Loading

Journal of Chronic Diseases and Management

Exercise for Neurodegeneration-Related Disorders

Short Note | Open Access | Volume 1 | Issue 1

  • 1. Department of Psychology, University of Gothenburg, Sweden
  • 2. Centre for Empowerment and Well-being, University of Gothenburg, Sweden
+ Show More - Show Less
Corresponding Authors
Trevor Archer, Department of Psychology, University of Gothenburg, Sweden, Tel: 0704-668623
Abstract

Neurodenerative processes associated with ageing or retarded normal neurodevelopment compromise several domains of health, well-being and the functional capacity of individuals, particularly those of advanced age. Physical exercise has provided a plethora of improvements in functional capacity, neurocognitive ability, neuroaffective status and brain plasticity. Despite all these achievements, further effort requires to be invested in order to challenge one current conviction that there exist no effective treatments, or even a paucity, of intervention, e.g. exercise, available to retard or hinder or reverse the Neurodegeneration processes afflicting the diseased brain.

Keywords

•    Neurodegeneration
•    Exercise
•    Intervention
•    Improvement
•    Health

Citation

Archer T, Jacobsson C (2016) Exercise for Neurodegeneration-Related Disorders. J Chronic Dis Manag 1(1): 1001.

INTRODUCTION

The regular practice of physical exercise, whether engaged upon for the maintenance of accustomed health condition or acceded to under pressures of an unhealthy or diseased condition, invariably advances well-being and structural and functional integrity through: the advancement of functional and biomarker manifestations during ageing and cellular senescence, the amelioration of cognitive performance deficits by optimal augmentation of cerebral plasticity and the enrichment of individuals’ proclivities for advantageous ontogenetic and epigenetic dispositions. The notion of ‘organismal robustness’, through which ‘dormant’, or otherwise, genetic predispositions will translate into disease in individuals with decreased organismal robustness [1], offers a growing, as yet underestimated, preventional/interventional aspect of decelerating ageing-related debilities and is bolstered by the concomitant aspect of ‘organismal resilience’; regular exercise/ activity has been shown to offer consistent benefit for maximizing organismal resilience against a broad range of extrinsic and intrinsic stressors, such as infections, injury/surgery, wound-healing, toxicants, genetic predispositions and frailty [2]. The benefits of exercise for specific neurodegeneration-related disorders are consistently documented: In Parkinson’s disease patients exercise programs generally increase quality-of-life and fitness condition [3,4] and motor performance [5,6] but not always in restoring the loss of dopamine (DA) innervation [7]. Nevertheless Shi et al. [8], have demonstrated that physical exercise induced neuroprotective-restorative effects by reducing the degeneration of the nigrostriatal DA system and curtailing the abnormal neuronal spike firing in parkinsonian striatum. Furthermore, exercise interventions in PD increased both trophic factors and functional capacity, e.g. brain-derived neurotrophic factor (BDNF), and neuroplasticity of DA neurons [9-12]. Under conditions of both normal ageing, Alzheimer’s disease (AD) and other types of dementia, physical exercise improves physical health and capacity, quality-of-life, brain plasticity, increasing cognition and reducing the risk of cognitive decline and dementia in later life, as well as greater integrity at different levels of neuronal and brain regional organization [13-16]. The overall purpose of the present treatise is to outline several domains of physical exercise intervention may induce improvements in motor performance and daily activity capacity, neurocognitive functioning and biomarkers of functional and healthy ageing.

Lifestyle and non-invasive, e.g. exercise and dietary considerations, interventions are employed with increasing frequency and efficacy in the facilitation of healthy neurocognitive and biological aging [17-20], particularly since the avoidance of a sedentary existence bears with it essential ingredients for health promotion, necessarily brain health, and prevention of lifestyle-related diseases [21]. Exercise interventions have been shown to be neurorestorative: In MPTP-treated mice showing procedural and working memory impairments and dopamine D2 receptor hypersensitivity, horizontal treadmill running over six weeks ameliorated these deficits [22]. The range of adaptive response to regular physical exercise incorporates several neuroprotection, anti-neurodegenerative and neurorestorative manifestations pertaining to function and biomarker integrity [23-27]; these benefits include also the up-regulation of the enzymatic antioxidant systems and modulation of oxidative damage [28]. For example, among individuals presenting cerebral palsy, whether as children, adolescents or younger/older adults, the recommendation for activity and reduction of sedentary behavior are a burgeoning necessity [29]. Exercise, through the modulation of intensity-dependent metabolism and/or directly-activated reactive oxygen species-generating enzymes, coordinates the cellular redox state of the ageing/degenerating brain. Reactive oxygen species contribute towards the self-renewing, proliferating and differentiating aspects of neuronal stem cells as well as exercise-mediated neurogenesis and neurorestorative effects [30]. The promotion and maintenance of physical exercise habits among older people demands greater consideration and diligence regarding ”activeness” as an ’health-attitude’ and present-day lifestyle within a workable social context, this requires initiatives that galvanize a broader range activity habits rather than the limitations of discrete activities [31,32].

Physical exercise forestalls both cellular senescence and immunosenescence with active elderly individuals at lower risk for deterioration through a multitude of malignancies including cancer forms, e.g. prostate and colon, osteoporosis, depression and dementia [33], in many cases extending life-expectancy by several years and patients’ quality-of-life [34,35], and metabolic processes [36,37]. Activity programs induce major effects on the neuroimmune system functioning and alter dramatically cytokine production, particularly IL-6, IL-1, TNF-α, IL-18 and IFN gamma, which are involved actively in the modulation of synaptic plasticity and neurogenesis [38]. These cytokines may contribute also to reactive oxygen species production through which alterations affect the availability of lipids, proteins, and DNA and regulate directly brain function and integrity [30]. Zimmer et al. [39], have described the influence of physical activity upon objective and subjective cancer-related cognitive impairments in 19 studies involving both humans and laboratory rodents. They observed patient/rodent improvements in both types of studies and posited the general conclusion that the activity-exercise programs reduced inflammation and provided partial benefits for cancer-related cognitive performances. Among chronic diabetic patients, prevention programs are imperative for minimizing the risk of onset of neurodegenerative diseases since a single bout of exercise was found to be efficacious in obese, glucose-intolerant laboratory rodents [40]. Cerebrovascular complications, caused by inflammatory, oxidative, and metabolic changes expressed in diabetes type II patients may induce blood-brain-barrier breakdown may allow peripherally-located pro-inflammatory molecules, e.g. ceramides, to infiltrate thereby activating stress pathways with subsequent promotion of several neuropathological features of dementia including brain insulin resistance, mitochondrial dysfunction, and accumulation of neurotoxic beta-amyloid oligomers, with consequential and subsequent synaptic loss, neuronal dysfunction, and cell death [41]. Physical exercise augmented antioxidative capacity, reduced oxidative stress, and induced anti-inflammatory effects buttressing endothelial function with accompanying elevations of brain capillarization and angiogenesis. Exercise also counteracted dyslipidemia and reduced the increased levels of ceramide and enhanced beta-amyloid clearance through up-regulation of beta-amyloid transporters, elevated basal testosterone, reduced in diabetes II, and promoted neurogenesis.

The influences of physical exercise parameters, such as whether endurance, i.e. aerobic, or resistance, intensity, duration, frequency, type of muscular contraction, extent of exertion and solicited energetic metabolism upon neuroprotective expressions over a range of neurodegenerative disorders has been explored to a limited extent. Applying mouse models of spinal muscular atrophy, high intensity swimming and low intensity running activities have provided behavioural, biochemical and cellular markers of ameliorative manifestations [42,43]. Spinal muscular atrophy presents a collection of autosomal recessive neurodegenerative diseases that differ with regard to clinical outcome, characterized by the specific loss of spinal motor neurons, caused by insufficient level of expression of the protein survival of motor neuron. Chali et al. [44], have shown that both types of exercise, swimming and running, enhanced markedly motor neuron integrity and survival, independent of disorder expression, thereby promulgating the maintenance of neuromuscular junctions and skeletal muscle phenotypes, with particular regard to the soleus, plantaris and tibialis of the exercised mice. Critically, both types of exercises improved dramatically the properties of neuromuscular excitability. Additionally, all of the exercise-activity engender benefits were both quantitatively and qualitatively associated with the specific characteristics of each type of exercise, which implies that the correspondent neuroprotective effects were strongly dependent on the specific activation of certain motor neuron subpopulations. Normal aging, accompanied by energy process dysregulation, directs microglia towards a pro-inflammatory phenotype with subsequent release of IL-1β and IL-6 [45-47], whereas exercise exerts an anti-inflammatory effect [48-51]. Littlefield et al. [50], observed that voluntary running wheel exercise bolstered the induction of a neuroprotective microglia phenotype against pro-inhlammatory reductions in hippocampal neurogenesis in aged rat brains.

