Loading

Commentary Open Access
Volume 4 | Issue 1 | DOI: https://doi.org/10.33696/rehabilitation.4.024

Exercise Benefits on Alzheimer’s Disease

  • 1Department of Pharmacology, Laboratory of Aging and Nervous Diseases (SZS0703), Jiangsu Key Laboratory of Neuropsychiatric Disease, College of Pharmaceutical Sciences, Soochow University, Suzhou 215123, China
+ Affiliations - Affiliations

*Corresponding Author

Fang Lin, linfang-1@suda.edu.cn

Received Date: November 02, 2021

Accepted Date: December 09, 2021

Abstract

Alzheimer’s disease (AD) is a major threat to the health of the elderly, and there are few drugs that can completely cure AD. Here we find that longterm exercise improves cognitive impairment and motor coordination in AD mice, maintains lysosomal Golgi morphology, promotes lysosomal maturation to enhance lysosomal function, stimulates enhanced effects of TFEB and AMPK mediated acetyl-CoA synthetase2 (ACSS2) for lysosomal biogenesis thereby increasing amyloid degradation and reducing its accumulation in the hippocampus and cortex.

 

Keywords

Exercise, Lysosome function, Alzheimer’s disease

Introduction

Alzheimer’s disease (AD) is an age-related neurodegenerative disorder [1] and the most common cause of human dementia, accounting for approximately 60%−80% of cases. It is estimated that more than 30 million AD patients, and the number likely to increase to over 100 million by 2050 because of the increase of the elderly population [2]. Its main clinical manifestations are memory disorder, personality abnormality, apraxia, visual space abnormality, executive dysfunction and neuropsychiatric symptoms [3,4]. Alzheimer’s disease is characterized by accumulation of abnormal protein aggregates including amyloid plaques (composed of betaamyloid (Aβ) peptides) and neurofibrillary tangles (formed by hyper-phosphorylated tau protein) [5,6]. Fibrillar aggregates of the amyloid-β protein (Aβ) are the main components of the senile plaques found in brains of Alzheimer’s disease patients [7]. Synaptic plasticity [8], neuroinflammation [9], calcium signaling etc. also show dysfunction in AD patients [2]. Lysosome is the degradation center and signaling hubs in cells and play important roles in cellular homeostasis, development, and aging [10,11]. In neurons, Aβ accumulates in endo-lysosomal vesicles at low pH [12]. It is important to maintain the normal function of lysosome for Aβ degradation.

It is well known that exercise is beneficial to physical and mental health and can relieve the occurrence and development of neurodegenerative diseases to a certain extent. Therefore, in this commentary, we conducted regular running on 5-month-old APP/PS1 mice at 18r/min per day for 5 months and found that exercise can relieve Alzheimer’s disease by activating lysosome function.

Exercise Alleviates Cognitive Impairment through Different Brain Regions

Exercise has been shown to benefit brain structure and function, especially in aging populations. Various modes, frequencies, intensities, and durations of exercise might elicit different pathways and thus have differing effects on brain health outcomes [13]. Studies have shown that moderate exercise improves cognition including processing speed, memory, and executive function [14]. Regular physical exercise has proven to be beneficial for traditional cardiovascular risk factors (e.g., reduced vascular flow, diabetes) involved in the pathogenesis of Alzheimer’s disease, which exerts antiinflammatory effects and improves the brain redox status. Otherwise, exercise-induced metabolic factors, ketone bodies and lactate, and muscle-derived myokines, such as cathepsin-B and irisin, can stimulate the secretion of brainderived neurotrophic factor and promote neurogenesis [15]. In summary, physical activity has a relation to areas of the brain that support complex cognitive processes during laboratory tasks [16].

Hippocampus plays an important role in learning and higher order cognition [17]. Together with its medial temporal lobe and subcortical circuits, the hippocampus is compromised early in ageing and neurodegenerative conditions, most notably in Alzheimer’s disease and frontotemporal dementias [18]. Hippocampus comprises three main subfields: the dentate gyrus (DG), area CA3, and area CA1. The DG is unique in its ability to generate new neurons in mammals, which can be doubled or tripled by exercise in rodents [19-21]. Facilitated plasticity is most evident in the dentate gyrus (DG), where exercise enhances both short-term potentiation and longterm potentiation (LTP) [22].

