Exercise Intervention for Alzheimer’s Disease: Unraveling Neurobiological Mechanisms and Assessing Effects
Abstract
:1. Introduction
2. Exercise Regulates Bone-Derived Factors to Improve Symptoms of Alzheimer’s Disease through “Bone–Brain Crosstalk”
- OCN produced by osteoblasts enhances the synthesis and secretion levels of monoamine neurotransmitters (MN) while inhibiting the secretion and synthesis of γ-aminobutyric acid (GABA) [27], thereby improving learning, memory, and brain metabolic function. OCN also increases the secretion of brain-derived neurotrophic factor (BDNF), which subsequently improves the impact on neurodevelopment and function, reduces inflammatory responses, inhibits cell apoptosis, and suppresses anxiety and depressive behaviors. Exercise-induced skeletal stimulation can regulate OCN secretion levels, allowing it to accumulate in the brainstem, thalamus, and hypothalamus through the blood–brain barrier [28]. It binds specifically to neurons in the brain, influencing neurotransmitter synthesis and signal transmission. OCN further promotes the organism’s learning and memory abilities, increases neurotransmitter synthesis, and improves hippocampal development, thus enhancing cognitive function and inhibiting the development of anxiety emotions [29]. Additionally, OCN directly prevents neuronal apoptosis in the hippocampus, thereby protecting cognitive functions such as spatial learning and memory [30]. The binding of OCN and Gpr158 in CA3 pyramidal neurons of the hippocampus enhances the synthesis and secretion of neurotransmitters such as 5-hydroxytryptamine (5-HT), dopamine (DA), and norepinephrine (NE), while inhibiting GABA synthesis, thereby improving spatial learning and memory abilities [31]. The total osteocalcin (tOCN) content in the body primarily consists of carboxylated osteocalcin (cOCN) and undercarboxylated osteocalcin (uOCN), which is biologically active. Exercise stimulation can increase the overall activity level of uOCN. Additionally, exercise promotes the production of active uOCN by increasing muscle secretion of interleukin-6 (IL-6) [32,33]. Higher circulating OCN levels can significantly regulate the prevention of age-related cognitive decline. Exercise improves skeletal secretion capacity, increases circulating OCN levels in the body, and induces neuronal plasticity. Improved cognitive function can be achieved through OCN signaling [34,35,36,37].
- The SOST, synthesized by osteocytes, binds to LDH receptor-related proteins 4/5/6 (Lrp4/5/6) to antagonize the Wnt/β-catenin signaling pathway. Intracerebral Wnt/β-catenin signaling is involved in maintaining neurogenesis, synaptic plasticity, and blood–brain barrier integrity. Wnt/β-catenin signaling regulates synaptic plasticity, and memory processes, inhibits neurotoxicity caused by Aβ, and participates in tau protein phosphorylation and learning and memory. Dysfunction of Wnt/β-catenin can lead to the production and aggregation of Aβ, thereby triggering the onset of AD [38,39]. Wnt/β-catenin signaling is considered a potential mechanism for treating AD. Among the mechanisms that affect brain Wnt/β-catenin signaling, Dickkopf-related protein 1 (Dkk1) can inhibit Wnt signaling by inducing LRP5/6, thereby blocking the Wnt signal-induced synaptic disassembly process [40]. Dkk1 is overexpressed in AD patients and in the brains of AD mice, and its expression levels can be effectively reduced through exercise [41]. Exercise stimulation activates the Wnt signaling pathway in APP/PS1 rats, improving synaptic dysfunction and promoting synaptic plasticity and neurogenesis in the hippocampus [42,43]. In the brain mechanism, Aβ activates glycogen synthase kinase-3β (GSK-3β) and reduces β-catenin activity, blocking the Wnt/β-catenin signaling pathway. Exercise attenuates the secretion of SOST from the bone, thereby effectively improving the occurrence and development of AD through the Wnt/β-catenin pathway. Research has found that the expression levels of SOST significantly decrease in 8-week-old mice after 5 weeks of exercise. In human studies, both highly active males and females exhibit lower levels of SOST secretion and circulation [44]. Exercise activates Wnt/β-catenin signaling in the brain, leading to an increase in β-catenin levels within brain cells, which tends to stabilize. Rats subjected to treadmill exercise for 30 min/day, 5 days/week, for a total of 12 weeks, show increased expression levels of Wnt3, reduced expression of GSK-3β, activation of the Wnt signaling pathway, increased neurogenesis, and alleviated memory loss associated with AD [45]. After exercise, the secretion of SOST from the bone decreases, resulting in a reduced amount circulating through the blood–brain barrier. This reduction weakens its binding with Lrp4/5/6, further activating the Wnt/β-catenin signaling pathway, ultimately promoting synaptic plasticity and neurogenesis, and mitigating AD levels and biological mechanisms [46].
- OPN is a matrix cell immune regulator highly expressed by monocytes in the bone marrow, and it can regulate immune cell migration while responding to brain injuries [47]. Compared to chronic patients, AD patients have higher levels of OPN protein in their cerebrospinal fluid and plasma, suggesting that OPN plays a role in protecting neurons regulating brain diseases, and repairing neurodegenerative diseases [41]. OPN is involved in the process of brain remodeling, promoting myelination formation and regeneration [48]. Moderate-intensity (85% VO2 max) treadmill and weight-bearing running interventions for 5 weeks in 2-month-old male C57BL/6 mice can improve mouse bone mineral density (BMD), cortical bone mass, and osteogenic ability, as well as increase the expression and secretion levels of OCN and OPN in osteoblasts [49,50]. OPN enters the brain through the blood–brain barrier for regulation. OPN is mainly involved in AD neuron loss, degeneration, and the death of neurons. It plays a role in AD neuron abnormalities and re-entry into the cell cycle and/or myelin regenerative processes [51]. Studies have found that the increased expression of OPN is closely related to Aβ deposition in the cone neurons of AD patients and the brains of APP/PS1 mice [52]. In the process of OPN regulating AD, OPN binds to downstream receptor CD44 to further exert neuroprotective and remodeling activities. Due to the important role of the OPN–CD44 complex in neuroprotection and remodeling, enhancing OPN expression can inhibit neuro-damaging phenomena in AD [53,54]. Exercise stimulates OPN secretion and increases its expression levels in the bone. Strengthening OPN expression can better inhibit neuro damage in AD. In the Aβ clearance mechanism of AD, OPN can also regulate macrophage immune resistance to Aβ deposition. OPN promotes the phagocytosis of Aβ fibrils and related receptors, changes cell morphology, reduces inducible nitric oxide synthase (iNOS) levels, and enhances the anti-inflammatory effects of interleukin-10(IL-10) and matrix metalloproteinase 9 (MMP-9) [47] (Figure 1).
3. Exercise-Induced Modulation of Muscle Factors Improves Alzheimer’s Disease Symptoms through “Muscle–Brain Crosstalk”
- BDNF, a neurotrophic factor, plays a crucial role in neurogenesis and synaptic plasticity. Its low levels are linked to AD, with studies indicating reduced BDNF in AD patients and animal models [55,56,57]. BDNF can cross the blood–brain barrier, enhancing neurotrophic production in the hippocampus and supporting cognitive function [58,59]. Exercise has been shown to increase BDNF secretion in muscle tissue, correlating with exercise intensity and leading to cognitive improvements in both humans and animal models of AD [60,61,62]. Long-term exercise can elevate baseline BDNF levels, with evidence of hippocampal growth and better spatial memory after a year of aerobic exercise [63,64]. BDNF acts through the TrkB receptor, activating pathways like MAPK and PI3K, which are important for neuronal survival and plasticity [65]. It also modulates amyloid-beta (Aβ) production by enhancing alpha-secretase activity and reducing BACE1 levels, mitigating Aβ-induced toxicity [66,67,68,69,70]. Thus, exercise-induced BDNF not only offers direct neuroprotective effects but may also contribute to the reduction in AD pathology. These findings illustrate that BDNF plays a dual role in maintaining brain health and combating AD. By promoting neurogenesis and modulating pathological processes associated with AD, BDNF emerges as a promising therapeutic target. The research underscores the importance of long-term exercise, not only for its direct elevation of BDNF levels but also for its potential benefits in cognitive enhancement and the deceleration of AD pathology. Future studies may focus on how to maximize the impact of exercise on BDNF and how to translate these findings into concrete preventative and therapeutic strategies.
- Irisin, a myokine released during exercise, is produced by the cleavage of FNDC5 and affects energy metabolism and neuroprotection [71]. It promotes the browning of white adipose tissue, enhances insulin sensitivity, and is implicated in the pathophysiology of AD, with lower levels observed in AD models [62,72,73]. Exercise activates PGC-1α, up-regulating FNDC5/Irisin and reducing amyloid-beta (Aβ) production [74,75]. Irisin levels rise in response to exercise, potentially improving cognitive functions via various mechanisms, including promoting neuronal growth and reducing inflammation [76,77,78,79,80,81,82,83,84,85,86,87,88,89]. Irisin also influences the production of BDNF, a factor crucial for neuronal health [90,91,92]. Considering AD’s metabolic aspects, often termed “type 3 diabetes,” Irisin’s role in energy homeostasis and insulin sensitivity is of particular interest, offering a potential therapeutic avenue for AD and diabetes [93,94,95,96]. In summary, Irisin emerges as a promising molecule linking exercise to metabolic and cognitive health. Its multifaceted role in energy regulation, neuroprotection, and potential to mitigate AD symptoms underscores the need for further research. Future studies should focus on the therapeutic potential of Irisin, aiming to harness its benefits for treating metabolic and neurodegenerative disorders in aging populations.
- IL-6, a cytokine released from muscles during exercise, plays complex roles in the body and is linked to AD. High baseline levels of IL-6 are associated with increased risk of AD and cognitive decline in the elderly [97,98,99,100,101]. This cytokine has dual effects in AD: it can exacerbate neuronal damage by enhancing APP synthesis and Aβ toxicity, yet it also supports neurogenesis and gliogenesis through specific signaling pathways [102,103,104]. Acute exercise raises IL-6 levels, which may help regulate inflammation and promote neuroprotective responses [105,106]. Chronic exercise, conversely, is associated with reduced resting IL-6 levels and may prevent its detrimental effects in the brain [107,108,109]. In essence, while acute exercise triggers a beneficial IL-6 response that protects the brain, chronic exercise lowers the baseline of IL-6, potentially reducing chronic inflammation and the risk of neurodegenerative diseases. This suggests that exercise, both acute and chronic, can be a strategic approach to modulate IL-6 levels for brain health, emphasizing the importance of physical activity in the prevention and management of AD.