Neuroprotective effects have been disclosed repeatedly among ageing individual as most generally obtained in laboratory studies. Due to the induction of neuroprotective mechanisms, e.g. neurotrophic factors and angiogenesis, exercise exerts a neuroprotective effect upon the progression of manifest dementia [52,53]. In aged rats (27 month-old), swimming exercise combined with diselenide-supplemented diet rendered marked neuroprotective effects as displayed by reduction of apoptosis and glial cell activation [54]. Within the context of traumatic brain injuries, such as stroke, Otsuka et al. [55] have demonstrated that preconditioning exercise schedules enhanced the levels of expression of midkine, brain-derived neurotrophic factor, glial fibrillary acidic protein, modulating cell communication and regulating the blood-brain barrier, and platelet endothelial cell adhesion molecule, involved in leucocyte transmigration and angiogenesis, in the Exercised group compared with the expression levels in the Non-exercised group following brain ischemia. In contrast, the expression levels of activated caspase 3 and NT were reduced in the area surrounding the necrotic lesion thereby reducing neuronal apoptosis and oxidative stress. Physical exercise antagonized abnormal activations of the RhoA/Rho kinase pathway, involved in neuroinflammatory and pro-oxidative responses, axonal retraction, and apoptosis; the pathway is linked to aging-related neurodegenerative mechanisms, thereby providing a marked extent of neuroprotection in aged rats [56]. In this context, the issue of whether or not an extended exercise regime offers long-lasting resistance, i.e. neuroprotective alterations, to beta-amyloid-induced network dysfunction in hippocampal cell population activity poses a relevant question. Isla et al. [57], observed that hippocampal cell populations’ activity that was recorded in slices obtained from voluntarily-exercised mice that were provided with free access to a running wheel over a period of 21 days displayed greater power and faster frequency composition than those hippocampal slices obtained from sedentary animals. Hippocampal networks from exercising mice that were rendered insensitive to beta-amyloid-induced inhibition of spontaneous population activity prompting the conclusion that voluntary exercise produced a long-lasting neuroprotective influence upon the hippocampal tissue. Trivino-Paredes et al. [58], have provided a comprehensive description of the interactive influences of gonadal hormones, stress hormones and metabolic hormones upon hippocampal structural plasticity with regard to the mediatory role of physical exercise parameters, frequency, duration and intensity and training regimes.

One hallmark of the pathophysiological progression of Alzheimer’s disease and dementia conditions is observed in the severe hippocampal atrophy brought about by inexorable neuronal loss. Long-term physical exrcise diminished hippocampal CA1 neuron loss linked with the complete abolishment of spatial memory deficits [59]. These influences of exercise upon the integrity of hippocampal and other brain regions involved in higher levels of functioning underline the multidomain importance of exercise interventions for prevention of cognitive decline and somatic concomitants of deterioration [60]. It is increasingly evident that globally structured exercise programs/schedules ought to be designed to alleviate different aspects of psychophysiological function in elderly populations with the chosen activity regimes varying with ‘training-volume’ in relation to age, gender, exercise background [61]. According to the notions of Laitman and John [62] age-related cognitive decline is driven by CNS structural and functional deterioration, neurovascular decline and pro-inflammatory (microglia) reactivity; in this context, physical exercise, through reduction of systemic inflammation, promotion of angiogenesis and neurogenesis, provides both neuroprotective and neurorestorative manifestations. Lancioni et al. [63], observed that patients’ improved commitment and indications of positive personal affected strongly their applicability and potential benefits of the program in daily contexts. Similarly, exercise interventions effectively minimized the decline in activities of daily living in patients diagnosed with dementia [64].

Future considerations need to appraise more systematically the relationships between functional decline and the neurorestorative properties of different types of exercise interventions. Critically, efforts must be made to challenge one current conviction that there exist no effective treatments, or even a paucity, of intervention, e.g. exercise, available to retard or hinder or reverse the neurodegeneration processes afflicting the diseased brain.

REFERENCES

1. Queitsch C, Carlson KD, Girirajan S. Lessons from model organisms: phenotypic robustness and missing heritability in complex disease. PLoS Genet. 2012; 8: 1003041.

2. Huffman DM, Schafer MJ, LeBrasseur NK. Energetic Interventions for healthspan and resiliency with aging. Exp Gerontol. 2016; 0531-5565: 30147-30154.

3. Ayan C, Varela S, Vila MH, Seijo M, Cancela JM. Treadmill training combined with water and land-based exercise programs: Effects on Parkinson’s disease patients. Neuro Rehabilitation. 2016; 30.

4. Lee J, Choi M, Yoo Y. A Meta-Analysis of Nonpharmacological Interventions for People With Parkinson’s Disease. Clin Nurs Res. 2016.

5. Mohammadi-Abdar H, Ridgel AL, Discenzo FM, Loparo KA. Design and Development of a Smart Exercise Bike for Motor Rehabilitation in Individuals with Parkinson’s Disease. IEEE ASME Trans Mechatron. 2016; 21: 1650-1658.

6. Sedaghati P, Daneshmandi H, Karimi N, Barati AH. A Selective Corrective Exercise to Decrease Falling and Improve Functional Balance in Idiopathic Parkinson’s Disease. Trauma Mon. 2016; 21: 23573.

7. Hood RL, Liguore WA, Moore C, Pflibsen L, Meshul CK. Exercise intervention increases spontaneous locomotion but fails to attenuate dopaminergic system loss in a progressive MPTP model in aged mice. Brain Res. 2016; 0006-8993: 30456-30465.