While the hippocampus is critical for spatial memory formation, it does not store long-term memories, and studies have shown that the prefrontal and anterior cingulate cortex are critical for storage and retrieval of remote spatial memories. Consolidation of spatial memory requires a time-dependent hippocampal-cortical dialogue, ultimately enabling widespread cortical networks to mediate effortful recall and use of cortically stored remote memories independently [23,24]. The prefrontal cortex is one of the most vulnerable brain regions during aging, and previous studies have linked PFC atrophy to AD [25].

 

Long Term Exercise Activates Lysosomal Function

In our study, we tested cognition-related behavioral, water mazes and new object recognition, and found that 40 minutes of exercise per day from 5 months to 10 months of age, enhanced learning ability in APP/PS1 mice. Behavioral studies related to balance including rotarod and balance beam showed that exercise saved coordination in AD mice. Long term exercise also reduced amyloid deposition and phosphorylation of Tau in the hippocampal cortex. We also found that the decrease of Aβ was not because exercise affected Aβ production but increased degradation.

Lysosomes are cytoplasmic membrane-enclosed organelles that degrade macromolecules and cell components [26], which are found in all eukaryotic cell types, except for erythrocytes. Lysosomes are acidic organelles with an internal pH of 4.5- 5.5 [27], maintained by the presence of the vacuolar-type H+ ATPase(V-ATPase) on the lysosomal membrane [28]. Lysosomes are involved in catabolism (autophagy, heterophagy), cell signaling (calcium storage, mTOR function, amino acid release) and cell death(cancer therapy, antibacterial infection) [29].

Exercise improve the maturation of lysosomal enzymes

Lysosomes contain more than 70 hydrolases and more than 200 membrane proteins [30] that control the distribution, number, size and activity of lysosomes, and the specificity of cargo flux and degradation time (initiation and termination) [31]. In our study, we found that the lysosome morphology in the CA1 region of APP/PS1 mice was abnormally enlarged and contained the undegraded contents, while the lysosome morphology in AD mice after long-term wheel running was normal. In addition, lysosomal membrane LAMP1, was significantly increased in the hippocampus and cortex of AD mice by western blot, which consisted of enlarged lysosomes in AD mice, and it was reversed after exercise. With long-term exercise, Cathepsin L and Cathepsin D levels increased in the hippocampus and cortex, and autophagy flow was smooth. The result of immunofluorescence showed that Aβ was transported to the lysosome for degradation. These results indicate that lysosome function is enhanced in AD mice with long-term exercise.

Lysosomal enzymes are synthesized in the endoplasmic reticulum and transported to the endosomal system via Golgi complexes [32]. On the one hand, the morphology and function of Golgi in CA1 region were impaired in AD mice, on the other hand, the lysosomal modification of Golgi was damaged when transported to the lysosome through vesicles, which both caused the accumulation of unmatured cathepsins in AD mice. Rab7 located in late endosomal lysosomes can promote the maturation of autophagosomes [33,34]. Although the fusion of autophagosomes with early endosomes and multivesicular bodies can still occur in Rab7 knockdown cells, but fusion with lysosomes is blocked [35,36].

Exercise enhance the lysosomal biogenesis

Transcription factor EB (TFEB) is a master regulator of autophagy and lysosomal biogenesis [37,38]. Autophagy is promoted by AMP activated protein kinase (AMPK), which is a key energy sensor and regulates cellular metabolism to maintain energy homeostasis [39]. In our study, we found that exercise enhanced phosphorylation of AMPK. AMPK is involved in ULK1(Unc-51 like autophagy activating kinase 1) [40] activation and lysosomal biogenesis [41]. AMPK phosphorylates ACSS2, resulting in nuclear translocation. In nuclear ACSS2 forms a complex with TFEB, which up-regulates lysosomal and autophagosomal genes by locally producing acetyl-CoA for histone H3 acetylation in the promoter regions of these genes by utilizing acetate generated from the turnover of histone acetylation [42,43]. In our study, exercise enhances nucleation of ACSS2. Finally, the enhanced action of ACSS2 and TFEB ultimately promotes lysosomal biogenesis.