- IGF-1 plays a crucial role in CNS function by enhancing synaptic plasticity and density, with a decline in IGF-1 associated with AD cognitive symptoms [110,111]. Exercise-induced IGF-1 secretion from muscles benefits brain health, potentially reducing AD pathology through mechanisms like inhibiting Aβ production and tau phosphorylation via the IRS1/PI3K/Akt/mTOR pathway [112]. It also supports hippocampal neurogenesis and BDNF regulation, integral to neuronal growth and differentiation [113,114,115]. Similarly, VEGF, a key angiogenesis regulator, promotes vascular and neural health. Its overexpression in rodent models boosts hippocampal angiogenesis and neurogenesis, while its inhibition can negate exercise-induced neurogenic benefits, highlighting the importance of muscle–brain crosstalk [116,117,118,119]. These studies highlight how exercise promotes brain health by stimulating the secretion of molecules such as IGF-1 and VEGF, particularly with respect to neuroprotection and neurogenesis associated with AD. These findings reveal the biochemical links between physical activity and brain health, offering potential strategies for the prevention and treatment of AD. By enhancing the health of the vascular and nervous systems, exercise not only aids in improving cognitive functions but may also slow the progression of AD (Figure 1).
4. Exercise Regulates the Gut Microbiota and Improves Symptoms of Alzheimer’s Disease through “Gut–Brain Crosstalk”
5. Effects of Different Exercise Intervention Programs on Symptom Improvement in Alzheimer’s Disease
5.1. Effects of Different Types of Exercise on Symptom Improvement in Alzheimer’s Disease
5.1.1. Aerobic Exercise
5.1.2. Resistance Exercise
5.1.3. Multimodal Exercises Combination
5.2. Effects of Different Exercise Intensities and Frequencies on Symptom Improvement in Alzheimer’s Disease
5.3. Characteristics and Risk of Bias in Exercise Intervention Studies for Alzheimer’s Disease
6. The Role of Artificial Intelligence and Neuroimaging Technologies in Exercise Intervention for Alzheimer’s Disease
7. Discussion
8. Conclusions
Author Contributions
Funding
Institutional Review Board Statement
Informed Consent Statement
Data Availability Statement
Conflicts of Interest
References
- Breijyeh, Z.; Karaman, R. Comprehensive Review on Alzheimer’s Disease: Causes and Treatment. Molecules 2020, 25, 5789. [Google Scholar] [CrossRef]
- Long, J.M.; Holtzman, D.M. Alzheimer Disease: An Update on Pathobiology and Treatment Strategies. Cell 2019, 179, 312–339. [Google Scholar] [CrossRef]
- De la Rosa, A.; Olaso-Gonzalez, G.; Arc-Chagnaud, C.; Millan, F.; Salvador-Pascual, A.; Garcia-Lucerga, C.; Blasco-Lafarga, C.; Garcia-Dominguez, E.; Carretero, A.; Correas, A.G.; et al. Physical exercise in the prevention and treatment of Alzheimer’s disease. J. Sport Health Sci. 2020, 9, 394–404. [Google Scholar] [CrossRef] [PubMed]
- Weidling, I.W.; Swerdlow, R.H. Mitochondria in Alzheimer’s disease and their potential role in Alzheimer’s proteostasis. Exp. Neurol. 2020, 330, 113321. [Google Scholar] [CrossRef] [PubMed]
- Clay, E.; Zhou, J.; Yi, Z.M.; Zhai, S.; Toumi, M. Economic burden for Alzheimer’s disease in China from 2010 to 2050: A modelling study. J. Mark. Access Health Policy 2019, 7, 1667195. [Google Scholar] [CrossRef] [PubMed]
- Jia, J.; Wei, C.; Chen, S.; Li, F.; Tang, Y.; Qin, W.; Zhao, L.; Jin, H.; Xu, H.; Wang, F.; et al. The cost of Alzheimer’s disease in China and re-estimation of costs worldwide. Alzheimer’s Dement. 2018, 14, 483–491. [Google Scholar] [CrossRef] [PubMed]
- 2023 Alzheimer’s disease facts and figures. Alzheimer’s Dement. 2023, 19, 1598–1695. [CrossRef] [PubMed]
- Chiti, F.; Dobson, C.M. Protein Misfolding, Amyloid Formation, and Human Disease: A Summary of Progress over the Last Decade. Annu. Rev. Biochem. 2017, 86, 27–68. [Google Scholar] [CrossRef]
- Berriman, J.; Serpell, L.C.; Oberg, K.A.; Fink, A.L.; Goedert, M.; Crowther, R.A. Tau filaments from human brain and from in vitro assembly of recombinant protein show cross-beta structure. Proc. Natl. Acad. Sci. USA 2003, 100, 9034–9038. [Google Scholar] [CrossRef]
- Lyketsos, C.G.; Carrillo, M.C.; Ryan, J.M.; Khachaturian, A.S.; Trzepacz, P.; Amatniek, J.; Cedarbaum, J.; Brashear, R.; Miller, D.S. Neuropsychiatric symptoms in Alzheimer’s disease. Alzheimer’s Dement. 2011, 7, 532–539. [Google Scholar] [CrossRef]
- Martorana, A.; Koch, G. “Is dopamine involved in Alzheimer’s disease?”. Front. Aging Neurosci. 2014, 6, 252. [Google Scholar] [CrossRef]
- Luebke, J.I.; Weaver, C.M.; Rocher, A.B.; Rodriguez, A.; Crimins, J.L.; Dickstein, D.L.; Wearne, S.L.; Hof, P.R. Dendritic vulnerability in neurodegenerative disease: Insights from analyses of cortical pyramidal neurons in transgenic mouse models. Brain Struct. Funct. 2010, 214, 181–199. [Google Scholar] [CrossRef]
- Calsolaro, V.; Edison, P. Neuroinflammation in Alzheimer’s disease: Current evidence and future directions. Alzheimer’s Dement. 2016, 12, 719–732. [Google Scholar] [CrossRef]
- Motta, C.; Finardi, A.; Toniolo, S.; Di Lorenzo, F.; Scaricamazza, E.; Loizzo, S.; Mercuri, N.B.; Furlan, R.; Koch, G.; Martorana, A. Protective Role of Cerebrospinal Fluid Inflammatory Cytokines in Patients with Amnestic Mild Cognitive Impairment and Early Alzheimer’s Disease Carrying Apolipoprotein E4 Genotype. J. Alzheimer’s Dis. 2020, 76, 681–689. [Google Scholar] [CrossRef] [PubMed]
- Mahley, R.W.; Weisgraber, K.H.; Huang, Y. Apolipoprotein E4: A causative factor and therapeutic target in neuropathology, including Alzheimer’s disease. Proc. Natl. Acad. Sci. USA 2006, 103, 5644–5651. [Google Scholar] [CrossRef] [PubMed]
- Cummings, J.; Zhou, Y.; Lee, G.; Zhong, K.; Fonseca, J.; Cheng, F. Alzheimer’s disease drug development pipeline: 2023. Alzheimer’s Dement. 2023, 9, e12385. [Google Scholar] [CrossRef] [PubMed]
- McKhann, G.M.; Knopman, D.S.; Chertkow, H.; Hyman, B.T.; Jack, C.R., Jr.; Kawas, C.H.; Klunk, W.E.; Koroshetz, W.J.; Manly, J.J.; Mayeux, R.; et al. The diagnosis of dementia due to Alzheimer’s disease: Recommendations from the National Institute on Aging-Alzheimer’s Association workgroups on diagnostic guidelines for Alzheimer’s disease. Alzheimer’s Dement. 2011, 7, 263–269. [Google Scholar] [CrossRef] [PubMed]
- Hampel, H.; Mesulam, M.M.; Cuello, A.C.; Farlow, M.R.; Giacobini, E.; Grossberg, G.T.; Khachaturian, A.S.; Vergallo, A.; Cavedo, E.; Snyder, P.J.; et al. The cholinergic system in the pathophysiology and treatment of Alzheimer’s disease. Brain 2018, 141, 1917–1933. [Google Scholar] [CrossRef]
- Agrawal, M.; Saraf, S.; Saraf, S.; Antimisiaris, S.G.; Chougule, M.B.; Shoyele, S.A.; Alexander, A. Nose-to-brain drug delivery: An update on clinical challenges and progress towards approval of anti-Alzheimer drugs. J. Control. Release 2018, 281, 139–177. [Google Scholar] [CrossRef]
- Ren, J.; Xiao, H. Exercise for Mental Well-Being: Exploring Neurobiological Advances and Intervention Effects in Depression. Life 2023, 13, 1505. [Google Scholar] [CrossRef]
- Song, D.; Yu, D. Effects of a moderate-intensity aerobic exercise programme on the cognitive function and quality of life of community-dwelling elderly people with mild cognitive impairment: A randomised controlled trial. Int. J. Nurs. Stud. 2019, 93, 97–105. [Google Scholar] [CrossRef]
- Baatile, J.; Langbein, W.E.; Weaver, F.; Maloney, C.; Jost, M.B. Effect of exercise on perceived quality of life of individuals with Parkinson’s disease. J. Rehabil. Res. Dev. 2000, 37, 529–534. [Google Scholar] [PubMed]
- Zarza-Rebollo, J.A.; Molina, E.; López-Isac, E.; Pérez-Gutiérrez, A.M.; Gutiérrez, B.; Cervilla, J.A.; Rivera, M. Interaction Effect between Physical Activity and the BDNF Val66Met Polymorphism on Depression in Women from the PISMA-ep Study. Int. J. Environ. Res. Public Health 2022, 19, 2068. [Google Scholar] [CrossRef] [PubMed]
- López-Ortiz, S.; Pinto-Fraga, J.; Valenzuela, P.L.; Martín-Hernández, J.; Seisdedos, M.M.; García-López, O.; Toschi, N.; Di Giuliano, F.; Garaci, F.; Mercuri, N.B.; et al. Physical Exercise and Alzheimer’s Disease: Effects on Pathophysiological Molecular Pathways of the Disease. Int. J. Mol. Sci. 2021, 22, 2897. [Google Scholar] [CrossRef] [PubMed]
- Iso-Markku, P.; Kujala, U.M.; Knittle, K.; Polet, J.; Vuoksimaa, E.; Waller, K. Physical activity as a protective factor for dementia and Alzheimer’s disease: Systematic review, meta-analysis and quality assessment of cohort and case-control studies. Br. J. Sports Med. 2022, 56, 701–709. [Google Scholar] [CrossRef]
- Meng, Q.; Lin, M.S.; Tzeng, I.S. Relationship Between Exercise and Alzheimer’s Disease: A Narrative Literature Review. Front. Neurosci. 2020, 14, 131. [Google Scholar] [CrossRef]
