Back to Journals » Clinical Interventions in Aging » Volume 20
Understanding Cognitive Decline in Aging: Mechanisms and Mitigation Strategies – A Narrative Review
Received 6 December 2024
Accepted for publication 15 March 2025
Published 15 April 2025 Volume 2025:20 Pages 459—469
DOI https://doi.org/10.2147/CIA.S510670
Checked for plagiarism Yes
Review by Single anonymous peer review
Peer reviewer comments 2
Editor who approved publication: Dr Nandu Goswami
Zbigniew Jost,1 Sylwester Kujach2,3
1Department of Biochemistry, Gdansk University of Physical Education and Sport, Gdansk, Poland; 2Department of Physiology, Medical University of Gdansk, Gdansk, Poland; 3Department of Physiology, Gdansk University of Physical Education and Sport, Gdansk, Poland
Correspondence: Zbigniew Jost, Email [email protected]
Abstract: Cognitive decline is a natural process that accompanies aging. In some cases, such as in sarcopenia-burdened or diseased older adults, the disease course may be more rapid. Declining cognitive function is associated with changes in the central nervous system per se or peripheral triggers that impair cognition. This review discusses issues related to central, central-peripheral, and peripheral factors that enhance cognitive deterioration, such as cortical thickness, cerebral white matter structure and function, blood-brain barrier (BBB) disruption, insulin resistance, inflammation, and vascular dysfunction. BBB permeability appears to be a critical point for factors associated with aging that may accelerate cognitive decline. Thus, we provide an in-depth analysis of the central-peripheral crosstalk. Additionally, we discuss high-intensity interval training (HIIT) as a promising strategy to counteract changes that accompany the aging process. Resistance (RHIIT) and aerobic (AHIIT) may be beneficial for cognitive health among the elderly, but their lack of empirical confirmation is a huge gap in the research.
Keywords: aging, adipose tissue, central nervous system, cognition, exercise, inflammation
Introduction
Healthy aging and quality of life are becoming the goals of gerontological research, as extending the health span and delaying the development of disabilities and chronic diseases are the expectations of aging societies.1 Unfortunately, by 2020, 1 billion people worldwide were aged 60 years or over and by 2050 that number will exceed 2 billion.2 Moreover, projections of dementia onset will triple over the next 30 years.3 This has led to an intensified search for anti-aging treatments.
Aging is a multifactorial process based on cellular senescence,4 which, through its influence on specific signalling pathways, leads to a progressive decline in tissue and organ function among older people.5 In addition, negative age-related changes have been observed in the brain tissue, where structural changes have an important impact on cognitive functioning.
Among others, deterioration of skeletal muscles (sarcopenia), attenuation of cardiovascular functions, and pathophysiological metabolic states, such as obesity and/or type 2 diabetes, have been listed as risk factors for cognitive health.4–6 The key factors associated with cognitive decline are sleep apnea,7 hearing deficits,8 atrial fibrillation,9 vitamin B12 deficiency,10 but physical activity, in turn, can delay cognitive decline.11 It has been shown that most affected cognitive domains include short- and long-term memory as well as executive functions which are crucial for everyday functioning.12
Moreover, a progressive decline in cognitive function may predict the development of neurodegenerative diseases such as Alzheimer’s disease and related dementias,13 chronic mental health conditions such as depression or anxiety.14 Furthermore, low-grade inflammation originating from adipose tissue throughout life may accelerate this process.15
Interestingly, healthy habits, such as physical activity, balanced nutrition, and social interactions, can delay and prevent negative changes in the body.16 Moreover, the approach to healthy aging includes both physical and mental aspects, which are partly established.17,18 However, therapeutic interventions targeting cognitive decline during aging require a comprehensive understanding of the underlying functional and structural changes, which would enable formulation of more precise and effective recommendations.
Consequently, the aim of this review was to shed light on current knowledge regarding the mechanisms related to cognitive decline accompanying aging. Simultaneously, high-intensity interval training (HIIT) is also discussed as a promising exercise strategy that has the potential to slow cognitive decline in aging.
Potential Mechanisms of Cognitive Decline
Cognitive decline during aging is a multidimensional issue that deserves a broader perspective. Hence, the division into central, central-peripheral, and peripheral factors/mechanisms seems to be the most appropriate and is summarized in Figure 1.
![]() |
Figure 1 Factors affecting cognitive decline in aging. |
Central Mechanisms
Cortical Thickness
The cerebral cortex, particularly the prefrontal cortex, is a region in which cognition-related processes occur.19 Hence, the greater the thickness of the cerebral cortex, the better is the cognitive performance throughout life.20 On the other hand, cortical thinning or shrinkage is a natural process that occurs with aging,21 but the thinning of some regions of the cerebral cortex is accelerated with age (lateral occipital, lingual), and others are slowed down (posteriorcingulate, lateral orbitofrontal).22 In addition, some cortical regions undergo thinning dependent on genetic and environmental factors.23 The findings indicate that lifestyle, including moderate to vigorous physical activity, was associated with greater thickness of the cerebral cortex.24 In addition, a meta-analysis provided by Afanador-Restrepo et al indicated that high-intensity training interventions are sufficient to improve or at least delay the decline in global cognition.25 Unfortunately, the lack of cortical thickness analyses does not directly indicate a cause-and-effect relationship.
On the contrary, a sedentary lifestyle and increased fat accumulation can enhance cortical thinning. A longitudinal study by Shaw et al examined nearly 400 mildly overweight older adults (60–66 years of age) and observed that the cortical thickness decreased with age by 0.3% per year over a 12-year study period.26 Furthermore, a longitudinal study of overweight and obese individuals found that a 1% annual increase in BMI was associated with cortical thinning of up to 0.5%.27 Similarly, another longitudinal study demonstrated that BMI trajectories over four decades predict a pattern of cortical thinning. Moreover, the authors concluded that a steeper increase in BMI during midlife was linked to a thinner cortex later in life.28 Several mechanisms may link excessive midlife BMI to neurodegenerative damage, likely through interconnected processes such as vascular and metabolic pathways, inflammatory responses, and potential genetic influences. Therefore, maintaining a healthy body weight (BMI < 24.9) and avoiding excessive adiposity are crucial for long-term health, not just in later life, but at every stage.
Cerebral White Matter
The white matter of the brain integrates areas responsible for cognition. Fluidity-based measurements and crystallization, which characterize speed and knowledge, respectively, differ in terms of aging changes.29 A few studies have indicated that aging is strictly related to white matter decline and cortical disconnectivity.30–32 Additionally, variations in BMI during the lifespan are associated with a reduction in white matter integrity, indicating that deteriorated physical health (increased global adiposity) can affect brain function.33 A significant number of studies have confirmed this hypothesis.34–36 Moreover, similar conclusions have been drawn from studies involving older adults who were overweight or obese.37,38 Nevertheless, the mechanisms responsible for these changes are not fully understood. However, some researchers have claimed that it can be associated with low-grade systemic inflammation owing to excessive adiposity or abnormal cardiometabolic and cardiovascular factor levels.39,40
Therefore, central mechanisms are involved in shaping the grey and white matter of the brain, which is crucial for cognition in the elderly. Structural, functional, and endocrine changes in peripheral systems and tissues, including the blood-brain barrier (BBB), insulin resistance, adipose tissue-derived inflammation, and vascular function, should also be considered when explaining the mechanisms responsible for the decrease in cognitive performance in older adults.