 

Lysosome in Glia

Microglia are the resident phagocytotic immune cells of the CNS [44] and have highly dynamic processes. It plays important roles in investigating brain pathogens, contacting neurons, shaping synaptic connections, etc. The lysosome contributes to these functions, playing a role in the exocytosis of extracellular matrix proteases, endocytosis and phagocytosis of myelin debris, extracellular aggregates, and pathogens [45]. Sphingolipids are mainly degraded in lysosomes, and the disruption of the sphingolipid degradation pathway in neurosphingolipid storage diseases affects the normal homeostasis of microglia. The abnormal lysosomal function causes the formation of destructive pro-inflammatory phenotypes in microglia and exacerbates the disease process [46]. When microglia overexpress deacetylated TFEB, which accelerates the degradation of intracellular fibrillar Aβ by stimulating lysosome biogenesis and greatly reduced the deposited amyloid plaques in the brain slices of APP/PS1 transgenic mice [47].

In vitro, overexpression of exogenous TFEB in primary astrocytes enhances Tau fiber absorption and lysosomal activity; In vivo, the induced TFEB expression in astrocytes reduces pathology in the hippocampus of PS19 tauopathy mice, as well as prominently attenuates tau spreading from the ipsilateral to the contralateral hippocampus in a mouse model of tau spreading [48]. Proteins related to Parkinson’s disease genes and genetic risk factors of Parkinson’s disease are highly expressed in glial cells. Damage of endo-lysosome causes in glial cells causes the phagocytic substances are unable to be degraded. The undigested substances in cells in turn interfere with the homeostasis of glial cells and affects neurological health [49].

 

Conclusions

Physical exercise can increase metabolic activity in skeletal muscles, enhance antioxidant systems, and reduce inflammation [50]. At the meanwhile, we also found that longterm regular exercise can affect different brain regions and delay the process of the cognitive dysfunction in the APP/ PS1 mice by promoting the lysosomal biogenesis and lysosomal enzyme maturation, which subsequently enhance the degradation of amyloid protein and keep the cell homeostasis both in neurons and glia. Therefore, it is necessary for Alzheimer’s patients and healthy adults to maintain regular exercise.

References

1. Yu H, Zhang C, Xia J, Xu B. Treadmill Exercise Ameliorates Adult Hippocampal Neurogenesis Possibly by Adjusting the APP Proteolytic Pathway in APP/PS1 Transgenic Mice. Int J Mol Sci. 2021;22.

2. Zhang Z, Yang X, Song YQ, Tu J. Autophagy in Alzheimer’s disease pathogenesis: Therapeutic potential and future perspectives. Ageing Res Rev. 2021;72:101464.

3. Bai H, Zhang Q. Activation of NLRP3 Inflammasome and Onset of Alzheimer’s Disease. Front Immunol. 2021;12:701282.

4. Yamazaki Y, Zhao N, Caulfield TR, Liu CC, Bu G. Apolipoprotein E and Alzheimer disease: pathobiology and targeting strategies. Nat Rev Neurol. 2019;15:501-18.

5. Song XJ, Zhou HY, Sun YY, Huang HC. Phosphorylation and Glycosylation of Amyloid-β Protein Precursor:The Relationship to Trafficking and Cleavage in Alzheimer’s Disease. J Alzheimers Dis. 2021.

6. Xiong Y, Lim CS. Understanding the Modulatory Effects of Cannabidiol on Alzheimer’s Disease. Brain Sci. 2021;11.

7. Gremer L, Scholzel D, Schenk C, Reinartz E, Labahn J, Ravelli RBG, et al. Fibril structure of amyloid-beta(1-42) by cryo-electron microscopy. Science. 2017;358:116-9.

8. Ghatak S, Dolatabadi N, Gao R, Wu Y, Scott H, Trudler D, et al. NitroSynapsin ameliorates hypersynchronous neural network activity in Alzheimer hiPSC models. Mol Psychiatry. 2020.

9. Dekens DW, Eisel ULM, Gouweleeuw L, Schoemaker RG, De Deyn PP, Naudé PJW. Lipocalin 2 as a link between ageing, risk factor conditions and age-related brain diseases. Ageing Res Rev. 2021;70:101414.

10. Yang C, Wang X. Lysosome biogenesis:Regulation and functions. J Cell Biol. 2021;220.

11. López-Hernández T, Puchkov D, Krause E, Maritzen T, Haucke V. Endocytic regulation of cellular ion homeostasis controls lysosome biogenesis. Nat Cell Biol. 2020;22:815-27.

