Treino de baixa intensidade mantem a funcionalidade e fibrose em camundongos mdx

Autores

DOI:

https://doi.org/10.34024/rnc.2022.v30.13541

Palavras-chave:

modelo mdx, diafragma, músculo cardíaco, fibrose, vo2max., Duchenne

Resumo

Objetivo. O presente estudo investigou os efeitos de um protocolo de treinamento de baixa intensidade sobre parâmetros funcionais, aeróbios, morfológicos e morfométricos do diafragma e músculo cardíaco de camundongos mdx. Método. Camundongos mdx machos com 8 semanas de idadeforam submetidos a um protocolo curto (4 semanas) ou longo(8 semanas) em esteira horizontal (9m/min,3x/semana,30min/dia). Os camundongos mdx foram distribuídos aleatoriamente no grupo treinado (mdxT, n=8) ou não treinado (mdxNT, n=8). Medidas de força, equilíbrio, análise do VO2 máximo, tempo de exaustão dos animais e histomorfometria das fibras de colágeno intramuscular foram avaliadas ao longo do tempo do protocolo no diafragma e músculo cardíaco. Resultados. Não houve diferenças no teste de força de preensão e Rotarod ou na deposição de fibrose do musculo cardíaco. No musculo diafragma houve um aumento na porcentagem de fibrose intramuscular no grupo mdxT no T4, aumento da fibrose tanto do grupo mdxT como mdxNT no T8 quando comparados ao grupo controle T0. Nas variáveis VO2máx e tempo de exaustão, não houve um desempenho significante mesmo com o fator tempo e treinamento. Os achados histopatológicos são observados com maior frequência no final do protocolo longo (8 semanas) tanto no grupo mdxT como mdxNT, principalmente no músculo diafragma. Conclusão. O treino de baixa intensidade em esteira não influenciou na deposição de fibrose no músculo cardíaco e diafragma mdx ao longo do tempo mantendo a força de preensão e a capacidade aeróbia dos camundongos mdx durante o protocolo.

Downloads

Não há dados estatísticos.

Métricas

Carregando Métricas ...

Referências

Chakkalakal JV, Thompson J, Parks RJ, Jasmin BJ. Molecular, cellular, and pharmacological therapies for Duchenne/Becker muscular dystrophies. FASEB J 2005;19:880-91. https://doi.org/10.1096/fj.04-1956rev

Eagle M. Report on the Muscular Dystrophy Campaign workshop: Exercise in neuromuscular diseases Newcastle, January 2002. Neuromuscul Disord 2002;12:975-83.

https://doi.org/10.1016/s0960-8966(02)00136-0

Morrison J, Lu QL, Pastoret C, Partridge T, Bou-Gharios G. T-cell-dependent fibrosis in the mdx dystrophic mouse. Lab Investig 2000;80:881-91. https://doi.org/10.1038/labinvest.3780092

Araujo APQC, Nardes F, Fortes CPDD, Pereira JA, Rebel MF, Dias CM, et al. Brazilian consensus on duchenne muscular dystrophy. Part 2: Rehabilitation and systemic care. Arq Neuropsiquiatr 2018;76:481-9. https://doi.org/10.1590/0004-282X20180062

Lessa TB, Carvalho RC, Spagnolo JD, Silva LCLC, Cortopassi SRG, Ambrósio CE. Laparoscopic guided local injection in the X-linked muscular dystrophy mouse (MDX) diaphragm. an advance in experimental therapies for duchenne muscular dystrophy1. Acta Cir Bras 2014;29:715-20. https://doi.org/10.1590/S0102-86502014001800004

Bendixen RM, Lott DJ, Senesac C, Mathur S, Vandenborne K. Participation in daily life activities and its relationship to strength and functional measures in boys with Duchenne muscular dystrophy. Disabil Rehabil 2014;36:1918-23.