Blood-Brain Barrier as a Central-Peripheral Mechanism
Cross-environmental homeostasis between the central nervous system (CNS) and blood that connects the rest of the body is rigorously protected by specialized endothelial cells of pericytes, astrocytes, and cerebral microvessels called the BBB.41 The influx and efflux of energy substrates, metabolites, ions, and other serum-derived factors from the CNS through protein complexes, tight junctions, and glycocalixes are regulated by the BBB to maintain physiological neuronal function.42 Unfortunately, some pathophysiological conditions, such as diseases of the nervous system, including neurodegenerative and metabolic conditions, cause BBB disruption and increase the permeability to neurotoxic compounds.43,44 Moreover, impaired efflux, accumulation, and aggregation of amyloid-β (Aβ) in the brain also causes cognitive impairment.45 However, other factors such as aging and obesity are associated with BBB influx-efflux disruption.46,47
Some studies using magnetic resonance imaging (MRI) have shown that BBB disruption is most pronounced in the hippocampus48 and in the grey and white matter of the brain.49 It was also indicated that over 12 years, BBB leakage in the white and grey matter was significantly associated with memory retrieval decline.50 Unfortunately, aging and excess adipose tissue aggregation, that is, in overweight and obese individuals, show synergistic and negative health effects.51–53 Low-grade systemic inflammation induced by adipose tissue-derived cytokines can exacerbate the activation of immune cells, such as microglia, causing neuroinflammation and further BBB disruption,51,54 as summarized in the review by Takata et al.55 Therefore, obesity-induced chronic peripheral inflammation is associated with impaired hippocampal plasticity in an animal model.56,57 Interestingly, high-fat diet (HFD)-fed animals by reducing mRNA expression of proteins such as claudin-5 and −12 resulted in impaired BBB integrity. Increased BBB permeability was observed mainly in the hippocampal area, which appears to be particularly vulnerable to BBB disruption.58 Moreover, HFD-induced obesity interacts with aging, causing neuroinflammation and cognitive decline.59 HFD also amplifies the negative impact of aging through neuroinflammation-induced increase in microglial activation.60 Therefore, BBB disturbances caused by peripheral pathophysiological changes such as excessive fat accumulation during aging can pose a danger to cognitive health.
On the other hand, exogenous supply of substances with antioxidant, anti-inflammatory and prebiotic properties, such as polyphenols, may contribute to strengthening the integrity of the BBB through changes in the composition of the gut microbiome.61,62 In an association study, elderly people with cognitive impairment had lower abundance of the anti-inflammatory bacteria E. rectale and B. fragilis, and higher presence of the pro-inflammatory bacteria Escherichia/Shigella than those without cerebral amyloidosis - one of the causes of the development of Alzheimer’s disease.63 Other gut bacteria such as Odoribacter has been positively associated with white matter volume and the right hippocampus, and Bacteroides enrichment has been linked to better cognitive performance among older adults.64 Dietary polyphenols present in tea and grapes, among others, can improve the composition of the intestinal microbiota, including increasing the abundance of Bacteroidetes phyla.65–67 Therefore, increasing the presence of anti-inflammatory gut bacteria may reduce peripheral inflammation,68 contributing to a decrease in BBB permeability to inflammatory factors, thereby protecting CNS structure and function. Nevertheless, the exact mechanism underlying the changes in BBB permeability following dietary polyphenol supply requires further experimental studies.
Peripheral Mechanisms
Despite the central mechanisms mentioned above, special attention should also be paid to body-brain crosstalk and its impact on aging-associated cognitive performance decline. Peripheral mechanisms include insulin resistance, low-grade systemic inflammation, and vascular dysfunction.69–71
Insulin Resistance
Insulin is a hormone secreted by pancreatic β-cells that plays a key role in the regulation of glucose levels in the body.72 Interestingly, insulin is also involved in neuronal cell signalling and communication by regulating neurotransmitter secretion in the CNS.73 Insulin transported from the periphery across the BBB to the CNS influences synaptogenesis and nerve growth,74 and the consolidation of short- and long-term memory (at least in animal models) is associated with insulin receptor (IR) levels.75 However, weakening of excitability in response to insulin due to dysfunctions/low levels of IRs, that is, insulin resistance, leads to cognitive deterioration.76
Insulin resistance has been linked to memory deficits such as dementia, vascular dementia, and other cognitive dysfunctions such as Alzheimer’s disease.77 However, some studies have attempted to link insulin resistance with cognitive decline as a normal aging-associated process in humans. Indeed, a recently published study by Wei et al showed that the triglyceride glucose (TyG) index of over 660 older adults was strongly correlated with low cognitive function,78 which also confirmed a previous report.79 Moreover, data from nearly 3,000 older adults aged > 60 years showed that among participants without obesity, homeostasis model assessment of insulin resistance (HOMA-IR) and insulin were negatively correlated with the Consortium to Establish a Registry for Alzheimer’s Disease Immediate Recall (CERAD-IR). In addition, the odds of a low score on the Digit Symbol Substitution Test (DSST), one of a cognitive function test) increased with the number of glucose metabolic risk factors.80
Finally, the largest study to date, conducted by Wang et al, with 4,420 participants, found that the TyG index was significantly associated with an increased risk of global cognitive decline, but only in men over 60 years of age (n = 2062).81 Therefore, a weakened insulin response (insulin resistance) may increase the risk or accelerate a decline in cognitive function during aging.
However, the connection between these factors - low-grade systemic inflammation, reduced IRs, BBB disruption, and increased microglial cytotoxicity along with interventions that enhance insulin responsiveness to elevated glucose levels warrants further investigation in the context of cognitive aging.