12. Schutzmann MP, Hasecke F, Bachmann S, Zielinski M, Hansch S, Schroder GF, et al. Endo-lysosomal Abeta concentration and pH trigger formation of Abeta oligomers that potently induce Tau missorting. Nat Commun. 2021;12:4634.

13. Stillman CM, Esteban-Cornejo I, Brown B, Bender CM, Erickson KI. Effects of Exercise on Brain and Cognition Across Age Groups and Health States. Trends Neurosci. 2020;43:533-43.

14. Erickson KI, Hillman C, Stillman CM, Ballard RM, Bloodgood B, Conroy DE, et al. Physical Activity, Cognition, and Brain Outcomes: A Review of the 2018 Physical Activity Guidelines. Medicine & Science in Sports & Exercise. 2019;51:1242-51.

15. Valenzuela PL, Castillo-Garcia A, Morales JS, de la Villa P, Hampel H, Emanuele E, et al. Exercise benefits on Alzheimer’s disease: Stateof- the-science. Ageing Res Rev. 2020;62:101108.

16. Donnelly JE, Hillman CH, Castelli D, Etnier JL, Lee S, Tomporowski P, et al. Physical Activity, Fitness, Cognitive Function, and Academic Achievement in Children: A Systematic Review. Med Sci Sports Exerc. 2016;48:1197-222.

17. Gomez-Pinilla F, Hillman C. The influence of exercise on cognitive abilities. Compr Physiol. 2013;3:403-28.

18. Duzel E, van Praag H, Sendtner M. Can physical exercise in old age improve memory and hippocampal function? Brain. 2016;139:662- 73.

19. van Praag H. Neurogenesis and exercise:past and future directions. Neuromolecular Med. 2008;10:128-40.

20. van Praag H, Kempermann G, Gage FH. Running increases cell proliferation and neurogenesis in the adult mouse dentate gyrus. Nat Neurosci. 1999;2:266-70.

21. Voss MW, Vivar C, Kramer AF, van Praag H. Bridging animal and human models of exercise-induced brain plasticity. Trends Cogn Sci. 2013;17:525-44.

22. Cotman CW, Berchtold NC, Christie LA. Exercise builds brain health:key roles of growth factor cascades and inflammation. Trends Neurosci. 2007;30:464-72.

23. Maviel T, Durkin TP, Menzaghi F, Bontempi B. Sites of neocortical reorganization critical for remote spatial memory. Science. 2004;305:96-9.

24. Wiltgen BJ, Brown RA, Talton LE, Silva AJ. New circuits for old memories:the role of the neocortex in consolidation. Neuron. 2004;44:101-8.

25. Zhu L, Fan JH, Chao FL, Zhou CN, Jiang L, Zhang Y, et al. Running exercise protects spinophilin-immunoreactive puncta and neurons in the medial prefrontal cortex of APP/PS1 transgenic mice. J Comp Neurol. 2021.

26. Wang F, Gomez-Sintes R, Boya P. Lysosomal membrane permeabilization and cell death. Traffic. 2018;19:918-31.

27. Chin MY, Patwardhan AR, Ang KH, Wang AL, Alquezar C, Welch M, et al. Genetically Encoded, pH-Sensitive mTFP1 Biosensor for Probing Lysosomal pH. ACS Sens. 2021;6:2168-80.

28. Rafiq S, McKenna SL, Muller S, Tschan MP, Humbert M. Lysosomes in acute myeloid leukemia:potential therapeutic targets? Leukemia. 2021;35:2759-70.

29. Gomez-Sintes R, Ledesma MD, Boya P. Lysosomal cell death mechanisms in aging. Ageing Res Rev 2016;32:150-68.

30. Saftig P, Puertollano R. How Lysosomes Sense, Integrate, and Cope with Stress. Trends in Biochemical Sciences. 2021;46:97-112.

31. Xu H, Ren D. Lysosomal physiology. Annual Review of Physiology. 2015;77:57-80.

32. di Ronza A, Bajaj L, Sharma J, Sanagasetti D, Lotfi P, Adamski CJ, et al. CLN8 is an endoplasmic reticulum cargo receptor that regulates lysosome biogenesis. Nature Cell Biology. 2018;20:1370-7.

33. Gao J, Langemeyer L, Kummel D, Reggiori F, Ungermann C. Molecular mechanism to target the endosomal Mon1-Ccz1 GEF complex to the pre-autophagosomal structure. Elife. 2018;7.

34. Nordmann M, Cabrera M, Perz A, Brocker C, Ostrowicz C, Engelbrecht-Vandre S, et al. The Mon1-Ccz1 complex is the GEF of the late endosomal Rab7 homolog Ypt7. Curr Biol. 2010;20:1654-9.

35. Jager S, Bucci C, Tanida I, Ueno T, Kominami E, Saftig P, et al. Role for Rab7 in maturation of late autophagic vacuoles. J Cell Sci. 2004;117:4837-48.

36. Gutierrez MG, Munafo DB, Beron W, Colombo MI. Rab7 is required for the normal progression of the autophagic pathway in mammalian cells. J Cell Sci. 2004;117:2687-97.

37. Chen X, Guan Y, Zhang Y, Jia Y, Li W, Guo C, et al. Programmed cell death 4 modulates lysosomal function by inhibiting TFEB translation. Cell Death Differ. 2021;28:1237-50.

38. Guo H, Pu M, Tai Y, Chen Y, Lu H, Qiao J, et al. Nuclear miR-30b-5p suppresses TFEB-mediated lysosomal biogenesis and autophagy. Cell Death Differ. 2021;28:320-36.

39. Kim J, Kundu M, Viollet B, Guan KL. AMPK and mTOR regulate autophagy through direct phosphorylation of Ulk1. Nat Cell Biol. 2011;13:132-41.

40. Vahsen BF, Ribas VT, Sundermeyer J, Boecker A, Dambeck V, Lenz C, et al. Inhibition of the autophagic protein ULK1 attenuates axonal degeneration in vitro and in vivo, enhances translation, and modulates splicing. Cell Death Differ. 2020;27:2810-27.

41. Laker RC, Drake JC, Wilson RJ, Lira VA, Lewellen BM, Ryall KA, et al. Ampk phosphorylation of Ulk1 is required for targeting of mitochondria to lysosomes in exercise-induced mitophagy. Nat Commun. 2017;8:548.

42. Li X, Yu W, Qian X, Xia Y, Zheng Y, Lee JH, et al. Nucleus- Translocated ACSS2 Promotes Gene Transcription for Lysosomal Biogenesis and Autophagy. Mol Cell. 2017;66:684-97 e9.

43. Li G, Kidd J, Kaspar C, Dempsey S, Bhat OM, Camus S, et al. Podocytopathy and Nephrotic Syndrome in Mice with Podocyte-Specific Deletion of the Asah1 Gene: Role of Ceramide Accumulation in Glomeruli. The American Journal of Pathology. 2020 Mar 16.

44. Bisht K, Okojie KA, Sharma K, Lentferink DH, Sun YY, Chen HR, et al. Capillary-associated microglia regulate vascular structure and function through PANX1-P2RY12 coupling in mice. Nat Commun. 2021;12:5289.

45. Kreher C, Favret J, Maulik M, Shin D. Lysosomal Functions in Glia Associated with Neurodegeneration. Biomolecules. 2021;11.

46. Allende ML, Zhu H, Kono M, Hoachlander-Hobby LE, Huso VL, Proia RL. Genetic defects in the sphingolipid degradation pathway and their effects on microglia in neurodegenerative disease. Cell Signal. 2021;78:109879.

47. Bao J, Zheng L, Zhang Q, Li X, Zhang X, Li Z, et al. Deacetylation of TFEB promotes fibrillar Abeta degradation by upregulating lysosomal biogenesis in microglia. Protein Cell. 2016;7:417-33.

48. Martini-Stoica H, Cole AL, Swartzlander DB, Chen F, Wan YW, Bajaj L, et al. TFEB enhances astroglial uptake of extracellular tau species and reduces tau spreading. J Exp Med. 2018;215:2355-77.

49. Tremblay ME, Cookson MR, Civiero L. Glial phagocytic clearance in Parkinson’s disease. Mol Neurodegener. 2019;14:16.

50. Belaya I, Kucharikova N, Gorova V, Kysenius K, Hare DJ, Crouch PJ, et al. Regular Physical Exercise Modulates Iron Homeostasis in the 5xFAD Mouse Model of Alzheimer’s Disease. Int J Mol Sci. 2021;22.

Author Information X