- Oury, F.; Khrimian, L.; Denny, C.A.; Gardin, A.; Chamouni, A.; Goeden, N.; Huang, Y.Y.; Lee, H.; Srinivas, P.; Gao, X.B.; et al. Maternal and offspring pools of osteocalcin influence brain development and functions. Cell 2013, 155, 228–241. [Google Scholar] [CrossRef] [PubMed]
- Shan, C.; Ghosh, A.; Guo, X.Z.; Wang, S.M.; Hou, Y.F.; Li, S.T.; Liu, J.M. Roles for osteocalcin in brain signalling: Implications in cognition- and motor-related disorders. Mol. Brain 2019, 12, 23. [Google Scholar] [CrossRef]
- Lu, B.; Zhang, T.; Yang, F. “Bone” in the Brain? Osteocalcin-Expressing Neurons in Adult Hippocampus Promote Neurogenesis and Suppress Anxiety. Biol. Psychiatry 2021, 89, 539–540. [Google Scholar] [CrossRef]
- Park, S.S.; Park, H.S.; Kim, C.J.; Kang, H.S.; Kim, D.H.; Baek, S.S.; Kim, T.W. Physical exercise during exposure to 40-Hz light flicker improves cognitive functions in the 3xTg mouse model of Alzheimer’s disease. Alzheimer’s Res. Ther. 2020, 12, 62. [Google Scholar] [CrossRef]
- Zhao, Y.; Suo, Y.; Yang, Z.; Hao, Y.; Li, W.; Su, Y.; Shi, Y.; Gao, Y.; Song, L.; Yin, X.; et al. Inspiration for the prevention and treatment of neuropsychiatric disorders: New insight from the bone-brain-axis. Brain Res. Bull. 2021, 177, 263–272. [Google Scholar] [CrossRef] [PubMed]
- Shimizu, N.; Saito, T.; Wada, N.; Hashimoto, M.; Shimizu, T.; Kwon, J.; Cho, K.J.; Saito, M.; Karnup, S.; de Groat, W.C.; et al. Molecular Mechanisms of Neurogenic Lower Urinary Tract Dysfunction after Spinal Cord Injury. Int. J. Mol. Sci. 2023, 24, 7885. [Google Scholar] [CrossRef]
- Nicolini, C.; Fahnestock, M.; Gibala, M.J.; Nelson, A.J. Understanding the Neurophysiological and Molecular Mechanisms of Exercise-Induced Neuroplasticity in Cortical and Descending Motor Pathways: Where Do We Stand? Neuroscience 2021, 457, 259–282. [Google Scholar] [CrossRef] [PubMed]
- Ahn, N.; Kim, K. Effects of 12-week exercise training on osteocalcin, high-sensitivity C-reactive protein concentrations, and insulin resistance in elderly females with osteoporosis. J. Phys. Ther. Sci. 2016, 28, 2227–2231. [Google Scholar] [CrossRef] [PubMed]
- Baria, M.R.; Miller, M.M.; Borchers, J.; Desmond, S.; Onate, J.; Magnussen, R.; Vasileff, W.K.; Flanigan, D.; Kaeding, C.; Durgam, S. High Intensity Interval Exercise Increases Platelet and Transforming Growth Factor-beta Yield in Platelet-Rich Plasma. PM&R 2020, 12, 1244–1250. [Google Scholar] [CrossRef]
- Nicolini, C.; Michalski, B.; Toepp, S.L.; Turco, C.V.; D’Hoine, T.; Harasym, D.; Gibala, M.J.; Fahnestock, M.; Nelson, A.J. A Single Bout of High-intensity Interval Exercise Increases Corticospinal Excitability, Brain-derived Neurotrophic Factor, and Uncarboxylated Osteolcalcin in Sedentary, Healthy Males. Neuroscience 2020, 437, 242–255. [Google Scholar] [CrossRef] [PubMed]
- Deemer, S.E.; Castleberry, T.J.; Irvine, C.; Newmire, D.E.; Oldham, M.; King, G.A.; Ben-Ezra, V.; Irving, B.A.; Biggerstaff, K.D. Pilot study: An acute bout of high intensity interval exercise increases 12.5 h GH secretion. Physiol. Rep. 2018, 6, e13563. [Google Scholar] [CrossRef]
- Bien-Ly, N.; Boswell, C.A.; Jeet, S.; Beach, T.G.; Hoyte, K.; Luk, W.; Shihadeh, V.; Ulufatu, S.; Foreman, O.; Lu, Y.; et al. Lack of Widespread BBB Disruption in Alzheimer’s Disease Models: Focus on Therapeutic Antibodies. Neuron 2015, 88, 289–297. [Google Scholar] [CrossRef]
- Inestrosa, N.C.; Tapia-Rojas, C.; Cerpa, W.; Cisternas, P.; Zolezzi, J.M. WNT Signaling Is a Key Player in Alzheimer’s Disease. In Pharmacology of the WNT Signaling System; Handbook of Experimental Pharmacology; Springer: Cham, Switzerland, 2021; Volume 269, pp. 357–382. [Google Scholar] [CrossRef]
- Purro, S.A.; Dickins, E.M.; Salinas, P.C. The secreted Wnt antagonist Dickkopf-1 is required for amyloid beta-mediated synaptic loss. J. Neurosci. 2012, 32, 3492–3498. [Google Scholar] [CrossRef]
- Gerosa, L.; Lombardi, G. Bone-to-Brain: A Round Trip in the Adaptation to Mechanical Stimuli. Front. Physiol. 2021, 12, 623893. [Google Scholar] [CrossRef]
- Vargas, J.Y.; Fuenzalida, M.; Inestrosa, N.C. In vivo activation of Wnt signaling pathway enhances cognitive function of adult mice and reverses cognitive deficits in an Alzheimer’s disease model. J. Neurosci. 2014, 34, 2191–2202. [Google Scholar] [CrossRef]
- Xiao, H.H.; Chen, J.C.; Li, H.; Li, R.H.; Wang, H.B.; Song, H.P.; Li, H.Y.; Shan, G.S.; Tian, Y.; Zhao, Y.M.; et al. Icarisid II rescues cognitive dysfunction via activation of Wnt/beta-catenin signaling pathway promoting hippocampal neurogenesis in APP/PS1 transgenic mice. Phytother. Res. 2022, 36, 2095–2108. [Google Scholar] [CrossRef]
- Gardinier, J.D.; Rostami, N.; Juliano, L.; Zhang, C. Bone adaptation in response to treadmill exercise in young and adult mice. Bone Rep. 2018, 8, 29–37. [Google Scholar] [CrossRef] [PubMed]
- Kim, D.Y.; Jung, S.Y.; Kim, K.; Kim, C.J. Treadmill exercise ameliorates Alzheimer disease-associated memory loss through the Wnt signaling pathway in the streptozotocin-induced diabetic rats. J. Exerc. Rehabil. 2016, 12, 276–283. [Google Scholar] [CrossRef] [PubMed]
- Xiang, T.; Zhang, S.; Cheng, N.; Ge, S.; Wen, J.; Xiao, J.; Wu, X. Oxidored-nitro domain-containing protein 1 promotes liver fibrosis by activating the Wnt/beta-catenin signaling pathway in vitro. Mol. Med. Rep. 2017, 16, 5050–5054. [Google Scholar] [CrossRef] [PubMed]
- Rentsendorj, A.; Sheyn, J.; Fuchs, D.T.; Daley, D.; Salumbides, B.C.; Schubloom, H.E.; Hart, N.J.; Li, S.; Hayden, E.Y.; Teplow, D.B.; et al. A novel role for osteopontin in macrophage-mediated amyloid-beta clearance in Alzheimer’s models. Brain Behav. Immun. 2018, 67, 163–180. [Google Scholar] [CrossRef] [PubMed]
- Carecchio, M.; Comi, C. The role of osteopontin in neurodegenerative diseases. J. Alzheimer’s Dis. 2011, 25, 179–185. [Google Scholar] [CrossRef] [PubMed]
- Cheng, L.; Khalaf, A.T.; Lin, T.; Ran, L.; Shi, Z.; Wan, J.; Zhou, X.; Zou, L. Exercise Promotes the Osteoinduction of HA/beta-TCP Biomaterials via the Wnt Signaling Pathway. Metabolites 2020, 10, 90. [Google Scholar] [CrossRef] [PubMed]
- Zhang, L.; Yuan, Y.; Wu, W.; Sun, Z.; Lei, L.; Fan, J.; Gao, B.; Zou, J. Medium-Intensity Treadmill Exercise Exerts Beneficial Effects on Bone Modeling Through Bone Marrow Mesenchymal Stromal Cells. Front. Cell Dev. Biol. 2020, 8, 600639. [Google Scholar] [CrossRef]
- Wung, J.K.; Perry, G.; Kowalski, A.; Harris, P.L.; Bishop, G.M.; Trivedi, M.A.; Johnson, S.C.; Smith, M.A.; Denhardt, D.T.; Atwood, C.S. Increased expression of the remodeling- and tumorigenic-associated factor osteopontin in pyramidal neurons of the Alzheimer’s disease brain. Curr. Alzheimer Res. 2007, 4, 67–72. [Google Scholar] [CrossRef]
- Comi, C.; Carecchio, M.; Chiocchetti, A.; Nicola, S.; Galimberti, D.; Fenoglio, C.; Cappellano, G.; Monaco, F.; Scarpini, E.; Dianzani, U. Osteopontin is increased in the cerebrospinal fluid of patients with Alzheimer’s disease and its levels correlate with cognitive decline. J. Alzheimer’s Dis. 2010, 19, 1143–1148. [Google Scholar] [CrossRef] [PubMed]
- Iida, T.; Furuta, A.; Nishioka, K.; Nakabeppu, Y.; Iwaki, T. Expression of 8-oxoguanine DNA glycosylase is reduced and associated with neurofibrillary tangles in Alzheimer’s disease brain. Acta Neuropathol. 2002, 103, 20–25. [Google Scholar] [CrossRef] [PubMed]
- Jeynes, B.; Provias, J. P-Glycoprotein Altered Expression in Alzheimer’s Disease: Regional Anatomic Variability. J. Neurodegener. Dis. 2013, 2013, 257953. [Google Scholar] [CrossRef] [PubMed]
- Kim, S.; Choi, J.Y.; Moon, S.; Park, D.H.; Kwak, H.B.; Kang, J.H. Roles of myokines in exercise-induced improvement of neuropsychiatric function. Pflug. Arch. Eur. J. Physiol. 2019, 471, 491–505. [Google Scholar] [CrossRef] [PubMed]
- Wang, T.; Li, T.; Hao, S.; Han, Y.; Cai, Y. Association of plasma BDNF levels with different stages of Alzheimer’s disease: A cross-sectional study. Neurol. Res. 2023, 45, 234–240. [Google Scholar] [CrossRef]
- Balietti, M.; Giuli, C.; Conti, F. Peripheral Blood Brain-Derived Neurotrophic Factor as a Biomarker of Alzheimer’s Disease: Are There Methodological Biases? Mol. Neurobiol. 2018, 55, 6661–6672. [Google Scholar] [CrossRef]
- Swift, D.L.; Johannsen, N.M.; Myers, V.H.; Earnest, C.P.; Smits, J.A.; Blair, S.N.; Church, T.S. The effect of exercise training modality on serum brain derived neurotrophic factor levels in individuals with type 2 diabetes. PLoS ONE 2012, 7, e42785. [Google Scholar] [CrossRef]
- Ferris, L.T.; Williams, J.S.; Shen, C.L. The effect of acute exercise on serum brain-derived neurotrophic factor levels and cognitive function. Med. Sci. Sports Exerc. 2007, 39, 728–734. [Google Scholar] [CrossRef]