Inflammation
Few reviews have been released concerning the association between peripheral inflammation and CNS ie brain and its function.82–84 In a longitudinal study conducted over 4.5 years, low-grade systemic inflammation originating in the periphery has led to unfavourable changes in brain morphology in older adults. Doubling of circulating interleukin-6 (IL-6) was associated with lower total brain volume and, what is more fascinating, with doubled aging equivalent (9 years).85 In support of this, a study investigating the risk factors for brain aging in individuals aged 64–100 years found that elevated levels of IL-6 were associated with an increased likelihood of accelerated brain aging. Moreover, in the same study, obesity was associated with brain aging, which seems to demonstrate the synergy between these two factors,86 confirming the findings of previous studies.87,88 Hence, body–brain crosstalk, which refers to the communication between factors secreted by peripheral tissues and cells, particularly cytokines released by adipose tissue as well as neuroprotective molecules produced and/or released from skeletal muscles, as well as from the brain, deserves special attention. Indeed, in cognitively unimpaired adults aged slightly above 85 years, higher IL-6 and soluble tumor necrosis factor receptor 2 (sTNFr2) levels are positively associated with greater Aβ deposition in the brain.89
However, not all scientific evidence unequivocally supports the involvement of pro-inflammatory cytokines in cognitive decline. In a study by Mendelson et al, which analyzed data from nearly 40,000 participants aged 40–70 years from the UK Biobank, a weak association was found between brain structure and levels of C-reactive protein (CRP) in these individuals. In addition, CRP levels were weakly, but negatively, correlated with cognitive function.90
Nevertheless, when assessing the relationship between brain function and aging, the total profile of pro-inflammatory cytokines should be considered, as well as single markers that cannot fully demonstrate the association between inflammation and cognitive decline.91
Vascular Dysfunction
The mechanisms of vascular aging are likely multifaceted. Vascular dysfunction is usually the main consequence of increased oxidative stress due to decreased mitochondrial density as well as the weakened process of biogenesis observed during aging.92,93 Elevated levels of reactive oxygen species (ROS), such as superoxide (O2-), are produced by the mitochondrial electron transport chain, resulting in increased expression and activity of NADPH oxidase.94 Peroxides combine with nitric oxide (NO), a precursor of vasodilation in blood vessels, to form peroxynitrates, which are molecules with strong oxidative effects.95,96 Moreover, perivascular adiposity in overweight and obese individuals is a source of tumor necrosis factor-α (TNF-α), a molecule that can initiate endothelial oxidative stress signalling, leading to reduced vasorelaxation.97 Therefore, excess perivascular and global adipose tissue, and aging have both synergistic and negative effects. They trigger an increase in oxidative and nitro-oxidative stress, causing early vascular aging through inflammation, leading to vascular dysfunction.98
Vascular inflammation leads to negative structural and functional consequences in the blood and cerebrovascular vessels, such as atherogenesis, aneurysm, vascular rarefaction, and hemorrhage.99 Therefore, aging induced by excessive adiposity-derived inflammation can be dangerous for cognitive health due to cerebromicrovascular rarefaction, which promotes the dysregulation of cerebral blood flow.100 Indeed, a decrease in the cerebromicrovasculature causes hypoperfusion of the white matter of the brain, leading to metabolic and functional changes in neuronal dysregulation as well as cognitive decline.101 Moreover, a much more severe decline in cognitive performance due to cerebral hypoperfusion has been observed in individuals with higher adipose tissue deposition in patients with heart failure,102,103 and healthy individuals.104,105 Hence, the search for interventions that induce blood perfusion through organs, including the brain, while from the long-term perspective, reducing low-grade systemic inflammation seems to be a promising direction of research in aged overweight/obese people.
Moreover, focusing on strategies that reduce the resting level of pro-inflammatory cytokines and interventions that slow down aging, that is, maintaining the number and density of mitochondria and increasing the level of anti-inflammatory cytokines, myokines, trophic factors, and neuroprotective metabolites, should be of interest to researchers.
Physical Exercise as a Deaccelerating Strategy in Aging
Strategies to Improve Cognition Through Peripheral Mechanisms – What’s Known?
Sarcopenia, along with accompanying changes in body composition (ie, increased adiposity) sometimes referred to as sarcobesity,106 are part of the aging population. Muscle loss, as well as fat accumulation, promotes the development of pathological conditions such as insulin resistance, inflammation and vascular dysfunction (see above subsections). Since skeletal muscle is known as the largest organ sensitive to insulin and one that secretes pro- and anti-inflammatory factors and has significant vascularization, strategies to enhance the anabolic response are intensively sought. Resistance training (RT) preserves and/or increases skeletal muscle mass by stimulating the protein kinase B/mammalian target of rapamycin (Akt/mTOR) signalling pathway among people of all ages.107,108 As a result, higher muscle mass can absorb larger amounts of glucose through upregulated IRs sensitivity to insulin, which is often dysfunctional in elderlies. Hence, lowering plasma glucose levels not only promotes a reduction in hyperglycaemia, but also, in the long term, inflammation. Low-grade systemic inflammation caused pro-inflammatory cytokines can be lowered by RT.109 In addition, contracting skeletal muscles following RT secrete anti-inflammatory factors and exerkines such as brain-derived neurotrophic factor, cathepsin B and irisin, which can cross the BBB.110 By modulating synaptic plasticity and neuronal migration, exerkines appear to be a key mediator of the crosstalk between peripheral and CNS factors shaping cognition among the elderly. Hence, RT is a promising strategy against cognitive decline.
Moreover, the combination of RT with aerobic training (AT) ie concurrent training (CT) is as effective with its cardiorespiratory fitness (CRF) enhancing effect as AT alone.111 Furthermore, the increase in CRF is closely linked to improvements in global white matter volume and local integrity,112 which together promote the preservation of cognitive function. Although it is known that AT and RT are hugely beneficial for brain health in elderly, current recommendations do not specify the nature of the exercises performed in the first place.
Physical Exercise – Facts and Recommendations
Scientific reports have clearly indicated that physical exercise leads to the improvement or preservation of physical fitness components, especially during aging.113 However, the proper implementation of aerobic- or strength-based protocols needs to be adapted to the level of physical and cognitive fitness of older people.114,115 In addition, many positions/recommendations for prescribing training programs can be found in literature,116,117 but the specifics are not fully standardized. At the same time, the World Health Organization guidelines assume that at least 150–300 minutes of moderate-intensity or 75–150 minutes weekly of vigorous-intensity aerobic physical activity should be performed by adults aged > 65 years.118 Studies indicate that most adults are aware of the importance of physical activity on most days of the week, but do not meet the minimum guidelines, mainly due to lack of time.119,120
Furthermore, the often-recommended traditional endurance exercises may be perceived as boring and consequently discourage further exercise activities. Recently, increasing attention has been paid to HIIT, which is supposed to be a non-pharmacological, decelerative cognitive decline treatment for aging.121
HIIT as a Remedy to Aging – A Future Direction
HIIT is a strategy that was originally compared in terms of adaptation to classic, moderate-intensity continuous endurance training (MICT),122 as well as in terms of cell signalling,123 and metabolic changes.124 Similar training adaptations following HIIT and MICT have prompted a cascade of subsequent experiments on HIIT and its effects on individuals with type I and II diabetes,125,126 cardiovascular system dysfunction,127,128 and its impact on cognitive function.129,130
Safety issues of interventions that induce significant physiological and metabolic stress often raise questions. HIIT is highly safe among older adults, as summarized in a recent review paper.131 Moreover, even among patients with coronary heart disease,132 and chronic stroke,133 HIIT is very well tolerated, and the number of participants withdrawing from continuation of the experiment, including personal reasons, is similar to the groups performing lower-intensity workouts and controls.
To date, only the impact of classical resistance training on physical fitness among older adults has been studied.134,135 It is also believed that dividing HIIT into two models, depending on the nature of the work undertaken, resistance (RHIIT) and aerobic (AHIIT), can be beneficial for cognition. RHIIT may inhibit sarcopenia by activating the mTOR/insulin-like growth factor-1-dependent signalling pathways, resulting in a hypertrophic response in the skeletal muscle. Preserving and/or increasing muscle mass and postural muscle and limb muscle balance responsible for locomotion leads to an increase in quality of life in older adults.136 RHIIT also leads to improvements in aerobic capacity due to adaptations to physical training at the skeletal muscle level in older adults.137 However, an increase in aerobic capacity is the main outcome of AHIIT,138 and may lead to an increase in cognitive performance. Thus, both RHIIT and AHIIT appear to be promising interventions for aging; however, many issues remain unclear.