https://doi.org/10.3109/09638288.2014.883444

Grounds MD, Radley HG, Lynch GS, Nagaraju K, De Luca A. Towards developing standard operating procedures for pre-clinical testing in the mdx mouse model of Duchenne muscular dystrophy. Neurobiol Dis 2008;31:1-19. https://doi.org/10.1016/j.nbd.2008.03.008

Vainzof M, Ayub-Guerrieri D, Onofre PCG, Martins PCM, Lopes VF, Zilberztajn D, et al. Animal models for genetic neuromuscular diseases. J Mol Neurosci 2008;34:241-8. https://doi.org/10.1007/s12031-007-9023-9

Selsby JT, Acosta P, Sleeper MM, Barton ER, Sweeney HL. Long-term wheel running compromises diaphragm function but improves cardiac and plantarflexor function in the mdx mouse. J Appl Physiol 2013;115:660-6. https://doi.org/10.1152/japplphysiol.00252.2013

Nigro G, Comi LI, Politano L, Bain RJI. The incidence and evolution of cardiomyopathy in Duchenne muscular dystrophy. Int J Cardiol 1990;26:271-7. https://doi.org/10.1016/0167-5273(90)90082-g

Benigni R, Bossa C, Tcheremenskaia O, Giuliani A. Alternatives to the carcinogenicity bioassay: In silico methods, and the in vitro and in vivo mutagenicity assays. Expert Opin Drug Metab Toxicol 2010;6:809-19. https://doi.org/10.1517/17425255.2010.486400

Laflamme MA, Murry CE. Heart regeneration. Nature 2011;473:326-35. https://doi.org/10.1038/nature10147

Shete AN, Bute SS, Deshmukh PR. A study of VO2 max and body fat percentage in female athletes. J Clin Diagnostic Res 2014;8:BC01-3. https://doi.org/10.7860/JCDR/2014/10896.5329

Ranković G, Mutavdžić V, Toskić D, Preljević A, Kocić M, Nedin-Ranković G, et al. Aerobic capacity as an indicator in different kinds of sports. Bosn J Basic Med Sci 2010;10:44-8. https://doi.org/10.17305/bjbms.2010.2734

Kodama S. CLINICIAN’S CORNER Cardiorespiratory Fitness as a Quantitative Predictor of All-Cause Mortality and Cardiovascular Events. J Am Med Assoc 2009;301:2024-35. https://doi.org/10.1001/jama.2009.681

Hyzewicz J, Ruegg UT, Takeda S. Comparison of Experimental Protocols of Physical Exercise for mdx Mice and Duchenne Muscular Dystrophy Patients. J Neuromuscul Dis 2015;2:325-42. https://doi.org/10.3233/JND-150106

Kaczor JJ, Hall JE, Payne E, Tarnopolsky MA. Low intensity training decreases markers of oxidative stress in skeletal muscle of mdx mice. Free Radic Biol Med 2007;43:145-54.

https://doi.org/10.1016/j.freeradbiomed.2007.04.003

Hyzewicz J, Tanihata J, Kuraoka M, Ito N, Miyagoe-Suzuki Y, Takeda S. Low intensity training of mdx mice reduces carbonylation and increases expression levels of proteins involved in energy metabolism and muscle contraction. Free Radic Biol Med 2015;82:122-36. http://dx.doi.org/10.1016/j.freeradbiomed.2015.01.023

du Sert NP, Hurst V, Ahluwalia A, Alam S, Avey MT, Baker M, et al. The arrive guidelines 2.0: Updated guidelines for reporting animal research. PLoS Biol 2020;18:1-12.

https://doi.org/10.1371/journal.pbio.3000410

Luca A. Use of treadmill and wheel exercise for impact on mdx mice phenotype. Neuromuscul Netw Protoc 2011;(Id):1-11. https://treat-nmd.org/wp-content/uploads/2016/08/MDX-DMD_M.2.1.001.pdf

van Putten M, Winter C, van Roon-Mom W, van Ommen GJ, ’t Hoen PAC, Aartsma-Rus A. A 3 months mild functional test regime does not affect disease parameters in young mdx mice. Neuromuscul Disord 2010;20:273-80. http://dx.doi.org/10.1016/j.nmd.2010.02.004