- Choi, S.H.; Bylykbashi, E.; Chatila, Z.K.; Lee, S.W.; Pulli, B.; Clemenson, G.D.; Kim, E.; Rompala, A.; Oram, M.K.; Asselin, C.; et al. Combined adult neurogenesis and BDNF mimic exercise effects on cognition in an Alzheimer’s mouse model. Science 2018, 361, eaan8821. [Google Scholar] [CrossRef]
- Maekawa, T.; Ogasawara, R.; Tsutaki, A.; Lee, K.; Nakada, S.; Nakazato, K.; Ishii, N. Electrically evoked local muscle contractions cause an increase in hippocampal BDNF. Appl. Physiol. Nutr. Metab. 2018, 43, 491–496. [Google Scholar] [CrossRef]
- Jin, Y.; Sumsuzzman, D.M.; Choi, J.; Kang, H.; Lee, S.R.; Hong, Y. Molecular and Functional Interaction of the Myokine Irisin with Physical Exercise and Alzheimer’s Disease. Molecules 2018, 23, 3229. [Google Scholar] [CrossRef] [PubMed]
- Sayal, N. Exercise training increases size of hippocampus and improves memory PNAS (2011) vol. 108|no. 7|3017–3022. Ann. Neurosci. 2015, 22, 107. [Google Scholar] [CrossRef] [PubMed]
- Erickson, K.I.; Voss, M.W.; Prakash, R.S.; Basak, C.; Szabo, A.; Chaddock, L.; Kim, J.S.; Heo, S.; Alves, H.; White, S.M.; et al. Exercise training increases size of hippocampus and improves memory. Proc. Natl. Acad. Sci. USA 2011, 108, 3017–3022. [Google Scholar] [CrossRef] [PubMed]
- MacPherson, R. Filling the void: A role for exercise-induced BDNF and brain amyloid precursor protein processing. Am. J. Physiol.-Regul. Integr. Comp. Physiol. 2017, 313, R585–R593. [Google Scholar] [CrossRef]
- Matsuzaki, M.; Yokoyama, M.; Yoshizawa, Y.; Kaneko, N.; Naito, H.; Kobayashi, H.; Korenaga, A.; Sekiya, S.; Ikemura, K.; Opoku, G.; et al. ADAMTS4 is involved in the production of the Alzheimer disease amyloid biomarker APP669-711. Mol. Psychiatry 2023, 28, 1802–1812. [Google Scholar] [CrossRef]
- Ballesteros-Alvarez, J.; Nguyen, W.; Sivapatham, R.; Rane, A.; Andersen, J.K. Urolithin A reduces amyloid-beta load and improves cognitive deficits uncorrelated with plaque burden in a mouse model of Alzheimer’s disease. Geroscience 2023, 45, 1095–1113. [Google Scholar] [CrossRef]
- Vasconcelos-Filho, F.S.L.; da Rocha-E-Silva, R.C.; Martins, J.E.R.; Godinho, W.D.N.; da Costa, V.V.; Ribeiro, J.K.C.; da Silva, C.A.; Ceccatto, V.M.; Soares, P.M.; Evangelista, J.S.A.M. Neuroprotector Effect of Daily 8-Minutes of High-Intensity Interval Training in Rat Abeta(1-42) Alzheimer Disease Model. Curr. Alzheimer Res. 2020, 17, 1320–1333. [Google Scholar] [CrossRef]
- Rosa, J.M.; Pazini, F.L.; Olescowicz, G.; Camargo, A.; Moretti, M.; Gil-Mohapel, J.; Rodrigues, A. Prophylactic effect of physical exercise on Abeta(1-40)-induced depressive-like behavior: Role of BDNF, mTOR signaling, cell proliferation and survival in the hippocampus. Prog. Neuro-Psychopharmacol. Biol. Psychiatry 2019, 94, 109646. [Google Scholar] [CrossRef]
- Nigam, S.M.; Xu, S.; Kritikou, J.S.; Marosi, K.; Brodin, L.; Mattson, M.P. Exercise and BDNF reduce Abeta production by enhancing alpha-secretase processing of APP. J. Neurochem. 2017, 142, 286–296. [Google Scholar] [CrossRef]
- Bostrom, P.; Wu, J.; Jedrychowski, M.P.; Korde, A.; Ye, L.; Lo, J.C.; Rasbach, K.A.; Bostrom, E.A.; Choi, J.H.; Long, J.Z.; et al. A PGC1-alpha-dependent myokine that drives brown-fat-like development of white fat and thermogenesis. Nature 2012, 481, 463–468. [Google Scholar] [CrossRef]
- Lourenco, M.V.; Frozza, R.L.; de Freitas, G.B.; Zhang, H.; Kincheski, G.C.; Ribeiro, F.C.; Goncalves, R.A.; Clarke, J.R.; Beckman, D.; Staniszewski, A.; et al. Exercise-linked FNDC5/irisin rescues synaptic plasticity and memory defects in Alzheimer’s models. Nat. Med. 2019, 25, 165–175. [Google Scholar] [CrossRef] [PubMed]
- Gronwald, T.; de Bem Alves, A.C.; Murillo-Rodriguez, E.; Latini, A.; Schuette, J.; Budde, H. Standardization of exercise intensity and consideration of a dose-response is essential. Commentary on “Exercise-linked FNDC5/irisin rescues synaptic plasticity and memory defects in Alzheimer’s models”, by Lourenco et al., published 2019 in Nature Medicine. J. Sport Health Sci. 2019, 8, 353–354. [Google Scholar] [CrossRef] [PubMed]
- Shirvani, H.; Arabzadeh, E. Metabolic cross-talk between skeletal muscle and adipose tissue in high-intensity interval training vs. moderate-intensity continuous training by regulation of PGC-1alpha. Eat. Weight Disord.-Stud. Anorex. Bulim. Obes. 2020, 25, 17–24. [Google Scholar] [CrossRef] [PubMed]
- Noda, Y.; Kuzuya, A.; Tanigawa, K.; Araki, M.; Kawai, R.; Ma, B.; Sasakura, Y.; Maesako, M.; Tashiro, Y.; Miyamoto, M.; et al. Fibronectin type III domain-containing protein 5 interacts with APP and decreases amyloid beta production in Alzheimer’s disease. Mol. Brain 2018, 11, 61. [Google Scholar] [CrossRef] [PubMed]
- Brenmoehl, J.; Albrecht, E.; Komolka, K.; Schering, L.; Langhammer, M.; Hoeflich, A.; Maak, S. Irisin is elevated in skeletal muscle and serum of mice immediately after acute exercise. Int. J. Biol. Sci. 2014, 10, 338–349. [Google Scholar] [CrossRef] [PubMed]
- Notaristefano, G.; Merola, A.; Scarinci, E.; Ubaldi, N.; Ranalli, M.; Tropea, A.; Diterlizzi, A.; Fabozzi, S.M.; Alesiani, O.; Silvestrini, A.; et al. Circulating irisin levels in functional hypothalamic amenorrhea: A new bone damage index? A pilot study. Endocrine 2022, 77, 168–176. [Google Scholar] [CrossRef] [PubMed]
- Pan, J.A.; Zhang, H.; Yu, Q.; Zhang, J.F.; Wang, C.Q.; Gu, J.; Chen, K. Association of Circulating Irisin Levels and the Characteristics and Prognosis of Coronary Artery Disease. Am. J. Med. Sci. 2021, 362, 63–71. [Google Scholar] [CrossRef]
- Miazgowski, T.; Miazgowski, B.; Kaczmarkiewicz, A.; Kopec, J. Associations of circulating irisin with 24-h blood pressure, total and visceral fat, and metabolic parameters in young adult hypertensives. Arch. Endocrinol. Metab. 2021, 65, 137–143. [Google Scholar] [CrossRef]
- Huh, J.Y.; Panagiotou, G.; Mougios, V.; Brinkoetter, M.; Vamvini, M.T.; Schneider, B.E.; Mantzoros, C.S. FNDC5 and irisin in humans: I. Predictors of circulating concentrations in serum and plasma and II. mRNA expression and circulating concentrations in response to weight loss and exercise. Metabolism 2012, 61, 1725–1738. [Google Scholar] [CrossRef]
- Waseem, R.; Shamsi, A.; Mohammad, T.; Hassan, M.I.; Kazim, S.N.; Chaudhary, A.A.; Rudayni, H.A.; Al-Zharani, M.; Ahmad, F.; Islam, A. FNDC5/Irisin: Physiology and Pathophysiology. Molecules 2022, 27, 1118. [Google Scholar] [CrossRef]
- Zhou, W.; Shi, Y.; Wang, H.; Chen, L.; Yu, C.; Zhang, X.; Yang, L.; Zhang, X.; Wu, A. Exercise-induced FNDC5/irisin protects nucleus pulposus cells against senescence and apoptosis by activating autophagy. Exp. Mol. Med. 2022, 54, 1038–1048. [Google Scholar] [CrossRef] [PubMed]
- Pignataro, P.; Dicarlo, M.; Zerlotin, R.; Zecca, C.; Dell’Abate, M.T.; Buccoliero, C.; Logroscino, G.; Colucci, S.; Grano, M. FNDC5/Irisin System in Neuroinflammation and Neurodegenerative Diseases: Update and Novel Perspective. Int. J. Mol. Sci. 2021, 22, 1605. [Google Scholar] [CrossRef] [PubMed]
- Zhang, X.; Hu, C.; Kong, C.Y.; Song, P.; Wu, H.M.; Xu, S.C.; Yuan, Y.P.; Deng, W.; Ma, Z.G.; Tang, Q.Z. FNDC5 alleviates oxidative stress and cardiomyocyte apoptosis in doxorubicin-induced cardiotoxicity via activating AKT. Cell Death Differ. 2020, 27, 540–555. [Google Scholar] [CrossRef]
- Jung, K.H.; Chu, K.; Lee, S.T.; Kim, S.J.; Sinn, D.I.; Kim, S.U.; Kim, M.; Roh, J.K. Granulocyte colony-stimulating factor stimulates neurogenesis via vascular endothelial growth factor with STAT activation. Brain Res. 2006, 1073–1074, 190–201. [Google Scholar] [CrossRef]
- Moon, H.S.; Dincer, F.; Mantzoros, C.S. Pharmacological concentrations of irisin increase cell proliferation without influencing markers of neurite outgrowth and synaptogenesis in mouse H19-7 hippocampal cell lines. Metabolism 2013, 62, 1131–1136. [Google Scholar] [CrossRef] [PubMed]
- Li, D.J.; Li, Y.H.; Yuan, H.B.; Qu, L.F.; Wang, P. The novel exercise-induced hormone irisin protects against neuronal injury via activation of the Akt and ERK1/2 signaling pathways and contributes to the neuroprotection of physical exercise in cerebral ischemia. Metabolism 2017, 68, 31–42. [Google Scholar] [CrossRef] [PubMed]
- Peng, J.; Deng, X.; Huang, W.; Yu, J.H.; Wang, J.X.; Wang, J.P.; Yang, S.B.; Liu, X.; Wang, L.; Zhang, Y.; et al. Irisin protects against neuronal injury induced by oxygen-glucose deprivation in part depends on the inhibition of ROS-NLRP3 inflammatory signaling pathway. Mol. Immunol. 2017, 91, 185–194. [Google Scholar] [CrossRef]
- Wang, K.; Li, H.; Wang, H.; Wang, J.H.; Song, F.; Sun, Y. Irisin Exerts Neuroprotective Effects on Cultured Neurons by Regulating Astrocytes. Mediat. Inflamm. 2018, 2018, 9070341. [Google Scholar] [CrossRef]
- Malekloo, R.; Nematollahi, S.; Vafaei, A.A.; Rashidy-Pour, A. Effects of different intensities of treadmill exercise on cued fear extinction failure, hippocampal BDNF decline, and Bax/Bcl-2 ratio alteration in chronic-morphine treated male rats. Behav. Brain Res. 2022, 421, 113732. [Google Scholar] [CrossRef]
- Yakhkeshi, R.; Roshani, F.; Akhoundzadeh, K.; Shafia, S. Effect of treadmill exercise on serum corticosterone, serum and hippocampal BDNF, hippocampal apoptosis and anxiety behavior in an ovariectomized rat model of post-traumatic stress disorder (PTSD). Physiol. Behav. 2022, 243, 113629. [Google Scholar] [CrossRef]
- Wrann, C.D.; White, J.P.; Salogiannnis, J.; Laznik-Bogoslavski, D.; Wu, J.; Ma, D.; Lin, J.D.; Greenberg, M.E.; Spiegelman, B.M. Exercise induces hippocampal BDNF through a PGC-1alpha/FNDC5 pathway. Cell Metab. 2013, 18, 649–659. [Google Scholar] [CrossRef]
- Zhang, S.; Zhang, Y.; Wen, Z.; Yang, Y.; Bu, T.; Bu, X.; Ni, Q. Cognitive dysfunction in diabetes: Abnormal glucose metabolic regulation in the brain. Front. Endocrinol. 2023, 14, 1192602. [Google Scholar] [CrossRef]
- Panov, A.; Mayorov, V.I.; Dikalov, S. Metabolic Syndrome and beta-Oxidation of Long-Chain Fatty Acids in the Brain, Heart, and Kidney Mitochondria. Int. J. Mol. Sci. 2022, 23, 4047. [Google Scholar] [CrossRef] [PubMed]
- Maciejczyk, M.; Zebrowska, E.; Chabowski, A. Insulin Resistance and Oxidative Stress in the Brain: What’s New? Int. J. Mol. Sci. 2019, 20, 874. [Google Scholar] [CrossRef] [PubMed]
- Bruehl, H.; Wolf, O.T.; Sweat, V.; Tirsi, A.; Richardson, S.; Convit, A. Modifiers of cognitive function and brain structure in middle-aged and elderly individuals with type 2 diabetes mellitus. Brain Res. 2009, 1280, 186–194. [Google Scholar] [CrossRef] [PubMed]
- Olsen, A.A.; Strandby, R.B.; Nerup, N.; Johansson, P.I.; Svendsen, L.B.; Achiam, M.P. Severe mesenteric traction syndrome is associated with increased systemic inflammatory response, endothelial dysfunction, and major postoperative morbidity. Langenbeck’s Arch. Surg. 2021, 406, 2457–2467. [Google Scholar] [CrossRef] [PubMed]
- Lin, C.; Arevalo, Y.A.; Nanavati, H.D.; Lin, D.M. Racial differences and an increased systemic inflammatory response are seen in patients with COVID-19 and ischemic stroke. Brain Behav. Immun. Health 2020, 8, 100137. [Google Scholar] [CrossRef]
- Honma, T.; Hatta, K.; Hitomi, Y.; Kambayashi, Y.; Hibino, Y.; Konoshita, T.; Nakamura, H. Increased systemic inflammatory interleukin-1ss and interleukin-6 during agitation as predictors of Alzheimer’s disease. Int. J. Geriatr. Psychiatry 2013, 28, 233–241. [Google Scholar] [CrossRef]
- Kim, Y.S.; Lee, K.J.; Kim, H. Serum tumour necrosis factor-alpha and interleukin-6 levels in Alzheimer’s disease and mild cognitive impairment. Psychogeriatrics 2017, 17, 224–230. [Google Scholar] [CrossRef]
- Cestari, J.A.; Fabri, G.M.; Kalil, J.; Nitrini, R.; Jacob-Filho, W.; de Siqueira, J.T.; Siqueira, S.R. Oral Infections and Cytokine Levels in Patients with Alzheimer’s Disease and Mild Cognitive Impairment Compared with Controls. J. Alzheimer’s Dis. 2016, 52, 1479–1485. [Google Scholar] [CrossRef]
- Qiu, Z.; Gruol, D.L. Interleukin-6, beta-amyloid peptide and NMDA interactions in rat cortical neurons. J. Neuroimmunol. 2003, 139, 51–57. [Google Scholar] [CrossRef]
- Velloso, F.J.; Wadhwa, A.; Kumari, E.; Carcea, I.; Gunal, O.; Levison, S.W. Modestly increasing systemic interleukin-6 perinatally disturbs secondary germinal zone neurogenesis and gliogenesis and produces sociability deficits. Brain Behav. Immun. 2022, 101, 23–36. [Google Scholar] [CrossRef] [PubMed]
- Islam, O.; Gong, X.; Rose-John, S.; Heese, K. Interleukin-6 and neural stem cells: More than gliogenesis. Mol. Biol. Cell 2009, 20, 188–199. [Google Scholar] [CrossRef] [PubMed]
- Rothaug, M.; Becker-Pauly, C.; Rose-John, S. The role of interleukin-6 signaling in nervous tissue. Biochim. Biophys. Acta 2016, 1863, 1218–1227. [Google Scholar] [CrossRef] [PubMed]
- Funk, J.A.; Gohlke, J.; Kraft, A.D.; McPherson, C.A.; Collins, J.B.; Jean Harry, G. Voluntary exercise protects hippocampal neurons from trimethyltin injury: Possible role of interleukin-6 to modulate tumor necrosis factor receptor-mediated neurotoxicity. Brain Behav. Immun. 2011, 25, 1063–1077. [Google Scholar] [CrossRef]
- Hoekstra, S.P.; Bishop, N.C.; Leicht, C.A. Can intervals enhance the inflammatory response and enjoyment in upper-body exercise? Eur. J. Appl. Physiol. 2017, 117, 1155–1163. [Google Scholar] [CrossRef] [PubMed]
- Sponder, M.; Campean, I.A.; Emich, M.; Fritzer-Szekeres, M.; Litschauer, B.; Bergler-Klein, J.; Graf, S.; Strametz-Juranek, J. Long-term endurance training increases serum cathepsin S and decreases IL-6 and hsCRP levels. J. Sports Sci. 2017, 35, 2129–2134. [Google Scholar] [CrossRef] [PubMed]
- Abd El-Kader, S.M.; Al-Jiffri, O.H. Aerobic exercise improves quality of life, psychological well-being and systemic inflammation in subjects with Alzheimer’s disease. Afr. Health Sci. 2016, 16, 1045–1055. [Google Scholar] [CrossRef]
- Fernandez, A.M.; Torres-Aleman, I. The many faces of insulin-like peptide signalling in the brain. Nat. Rev. Neurosci. 2012, 13, 225–239. [Google Scholar] [CrossRef]
- Trueba-Sáiz, A.; Cavada, C.; Fernandez, A.M.; Leon, T.; González, D.A.; Fortea Ormaechea, J.; Lleó, A.; Del Ser, T.; Nuñez, A.; Torres-Aleman, I. Loss of serum IGF-I input to the brain as an early biomarker of disease onset in Alzheimer mice. Transl. Psychiatry 2013, 3, e330. [Google Scholar] [CrossRef]
- Lin, T.W.; Tsai, S.F.; Kuo, Y.M. Physical Exercise Enhances Neuroplasticity and Delays Alzheimer’s Disease. Brain Plast. 2018, 4, 95–110. [Google Scholar] [CrossRef]
- Kovacevic, A.; Fenesi, B.; Paolucci, E.; Heisz, J.J. The effects of aerobic exercise intensity on memory in older adults. Appl. Physiol. Nutr. Metab. 2020, 45, 591–600. [Google Scholar] [CrossRef]
- Jeon, Y.K.; Ha, C.H. The effect of exercise intensity on brain derived neurotrophic factor and memory in adolescents. Environ. Health Prev. Med. 2017, 22, 27. [Google Scholar] [CrossRef] [PubMed]
- Maass, A.; Duzel, S.; Brigadski, T.; Goerke, M.; Becke, A.; Sobieray, U.; Neumann, K.; Lovden, M.; Lindenberger, U.; Backman, L.; et al. Relationships of peripheral IGF-1, VEGF and BDNF levels to exercise-related changes in memory, hippocampal perfusion and volumes in older adults. Neuroimage 2016, 131, 142–154. [Google Scholar] [CrossRef] [PubMed]
- Licht, T.; Goshen, I.; Avital, A.; Kreisel, T.; Zubedat, S.; Eavri, R.; Segal, M.; Yirmiya, R.; Keshet, E. Reversible modulations of neuronal plasticity by VEGF. Proc. Natl. Acad. Sci. USA 2011, 108, 5081–5086. [Google Scholar] [CrossRef] [PubMed]
- Religa, P.; Cao, R.; Religa, D.; Xue, Y.; Bogdanovic, N.; Westaway, D.; Marti, H.H.; Winblad, B.; Cao, Y. VEGF significantly restores impaired memory behavior in Alzheimer’s mice by improvement of vascular survival. Sci. Rep. 2013, 3, 2053. [Google Scholar] [CrossRef] [PubMed]
- Rich, B.; Scadeng, M.; Yamaguchi, M.; Wagner, P.D.; Breen, E.C. Skeletal myofiber vascular endothelial growth factor is required for the exercise training-induced increase in dentate gyrus neuronal precursor cells. J. Physiol. 2017, 595, 5931–5943. [Google Scholar] [CrossRef] [PubMed]
- Kannangara, T.S.; Vani, M.A. The muscles’ grip on neurogenesis: Contributions of skeletal muscle-derived vascular endothelial growth factor to running-induced stem cell proliferation. J. Physiol. 2017, 595, 6821–6822. [Google Scholar] [CrossRef]
- Zheng, D.; Liwinski, T.; Elinav, E. Interaction between microbiota and immunity in health and disease. Cell Res. 2020, 30, 492–506. [Google Scholar] [CrossRef]
- Dinan, T.G.; Cryan, J.F. The Microbiome-Gut-Brain Axis in Health and Disease. Gastroenterol. Clin. N. Am. 2017, 46, 77–89. [Google Scholar] [CrossRef]
- Asadi, A.; Shadab Mehr, N.; Mohamadi, M.H.; Shokri, F.; Heidary, M.; Sadeghifard, N.; Khoshnood, S. Obesity and gut-microbiota-brain axis: A narrative review. J. Clin. Lab. Anal. 2022, 36, e24420. [Google Scholar] [CrossRef] [PubMed]
- Morais, L.H.; Schreiber, H.L., IV; Mazmanian, S.K. The gut microbiota-brain axis in behaviour and brain disorders. Nat. Rev. Microbiol. 2021, 19, 241–255. [Google Scholar] [CrossRef] [PubMed]
- Clark, A.; Mach, N. Exercise-induced stress behavior, gut-microbiota-brain axis and diet: A systematic review for athletes. J. Int. Soc. Sports Nutr. 2016, 13, 43. [Google Scholar] [CrossRef]
- Wang, H.X.; Wang, Y.P. Gut Microbiota-brain Axis. Chin. Med. J. 2016, 129, 2373–2380. [Google Scholar] [CrossRef]