Conclusion
In conclusion, at least in theory, RHIIT and AHIIT may be beneficial for cognitive health among older adults. The lack of experimental studies comparing the effects of the two forms of HIIT is a major gap in the research. In this review we highlight the mechanisms underlying cognitive aging that may be improved following different types of HIIT. However, validating these innovative ideas require providing empirical evidence.
Funding
This research was supported by a grant from the National Science Center (Poland) under grant Opus no: 2019/33/B/NZ7/01980.
Disclosure
The authors report no conflicts of interest in this work.
References
1. Cai J, Hu W, Yang Y. et al. Healthy life expectancy for 202 countries up to 2030: projections with a Bayesian model ensemble. J Glob Health. 2023;13:04185. doi:10.7189/jogh.13.04185
2. WHO. Mental health of older adults. 2023. Available from: https://www.who.int/news-room/fact-sheets/detail/mental-health-of-older-adults.
3. Lancet T. A global assessment of dementia, now and in the future. The Lancet. 2015;386(9997):931. doi:10.1016/S0140-6736(15)00117-8
4. Przybycien-Gaweda PM, Gwee X, Gao Q, Chua DQL, Fam J, Ng TP. Metabolic syndrome and cognition: follow-up study of Chinese over-55-year-olds. Dement Geriatr Cogn Disord. 2020;49(2):129–137. doi:10.1159/000509124
5. Joly L. Arterial stiffness and cognitive function. Gériatrie Et Psychologie Neuropsychiatrie du Vieillissement. 2017;15(1):83–88. doi:10.1684/pnv.2017.0655
6. Sui SX, Williams LJ, Holloway-Kew KL, Hyde NK, Pasco JA. Skeletal muscle health and cognitive function: a narrative review. Int J mol Sci. 2020;22(1):255. doi:10.3390/ijms22010255
7. Kerner NA, Roose SP. Obstructive sleep apnea is linked to depression and cognitive impairment: evidence and potential mechanisms. Am J Geriatric Psychiatry. 2016;24(6):496–508. doi:10.1016/j.jagp.2016.01.134
8. Loughrey DG, Kelly ME, Kelley GA, Brennan S, Lawlor BA. Association of age-related hearing loss with cognitive function, cognitive impairment, and dementia. JAMA Otolaryngol–Head Neck Surgery. 2018;144(2):115. doi:10.1001/jamaoto.2017.2513
9. Ding M, Qiu C. Atrial fibrillation, cognitive decline, and dementia: an epidemiologic review. Curr Epidemiol Rep. 2018;5(3):252–261. doi:10.1007/s40471-018-0159-7
10. Morley JE. Can we improve care for patients with dementia? J Nutr Health Aging. 2011;15(7):523–526. doi:10.1007/s12603-011-0141-2
11. Zhao Y, Li Y, Wang L, et al. Physical activity and cognition in sedentary older adults: a systematic review and meta-analysis. J Alzheimer’s Dis. 2022;87(3):957–968. doi:10.3233/JAD-220073
12. Morley JE. An overview of cognitive impairment. Clin Geriatr Med. 2018;34(4):505–513. doi:10.1016/j.cger.2018.06.003
13. Gonzales MM, Garbarino VR, Pollet E, et al. Biological aging processes underlying cognitive decline and neurodegenerative disease. J Clin Invest. 2022;132(10). doi:10.1172/JCI158453
14. Fisher JE, Zhou J, Liu AG, Fullerton CS, Ursano RJ, Cozza SJ. Effect of comorbid anxiety and depression in complicated grief on perceived cognitive failures. Depress Anx. 2020;37(1):54–62. doi:10.1002/da.22943
15. Letra L, Santana I. The influence of adipose tissue on brain development, cognition, and risk of neurodegenerative disorders. Adv Neurobiol. 2017;19:151–161. doi:10.1007/978-3-319-63260-5_6
16. Daniel KM. Best practices for promoting healthy aging. Clin Geriatr Med. 2020;36(4):713–718. doi:10.1016/j.cger.2020.06.012
17. Eckstrom E, Neukam S, Kalin L, Wright J. Physical activity and healthy aging. Clin Geriatr Med. 2020;36(4):671–683. doi:10.1016/j.cger.2020.06.009
18. Estebsari F, Dastoorpoor M, Khalifehkandi ZR, et al. The concept of successful aging: a review article. Curr Aging Sci. 2020;13(1):4–10. doi:10.2174/1874609812666191023130117
19. Miller EK. The prefrontal cortex and cognitive control. Nat Rev Neurosci. 2000;1(1):59–65. doi:10.1038/35036228
20. Chen C, Omiya Y. Brain asymmetry in cortical thickness is correlated with cognitive function. Front Hum Neurosci. 2014;8:877. doi:10.3389/fnhum.2014.00877
21. MacDonald ME, Pike GB. MRI of healthy brain aging: a review. NMR Biomed. 2021;34(9):e4564. doi:10.1002/nbm.4564
22. Pfefferbaum A, Rohlfing T, Rosenbloom MJ, Chu W, Colrain IM, Sullivan EV. Variation in longitudinal trajectories of regional brain volumes of healthy men and women (ages 10 to 85 years) measured with atlas-based parcellation of MRI. Neuroimage. 2013;65:176–193. doi:10.1016/j.neuroimage.2012.10.008
23. Strike LT, Hansell NK, Couvy-Duchesne B, et al. Genetic complexity of cortical structure: differences in genetic and environmental factors influencing cortical surface area and thickness. Cerebral Cortex. 2019;29(3):952–962. doi:10.1093/cercor/bhy002
24. Falck RS, Hsu CL, Best JR, Li LC, Egbert AR, Liu-Ambrose T. Not just for joints: the associations of moderate-to-vigorous physical activity and sedentary behavior with brain cortical thickness. Med Sci Sports Exerc. 2020;52(10):2217–2223. doi:10.1249/MSS.0000000000002374
25. Afanador-Restrepo DF, Casanova-Correa A, Martín-Ojeda RI, et al. Dose-response relationship of high-intensity training on global cognition in older adults with mild cognitive impairment or dementia: a systematic review with meta-analysis - the ACHIEVE-Study. Europ Rev Aging Phys Act. 2024;21(1):23. doi:10.1186/s11556-024-00358-3
26. Shaw ME, Sachdev PS, Anstey KJ, Cherbuin N. Age-related cortical thinning in cognitively healthy individuals in their 60s: the PATH through life study. Neurobiol Aging. 2016;39:202–209. doi:10.1016/j.neurobiolaging.2015.12.009
27. Shaw ME, Sachdev PS, Abhayaratna W, Anstey KJ, Cherbuin N. Body mass index is associated with cortical thinning with different patterns in mid- and late-life. Int J Obes. 2018;42(3):455–461. doi:10.1038/ijo.2017.254
28. Franz CE, Xian H, Lew D, et al. Body mass trajectories and cortical thickness in middle-aged men: a 42-year longitudinal study starting in young adulthood. Neurobiol Aging. 2019;79:11–21. doi:10.1016/j.neurobiolaging.2019.03.003