Aartsma-Rus A, van Putten M. Assessing functional performance in the Mdx mouse model. J Vis Exp 2014;85:1-11. https://doi.org/10.3791/51303

Rocco AB, Levalley JC, Eldridge JA, Marsh SA, Rodgers BD. A novel protocol for assessing exercise performance and dystropathophysiology in the mdx mouse. Muscle and Nerve 2014;50:541-8. https://doi.org/10.1002/mus.24184

Høydal MA, Wisløff U, Kemi OJ, Ellingsen Ø. Running speed and maximal oxygen uptake in rats and mice: Practical implications for exercise training. Eur J Prev Cardiol 2007;14:753-60.

https://doi.org/10.1097/HJR.0b013e3281eacef1

Melo DS, Costa-Pereira LV, Santos CS, Mendes BF, Costa KB, Santos CFF, et al. Severe calorie restriction reduces cardiometabolic risk factors and protects rat hearts from ischemia/reperfusion injury. Front Physiol 2016;7:1-8. https://doi.org/10.3389/fphys.2016.00106

Smith LR, Barton ER. Collagen content does not alter the passive mechanical properties of fibrotic skeletal muscle in mdx mice. Am J Physiol Cell Physiol 2014;306:C889-98.

https://doi.org/10.1152/ajpcell.00383.2013

Capogrosso RF, Mantuano P, Cozzoli A, Sanarica F, Massari AM, Conte E, et al. Contractile efficiency of dystrophic mdx mouse muscle: In vivo and ex vivo assessment of adaptation to exercise of functional end points. J Appl Physiol 2017;122:828-43.

https://doi.org/10.1152/japplphysiol.00776.2015

Bassaglia Y, Gautron J. Fast and slow rat muscles degenerate and regenerate differently after whole crush injury. J Muscle Res Cell Motil 1995;16:420-9. https://doi.org/10.1007/BF00114507

Delaney K, Kasprzycka P, Ciemerych MA, Zimowska M. The role of TGF-β1 during skeletal muscle regeneration. Cell Biol Int 2017;41:706-15. https://doi.org/10.1002/cbin.10725

Yucel N, Chang AC, Day JW, Rosenthal N, Blau HM. Humanizing the mdx mouse model of DMD: the long and the short of it. NPJ Regen Med 2018;3:4. https://doi.org/10.1038/s41536-018-0045-4

Corrado G, Lissoni A, Beretta S, Terenghi L, Tadeo G, Foglia-Manzillo G, et al. Prognostic value of electrocardiograms, ventricular late potentials, ventricular arrhythmias, and left ventricular systolic dysfunction in patients with Duchenne muscular dystrophy. Am J Cardiol 2002;89:838-41. https://doi.org/10.1016/s0002-9149(02)02195-1

Kamogawa Y, Biro S, Maeda M, Setoguchi M, Hirakawa T, Yoshida H, et al. Dystrophin-deficient myocardium is vulnerable to pressure overload in vivo. Cardiovasc Res 2001;50:509-15.

https://doi.org/10.1016/s0008-6363(01)00205-x

Finsterer J, Stöllberger C. The heart in human dystrophinopathies. Cardiology 2003;99:1-19. https://doi.org/10.1159/000068446

Costas JM, Nye DJ, Henley JB, Plochocki JH. Voluntary exercise induces structural remodeling in the hearts of dystrophin-deficient mice. Muscle Nerve 2010;42:881-5.

https://doi.org/10.1002/mus.21783

Nakamura A. X-linked dilated cardiomyopathy: A cardiospecific phenotype of dystrophinopathy. Pharmaceuticals 2015;8:303-20. https://doi.org/10.3390/ph8020303

Ferry A, