tarantini S, Kiss T, Balasubramanian P, Csiszar A, Ungvari Z, Yabluchanskiy A. Short-term weight loss reverses obesity-induced microvascular endothelial dysfunction. Geroscience 40: 337–346, 2018. doi: 10.1007/s11357-018-0028-9. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 235.Pasqualini L, Schillaci G, Innocente S, Pucci G, Coscia F, Siepi D, Lupattelli G, Ciuffetti G, Mannarino E. Lifestyle intervention improves microvascular reactivity and increases serum adiponectin in overweight hypertensive patients. Nutr Metab Cardiovasc Dis 20: 87–92, 2010. doi: 10.1016/j.numecd.2009.03.002. [DOI] [PubMed] [Google Scholar]
- 236.Tabak AG, Herder C, Rathmann W, Brunner EJ, Kivimaki M. Prediabetes: a high-risk state for diabetes development. Lancet 379: 2279–2290, 2012. doi: 10.1016/S0140-6736(12)60283-9. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 237.Cooper C, Sommerlad A, Lyketsos CG, Livingston G. Modifiable predictors of dementia in mild cognitive impairment: a systematic review and meta-analysis. Am J Psychiatry 172: 323–334, 2015. doi: 10.1176/appi.ajp.2014.14070878. [DOI] [PubMed] [Google Scholar]
- 238.Tuligenga RH, Dugravot A, Tabak AG, Elbaz A, Brunner EJ, Kivimaki M, Singh-Manoux A. Midlife type 2 diabetes and poor glycaemic control as risk factors for cognitive decline in early old age: a post-hoc analysis of the Whitehall II cohort study. Lancet Diabetes Endocrinol 2: 228–235, 2014. doi: 10.1016/S2213-8587(13)70192-X. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 239.Xu W, Caracciolo B, Wang HX, Winblad B, Bäckman L, Qiu C, Fratiglioni L. Accelerated progression from mild cognitive impairment to dementia in people with diabetes. Diabetes 59: 2928–2935, 2010. doi: 10.2337/db10-0539. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 240.Ma F, Wu T, Miao R, Xiao YY, Zhang W, Huang G. Conversion of mild cognitive impairment to dementia among subjects with diabetes: a population-based study of incidence and risk factors with five years of follow-up. JAD 43: 1441–1449, 2014. doi: 10.3233/JAD-141566. [DOI] [PubMed] [Google Scholar]
- 241.Fu Z, Wu J, Nesil T, Li MD, Aylor KW, Liu Z. Long-term high-fat diet induces hippocampal microvascular insulin resistance and cognitive dysfunction. Am J Physiol Endocrinol Metab 312: E89–E97, 2017. doi: 10.1152/ajpendo.00297.2016. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 242.Hussain Y, Jain SK, Samaiya PK. Short-term westernized (HFFD) diet fed in adolescent rats: Effect on glucose homeostasis, hippocampal insulin signaling, apoptosis and related cognitive and recognition memory function. Behav Brain Res 361: 113–121, 2019. doi: 10.1016/j.bbr.2018.12.042. [DOI] [PubMed] [Google Scholar]
- 243.Grillo CA, Piroli GG, Lawrence RC, Wrighten SA, Green AJ, Wilson SP, Sakai RR, Kelly SJ, Wilson MA, Mott DD, Reagan LP. Hippocampal insulin resistance impairs spatial learning and synaptic plasticity. Diabetes 64: 3927–3936, 2015. doi: 10.2337/db15-0596. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 244.Begg DP, Mul JD, Liu M, Reedy BM, D'Alessio DA, Seeley RJ, Woods SC. Reversal of diet-induced obesity increases insulin transport into cerebrospinal fluid and restores sensitivity to the anorexic action of central insulin in male rats. Endocrinology 154: 1047–1054, 2013. doi: 10.1210/en.2012-1929. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 245.Kern W, Benedict C, Schultes B, Plohr F, Moser A, Born J, Fehm HL, Hallschmid M. Low cerebrospinal fluid insulin levels in obese humans. Diabetologia 49: 2790–2792, 2006. doi: 10.1007/s00125-006-0409-y. [DOI] [PubMed] [Google Scholar]