29. Craik FI, Salthouse TA. The Handbook of Aging and Cognition. Psychology press.; 2011.
30. Salat DH. The declining infrastructure of the aging brain. Brain Connect. 2011;1(4):279–293. doi:10.1089/brain.2011.0056
31. Marstaller L, Williams M, Rich A, Savage G, Burianová H. Aging and large-scale functional networks: white matter integrity, gray matter volume, and functional connectivity in the resting state. Neuroscience. 2015;290:369–378. doi:10.1016/j.neuroscience.2015.01.049
32. Gao Y, Zhao Y, Li M, et al. Functional alterations in bipartite network of white and grey matters during aging. Neuroimage. 2023;278:120277. doi:10.1016/j.neuroimage.2023.120277
33. Verstynen TD, Weinstein AM, Schneider WW, Jakicic JM, Rofey DL, Erickson KI. Increased body mass index is associated with a global and distributed decrease in white matter microstructural integrity. Psychosom Med. 2012;74(7):682–690. doi:10.1097/PSY.0b013e318261909c
34. Zhang R, Beyer F, Lampe L, et al. White matter microstructural variability mediates the relation between obesity and cognition in healthy adults. Neuroimage. 2018;172:239–249. doi:10.1016/j.neuroimage.2018.01.028
35. Dietze LMF, McWhinney SR, Radua J, Hajek T. Extended and replicated white matter changes in obesity: voxel-based and region of interest meta-analyses of diffusion tensor imaging studies. Front Nutr. 2023;10:1108360. doi:10.3389/fnut.2023.1108360
36. Cheng X, Wang W, Sun C, Sun Y, Zhou C. White matter integrity abnormalities in healthy overweight individuals revealed by whole brain meta-analysis of diffusion tensor imaging studies. J Obes. 2023;2023:7966540. doi:10.1155/2023/7966540
37. Bolzenius JD, Laidlaw DH, Cabeen RP, et al. Brain structure and cognitive correlates of body mass index in healthy older adults. Behavi Brain Res. 2015;278:342–347. doi:10.1016/j.bbr.2014.10.010
38. Repple J, Opel N, Meinert S, et al. Elevated body-mass index is associated with reduced white matter integrity in two large independent cohorts. Psychoneuroendocrinology. 2018;91:179–185. doi:10.1016/j.psyneuen.2018.03.007
39. Alfaro FJ, Gavrieli A, Saade-Lemus P, Lioutas V-A, Upadhyay J, Novak V. White matter microstructure and cognitive decline in metabolic syndrome: a review of diffusion tensor imaging. Metabolism. 2018;78:52–68. doi:10.1016/j.metabol.2017.08.009
40. Li Z, Wang W, Sang F, Zhang Z, Li X. White matter changes underlie hypertension-related cognitive decline in older adults. Neuroimage Clin. 2023;38:103389. doi:10.1016/j.nicl.2023.103389
41. Balasubramanian P, Kiss T, Tarantini S, et al. Obesity-induced cognitive impairment in older adults: a microvascular perspective. Am J Physiol Heart Circ Physiol. 2021;320(2):H740–H761. doi:10.1152/ajpheart.00736.2020
42. Blanchette M, Daneman R. Formation and maintenance of the BBB. Mech Dev. 2015;138(Pt 1):8–16. doi:10.1016/j.mod.2015.07.007
43. Sweeney MD, Zhao Z, Montagne A, Nelson AR, Zlokovic BV. Blood-brain barrier: from physiology to disease and back. Physiol Rev. 2019;99(1):21–78. doi:10.1152/physrev.00050.2017
44. Braun M, Iliff JJ. The impact of neurovascular, blood-brain barrier, and glymphatic dysfunction in neurodegenerative and metabolic diseases. Int Rev Neurobiol. 2020;154:413–436. doi:10.1016/bs.irn.2020.02.006
45. Jaeger LB, Dohgu S, Hwang MC, et al. Testing the neurovascular hypothesis of Alzheimer’s disease: LRP-1 antisense reduces blood-brain barrier clearance, increases brain levels of amyloid-beta protein, and impairs cognition. J Alzheimers Dis. 2009;17(3):553–570. doi:10.3233/JAD-2009-1074
46. Rhea EM, Salameh TS, Logsdon AF, Hanson AJ, Erickson MA, Banks WA. Blood-brain barriers in obesity. AAPS J. 2017;19(4):921–930. doi:10.1208/s12248-017-0079-3
47. Banks WA, Reed MJ, Logsdon AF, Rhea EM, Erickson MA. Healthy aging and the blood-brain barrier. Nat Aging. 2021;1(3):243–254. doi:10.1038/s43587-021-00043-5
48. Montagne A, Barnes SR, Sweeney MD, et al. Blood-brain barrier breakdown in the aging human hippocampus. Neuron. 2015;85(2):296–302. doi:10.1016/j.neuron.2014.12.032
49. Verheggen ICM, de Jong JJA, van Boxtel MPJ, et al. Increase in blood-brain barrier leakage in healthy, older adults. Geroscience. 2020;42(4):1183–1193. doi:10.1007/s11357-020-00211-2
50. Verheggen ICM, de Jong JJA, van Boxtel MPJ, et al. Imaging the role of blood-brain barrier disruption in normal cognitive ageing. Geroscience. 2020;42(6):1751–1764. doi:10.1007/s11357-020-00282-1
51. Tucsek Z, Toth P, Sosnowska D, et al. 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. 2014;69(10):1212–1226. doi:10.1093/gerona/glt177
52. Tucsek Z, Toth P, Tarantini S, et al. Aging exacerbates obesity-induced cerebromicrovascular rarefaction, neurovascular uncoupling, and cognitive decline in mice. J Gerontol a Biol Sci Med Sci. 2014;69(11):1339–1352. doi:10.1093/gerona/glu080
53. Valcarcel-Ares MN, Tucsek Z, Kiss T, et al. 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. 2019;74(3):290–298. doi:10.1093/gerona/gly127
54. Shigemoto-Mogami Y, Hoshikawa K, Sato K. Activated microglia disrupt the blood-brain barrier and induce chemokines and cytokines in a rat in vitro model. Front Cell Neurosci. 2018;12:494. doi:10.3389/fncel.2018.00494
55. Takata F, Nakagawa S, Matsumoto J, Dohgu S. Blood-brain barrier dysfunction amplifies the development of neuroinflammation: understanding of cellular events in brain microvascular endothelial cells for prevention and treatment of BBB dysfunction. Front Cell Neurosci. 2021;15:661838. doi:10.3389/fncel.2021.661838
56. Erion JR, Wosiski-Kuhn M, Dey A, et al. Obesity elicits interleukin 1-mediated deficits in hippocampal synaptic plasticity. J Neurosci. 2014;34(7):2618–2631. doi:10.1523/JNEUROSCI.4200-13.2014
57. Hao S, Dey A, Yu X, Stranahan AM. Dietary obesity reversibly induces synaptic stripping by microglia and impairs hippocampal plasticity. Brain Behav Immun. 2016;51:230–239. doi:10.1016/j.bbi.2015.08.023
58. Kanoski SE, Zhang Y, Zheng W, Davidson TL. The effects of a high-energy diet on hippocampal function and blood-brain barrier integrity in the rat. J Alzheimer’s Dis. 2010;21(1):207–219. doi:10.3233/JAD-2010-091414
59. 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. 2017;58:88–101. doi:10.1016/j.neurobiolaging.2017.06.014
60. Spencer SJ, Basri B, Sominsky L, et al. High-fat diet worsens the impact of aging on microglial function and morphology in a region-specific manner. Neurobiol Aging. 2019;74:121–134. doi:10.1016/j.neurobiolaging.2018.10.018
61. Grabska-Kobyłecka I, Szpakowski P, Król A, et al. Polyphenols and their impact on the prevention of neurodegenerative diseases and development. Nutrients. 2023;15(15):3454. doi:10.3390/nu15153454
62. Naomi R, Yazid MD, Teoh SH, et al. Dietary polyphenols as a protection against cognitive decline: evidence from animal experiments; mechanisms and limitations. Antioxidants. 2023;12(5):1054. doi:10.3390/antiox12051054
63. Cattaneo A, Cattane N, Galluzzi S, et al. Association of brain amyloidosis with pro-inflammatory gut bacterial taxa and peripheral inflammation markers in cognitively impaired elderly. Neurobiol Aging. 2017;49:60–68. doi:10.1016/j.neurobiolaging.2016.08.019
64. Liang X, Fu Y, ting CW, et al. Gut microbiome, cognitive function and brain structure: a multi-omics integration analysis. Transl Neurodegener. 2022;11(1):49. doi:10.1186/s40035-022-00323-z
65. Roopchand DE, Carmody RN, Kuhn P, et al. Dietary polyphenols promote growth of the gut bacterium akkermansia muciniphila and attenuate high-fat diet–induced metabolic syndrome. Diabetes. 2015;64(8):2847–2858. doi:10.2337/db14-1916
66. Li R, Wang GP, Whitlock JA, Zhao S, Yagiz Y, Gu L. Muscadine grapes (Vitis rotundifolia) and dealcoholized muscadine wine alleviated symptoms of colitis and protected against dysbiosis in mice exposed to dextran sulfate sodium. J Funct Foods. 2020;65:103746. doi:10.1016/j.jff.2019.103746
67. Liu YC, Li XY, Shen L. Modulation effect of tea consumption on gut microbiota. Appl Microbiol Biotechnol. 2020;104(3):981–987. doi:10.1007/s00253-019-10306-2
68. Molinari R, Merendino N, Costantini L. Polyphenols as modulators of pre‐established gut microbiota dysbiosis: state‐of‐the‐art. BioFactors. 2022;48(2):255–273. doi:10.1002/biof.1772
69. Kullmann S, Heni M, Hallschmid M, Fritsche A, Preissl H, Häring HU. Brain insulin resistance at the crossroads of metabolic and cognitive disorders in humans. Physiol Rev. 2016;96(4):1169–1209. doi:10.1152/physrev.00032.2015
70. Michaud M, Balardy L, Moulis G, et al. Proinflammatory cytokines, aging, and age-related diseases. J Am Med Dir Assoc. 2013;14(12):877–882. doi:10.1016/j.jamda.2013.05.009
71. Ungvari Z, Tarantini S, Sorond F, Merkely B, Csiszar A. Mechanisms of vascular aging, a geroscience perspective: JACC focus seminar. J Am Coll Cardiol. 2020;75(8):931–941. doi:10.1016/j.jacc.2019.11.061
72. Niswender KD. Basal insulin: physiology, pharmacology, and clinical implications. Postgrad Med. 2011;123(4):17–26. doi:10.3810/pgm.2011.07.2300
73. Ghasemi R, Haeri A, Dargahi L, Mohamed Z, Ahmadiani A. Insulin in the brain: sources, localization and functions. mol Neurobiol. 2013;47(1):145–171. doi:10.1007/s12035-012-8339-9
74. Nelson Thomas J, Sun MK, Hongpaisan J, Alkon DL. Insulin, PKC signaling pathways and synaptic remodeling during memory storage and neuronal repair. Eur J Pharmacol. 2008;585(1):76–87. doi:10.1016/j.ejphar.2008.01.051
75. Dou JT, Chen M, Dufour F, Alkon DL, Zhao WQ. Insulin receptor signaling in long-term memory consolidation following spatial learning. Learning & Memory. 2005;12(6):646–655. doi:10.1101/lm.88005
76. Lee J, Kim J, Shin SA, et al. Moderating effect of insulin resistance on the relationship between gray matter volumes and cognitive function. J Clin Med. 2018;7(11):413. doi:10.3390/jcm7110413
77. Kim H-G. Cognitive dysfunctions in individuals with diabetes mellitus. Yeungnam Univ J Med. 2019;36(3):183–191. doi:10.12701/yujm.2019.00255
78. Wei B, Dong Q, Ma J, Zhang A. The association between triglyceride-glucose index and cognitive function in nondiabetic elderly: NHANES 2011–2014. Lipids Health Dis. 2023;22(1):188. doi:10.1186/s12944-023-01959-0
79. Li S, Deng X, Zhang Y. The triglyceride-glucose index is associated with longitudinal cognitive decline in a middle-aged to elderly population: a cohort study. J Clin Med. 2022;11(23):7153. doi:10.3390/jcm11237153
80. He R, Zheng R, Li J, et al. Individual and combined associations of glucose metabolic components with cognitive function modified by obesity. Front Endocrinol. 2021:12. doi:10.3389/fendo.2021.769120.
81. Wang K, Xu L, Liu L, Zhan S, Wang S, Song Y. Sex differences in the association between the change in triglyceride-glucose index and cognitive decline: a population-based cohort study. J Affect Disord. 2022;316:42–49. doi:10.1016/j.jad.2022.08.014
82. Lim A, Krajina K, Marsland AL. Peripheral inflammation and cognitive aging. Mod Trends Pharmacopsych. 2013;28:175–187. doi:10.1159/000346362
83. Jin R, Chan AKY, Wu J, Lee TMC. Relationships between inflammation and age-related neurocognitive changes. Int J mol Sci. 2022;23(20):12573. doi:10.3390/ijms232012573
84. Tan S, Chen W, Kong G, Wei L, Xie Y. Peripheral inflammation and neurocognitive impairment: correlations, underlying mechanisms, and therapeutic implications. Front Aging Neurosci. 2023;15:1305790. doi:10.3389/fnagi.2023.1305790
85. Gu Y, Vorburger R, Scarmeas N, et al. Circulating inflammatory biomarkers in relation to brain structural measurements in a non-demented elderly population. Brain Behav Immun. 2017;65:150–160. doi:10.1016/j.bbi.2017.04.022
86. Merten N, Fischer ME, Pinto AA, Chappell RJ, Schubert CR. Lifestyle and factors of vascular and metabolic health and inflammation are associated with sensorineural-neurocognitive aging in older adults. Front Epidemiol. 2023;3:1299587. doi:10.3389/fepid.2023.1299587
87. Kalair A, Pavan M, Alpert N, Ghaffari S, Taioli E. Blood inflammatory markers and mortality in the US population: a health and retirement survey (HRS) analysis. PLoS One. 2023;18(10):e0293027. doi:10.1371/journal.pone.0293027
88. Stephan Y, Sutin AR, Luchetti M, Terracciano A. The prospective relationship between subjective aging and inflammation: evidence from the health and retirement study. Psychophysiology. 2023;60(2):e14177. doi:10.1111/psyp.14177
89. Oberlin LE, Erickson KI, Mackey R, et al. Peripheral inflammatory biomarkers predict the deposition and progression of amyloid-β in cognitively unimpaired older adults. Brain Behav Immun. 2021;95:178–189. doi:10.1016/j.bbi.2021.03.015
90. Mendelson D, Mizrahi R, Lepage M, Lavigne KM. C-Reactive protein and cognition: mediation analyses with brain morphology in the UK biobank. Brain Behav Immun Health. 2023;31:100664. doi:10.1016/j.bbih.2023.100664
91. Li X, Ma L. From biological aging to functional decline: insights into chronic inflammation and intrinsic capacity. Ageing Res Rev. 2024;93:102175. doi:10.1016/j.arr.2023.102175
92. Donato AJ, Morgan RG, Walker AE, Lesniewski LA. Cellular and molecular biology of aging endothelial cells. J mol Cell Cardiol. 2015;89(Pt B):122–135. doi:10.1016/j.yjmcc.2015.01.021
93. Jia G, Aroor AR, Jia C, Sowers JR. Endothelial cell senescence in aging-related vascular dysfunction. Biochim Biophys Acta mol Basis Dis. 2019;1865(7):1802–1809. doi:10.1016/j.bbadis.2018.08.008
94. Donato AJ, Machin DR, Lesniewski LA. Mechanisms of dysfunction in the aging vasculature and role in age-related disease. Circ Res. 2018;123(7):825–848. doi:10.1161/CIRCRESAHA.118.312563
95. Radi R. Peroxynitrite, a stealthy biological oxidant. J Biol Chem. 2013;288(37):26464–26472. doi:10.1074/jbc.R113.472936
96. Radi R. Oxygen radicals, nitric oxide, and peroxynitrite: redox pathways in molecular medicine. Proc Natl Acad Sci USA. 2018;115(23):5839–5848. doi:10.1073/pnas.1804932115
97. Virdis A, Duranti E, Rossi C, et al. Tumour necrosis factor-alpha participates on the endothelin-1/nitric oxide imbalance in small arteries from obese patients: role of perivascular adipose tissue. Eur Heart J. 2015;36(13):784–794. doi:10.1093/eurheartj/ehu072
98. Nilsson PM. Adiposity and vascular aging: indication for weight loss? Hypertension. 2015;66(2):270–272. doi:10.1161/HYPERTENSIONAHA.115.05621
99. Ungvari Z, Tarantini S, Donato AJ, Galvan V, Csiszar A. Mechanisms of Vascular Aging. Circ Res. 2018;123(7):849–867. doi:10.1161/CIRCRESAHA.118.311378
100. Selim M, Jones R, Novak P, Zhao P, Novak V. The effects of body mass index on cerebral blood flow velocity. Clin Auton Res. 2008;18(6):331–338. doi:10.1007/s10286-008-0490-z
101. Thorin-Trescases N, de Montgolfier O, Pinçon A, et al. Impact of pulse pressure on cerebrovascular events leading to age-related cognitive decline. Am J Physiol Heart Circ Physiol. 2018;314(6):H1214–H1224. doi:10.1152/ajpheart.00637.2017
102. Alosco ML, Spitznagel MB, Raz N, et al. Obesity interacts with cerebral hypoperfusion to exacerbate cognitive impairment in older adults with heart failure. Cerebrovasc Dis Extra. 2012;2(1):88–98. doi:10.1159/000343222
103. Alosco ML, Brickman AM, Spitznagel MB, et al. Higher BMI is associated with reduced brain volume in heart failure. BMC Obes. 2014;1(1):4. doi:10.1186/2052-9538-1-4
104. 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. 2011;19(5):1095–1097. doi:10.1038/oby.2011.16
105. Xing CY, Tarumi T, Liu J, et al. Distribution of cardiac output to the brain across the adult lifespan. J Cerebral Blood Flow Metabol. 2017;37(8):2848–2856. doi:10.1177/0271678X16676826
106. Parr EB, Coffey VG, Hawley JA. ‘Sarcobesity’: a metabolic conundrum. Maturitas. 2013;74(2):109–113. doi:10.1016/j.maturitas.2012.10.014
107. Wilkinson TJ, Watson EL, Vadaszy N, Baker LA, Viana JL, Smith AC. Response of the oxygen uptake efficiency slope to exercise training in patients with chronic kidney disease. Kidney Res Clin Pract. 2020;39(3):305–317. doi:10.23876/j.krcp.20.032
108. Zeng Z, Liang J, Wu L, Zhang H, Lv J, Chen N. Exercise-induced autophagy suppresses sarcopenia through Akt/mTOR and Akt/FoxO3a signal pathways and AMPK-mediated mitochondrial quality control. Front Physiol. 2020;11:11. doi:10.3389/fphys.2020.583478
109. Cheng X, Yang Z. Effect of resistance training on inflammatory markers in middle-aged and older adults: a meta-analysis. Arch Gerontol Geriatr. 2024;126:105536. doi:10.1016/j.archger.2024.105536
110. Vints WAJ, Levin O, Fujiyama H, Verbunt J, Masiulis N. Exerkines and long-term synaptic potentiation: mechanisms of exercise-induced neuroplasticity. Front Neuroendocrinol. 2022;66:100993. doi:10.1016/j.yfrne.2022.100993
111. Khalafi M, Sakhaei MH, Rosenkranz SK, Symonds ME. Impact of concurrent training versus aerobic or resistance training on cardiorespiratory fitness and muscular strength in middle-aged to older adults: a systematic review and meta-analysis. Physiol Behav. 2022;254:113888. doi:10.1016/j.physbeh.2022.113888
112. Maleki S, Hendrikse J, Chye Y, et al. Associations of cardiorespiratory fitness and exercise with brain white matter in healthy adults: a systematic review and meta-analysis. Brain Imaging Behav. 2022;16(5):2402–2425. doi:10.1007/s11682-022-00693-y
113. Chodzko-Zajko WJ, Proctor DN, Singh MA, et al.; American College of Sports Medicine. American college of sports medicine position stand. Exercise and physical activity for older adults. Med Sci Sports Exerc. 2009;41(7):1510–1530. doi:10.1249/MSS.0b013e3181a0c95c.
114. Lee PG, Jackson EA, Richardson CR. Exercise prescriptions in older adults. Am Fam Physician. 2017;95(7):425–432.
115. Northey JM, Cherbuin N, Pumpa KL, Smee DJ, Rattray B. Exercise interventions for cognitive function in adults older than 50: a systematic review with meta-analysis. Br J Sports Med. 2018;52(3):154–160. doi:10.1136/bjsports-2016-096587
116. Bangsbo J, Blackwell J, Boraxbekk CJ, et al. Copenhagen consensus statement 2019: physical activity and ageing. Br J Sports Med. 2019;53(14):856–858. doi:10.1136/bjsports-2018-100451
117. Izquierdo M, Merchant RA, Morley JE, et al. International exercise recommendations in older adults (ICFSR): expert consensus guidelines. J Nutr Health Aging. 2021;25(7):824–853. doi:10.1007/s12603-021-1665-8
118. World Health Organization. Physical activity. 2022. Available from: https://www.who.int/news-room/fact-sheets/detail/physical-activity.
119. Pan M, Ying B, Lai Y, Kuan G. Status and influencing factors of physical exercise among college students in china: a systematic review. Int J Environ Res Public Health. 2022;19(20):13465. doi:10.3390/ijerph192013465
120. Royse LA, Baker BS, Warne-Griggs MD, et al. “It’s not time for us to sit down yet”: how group exercise programs can motivate physical activity and overcome barriers in inactive older adults. Inter J Qual Stud Health Well-Being. 2023;18(1). doi:10.1080/17482631.2023.2216034
121. Gibala MJ. High-intensity interval training: a time-efficient strategy for health promotion? Curr Sports Med Rep. 2007;6(4):211–213.
122. Burgomaster KA, Howarth KR, Phillips SM, et al. Similar metabolic adaptations during exercise after low volume sprint interval and traditional endurance training in humans. J Physiol. 2008;586(1):151–160. doi:10.1113/jphysiol.2007.142109
123. Gibala MJ, McGee SL, Garnham AP, Howlett KF, Snow RJ, Hargreaves M. Brief intense interval exercise activates AMPK and p38 MAPK signaling and increases the expression of PGC-1alpha in human skeletal muscle. J Appl Physiol. 2009;106(3):929–934. doi:10.1152/japplphysiol.90880.2008
124. Little JP, Safdar A, Wilkin GP, Tarnopolsky MA, Gibala MJ. A practical model of low-volume high-intensity interval training induces mitochondrial biogenesis in human skeletal muscle: potential mechanisms. J Physiol. 2010;588(6):1011–1022. doi:10.1113/jphysiol.2009.181743
125. Larsen JK, Kruse R, Sahebekhtiari N, et al. High-throughput proteomics uncovers exercise training and type 2 diabetes–induced changes in human white adipose tissue. Sci Adv. 2023;9(48):eadi7548. doi:10.1126/sciadv.adi7548
126. Farrell CM, McNeilly AD, Hapca S, et al. High intensity interval training as a novel treatment for impaired awareness of hypoglycaemia in people with type 1 diabetes (HIT4HYPOS): a randomised parallel-group study. Diabetologia. 2024;67(2):392–402. doi:10.1007/s00125-023-06051-x
127. Sawyer BJ, Tucker WJ, Bhammar DM, Ryder JR, Sweazea KL, Gaesser GA. Effects of high-intensity interval training and moderate-intensity continuous training on endothelial function and cardiometabolic risk markers in obese adults. J Appl Physiol. 2016;121(1):279–288. doi:10.1152/japplphysiol.00024.2016
128. O’Brien MW, Johns JA, Robinson SA, Bungay A, Mekary S, Kimmerly DS. Impact of high-intensity interval training, moderate-intensity continuous training, and resistance training on endothelial function in older adults. Med Sci Sports Exerc. 2020;52(5):1057–1067. doi:10.1249/MSS.0000000000002226
129. Hendy AM, Andrushko JW, Della Gatta PA, Teo WP. Acute effects of high-intensity aerobic exercise on motor cortical excitability and inhibition in sedentary adults. Front Psychol. 2022;13:814633. doi:10.3389/fpsyg.2022.814633
130. Neva JL, Greeley B, Chau B, et al. Acute high-intensity interval exercise modulates corticospinal excitability in older adults. Med Sci Sports Exerc. 2022;54(4):673–682. doi:10.1249/MSS.0000000000002839
131. Marriott CFS, Petrella AFM, Marriott ECS, Boa Sorte Silva NC, Petrella RJ. High-intensity interval training in older adults: a scoping review. Sports Med Open. 2021;7(1):49. doi:10.1186/s40798-021-00344-4
132. Taylor JL, Holland DJ, Keating SE, et al. Short-term and long-term feasibility, safety, and efficacy of high-intensity interval training in cardiac rehabilitation. JAMA Cardiol. 2020;5(12):1382. doi:10.1001/jamacardio.2020.3511
133. Carl DL, Boyne P, Rockwell B, et al. Preliminary safety analysis of high-intensity interval training (HIIT) in persons with chronic stroke. Appl Phys Nutri Metabol. 2017;42(3):311–318. doi:10.1139/apnm-2016-0369
134. Izquierdo M, Ibañez J, HAkkinen K, Kraemer WJ, Larrión JL, Gorostiaga EM. Once weekly combined resistance and cardiovascular training in healthy older men. Med Sci Sports Exerc. 2004;36(3):435–443. doi:10.1249/01.mss.0000117897.55226.9a
135. Sillanpää E, Häkkinen A, Nyman K, et al. Body composition and fitness during strength and/or endurance training in older men. Med Sci Sports Exerc. 2008;40(5):950–958. doi:10.1249/MSS.0b013e318165c854
136. Morcillo-Losa JA, Díaz-Martínez MDP, Ceylan Hİ, Moreno-Vecino B, Bragazzi NL, Párraga Montilla J. Effects of high-intensity interval training on muscle strength for the prevention and treatment of sarcopenia in older adults: a systematic review of the literature. J Clin Med. 2024;13(5):1299. doi:10.3390/jcm13051299
137. Romero-Arenas S, Martínez-Pascual M, Alcaraz PE. Impact of resistance circuit training on neuromuscular, cardiorespiratory and body composition adaptations in the elderly. Aging Dis. 2013;4(5):256–263. doi:10.14336/AD.2013.0400256
138. Hood MS, Little JP, Tarnopolsky MA, Myslik F, Gibala MJ. Low-volume interval training improves muscle oxidative capacity in sedentary adults. Med Sci Sports Exerc. 2011;43(10):1849–1856. doi:10.1249/MSS.0b013e3182199834
© 2025 The Author(s). This work is published and licensed by Dove Medical Press Limited. The
full terms of this license are available at https://www.dovepress.com/terms.php
and incorporate the Creative Commons Attribution
- Non Commercial (unported, 4.0) License.
By accessing the work you hereby accept the Terms. Non-commercial uses of the work are permitted
without any further permission from Dove Medical Press Limited, provided the work is properly
attributed. For permission for commercial use of this work, please see paragraphs 4.2 and 5 of our Terms.