8. Shi K, Liu X, Qiao D, Hou L. Effects of Treadmill Exercise on Spontaneous Firing Activities of Striatal Neurons in a Rat Model of Parkinson’s Disease. Motor Control. 2016.

9. Angelucci F, Piermaria J, Gelfo F, Shofany J, Tramontano M, Fiore M, et al. The effects of motor rehabilitation training on clinical symptoms and serum BDNF levels in Parkinson’s disease subjects. Can J Physiol Pharmacol. 2016; 94: 455-461.

10. Campos C, Rocha NB, Lattari E, Paes F, Nardi AE, Machado S. Exercise-induced neuroprotective effects on neurodegenerative diseases: the key role of trophic factors. Expert Rev Neurother. 2016; 16: 723-734.

11. da Silva PG, Domingues DD, de Carvalho LA, Allodi S, Correa CL. Neurotrophic factors in Parkinson’s disease are regulated by exercise: Evidence-based practice. J Neurol Sci. 2016; 363: 5-15.

12. Shin MS, Jeong HY, An DI, Lee HY, Sung YH. Treadmill exercise facilitates synaptic plasticity on dopaminergic neurons and fibers in the mouse model with Parkinson’s disease. Neurosci Lett. 2016; 621: 28-33.

13. Bernardo TC, Marques-Aleixo I, Beleza J, Oliveira PJ, Ascensão A, Magalhães J. Physical exercise and brain mitochondrial fitness: The possible role againstalzheimer’s disease. Brain Pathol. 2016.

14. Herring A, Münster Y, Metzdorf J, Bolczek B, Krüssel S, Krieter D, et al. Late running is not too late against Alzheimer’s pathology. Neurobiol Dis. 2016; 94: 44-54.

15. Inskip M, Mavros Y, Sachdev PS, Fiatarone Singh MA. Exercise for Individuals with Lewy Body Dementia: A Systematic Review. PLoS One. 2016; 11: 0156520.

16. Sobol NA, Hoffmann K, Frederiksen KS, Vogel A, Vestergaard K, Brændgaard H, et al. Effect of aerobic exercise on physical performance in patients withAlzheimer’s disease. Alzheimers Dement. 2016; 1552- 5260, 30002-30009.

17. Archer T. Physical exercise alleviates debilities of normal aging and Alzheimer’s disease. Acta Neurol Scand. 2011; 123: 221-238.

18. Archer T, Fredriksson A, Schütz E, Kostrzewa RM. Influence of physical exercise on neuroimmunological functioning and health: aging and stress. Neurotox Res. 2011; 20: 69-83.

19. Archer T, Kostrzewa RM. Physical Exercise Alleviates Health Defects, Symptoms, and Biomarkers in Schizophrenia Spectrum Disorder. Neurotox Res. 2015; 28: 268-280.

20. Smith JC, Lancaster MA, Nielson KA, Woodard JL, Seidenberg M, Durgerian S, et al. Interactive effects of physical activity and APOE-ε4 on white matter tract diffusivity in healthy elders. Neuroimage. 2016; 131: 102-112.

21. Archer T. The influence of physical exercise on well-being and health. J Psychiat Psychol Res: Well-being, Empowerment Affect Profiles. 2016; 1.

22. Aguiar AS Jr, Lopes SC, Tristão FS, Rial D, de Oliveira G, da Cunha C, et al. Exercise Improves Cognitive Impairment and Dopamine Metabolism in MPTP-Treated Mice. Neurotox Res. 2016; 29: 118-125.

23. Archer T, Fredriksson A. Physical exercise attenuates MPTP-induced deficits in mice. Neurotox Res. 2010; 18: 313-327.

24. Archer T, Fredriksson A. Delayed exercise-induced functional and neurochemical partial restoration following MPTP. Neurotox Res. 2012; 21:210-221.

25. Archer T, Fredriksson A. The yeast product Milmed enhances the effect of physical exercise on motor performance and dopamine neurochemistry recovery in MPTP-lesioned mice. Neurotox Res. 2013; 24: 393-406.

26. Archer T, Garcia D, Fredriksson A. Restoration of MPTP-induced deficits by exercise and Milmed(®) co-treatment PeerJ. 2014; 2: 531.

27. Fredriksson A, Stigsdotter IM, Hurtig A, Ewalds-Kvist B, Archer T. Running wheel activity restores MPTP-induced functional deficits. J Neural Transm (Vienna). 2011; 118: 407-420.

28. Medeiros RF, Gaique TG, Bento-Bernardes T, Motta NA, Brito FC, Fernandes-Santos C, et al. Aerobic training prevents oxidative profile, improves nitric oxide and vascular reactivity in rats with cardiometabolic alteration. J Appl Physiol (1985). 2016.

29. Verschuren O, Peterson MD, Balemans AC, Hurvitz EA. Exercise and physical activity recommendations for people with cerebral palsy. Dev Med Child Neurol. 2016; 58: 798-808.

30. Radak Z, Suzuki K, Higuchi M, Balogh L, Boldogh I, Koltai E. Physical exercise, reactive oxygen species and neuroprotection. Free Radic Biol Med. 2016.

31. Barnett I, Guell C, Ogilvie D. How do couples influence each other’s physical activity behaviours in retirement? An exploratory qualitative study. BMC Public Health. 2013; 13:1197.

32. Guell C, Shefer G, Griffin S, Ogilvie D. ‘Keeping your body and mind active’: an ethnographic study of aspirations for healthy ageing. BMJ Open. 2016; 6: 009973.

33. Ahlskog JE, Geda YE, Graff-Radford NR, Petersen RC. Physical exercise as a preventive or disease-modifying treatment of dementia and brain aging. Mayo Clin Proc. 2011; 86: 876-884.

34. Dhutia H, Sharma S. Playing it safe: exercise and cardiovascular health. Practitioner. 2015; 259, 15-20.

35. Gerritsen JK, Vincent AJ. Exercise improves quality of life in patients with cancer: a systematic review and meta-analysis of randomized controlled trials. Br J Sports Med. 2016; 50: 796-803.

36. Joseph AM, Nguyen LM, Welter AE, Dominguez JM 2nd, Behnke BJ, Adhihetty PJ. Mitochondrial adaptations evoked with exercise are associated with a reduction in age-induced testicular atrophy in Fischer-344 rats. Biogerontology. 2014; 15: 517-534. 

37. Joseph AM, Adhihetty PJ, Leeuwenburgh C. Beneficial effects of exercise on age-related mitochondrial dysfunction and oxidative stress in skeletal muscle. J Physiol. 2015.

38. Archer T, Fredriksson A, Johansson B. Exercise alleviates Parkinsonism: clinical and laboratory evidence. Acta Neurol Scand. 2011; 123: 73-84.

39. Zimmer P, Baumann FT, Oberste M, Wright P, Garthe A, Schenk A, et al. Effects of Exercise Interventions and Physical Activity Behavior on Cancer Related Cognitive Impairments: A Systematic Review. Biomed Res Int. 2016; 2016:1820954.

40. MacPherson RE, Baumeister P, Peppler WT, Wright DC, Little JP. Reduced cortical BACE1 content with one bout of exercise is accompanied by declines in AMPK, Akt, and MAPK signaling in obese, glucose-intolerant mice. J Appl Physiol (1985).2015; 119: 1097-1104.

41. Bertram S, Brixius K, Brinkmann C. Exercise for the diabetic brain: how physical training may help prevent dementia and Alzheimer’s disease in T2DM patients. Endocrine. 2016.

42. Biondi O, Lopes P, Desseille C, Branchu J, Chali F, Ben Salah A, et al. Physical exercise reduces cardiac defects in type 2 spinal muscular atrophy-like mice. J Physiol. 2012; 590: 5907-5925.

43. Biondi O, Branchu J, Ben Salah A, Houdebine L, Bertin L, Chali F, et al. IGF-1R Reduction Triggers Neuroprotective Signaling Pathways in Spinal Muscular Atrophy Mice. J Neurosci. 2015; 35: 12063-12079.

44. Chali F, Desseille C, Houdebine L, Benoit E, Rouquet T, Bariohay B, et al. Long-term exercise-specific neuroprotection in spinal muscular atrophy-like mice. J Physiol. 2016; 594: 1931-1952.

45. Sierra A, Gottfried-Blackmore AC, McEwen BS, Bulloch K. Microglia derived from aging mice exhibit an altered inflammatory profile. Glia. 2007; 55: 412-424.

46. Sierra A, Beccari S, Diaz-Aparicio I, Encinas JM, Comeau S, Tremblay MÈ. Surveillance, phagocytosis, and inflammation: how never-resting microglia influence adult hippocampal neurogenesis. Neural Plast. 2014; 2014: 610343.

47. Yin F, Sancheti H, Patil I, Cadenas E. Energy metabolism and inflammation in brain aging and Alzheimer’s disease. Free Radic Biol Med. 2016; 0891-5849, 30216-30217.

48. Kohman RA. Aging microglia: relevance to cognition and neural plasticity. Methods Mol Biol. 2012; 934: 193-218.

49. Kohman RA, DeYoung EK, Bhattacharya TK, Peterson LN, Rhodes JS. Wheel running attenuates microglia proliferation and increases expression of a proneurogenic phenotype in the hippocampus of aged mice. Brain Behav Immun.2012; 26: 803-810.

50. Littlefield AM, Setti SE, Priester C, Kohman RA. Voluntary exercise attenuates LPS-induced reductions in neurogenesis and increases microglia expression of a proneurogenic phenotype in aged mice. J Neuroinflammation. 2015; 12: 138.

51. Marlatt MW, Bauer J, Aronica E, van Haastert ES, Hoozemans JJ, Joels M. Proliferation in the Alzheimer hippocampus is due to microglia, not astroglia, and occurs at sites of amyloid deposition. Neural Plast. 2014: 693851.

52. Hoffmann K, Frederiksen KS, Sobol NA, Beyer N, Vogel A, Simonsen AH, et al. Preserving cognition, quality of life, physical health and functional ability in Alzheimer’s disease: the effect of physical exercise (ADEX trial): rationale and design. Neuroepidemiology. 2013; 41: 198-207.

53. Hoffmann K, Sobol NA, Frederiksen KS, Beyer N, Vogel A, Vestergaard K, et al. Moderate-to-High Intensity Physical Exercise in Patients with Alzheimer’s Disease: A Randomized Controlled Trial. J Alzheimers Dis. 2015; 50: 443-453. 

54. Leite MR, Cechella JL, Pinton S, Nogueira CW, Zeni G. A Diphenyl Diselenide-Supplemented Diet and Swimming Exercise PromoteNeuroprotection, Reduced Cell Apoptosis and Glial Cell Activation in the Hypothalamus of Old Rats. Exp Gerontol. 2016; 82: 1-7.

55. Otsuka S, Sakakima H, Sumizono M, Takada S, Terashi T, Yoshida Y. The neuroprotective effects of preconditioning exercise on brain damage and neurotrophic factors after focal brain ischemia in rats. Behav Brain Res. 2016; 303: 9-18.

56. Muñoz A, Corrêa CL, Villar-Cheda B, Costa-Besada MA, Labandeira-Garcia JL. Aging-related Increase in Rho Kinase Activity in the Nigral Region Is Counteracted by Physical Exercise. J Gerontol A Biol Sci Med Sci. 2015.

57. Isla AG, Vázquez-Cuevas FG, Peña-Ortega F. Exercise Prevents Amyloid-β-Induced Hippocampal Network Disruption by Inhibiting GSK3β Activation. J Alzheimers Dis. 2016; 52: 333-343.

58. Triviño-Paredes J, Patten AR, Gil-Mohapel J, Christie BR. The effects of hormones and physical exercise on hippocampal structural plasticity. Front Neuroendocrinol. 2016; 41: 23-43.

59. Hüttenrauch M, Brauß A, Kurdakova A, Borgers H, Klinker F, Liebetanz D, et al. Physical activity delays hippocampal neurodegeneration and rescues memory deficits in an Alzheimer disease mouse model. Transl Psychiatry. 2016.

60. Mollenhauer B. Can we prevent and slow down neurodegeneration with diet and exercise? Mov Dis. 2015.

61. Mendonca GV, Pezarat-Correia P, Vaz JR, Silva L, Almeida ID, Heffernan KS. Impact of Exercise Training On Physiological Measures of Physical Fitness in the Elderly. Curr Aging Sci. 2016.

62. Laitman BM, John GR. Understanding how exercise promotes cognitive integrity in the aging brain. PLoS Biol. 2015; 13: 1002300.

63. Lancioni GE, Singh NN, O’Reilly MF, Sigafoos J, D’Amico F, Addante LM, et al. Persons With Advanced Alzheimer’s Disease Engage in Mild Leg Exercise Supported by Technology-Aided Stimulation and Prompts. Behav Modif. 2016.

64. Laver K, Dyer S, Whitehead C, Clemson L, Crotty M. Interventions to delay functional decline in people with dementia: a systematic review of systematic reviews. BMJ Open. 2016; 6: 010767.

Archer T, Jacobsson C (2016) Exercise for Neurodegeneration-Related Disorders. J Chronic Dis Manag 1(1): 1001

Received : 08 Jun 2016
Accepted : 09 Jul 2016
Published : 13 Jul 2016
Journals
Annals of Otolaryngology and Rhinology
ISSN : 2379-948X
Launched : 2014
JSM Schizophrenia
Launched : 2016
Journal of Nausea
Launched : 2020
JSM Internal Medicine
Launched : 2016
JSM Hepatitis
Launched : 2016
JSM Oro Facial Surgeries
ISSN : 2578-3211
Launched : 2016
Journal of Human Nutrition and Food Science
ISSN : 2333-6706
Launched : 2013
JSM Regenerative Medicine and Bioengineering
ISSN : 2379-0490
Launched : 2013
JSM Spine
ISSN : 2578-3181
Launched : 2016
Archives of Palliative Care
ISSN : 2573-1165
Launched : 2016
JSM Nutritional Disorders
ISSN : 2578-3203
Launched : 2017
Annals of Neurodegenerative Disorders
ISSN : 2476-2032
Launched : 2016
Journal of Fever
ISSN : 2641-7782
Launched : 2017
JSM Bone Marrow Research
ISSN : 2578-3351
Launched : 2016
JSM Mathematics and Statistics
ISSN : 2578-3173
Launched : 2014
Journal of Autoimmunity and Research
ISSN : 2573-1173
Launched : 2014
JSM Arthritis
ISSN : 2475-9155
Launched : 2016
JSM Head and Neck Cancer-Cases and Reviews
ISSN : 2573-1610
Launched : 2016
JSM General Surgery Cases and Images
ISSN : 2573-1564
Launched : 2016
JSM Anatomy and Physiology
ISSN : 2573-1262
Launched : 2016
JSM Dental Surgery
ISSN : 2573-1548
Launched : 2016
Annals of Emergency Surgery
ISSN : 2573-1017
Launched : 2016
Annals of Mens Health and Wellness
ISSN : 2641-7707
Launched : 2017
Journal of Preventive Medicine and Health Care
ISSN : 2576-0084
Launched : 2018
Annals of Vaccines and Immunization
ISSN : 2378-9379
Launched : 2014
JSM Heart Surgery Cases and Images
ISSN : 2578-3157
Launched : 2016
Annals of Reproductive Medicine and Treatment
ISSN : 2573-1092
Launched : 2016
JSM Brain Science
ISSN : 2573-1289
Launched : 2016
JSM Biomarkers
ISSN : 2578-3815
Launched : 2014
JSM Biology
ISSN : 2475-9392
Launched : 2016
Archives of Stem Cell and Research
ISSN : 2578-3580
Launched : 2014
Annals of Clinical and Medical Microbiology
ISSN : 2578-3629
Launched : 2014
JSM Pediatric Surgery
ISSN : 2578-3149
Launched : 2017
Journal of Memory Disorder and Rehabilitation
ISSN : 2578-319X
Launched : 2016
JSM Tropical Medicine and Research
ISSN : 2578-3165
Launched : 2016
JSM Head and Face Medicine
ISSN : 2578-3793
Launched : 2016
JSM Cardiothoracic Surgery
ISSN : 2573-1297
Launched : 2016
JSM Bone and Joint Diseases
ISSN : 2578-3351
Launched : 2017
JSM Bioavailability and Bioequivalence
ISSN : 2641-7812
Launched : 2017
JSM Atherosclerosis
ISSN : 2573-1270
Launched : 2016
Journal of Genitourinary Disorders
ISSN : 2641-7790
Launched : 2017
Journal of Fractures and Sprains
ISSN : 2578-3831
Launched : 2016
Journal of Autism and Epilepsy
ISSN : 2641-7774
Launched : 2016
Annals of Marine Biology and Research
ISSN : 2573-105X
Launched : 2014
JSM Health Education & Primary Health Care
ISSN : 2578-3777
Launched : 2016
JSM Communication Disorders
ISSN : 2578-3807
Launched : 2016
Annals of Musculoskeletal Disorders
ISSN : 2578-3599
Launched : 2016
Annals of Virology and Research
ISSN : 2573-1122
Launched : 2014
JSM Renal Medicine
ISSN : 2573-1637
Launched : 2016
Journal of Muscle Health
ISSN : 2578-3823
Launched : 2016
JSM Genetics and Genomics
ISSN : 2334-1823
Launched : 2013
JSM Anxiety and Depression
ISSN : 2475-9139
Launched : 2016
Clinical Journal of Heart Diseases
ISSN : 2641-7766
Launched : 2016
Annals of Medicinal Chemistry and Research
ISSN : 2378-9336
Launched : 2014
JSM Pain and Management
ISSN : 2578-3378
Launched : 2016
JSM Women's Health
ISSN : 2578-3696
Launched : 2016
Clinical Research in HIV or AIDS
ISSN : 2374-0094
Launched : 2013
Journal of Endocrinology, Diabetes and Obesity
ISSN : 2333-6692
Launched : 2013
Journal of Substance Abuse and Alcoholism
ISSN : 2373-9363
Launched : 2013
JSM Neurosurgery and Spine
ISSN : 2373-9479
Launched : 2013
Journal of Liver and Clinical Research
ISSN : 2379-0830
Launched : 2014
Journal of Drug Design and Research
ISSN : 2379-089X
Launched : 2014
JSM Clinical Oncology and Research
ISSN : 2373-938X
Launched : 2013
JSM Bioinformatics, Genomics and Proteomics
ISSN : 2576-1102
Launched : 2014
JSM Chemistry
ISSN : 2334-1831
Launched : 2013
Journal of Trauma and Care
ISSN : 2573-1246
Launched : 2014
JSM Surgical Oncology and Research
ISSN : 2578-3688
Launched : 2016
Annals of Food Processing and Preservation
ISSN : 2573-1033
Launched : 2016
Journal of Radiology and Radiation Therapy
ISSN : 2333-7095
Launched : 2013
JSM Physical Medicine and Rehabilitation
ISSN : 2578-3572
Launched : 2016
Annals of Clinical Pathology
ISSN : 2373-9282
Launched : 2013
Annals of Cardiovascular Diseases
ISSN : 2641-7731
Launched : 2016
Journal of Behavior
ISSN : 2576-0076
Launched : 2016
Annals of Clinical and Experimental Metabolism
ISSN : 2572-2492
Launched : 2016
Clinical Research in Infectious Diseases
ISSN : 2379-0636
Launched : 2013
JSM Microbiology
ISSN : 2333-6455
Launched : 2013
Journal of Urology and Research
ISSN : 2379-951X
Launched : 2014
Journal of Family Medicine and Community Health
ISSN : 2379-0547
Launched : 2013
Annals of Pregnancy and Care
ISSN : 2578-336X
Launched : 2017
JSM Cell and Developmental Biology
ISSN : 2379-061X
Launched : 2013
Annals of Aquaculture and Research
ISSN : 2379-0881
Launched : 2014
Clinical Research in Pulmonology
ISSN : 2333-6625
Launched : 2013
Journal of Immunology and Clinical Research
ISSN : 2333-6714
Launched : 2013
Annals of Forensic Research and Analysis
ISSN : 2378-9476
Launched : 2014
JSM Biochemistry and Molecular Biology
ISSN : 2333-7109
Launched : 2013
Annals of Breast Cancer Research
ISSN : 2641-7685
Launched : 2016
Annals of Gerontology and Geriatric Research
ISSN : 2378-9409
Launched : 2014
Journal of Sleep Medicine and Disorders
ISSN : 2379-0822
Launched : 2014
JSM Burns and Trauma
ISSN : 2475-9406
Launched : 2016
Chemical Engineering and Process Techniques
ISSN : 2333-6633
Launched : 2013
Annals of Clinical Cytology and Pathology
ISSN : 2475-9430
Launched : 2014
JSM Allergy and Asthma
ISSN : 2573-1254
Launched : 2016
Journal of Neurological Disorders and Stroke
ISSN : 2334-2307
Launched : 2013
Annals of Sports Medicine and Research
ISSN : 2379-0571
Launched : 2014
JSM Sexual Medicine
ISSN : 2578-3718
Launched : 2016
Annals of Vascular Medicine and Research
ISSN : 2378-9344
Launched : 2014
JSM Biotechnology and Biomedical Engineering
ISSN : 2333-7117
Launched : 2013
Journal of Hematology and Transfusion
ISSN : 2333-6684
Launched : 2013
JSM Environmental Science and Ecology
ISSN : 2333-7141
Launched : 2013
Journal of Cardiology and Clinical Research
ISSN : 2333-6676
Launched : 2013
JSM Nanotechnology and Nanomedicine
ISSN : 2334-1815
Launched : 2013
Journal of Ear, Nose and Throat Disorders
ISSN : 2475-9473
Launched : 2016
JSM Ophthalmology
ISSN : 2333-6447
Launched : 2013
Journal of Pharmacology and Clinical Toxicology
ISSN : 2333-7079
Launched : 2013
Annals of Psychiatry and Mental Health
ISSN : 2374-0124
Launched : 2013
Medical Journal of Obstetrics and Gynecology
ISSN : 2333-6439
Launched : 2013
Annals of Pediatrics and Child Health
ISSN : 2373-9312
Launched : 2013
JSM Clinical Pharmaceutics
ISSN : 2379-9498
Launched : 2014
JSM Foot and Ankle
ISSN : 2475-9112
Launched : 2016
JSM Alzheimer's Disease and Related Dementia
ISSN : 2378-9565
Launched : 2014
Journal of Addiction Medicine and Therapy
ISSN : 2333-665X
Launched : 2013
Journal of Veterinary Medicine and Research
ISSN : 2378-931X
Launched : 2013
Annals of Public Health and Research
ISSN : 2378-9328
Launched : 2014
Annals of Orthopedics and Rheumatology
ISSN : 2373-9290
Launched : 2013
Journal of Clinical Nephrology and Research
ISSN : 2379-0652
Launched : 2014
Annals of Community Medicine and Practice
ISSN : 2475-9465
Launched : 2014
Annals of Biometrics and Biostatistics
ISSN : 2374-0116
Launched : 2013
JSM Clinical Case Reports
ISSN : 2373-9819
Launched : 2013
Journal of Cancer Biology and Research
ISSN : 2373-9436
Launched : 2013
Journal of Surgery and Transplantation Science
ISSN : 2379-0911
Launched : 2013
Journal of Dermatology and Clinical Research
ISSN : 2373-9371
Launched : 2013
JSM Gastroenterology and Hepatology
ISSN : 2373-9487
Launched : 2013
Annals of Nursing and Practice
ISSN : 2379-9501
Launched : 2014
JSM Dentistry
ISSN : 2333-7133
Launched : 2013
Author Information X