- 246.Dimmeler S, Fleming I, Fisslthaler B, Hermann C, Busse R, Zeiher AM. Activation of nitric oxide synthase in endothelial cells by Akt-dependent phosphorylation. Nature 399: 601–605, 1999. doi: 10.1038/21224. [DOI] [PubMed] [Google Scholar]
- 247.Muniyappa R, Montagnani M, Koh KK, Quon MJ. Cardiovascular actions of insulin. Endocr Rev 28: 463–491, 2007. doi: 10.1210/er.2007-0006. [DOI] [PubMed] [Google Scholar]
- 248.Brooks SD, DeVallance E, d'Audiffret AC, Frisbee SJ, Tabone LE, Shrader CD, Frisbee JC, Chantler PD. Metabolic syndrome impairs reactivity and wall mechanics of cerebral resistance arteries in obese Zucker rats. Am J Physiol Heart Circ Physiol 309: H1846–H1859, 2015. doi: 10.1152/ajpheart.00691.2015. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 249.Takechi R, Lam V, Brook E, Giles C, Fimognari N, Mooranian A, Al-Salami H, Coulson SH, Nesbit M, Mamo JCL. Blood-brain barrier dysfunction precedes cognitive decline and neurodegeneration in diabetic insulin resistant mouse model: an implication for causal link. Front Aging Neurosci 9: 399, 2017. doi: 10.3389/fnagi.2017.00399. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 250.Yamamoto M, Guo DH, Hernandez CM, Stranahan AM. Endothelial Adora2a activation promotes blood-brain barrier breakdown and cognitive impairment in mice with diet-induced insulin resistance. J Neurosci 39: 4179–4192, 2019. doi: 10.1523/JNEUROSCI.2506-18.2019. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 251.Buford TW, Carter CS, VanDerPol WJ, Chen D, Lefkowitz EJ, Eipers P, Morrow CD, Bamman MM. Composition and richness of the serum microbiome differ by age and link to systemic inflammation. Geroscience 40: 257–268, 2018. doi: 10.1007/s11357-018-0026-y. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 252.Lim MY, Song EJ, Kang KS, Nam YD. Age-related compositional and functional changes in micro-pig gut microbiome. Geroscience 41: 935–944, 2019. doi: 10.1007/s11357-019-00121-y. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 253.Singh H, Torralba MG, Moncera KJ, DiLello L, Petrini J, Nelson KE, Pieper R. Gastro-intestinal and oral microbiome signatures associated with healthy aging. Geroscience 41: 907–921, 2019. doi: 10.1007/s11357-019-00098-8. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 254.Magnusson KR, Hauck L, Jeffrey BM, Elias V, Humphrey A, Nath R, Perrone A, Bermudez LE. Relationships between diet-related changes in the gut microbiome and cognitive flexibility. Neuroscience 300: 128–140, 2015. doi: 10.1016/j.neuroscience.2015.05.016. [DOI] [PubMed] [Google Scholar]
- 255.Rondanelli M, Giacosa A, Faliva MA, Perna S, Allieri F, Castellazzi AM. Review on microbiota and effectiveness of probiotics use in older. WJCC 3: 156–162, 2015. doi: 10.12998/wjcc.v3.i2.156. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 256.Ticinesi A, Tana C, Nouvenne A, Prati B, Lauretani F, Meschi T. Gut microbiota, cognitive frailty and dementia in older individuals: a systematic review. Clin Interv Aging 13: 1497–1511, 2018. doi: 10.2147/CIA.S139163. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 257.Cani PD, Bibiloni R, Knauf C, Waget A, Neyrinck AM, Delzenne NM, Burcelin R. Changes in gut microbiota control metabolic endotoxemia-induced inflammation in high-fat diet-induced obesity and diabetes in mice. Diabetes 57: 1470–1481, 2008. doi: 10.2337/db07-1403. [DOI] [PubMed] [Google Scholar]
- 258.Saji N, Niida S, Murotani K, Hisada T, Tsuduki T, Sugimoto T, Kimura A, Toba K, Sakurai T. Analysis of the relationship between the gut microbiome and dementia: a cross-sectional study conducted in. Sci Rep 9: 1008, 2019. doi: 10.1038/s41598-018-38218-7. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 259.Manderino L, Carroll I, Azcarate-Peril MA, Rochette A, Heinberg L, Peat C, Steffen K, Mitchell J, Gunstad J. Preliminary evidence for an association between the composition of the gut microbiome and cognitive function in neurologically healthy older adults. J Int Neuropsychol Soc 23: 700–705, 2017. doi: 10.1017/S1355617717000492. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 260.Zhang P, Yu Y, Qin Y, Zhou Y, Tang R, Wang Q, Li X, Wang H, Weston-Green K, Huang XF, Zheng K. Alterations to the microbiota-colon-brain axis in high-fat-diet-induced obese mice compared to diet-resistant mice. J Nutr Biochem 65: 54–65, 2019. doi: 10.1016/j.jnutbio.2018.08.016. [DOI] [PubMed] [Google Scholar]
- 261.Bruce-Keller AJ, Salbaum JM, Luo M, Blanchard E, Taylor CM, Welsh DA, Berthoud HR. Obese-type gut microbiota induce neurobehavioral changes in the absence of obesity. Biol Psychiatry 77: 607–615, 2015. doi: 10.1016/j.biopsych.2014.07.012. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 262.Braniste V, Al-Asmakh M, Kowal C, Anuar F, Abbaspour A, Tóth M, Korecka A, Bakocevic N, Ng LG, Guan NL, Kundu P, Gulyás B, Halldin C, Hultenby K, Nilsson H, Hebert H, Volpe BT, Diamond B, Pettersson S. The gut microbiota influences blood-brain barrier permeability in mice. Sci Transl Med 6: 263ra158, 2014. [Erratum in Sci Transl Med 6: 266er7, 2014]. doi: 10.1126/scitranslmed.3009759. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 263.Lam YY, Ha CW, Campbell CR, Mitchell AJ, Dinudom A, Oscarsson J, Cook DI, Hunt NH, Caterson ID, Holmes AJ, Storlien LH. Increased gut permeability and microbiota change associate with mesenteric fat inflammation and metabolic dysfunction in diet-induced obese mice. PLoS One 7: e34233, 2012. doi: 10.1371/journal.pone.0034233. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 264.Nishitsuji K, Xiao J, Nagatomo R, Umemoto H, Morimoto Y, Akatsu H, Inoue K, Tsuneyama K. Analysis of the gut microbiome and plasma short-chain fatty acid profiles in a spontaneous mouse model of metabolic syndrome. Sci Rep 7: 15876, 2017. doi: 10.1038/s41598-017-16189-5. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 265.Yoo DY, Kim DW, Kim MJ, Choi JH, Jung HY, Nam SM, Kim JW, Yoon YS, Choi SY, Hwang IK. Sodium butyrate, a histone deacetylase inhibitor, ameliorates SIRT2-induced memory impairment, reduction of cell proliferation, and neuroblast differentiation in the dentate gyrus. Neurol Res 37: 69–76, 2015. doi: 10.1179/1743132814Y.0000000416. [DOI] [PubMed] [Google Scholar]
- 266.Vaiserman AM, Pasyukova EG. Epigenetic drugs: a novel anti-aging strategy? Front Genet 3: 224, 2012. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 267.Baker DJ, Childs BG, Durik M, Wijers ME, Sieben CJ, Zhong J, Saltness RA, Jeganathan KB, Verzosa GC, Pezeshki A, Khazaie K, Miller JD, van Deursen JM. Naturally occurring p16(Ink4a)-positive cells shorten healthy lifespan. Nature 530: 184–189, 2016. doi: 10.1038/nature16932. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 268.Baker DJ, Petersen RC. Cellular senescence in brain aging and neurodegenerative diseases: evidence and perspectives. J Clin Invest 128: 1208–1216, 2018. doi: 10.1172/JCI95145. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 269.Bussian TJ, Aziz A, Meyer CF, Swenson BL, van Deursen JM, Baker DJ. Clearance of senescent glial cells prevents tau-dependent pathology and cognitive decline. Nature 562: 578–582, 2018. doi: 10.1038/s41586-018-0543-y. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 270.Campisi J. Aging, cellular senescence, and cancer. Annu Rev Physiol 75: 685–705, 2013. doi: 10.1146/annurev-physiol-030212-183653. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 271.Childs BG, Baker DJ, Wijshake T, Conover CA, Campisi J, van Deursen JM. Senescent intimal foam cells are deleterious at all stages of atherosclerosis. Science 354: 472–477, 2016. doi: 10.1126/science.aaf6659. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 272.Chinta SJ, Woods G, Rane A, Demaria M, Campisi J, Andersen JK. Cellular senescence and the aging brain. Exp Gerontol, 68: 3–7, 2015. doi: 10.1016/j.exger.2014.09.018. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 273.Kiss T, Nyul-Toth A, Balasubramanian P, Tarantini S, Ahire C, DelFavero J, Yabluchanskiy A, Csipo T, Farkas E, Wiley G, Garman L, Csiszar A, Ungvari Z. Single-cell RNA sequencing identifies senescent cerebromicrovascular endothelial cells in the aged mouse brain. Geroscience 42: 429–444, 2020. doi: 10.1007/s11357-020-00177-1. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 274.Lawrence I, Bene M, Nacarelli T, Azar A, Cohen JZ, Torres C, Johannes G, Sell C. Correlations between age, functional status, and the senescence-associated proteins HMGB2 and p16(INK4a). Geroscience 40: 193–199, 2018. doi: 10.1007/s11357-018-0015-1. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 275.Wang J, Uryga AK, Reinhold J, Figg N, Baker L, Finigan A, Gray K, Kumar S, Clarke M, Bennett M. Vascular smooth muscle cell senescence promotes atherosclerosis and features of plaque vulnerability. Circulation 132: 1909–1919, 2015. doi: 10.1161/CIRCULATIONAHA.115.016457. [DOI] [PubMed] [Google Scholar]
- 276.Baar MP, Brandt RMC, Putavet DA, Klein JDD, Derks KWJ, Bourgeois BRM, Stryeck S, Rijksen Y, van Willigenburg H, Feijtel DA, van der Pluijm I, Essers J, van Cappellen WA, van IJcken WF, Houtsmuller AB, Pothof J, de Bruin RWF, Madl T, Hoeijmakers JHJ, Campisi J, de Keizer PLJ. Targeted apoptosis of senescent cells restores tissue homeostasis in response to chemotoxicity and aging. Cell 169: 132–147, 2017. doi: 10.1016/j.cell.2017.02.031. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 277.Baker DJ, Wijshake T, Tchkonia T, LeBrasseur NK, Childs BG, van de Sluis B, Kirkland JL, van Deursen JM. Clearance of p16Ink4a-positive senescent cells delays ageing-associated disorders. Nature 479: 232–236, 2011. doi: 10.1038/nature10600. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 278.Farr JN, Xu M, Weivoda MM, Monroe DG, Fraser DG, Onken JL, Negley BA, Sfeir JG, Ogrodnik MB, Hachfeld CM, LeBrasseur NK, Drake MT, Pignolo RJ, Pirtskhalava T, Tchkonia T, Oursler MJ, Kirkland JL, Khosla S. Targeting cellular senescence prevents age-related bone loss in mice. Nat Med 23: 1072–1079, 2017. doi: 10.1038/nm.4385. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 279.Patil P, Dong Q, Wang D, Chang J, Wiley C, Demaria M, Lee J, Kang J, Niedernhofer LJ, Robbins PD, Sowa G, Campisi J, Zhou D, Vo N. Systemic clearance of p16. Aging Cell 18: e12927, 2019. doi: 10.1111/acel.12927. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 280.Xu M, Pirtskhalava T, Farr JN, Weigand BM, Palmer AK, Weivoda MM, Inman CL, Ogrodnik MB, Hachfeld CM, Fraser DG, Onken JL, Johnson KO, Verzosa GC, Langhi LGP, Weigl M, Giorgadze N, LeBrasseur NK, Miller JD, Jurk D, Singh RJ, Allison DB, Ejima K, Hubbard GB, Ikeno Y, Cubro H, Garovic VD, Hou X, Weroha SJ, Robbins PD, Niedernhofer LJ, Khosla S, Tchkonia T, Kirkland JL. Senolytics improve physical function and increase lifespan in old age. Nat Med 24: 1246–1256, 2018. doi: 10.1038/s41591-018-0092-9. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 281.Yamazaki Y, Baker DJ, Tachibana M, Liu CC, van Deursen JM, Brott TG, Bu G, Kanekiyo T. Vascular cell senescence contributes to blood-brain barrier breakdown. Stroke 47: 1068–1077, 2016. doi: 10.1161/STROKEAHA.115.010835. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 282.Burton DGA, Faragher RGA. Obesity and type-2 diabetes as inducers of premature cellular senescence and ageing. Biogerontology 19: 447–459, 2018. doi: 10.1007/s10522-018-9763-7. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 283.Maeda M, Hayashi T, Mizuno N, Hattori Y, Kuzuya M. Intermittent high glucose implements stress-induced senescence in human vascular endothelial cells: role of superoxide production by NADPH oxidase. PLoS One 10: e0123169, 2015. doi: 10.1371/journal.pone.0123169. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 284.Shosha E, Xu Z, Narayanan SP, Lemtalsi T, Fouda AY, Rojas M, Xing J, Fulton D, Caldwell RW, Caldwell RB. Mechanisms of diabetes-induced endothelial cell senescence: role of arginase 1. Int J Mol Sci 19: 1215, 2018. doi: 10.3390/ijms19041215. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 285.Yabluchanskiy A, Tarantini S, Balasubramanian P, Kiss T, Csipo T, Fülöp GA, Lipecz A, Ahire C, DelFavero J, Nyul-Toth A, Sonntag WE, Schwartzman ML, Campisi J, Csiszar A, Ungvari Z. Pharmacological or genetic depletion of senescent astrocytes prevents whole brain irradiation-induced impairment of neurovascular coupling responses protecting cognitive function in mice. Geroscience 42: 409–428, 2020. doi: 10.1007/s11357-020-00154-8. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 286.Wheeler MJ, Dunstan DW, Smith B, Smith KJ, Scheer A, Lewis J, Naylor LH, Heinonen I, Ellis KA, Cerin E, Ainslie PN, Green DJ. Morning exercise mitigates the impact of prolonged sitting on cerebral blood flow in older adults. J Appl Physiol (1985) 126: 1049–1055, 2019. doi: 10.1152/japplphysiol.00001.2019. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 287.Drigny J, Gremeaux V, Dupuy O, Gayda M, Bherer L, Juneau M, Nigam A. Effect of interval training on cognitive functioning and cerebral oxygenation in obese patients: a pilot study. J Rehabil Med 46: 1050–1054, 2014. doi: 10.2340/16501977-1905. [DOI] [PubMed] [Google Scholar]
- 288.Blondell SJ, Hammersley-Mather R, Veerman JL. Does physical activity prevent cognitive decline and dementia?: A systematic review and meta-analysis of longitudinal studies. BMC Public Health 14: 510, 2014. doi: 10.1186/1471-2458-14-510. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 289.Carvalho A, Rea IM, Parimon T, Bj C. Physical activity and cognitive function in individuals over 60 years of age: a systematic review. Clin Interv Aging 9: 661–682, 2014. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 290.Sofi F, Valecchi D, Bacci D, Abbate R, Gensini GF, Casini A, Macchi C. Physical activity and risk of cognitive decline: a meta-analysis of prospective studies. J Intern Med 269: 107–117, 2011. [DOI] [PubMed] [Google Scholar]
- 291.Dunsky A, Abu-Rukun M, Tsuk S, Dwolatzky T, Carasso R, Netz Y. The effects of a resistance vs. an aerobic single session on attention and executive functioning in adults. PLoS One 12: e0176092, 2017. [Erratum in PLoS One 12: e0179799, 2017]. doi: 10.1371/journal.pone.0176092. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 292.Alghadir AH, Gabr SA, Al-Eisa ES. Effects of moderate aerobic exercise on cognitive abilities and redox state biomarkers in older adults. Oxid Med Cell Longev 2016: 1–8, 2016. doi: 10.1155/2016/2545168. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 293.Guadagni V, Drogos LL, Tyndall AV, Davenport MH, Anderson TJ, Eskes GA, Longman RS, Hill MD, Hogan DB, Poulin MJ. Aerobic exercise improves cognition and cerebrovascular regulation in older adults. Neurology 94: e2245–e2257, 2020. [Erratum in Neurology 95: 890, 2020]. doi: 10.1212/WNL.0000000000009478. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 294.Alfini AJ, Weiss LR, Nielson KA, Verber MD, Smith JC. Resting cerebral blood flow after exercise training in mild cognitive impairment. JAD 67: 671–684, 2019. doi: 10.3233/JAD-180728. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 295.Chapman SB, Aslan S, Spence JS, Defina LF, Keebler MW, Didehbani N, Lu H. Shorter term aerobic exercise improves brain, cognition, and cardiovascular fitness in aging. Front Aging Neurosci 5: 75, 2013. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 296.Kleinloog JPD, Mensink RP, Ivanov D, Adam JJ, Uludağ K, Pj JORIS. Aerobic exercise training improves cerebral blood flow and executive function: a randomized, controlled cross-over trial in sedentary older men. Front Aging Neurosci 11: 333, 2019. doi: 10.3389/fnagi.2019.00333. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 297.Napoli N, Shah K, Waters DL, Sinacore DR, Qualls C, Villareal DT. Effect of weight loss, exercise, or both on cognition and quality of life in obese older adults. Am J Clin Nutr 100: 189–198, 2014. doi: 10.3945/ajcn.113.082883. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 298.Espeland MA, Luchsinger JA, Baker LD, Neiberg R, Kahn SE, Arnold SE, Wing RR, Blackburn GL, Bray G, Evans M, Hazuda HP, Jeffery RW, Wilson VM, Clark JM, Coday M, Demos-McDermott K, Foreyt JP, Greenway F, Hill JO, Horton ES, Jakicic JM, Johnson KC, Knowler WC, Lewis CE, Nathan DM, Peters A, Pi-Sunyer X, Pownall H, Wadden TA, Rapp SR; Look AHEAD Study Group. Effect of a long-term intensive lifestyle intervention on prevalence of cognitive impairment. Neurology 88: 2026–2035, 2017. doi: 10.1212/WNL.0000000000003955. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 299.Sink KM, Espeland MA, Castro CM, Church T, Cohen R, Dodson JA, Guralnik J, Hendrie HC, Jennings J, Katula J, Lopez OL, McDermott MM, Pahor M, Reid KF, Rushing J, Verghese J, Rapp S, Williamson JD, Investigators LS, LIFE Study Investigators. Effect of a 24-month physical activity intervention vs health education on cognitive outcomes in sedentary older adults: The LIFE randomized trial. JAMA 314: 781–790, 2015. doi: 10.1001/jama.2015.9617. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 300.Espeland MA, Lipska K, Miller ME, Rushing J, Cohen RA, Verghese J, McDermott MM, King AC, Strotmeyer ES, Blair SN, Pahor M, Reid K, Demons J, Kritchevsky SB, Investigators LS. Effects of physical activity intervention on physical and cognitive function in sedentary adults with and without diabetes. J Gerontol A Biol Sci Med Sci 72: 861–866, 2017. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 301.Soto I, Graham LC, Richter HJ, Simeone SN, Radell JE, Grabowska W, Funkhouser WK, Howell MC, Howell GR. APOE stabilization by exercise prevents aging neurovascular dysfunction and complement induction. PLoS Biol 13: e1002279, 2015. doi: 10.1371/journal.pbio.1002279. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 302.Latimer CS, Searcy JL, Bridges MT, Brewer LD, Popović J, Blalock EM, Landfield PW, Thibault O, Porter NM. Reversal of glial and neurovascular markers of unhealthy brain aging by exercise in middle-aged female mice. PLoS One 6: e26812, 2011. doi: 10.1371/journal.pone.0026812. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 303.Olver TD, McDonald MW, Klakotskaia D, Richardson RA, Jasperse JL, Melling CWJ, Schachtman TR, Yang HT, Emter CA, Laughlin MH. A chronic physical activity treatment in obese rats normalizes the contributions of ET-1 and NO to insulin-mediated posterior cerebral artery vasodilation. J Appl Physiol (1985) 122: 1040–1050, 2017. doi: 10.1152/japplphysiol.00811.2016. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 304.Abramson JL, Vaccarino V. Relationship between physical activity and inflammation among apparently healthy middle-aged and older US adults. Arch Intern Med 162: 1286–1292, 2002. doi: 10.1001/archinte.162.11.1286. [DOI] [PubMed] [Google Scholar]
- 305.Colbert LH, Visser M, Simonsick EM, Tracy RP, Newman AB, Kritchevsky SB, Pahor M, Taaffe DR, Brach J, Rubin S, Tb H. Physical activity, exercise, and inflammatory markers in older adults: findings from the Health, Aging and Body Composition Study. J Am Geriatr Soc 52: 1098–1104, 2004. doi: 10.1111/j.1532-5415.2004.52307.x. [DOI] [PubMed] [Google Scholar]
- 306.Witte AV, Fobker M, Gellner R, Knecht S, Flöel A. Caloric restriction improves memory in elderly humans. Proc Natl Acad Sci USA 106: 1255–1260, 2009. doi: 10.1073/pnas.0808587106. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 307.Horie NC, Serrao VT, Simon SS, Gascon MR, Dos Santos AX, Zambone MA, Del Bigio de Freitas MM, Cunha-Neto E, Marques EL, Halpern A, de Melo ME, Mancini MC, Cercato C. Cognitive effects of intentional weight loss in elderly obese individuals with mild cognitive impairment. J Clin Endocrinol Metab 101: 1104–1112, 2016. doi: 10.1210/jc.2015-2315. [DOI] [PubMed] [Google Scholar]
- 308.Prehn K, Jumpertz von Schwartzenberg R, Mai K, Zeitz U, Witte AV, Hampel D, Szela AM, Fabian S, Grittner U, Spranger J, Flöel A. Caloric restriction in older adults-differential effects of weight loss and reduced weight on brain structure and function. Cereb Cortex 27: 1765–1778, 2017. [DOI] [PubMed] [Google Scholar]
- 309.Espeland MA, Rapp SR, Bray GA, Houston DK, Johnson KC, Kitabchi AE, Hergenroeder AL, Williamson J, Jakicic JM, van Dorsten B, Kritchevsky SB; Subgroup AfHIDLAMaM and Group LAR. Long-term impact of behavioral weight loss intervention on cognitive function. J Gerontol A Biol Sci Med Sci 69: 1101–1108, 2014. doi: 10.1093/gerona/glu031. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 310.Peven JC, Jakicic JM, Rogers RJ, Lesnovskaya A, Erickson KI, Kang C, Zhou X, Porter A, Donofry SD, Watt JC, Stillman CM. The effects of a 12-month weight loss intervention on cognitive outcomes in adults with overweight and obesity. Nutrients 12: 2988, 2020. doi: 10.3390/nu12102988. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 311.Ooi TC, Meramat A, Rajab NF, Shahar S, Ismail IS, Azam AA, Sharif R. Intermittent fasting enhanced the cognitive function in older adults with mild cognitive impairment by inducing biochemical and metabolic changes: a 3-year progressive study. Nutrients 12: 2644, 2020. doi: 10.3390/nu12092644. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 312.Anton SD, Lee SA, Donahoo WT, McLaren C, Manini T, Leeuwenburgh C, Pahor M. The effects of time restricted feeding on overweight, older adults: a pilot study. Nutrients 11: 1500, 2019. doi: 10.3390/nu11071500. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 313.Brandt J, Buchholz A, Henry-Barron B, Vizthum D, Avramopoulos D, Cervenka MC. Preliminary report on the feasibility and efficacy of the modified atkins diet for treatment of mild cognitive impairment and early Alzheimer's disease. JAD 68: 969–981, 2019. doi: 10.3233/JAD-180995. [DOI] [PubMed] [Google Scholar]
- 314.Torres-Peña JD, Garcia-Rios A, Delgado-Casado N, Gomez-Luna P, Alcala-Diaz JF, Yubero-Serrano EM, Gomez-Delgado F, Leon-Acuña A, Lopez-Moreno J, Camargo A, Tinahones FJ, Delgado-Lista J, Ordovas JM, Perez-Martinez P, Lopez-Miranda J. Mediterranean diet improves endothelial function in patients with diabetes and prediabetes: A report from the CORDIOPREV study. Atherosclerosis 269: 50–56, 2018. doi: 10.1016/j.atherosclerosis.2017.12.012. [DOI] [PubMed] [Google Scholar]
- 315.Storniolo CE, Casillas R, Bulló M, Castañer O, Ros E, Sáez GT, Toledo E, Estruch R, Ruiz-Gutiérrez V, Fitó M, Martínez-González MA, Salas-Salvadó J, Mitjavila MT, Moreno JJ. A Mediterranean diet supplemented with extra virgin olive oil or nuts improves endothelial markers involved in blood pressure control in hypertensive women. Eur J Nutr 56: 89–97, 2017. doi: 10.1007/s00394-015-1060-5. [DOI] [PubMed] [Google Scholar]
- 316.Torres-Peña JD, Rangel-Zuñiga OA, Alcala-Diaz JF, Lopez-Miranda J, Delgado-Lista J. Mediterranean diet and endothelial function: a review of its effects at different vascular bed levels. Nutrients 12: 2212, 2020. doi: 10.3390/nu12082212. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 317.Alosco ML, Galioto R, Spitznagel MB, Strain G, Devlin M, Cohen R, Crosby RD, Mitchell JE, Gunstad J. Cognitive function after bariatric surgery: evidence for improvement 3 years after surgery. Am J Surg 207: 870–876, 2014. doi: 10.1016/j.amjsurg.2013.05.018. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 318.Alosco ML, Spitznagel MB, Strain G, Devlin M, Cohen R, Paul R, Crosby RD, Mitchell JE, Gunstad J. Improved memory function two years after bariatric surgery. Obesity (Silver Spring ) 22: 32–38, 2014. doi: 10.1002/oby.20494. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 319.Stolberg CR, Mundbjerg LH, Funch-Jensen P, Gram B, Bladbjerg EM, Juhl CB. Effects of gastric bypass surgery followed by supervised physical training on inflammation and endothelial function: a randomized controlled trial. Atherosclerosis 273: 37–44, 2018. doi: 10.1016/j.atherosclerosis.2018.04.002. [DOI] [PubMed] [Google Scholar]
- 320.Moreau KL, Hildreth KL, Klawitter J, Blatchford P, Kohrt WM. Decline in endothelial function across the menopause transition in healthy women is related to decreased estradiol and increased oxidative stress. Geroscience 42: 1699–1714, 2020. doi: 10.1007/s11357-020-00236-7. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 321.Georgakis MK, Beskou-Kontou T, Theodoridis I, Skalkidou A, Petridou ET. Surgical menopause in association with cognitive function and risk of dementia: a systematic review and meta-analysis. Psychoneuroendocrinology 106: 9–19, 2019. doi: 10.1016/j.psyneuen.2019.03.013. [DOI] [PubMed] [Google Scholar]
- 322.Kurita K, Henderson VW, Gatz M, St John J, Hodis HN, Karim R, Mack WJ. Association of bilateral oophorectomy with cognitive function in healthy, postmenopausal women. Fertil Steril 106: 749–756, 2016. doi: 10.1016/j.fertnstert.2016.04.033. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 323.Matyi JM, Rattinger GB, Schwartz S, Buhusi M, Tschanz JT. Lifetime estrogen exposure and cognition in late life: the Cache County Study. Menopause 26: 1366–1374, 2019. doi: 10.1097/GME.0000000000001405. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 324.Somani YB, Pawelczyk JAD, Souza MJ, Kris-Etherton PM, Proctor DN. Aging women and their endothelium: probing the relative role of estrogen on vasodilator function. Am J Physiol Heart Circ Physiol 317: H395–H404, 2019. [DOI] [PMC free article] [PubMed] [Google Scholar]