- Megur, A.; Baltriukienė, D.; Bukelskienė, V.; Burokas, A. The Microbiota-Gut-Brain Axis and Alzheimer’s Disease: Neuroinflammation Is to Blame? Nutrients 2020, 13, 37. [Google Scholar] [CrossRef]
- Jiang, C.; Li, G.; Huang, P.; Liu, Z.; Zhao, B. The Gut Microbiota and Alzheimer’s Disease. J. Alzheimer’s Dis. 2017, 58, 1–15. [Google Scholar] [CrossRef] [PubMed]
- Tsai, S.F.; Ku, N.W.; Wang, T.F.; Yang, Y.H.; Shih, Y.H.; Wu, S.Y.; Lee, C.W.; Yu, M.; Yang, T.T.; Kuo, Y.M. Long-Term Moderate Exercise Rescues Age-Related Decline in Hippocampal Neuronal Complexity and Memory. Gerontology 2018, 64, 551–561. [Google Scholar] [CrossRef]
- Rosa, J.M.; Pazini, F.L.; Camargo, A.; Wolin, I.A.V.; Olescowicz, G.; Eslabao, L.B.; Romero, O.B.; Winkelmann-Duarte, E.C.; Rodrigues, A.L.S. Prophylactic effect of physical exercise on Abeta(1-40)-induced depressive-like behavior and gut dysfunction in mice. Behav. Brain Res. 2020, 393, 112791. [Google Scholar] [CrossRef]
- Wang, G.; Zhou, H.H.; Luo, L.; Qin, L.Q.; Yin, J.; Yu, Z.; Zhang, L.; Wan, Z. Voluntary wheel running is capable of improving cognitive function only in the young but not the middle-aged male APPSwe/PS1De9 mice. Neurochem. Int. 2021, 145, 105010. [Google Scholar] [CrossRef]
- Abraham, D.; Feher, J.; Scuderi, G.L.; Szabo, D.; Dobolyi, A.; Cservenak, M.; Juhasz, J.; Ligeti, B.; Pongor, S.; Gomez-Cabrera, M.C.; et al. Exercise and probiotics attenuate the development of Alzheimer’s disease in transgenic mice: Role of microbiome. Exp. Gerontol. 2019, 115, 122–131. [Google Scholar] [CrossRef]
- Shamsipour, S.; Sharifi, G.; Taghian, F. Impact of interval training with probiotic (L. plantarum/Bifidobacterium bifidum) on passive avoidance test, ChAT and BDNF in the hippocampus of rats with Alzheimer’s disease. Neurosci. Lett. 2021, 756, 135949. [Google Scholar] [CrossRef]
- Gubert, C.; Kong, G.; Renoir, T.; Hannan, A.J. Exercise, diet and stress as modulators of gut microbiota: Implications for neurodegenerative diseases. Neurobiol. Dis. 2020, 134, 104621. [Google Scholar] [CrossRef] [PubMed]
- Wu, W.; Nicolazzo, J.A.; Wen, L.; Chung, R.; Stankovic, R.; Bao, S.S.; Lim, C.K.; Brew, B.J.; Cullen, K.M.; Guillemin, G.J. Expression of tryptophan 2,3-dioxygenase and production of kynurenine pathway metabolites in triple transgenic mice and human Alzheimer’s disease brain. PLoS ONE 2013, 8, e59749. [Google Scholar] [CrossRef] [PubMed]
- Guillemin, G.J.; Smythe, G.; Takikawa, O.; Brew, B.J. Expression of indoleamine 2,3-dioxygenase and production of quinolinic acid by human microglia, astrocytes, and neurons. Glia 2005, 49, 15–23. [Google Scholar] [CrossRef] [PubMed]
- Agus, A.; Planchais, J.; Sokol, H. Gut Microbiota Regulation of Tryptophan Metabolism in Health and Disease. Cell Host Microbe 2018, 23, 716–724. [Google Scholar] [CrossRef] [PubMed]
- Cervenka, I.; Agudelo, L.Z.; Ruas, J.L. Kynurenines: Tryptophan’s metabolites in exercise, inflammation, and mental health. Science 2017, 357, eaaf9794. [Google Scholar] [CrossRef] [PubMed]
- DiNatale, B.C.; Murray, I.A.; Schroeder, J.C.; Flaveny, C.A.; Lahoti, T.S.; Laurenzana, E.M.; Omiecinski, C.J.; Perdew, G.H. Kynurenic acid is a potent endogenous aryl hydrocarbon receptor ligand that synergistically induces interleukin-6 in the presence of inflammatory signaling. Toxicol. Sci. 2010, 115, 89–97. [Google Scholar] [CrossRef] [PubMed]
- Agudelo, L.Z.; Femenia, T.; Orhan, F.; Porsmyr-Palmertz, M.; Goiny, M.; Martinez-Redondo, V.; Correia, J.C.; Izadi, M.; Bhat, M.; Schuppe-Koistinen, I.; et al. Skeletal muscle PGC-1alpha1 modulates kynurenine metabolism and mediates resilience to stress-induced depression. Cell 2014, 159, 33–45. [Google Scholar] [CrossRef]
- Erny, D.; Hrabě de Angelis, A.L.; Jaitin, D.; Wieghofer, P.; Staszewski, O.; David, E.; Keren-Shaul, H.; Mahlakoiv, T.; Jakobshagen, K.; Buch, T.; et al. Host microbiota constantly control maturation and function of microglia in the CNS. Nat. Neurosci. 2015, 18, 965–977. [Google Scholar] [CrossRef]
- Colombo, A.V.; Sadler, R.K.; Llovera, G.; Singh, V.; Roth, S.; Heindl, S.; Sebastian Monasor, L.; Verhoeven, A.; Peters, F.; Parhizkar, S.; et al. Microbiota-derived short chain fatty acids modulate microglia and promote Abeta plaque deposition. eLife 2021, 10, e59826. [Google Scholar] [CrossRef] [PubMed]
- Allen, J.M.; Mailing, L.J.; Cohrs, J.; Salmonson, C.; Fryer, J.D.; Nehra, V.; Hale, V.L.; Kashyap, P.; White, B.A.; Woods, J.A. Exercise training-induced modification of the gut microbiota persists after microbiota colonization and attenuates the response to chemically-induced colitis in gnotobiotic mice. Gut Microbes 2018, 9, 115–130. [Google Scholar] [CrossRef]
- Barton, W.; Penney, N.C.; Cronin, O.; Garcia-Perez, I.; Molloy, M.G.; Holmes, E.; Shanahan, F.; Cotter, P.D.; O’Sullivan, O. The microbiome of professional athletes differs from that of more sedentary subjects in composition and particularly at the functional metabolic level. Gut 2018, 67, 625–633. [Google Scholar] [CrossRef]
- Carbajo-Pescador, S.; Porras, D.; Garcia-Mediavilla, M.V.; Martinez-Florez, S.; Juarez-Fernandez, M.; Cuevas, M.J.; Mauriz, J.L.; Gonzalez-Gallego, J.; Nistal, E.; Sanchez-Campos, S. Beneficial effects of exercise on gut microbiota functionality and barrier integrity, and gut-liver crosstalk in an in vivo model of early obesity and non-alcoholic fatty liver disease. Dis. Model. Mech. 2019, 12, dmm039206. [Google Scholar] [CrossRef]
- Zhang, N.; Mao, X.; Li, R.W.; Hou, E.; Wang, Y.; Xue, C.; Tang, Q. Neoagarotetraose protects mice against intense exercise-induced fatigue damage by modulating gut microbial composition and function. Mol. Nutr. Food Res. 2017, 61, 1600585. [Google Scholar] [CrossRef]
- Zhang, X.; He, Q.; Huang, T.; Zhao, N.; Liang, F.; Xu, B.; Chen, X.; Li, T.; Bi, J. Treadmill Exercise Decreases Abeta Deposition and Counteracts Cognitive Decline in APP/PS1 Mice, Possibly via Hippocampal Microglia Modifications. Front. Aging Neurosci. 2019, 11, 78. [Google Scholar] [CrossRef] [PubMed]
- Xiong, J.Y.; Li, S.C.; Sun, Y.X.; Zhang, X.S.; Dong, Z.Z.; Zhong, P.; Sun, X.R. Long-term treadmill exercise improves spatial memory of male APPswe/PS1dE9 mice by regulation of BDNF expression and microglia activation. Biol. Sport 2015, 32, 295–300. [Google Scholar] [CrossRef]
- Viloria, M.; Lara-Padilla, E.; Campos-Rodriguez, R.; Jarillo-Luna, A.; Reyna-Garfias, H.; Lopez-Sanchez, P.; Rivera-Aguilar, V.; Salas-Casas, A.; Berral de la Rosa, F.J.; Garcia-Latorre, E. Effect of moderate exercise on IgA levels and lymphocyte count in mouse intestine. Immunol. Investig. 2011, 40, 640–656. [Google Scholar] [CrossRef] [PubMed]
- Deng, Y.; Zhang, Q.; Luo, H.; Chen, X.; Han, Q.; Wang, F.; Huang, P.; Lai, W.; Guan, X.; Pan, X.; et al. Sustained elevation of NF-kappaB activity sensitizes offspring of maternal inflammation to hypertension via impairing PGC-1alpha recovery. Sci. Rep. 2016, 6, 32642. [Google Scholar] [CrossRef] [PubMed]
- Smith, S.M.; Vale, W.W. The role of the hypothalamic-pituitary-adrenal axis in neuroendocrine responses to stress. Dialogues Clin. Neurosci. 2006, 8, 383–395. [Google Scholar] [CrossRef]
- Lowder, T.; Padgett, D.A.; Woods, J.A. Moderate exercise early after influenza virus infection reduces the Th1 inflammatory response in lungs of mice. Exerc. Immunol. Rev. 2006, 12, 97–111. [Google Scholar]
- Xia, W.J.; Xu, M.L.; Yu, X.J.; Du, M.M.; Li, X.H.; Yang, T.; Li, L.; Li, Y.; Kang, K.B.; Su, Q.; et al. Antihypertensive effects of exercise involve reshaping of gut microbiota and improvement of gut-brain axis in spontaneously hypertensive rat. Gut Microbes 2021, 13, 1–24. [Google Scholar] [CrossRef]
- Friedman, R.; Tappen, R.M. The effect of planned walking on communication in Alzheimer’s disease. J. Am. Geriatr. Soc. 1991, 39, 650–654. [Google Scholar] [CrossRef] [PubMed]
- Venturelli, M.; Scarsini, R.; Schena, F. Six-month walking program changes cognitive and ADL performance in patients with Alzheimer. Am. J. Alzheimer’s Dis. 2011, 26, 381–388. [Google Scholar] [CrossRef] [PubMed]
- Arcoverde, C.; Deslandes, A.; Moraes, H.; Almeida, C.; de Araujo, N.B.; Vasques, P.E.; Silveira, H.; Laks, J. Treadmill training as an augmentation treatment for Alzheimer’s disease: A pilot randomized controlled study. Arq. Neuro-Psiquiatr. 2014, 72, 190–196. [Google Scholar] [CrossRef]
- Winsky-Sommerer, R.; de Oliveira, P.; Loomis, S.; Wafford, K.; Dijk, D.J.; Gilmour, G. Disturbances of sleep quality, timing and structure and their relationship with other neuropsychiatric symptoms in Alzheimer’s disease and schizophrenia: Insights from studies in patient populations and animal models. Neurosci. Biobehav. Rev. 2019, 97, 112–137. [Google Scholar] [CrossRef] [PubMed]
- McCurry, S.M.; Pike, K.C.; Vitiello, M.V.; Logsdon, R.G.; Larson, E.B.; Teri, L. Increasing walking and bright light exposure to improve sleep in community-dwelling persons with Alzheimer’s disease: Results of a randomized, controlled trial. J. Am. Geriatr. Soc. 2011, 59, 1393–1402. [Google Scholar] [CrossRef]
- Yang, S.Y.; Shan, C.L.; Qing, H.; Wang, W.; Zhu, Y.; Yin, M.M.; Machado, S.; Yuan, T.F.; Wu, T. The Effects of Aerobic Exercise on Cognitive Function of Alzheimer’s Disease Patients. CNS Neurol. Disord.-Drug Targets 2015, 14, 1292–1297. [Google Scholar] [CrossRef]
- Jensen, C.S.; Simonsen, A.H.; Siersma, V.; Beyer, N.; Frederiksen, K.S.; Gottrup, H.; Hoffman, K.; Hogh, P.; Frikke-Schmidt, R.; Sobol, N.A.; et al. Patients with Alzheimer’s disease who carry the APOE epsilon4 allele benefit more from physical exercise. Alzheimer’s Dement. 2019, 5, 99–106. [Google Scholar] [CrossRef]
- Hoffmann, K.; Sobol, N.A.; Frederiksen, K.S.; Beyer, N.; Vogel, A.; Vestergaard, K.; Braendgaard, H.; Gottrup, H.; Lolk, A.; Wermuth, L.; et al. Moderate-to-High Intensity Physical Exercise in Patients with Alzheimer’s Disease: A Randomized Controlled Trial. J. Alzheimer’s Dis. 2016, 50, 443–453. [Google Scholar] [CrossRef]
- Yu, F.; Vock, D.M.; Zhang, L.; Salisbury, D.; Nelson, N.W.; Chow, L.S.; Smith, G.; Barclay, T.R.; Dysken, M.; Wyman, J.F. Cognitive Effects of Aerobic Exercise in Alzheimer’s Disease: A Pilot Randomized Controlled Trial. J. Alzheimer’s Dis. 2021, 80, 233–244. [Google Scholar] [CrossRef]
- Cott, C.A.; Dawson, P.; Sidani, S.; Wells, D. The effects of a walking/talking program on communication, ambulation, and functional status in residents with Alzheimer disease. Alzheimer Dis. Assoc. Disord. 2002, 16, 81–87. [Google Scholar] [CrossRef]
- Ahn, N.; Kim, K. Effects of an elastic band resistance exercise program on lower extremity muscle strength and gait ability in patients with Alzheimer’s disease. J. Phys. Ther. Sci. 2015, 27, 1953–1955. [Google Scholar] [CrossRef] [PubMed]
- Ben Ayed, I.; Castor-Guyonvarch, N.; Amimour, S.; Naija, S.; Aouichaoui, C.; Ben Omor, S.; Tabka, Z.; El Massioui, F. Acute Exercise and Cognitive Function in Alzheimer’s Disease. J. Alzheimer’s Dis. 2021, 82, 749–760. [Google Scholar] [CrossRef] [PubMed]
- Sampaio, A.; Marques, E.A.; Mota, J.; Carvalho, J. Effects of a multicomponent exercise program in institutionalized elders with Alzheimer’s disease. Dementia 2019, 18, 417–431. [Google Scholar] [CrossRef]
- Rolland, Y.; Pillard, F.; Klapouszczak, A.; Reynish, E.; Thomas, D.; Andrieu, S.; Riviere, D.; Vellas, B. Exercise program for nursing home residents with Alzheimer’s disease: A 1-year randomized, controlled trial. J. Am. Geriatr. Soc. 2007, 55, 158–165. [Google Scholar] [CrossRef] [PubMed]
- Sanders, L.; Hortobagyi, T.; Karssemeijer, E.; Van der Zee, E.A.; Scherder, E.; van Heuvelen, M. Effects of low- and high-intensity physical exercise on physical and cognitive function in older persons with dementia: A randomized controlled trial. Alzheimer’s Res. Ther. 2020, 12, 28. [Google Scholar] [CrossRef]
- Toots, A.; Littbrand, H.; Bostrom, G.; Hornsten, C.; Holmberg, H.; Lundin-Olsson, L.; Lindelof, N.; Nordstrom, P.; Gustafson, Y.; Rosendahl, E. Effects of Exercise on Cognitive Function in Older People with Dementia: A Randomized Controlled Trial. J. Alzheimer’s Dis. 2017, 60, 323–332. [Google Scholar] [CrossRef]
- Lv, S.; Wang, Q.; Liu, W.; Zhang, X.; Cui, M.; Li, X.; Xu, Y. Comparison of various exercise interventions on cognitive function in Alzheimer’s patients: A network meta-analysis. Arch. Gerontol. Geriatr. 2023, 115, 105113. [Google Scholar] [CrossRef]
- Portugal, E.M.; Vasconcelos, P.G.; Souza, R.; Lattari, E.; Monteiro-Junior, R.S.; Machado, S.; Deslandes, A.C. Aging process, cognitive decline and Alzheimer;s disease: Can strength training modulate these responses? CNS Neurol. Disord.-Drug Targets 2015, 14, 1209–1213. [Google Scholar] [CrossRef]
- Liu, Y.; Chu, J.; Yan, T.; Zhang, Y.; Chen, Y.; Chang, R.; Wong, G. Short-term resistance exercise inhibits neuroinflammation and attenuates neuropathological changes in 3xTg Alzheimer’s disease mice. J. Neuroinflamm. 2020, 17, 4. [Google Scholar] [CrossRef]
- Tsai, C.L.; Pai, M.C.; Ukropec, J.; Ukropcova, B. Distinctive Effects of Aerobic and Resistance Exercise Modes on Neurocognitive and Biochemical Changes in Individuals with Mild Cognitive Impairment. Curr. Alzheimer Res. 2019, 16, 316–332. [Google Scholar] [CrossRef]
- Tsai, C.L.; Wang, C.H.; Pan, C.Y.; Chen, F.C.; Huang, T.H.; Chou, F.Y. Executive function and endocrinological responses to acute resistance exercise. Front. Behav. Neurosci. 2014, 8, 262. [Google Scholar] [CrossRef]
- Forte, R.; Boreham, C.A.; Leite, J.C.; De Vito, G.; Brennan, L.; Gibney, E.R.; Pesce, C. Enhancing cognitive functioning in the elderly: Multicomponent vs resistance training. Clin. Interv. Aging 2013, 8, 19–27. [Google Scholar] [CrossRef]
- Sanders, L.; Hortobagyi, T.; la Bastide-van Gemert, S.; van der Zee, E.A.; van Heuvelen, M. Dose-response relationship between exercise and cognitive function in older adults with and without cognitive impairment: A systematic review and meta-analysis. PLoS ONE 2019, 14, e0210036. [Google Scholar] [CrossRef]
- Nagamatsu, L.S.; Handy, T.C.; Hsu, C.L.; Voss, M.; Liu-Ambrose, T. Resistance training promotes cognitive and functional brain plasticity in seniors with probable mild cognitive impairment. Arch. Intern. Med. 2012, 172, 666–668. [Google Scholar] [CrossRef] [PubMed]
- Maillot, P.; Perrot, A.; Hartley, A. Effects of interactive physical-activity video-game training on physical and cognitive function in older adults. Psychol. Aging 2012, 27, 589–600. [Google Scholar] [CrossRef]
- Rogers, N.T.; Steptoe, A.; Cadar, D. Frailty is an independent predictor of incident dementia: Evidence from the English Longitudinal Study of Ageing. Sci. Rep. 2017, 7, 15746. [Google Scholar] [CrossRef] [PubMed]
- Braz de Oliveira, M.P.; Moreira Padovez, R.F.C.; Serrão, P.R.M.d.S.; de Noronha, M.A.; de Castro Cezar, N.O.; de Andrade, L.P. Effectiveness of physical exercise at improving functional capacity in older adults living with Alzheimer’s disease: A systematic review of randomized controlled trials. Disabil. Rehabil. 2023, 45, 391–402. [Google Scholar] [CrossRef]
- Özbeyli, D.; Sarı, G.; Özkan, N.; Karademir, B.; Yüksel, M.; Çilingir Kaya, Ö.T.; Kasımay Çakır, Ö. Protective effects of different exercise modalities in an Alzheimer’s disease-like model. Behav. Brain Res. 2017, 328, 159–177. [Google Scholar] [CrossRef]
- Northey, J.M.; Cherbuin, N.; Pumpa, K.L.; Smee, D.J.; Rattray, B. Exercise interventions for cognitive function in adults older than 50: A systematic review with meta-analysis. Br. J. Sports Med. 2018, 52, 154–160. [Google Scholar] [CrossRef] [PubMed]
- Thomas, R.; Zimmerman, S.D.; Yuede, K.M.; Cirrito, J.R.; Tai, L.M.; Timson, B.F.; Yuede, C.M. Exercise Training Results in Lower Amyloid Plaque Load and Greater Cognitive Function in an Intensity Dependent Manner in the Tg2576 Mouse Model of Alzheimer’s Disease. Brain Sci. 2020, 10, 88. [Google Scholar] [CrossRef] [PubMed]
- Zeng, B.; Zhao, G.; Liu, H.L. The Differential Effect of Treadmill Exercise Intensity on Hippocampal Soluble Abeta and Lipid Metabolism in APP/PS1 Mice. Neuroscience 2020, 430, 73–81. [Google Scholar] [CrossRef]
- Rashid, M.H.; Zahid, M.F.; Zain, S.; Kabir, A.; Hassan, S.U. The Neuroprotective Effects of Exercise on Cognitive Decline: A Preventive Approach to Alzheimer Disease. Cureus J. Med. Sci. 2020, 12, e6958. [Google Scholar] [CrossRef] [PubMed]
- Antunes, B.M.; Rossi, F.E.; Teixeira, A.M.; Lira, F.S. Short-time high-intensity exercise increases peripheral BDNF in a physical fitness-dependent way in healthy men. Eur. J. Sport Sci. 2020, 20, 43–50. [Google Scholar] [CrossRef] [PubMed]
- Boyne, P.; Meyrose, C.; Westover, J.; Whitesel, D.; Hatter, K.; Reisman, D.S.; Cunningham, D.; Carl, D.; Jansen, C.; Khoury, J.C.; et al. Exercise intensity affects acute neurotrophic and neurophysiological responses poststroke. J. Appl. Physiol. 2019, 126, 431–443. [Google Scholar] [CrossRef] [PubMed]
- Barnes, J.N. Exercise, cognitive function, and aging. Adv. Physiol. Educ. 2015, 39, 55–62. [Google Scholar] [CrossRef] [PubMed]
- Winchester, J.; Dick, M.B.; Gillen, D.; Reed, B.; Miller, B.; Tinklenberg, J.; Mungas, D.; Chui, H.; Galasko, D.; Hewett, L.; et al. Walking stabilizes cognitive functioning in Alzheimer’s disease (AD) across one year. Arch. Gerontol. Geriatr. 2013, 56, 96–103. [Google Scholar] [CrossRef]
- James, C.; Ranson, J.M.; Everson, R.; Llewellyn, D.J. Performance of Machine Learning Algorithms for Predicting Progression to Dementia in Memory Clinic Patients. JAMA Netw. Open 2021, 4, e2136553. [Google Scholar] [CrossRef]
- Ezzati, A.; Abdulkadir, A.; Jack, C.R., Jr.; Thompson, P.M.; Harvey, D.J.; Truelove-Hill, M.; Sreepada, L.P.; Davatzikos, C.; Lipton, R.B. Predictive value of ATN biomarker profiles in estimating disease progression in Alzheimer’s disease dementia. Alzheimer’s Dement. 2021, 17, 1855–1867. [Google Scholar] [CrossRef]
- Hammond, T.C.; Xing, X.; Wang, C.; Ma, D.; Nho, K.; Crane, P.K.; Elahi, F.; Ziegler, D.A.; Liang, G.; Cheng, Q.; et al. beta-amyloid and tau drive early Alzheimer’s disease decline while glucose hypometabolism drives late decline. Commun. Biol. 2020, 3, 352. [Google Scholar] [CrossRef]
- Lee, H.C.; Ryu, H.G.; Chung, E.J.; Jung, C.W. Prediction of Bispectral Index during Target-controlled Infusion of Propofol and Remifentanil: A Deep Learning Approach. Anesthesiology 2018, 128, 492–501. [Google Scholar] [CrossRef] [PubMed]
- van der Kleij, L.A.; Petersen, E.T.; Siebner, H.R.; Hendrikse, J.; Frederiksen, K.S.; Sobol, N.A.; Hasselbalch, S.G.; Garde, E. The effect of physical exercise on cerebral blood flow in Alzheimer’s disease. NeuroImage-Clin. 2018, 20, 650–654. [Google Scholar] [CrossRef] [PubMed]
- Colcombe, S.J.; Kramer, A.F.; Erickson, K.I.; Scalf, P.; McAuley, E.; Cohen, N.J.; Webb, A.; Jerome, G.J.; Marquez, D.X.; Elavsky, S. Cardiovascular fitness, cortical plasticity, and aging. Proc. Natl. Acad. Sci. USA 2004, 101, 3316–3321. [Google Scholar] [CrossRef] [PubMed]
- Voss, M.W.; Prakash, R.S.; Erickson, K.I.; Basak, C.; Chaddock, L.; Kim, J.S.; Alves, H.; Heo, S.; Szabo, A.N.; White, S.M.; et al. Plasticity of brain networks in a randomized intervention trial of exercise training in older adults. Front. Aging Neurosci. 2010, 2, 32. [Google Scholar] [CrossRef] [PubMed]
- Coudray, N.; Ocampo, P.S.; Sakellaropoulos, T.; Narula, N.; Snuderl, M.; Fenyo, D.; Moreira, A.L.; Razavian, N.; Tsirigos, A. Classification and mutation prediction from non-small cell lung cancer histopathology images using deep learning. Nat. Med. 2018, 24, 1559–1567. [Google Scholar] [CrossRef]
- Lee, E.E.; Torous, J.; De Choudhury, M.; Depp, C.A.; Graham, S.A.; Kim, H.C.; Paulus, M.P.; Krystal, J.H.; Jeste, D.V. Artificial Intelligence for Mental Health Care: Clinical Applications, Barriers, Facilitators, and Artificial Wisdom. Biol. Psychiatry-Cogn. Neurosci. Neuroimaging 2021, 6, 856–864. [Google Scholar] [CrossRef] [PubMed]
- Seifert, R.; Weber, M.; Kocakavuk, E.; Rischpler, C.; Kersting, D. Artificial Intelligence and Machine Learning in Nuclear Medicine: Future Perspectives. Semin. Nucl. Med. 2021, 51, 170–177. [Google Scholar] [CrossRef]
- Yedavalli, V.S.; Tong, E.; Martin, D.; Yeom, K.W.; Forkert, N.D. Artificial intelligence in stroke imaging: Current and future perspectives. Clin. Imaging 2021, 69, 246–254. [Google Scholar] [CrossRef]
- Mouridsen, K.; Thurner, P.; Zaharchuk, G. Artificial Intelligence Applications in Stroke. Stroke 2020, 51, 2573–2579. [Google Scholar] [CrossRef]
- Soun, J.E.; Chow, D.S.; Nagamine, M.; Takhtawala, R.S.; Filippi, C.G.; Yu, W.; Chang, P.D. Artificial Intelligence and Acute Stroke Imaging. Am. J. Neuroradiol. 2021, 42, 2–11. [Google Scholar] [CrossRef]
- Shafaat, O.; Bernstock, J.D.; Shafaat, A.; Yedavalli, V.S.; Elsayed, G.; Gupta, S.; Sotoudeh, E.; Sair, H.I.; Yousem, D.M.; Sotoudeh, H. Leveraging artificial intelligence in ischemic stroke imaging. J. Neuroradiol. 2022, 49, 343–351. [Google Scholar] [CrossRef] [PubMed]
- Martorana, A.; Di Lorenzo, F.; Belli, L.; Sancesario, G.; Toniolo, S.; Sallustio, F.; Sancesario, G.M.; Koch, G. Cerebrospinal Fluid Abeta42 Levels: When Physiological Become Pathological State. CNS Neurosci. Ther. 2015, 21, 921–925. [Google Scholar] [CrossRef]
- Clarke, J.R.; Lyra E Silva, N.M.; Figueiredo, C.P.; Frozza, R.L.; Ledo, J.H.; Beckman, D.; Katashima, C.K.; Razolli, D.; Carvalho, B.M.; Frazao, R.; et al. Alzheimer-associated Abeta oligomers impact the central nervous system to induce peripheral metabolic deregulation. EMBO Mol. Med. 2015, 7, 190–210. [Google Scholar] [CrossRef]
Author (s) | Exercise Type | Intensity | Frequency | Outcomes |
---|---|---|---|---|
Friedman, R. et al., 1991 [153] | walking | Low | 3 times/week × 30 min × 10 weeks | communication performance improved |
Venturelli, M. et al., 2011 [154] | walking | Moderate | 4 times/week × 30 min × 24 weeks | stabilize the progressive cognitive dysfunctions |
Arcoverde, C. et al., 2014 [155] | treadmill | Moderate | 2 times/week × 30 min × 16 weeks | improvement in the functional capacity |
McCurry, S.M. et al., 2011 [157] | walking | unclear | 3 times/week × 6 months | improving sleep |
Yang, S.Y. et al., 2015 [158] | cycling | Moderate | 3 times/week × 40 min × 3 months | improve cognitive function |
Hoffmann, K. et al., 2015 [160] | strength, cycling | Moderate-to-high | 3 times/week × 60 min × 16 weeks | reduced neuropsychiatric symptoms |
Yu, F. et al., 2021 [161] | cycling | Moderate-to-high | 3 times/week × 20–50 min × 6 months | reduce decline in global cognition |
Ahn, N. et al., 2015 [163] | resistance exercise | unclear | 3 times/week × 5 months | improved muscle strength and endurance, cardiovascular function, and gait speed |
Ben, A.I. et al., 2021 [164] | cycling, cognitive games | Moderate | Acute Exercise | improve cognitive functions |
Sampaio, A. et al., 2019 [165] | Multicomponent Training | unclear | 2 times/week × 45–55 min × 6 months | improve physical and cognitive functions |
Rolland, Y. et al., 2007 [166] | Collective exercise program | Low | 2 times/week × 60 min × 12 months | significantly slower decline in ADL score |
Sanders, L. et al., 2020 [167] | walking and lower limb strength training | Low and high | 3 times/week × 60 min × 24 weeks | no beneficial effects of the exercise vs. control group on cognitive function. |
Toots, A. et al., 2017 [168] | Functional Exercise program | High | 5 times/2 week × 45 min × 4 months | no superior effects on global cognition or executive function |
Author (s) | Study Design | Sample Size | Statistical Method | Consistency of Results | Risk of Bias |
---|---|---|---|---|---|
Friedman, R. et al., 1991 [153] | Randomized, non-blinded two-group experimental | 30 | MANOVA | High | Medium |
Venturelli, M. et al., 2011 [154] | Randomized controlled trial | 21 | ANOVA | High | Medium |
Arcoverde, C. et al., 2014 [155] | Randomized controlled trial | 20 | independent sample t-test | High | Medium |
McCurry, S.M. et al., 2011 [157] | Randomized, controlled trial with blinded assessors | 132 | unclear | High | Unclear |
Yang, S.Y. et al., 2015 [158] | Randomized controlled trial | 50 | paired samples t-test | High | Low |
Hoffmann, K. et al., 2015 [160] | Randomized controlled trial | 200 | linear regression models using generalized estimating equations | High | Low |
Yu, F. et al., 2021 [161] | Randomized controlled trial | 96 | 2-sided t-test | High | Low |
Ahn, N. et al., 2015 [163] | Randomized controlled trial | 23 | unclear | High | Unclear |
Ben, A.I. et al., 2021 [164] | Randomized controlled trial | 79 | ANOVA | High | Low |
Sampaio, A. et al., 2019 [165] | Non-randomized study | 37 | ANOVA | High | Medium |
Rolland, Y. et al., 2007 [166] | Randomized controlled trial | 134 | Multiple logistic regression analyses | High | Low |
Sanders, L. et al., 2020 [167] | Randomized controlled trial | 91 | ANCOVA | Low | Medium |
Toots, A. et al., 2017 [168] | Randomized controlled trial | 186 | Linear mixed models | Low | Medium |
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content. |
© 2023 by the authors. Licensee MDPI, Basel, Switzerland. This article is an open access article distributed under the terms and conditions of the Creative Commons Attribution (CC BY) license (https://creativecommons.org/licenses/by/4.0/).
Share and Cite
Ren, J.; Xiao, H. Exercise Intervention for Alzheimer’s Disease: Unraveling Neurobiological Mechanisms and Assessing Effects. Life 2023, 13, 2285. https://doi.org/10.3390/life13122285
Ren J, Xiao H. Exercise Intervention for Alzheimer’s Disease: Unraveling Neurobiological Mechanisms and Assessing Effects. Life. 2023; 13(12):2285. https://doi.org/10.3390/life13122285
Chicago/Turabian StyleRen, Jianchang, and Haili Xiao. 2023. "Exercise Intervention for Alzheimer’s Disease: Unraveling Neurobiological Mechanisms and Assessing Effects" Life 13, no. 12: 2285. https://doi.org/10.3390/life13122285
APA StyleRen, J., & Xiao, H. (2023). Exercise Intervention for Alzheimer’s Disease: Unraveling Neurobiological Mechanisms and Assessing Effects. Life, 13(12), 2285. https://doi.org/10.3390/life13122285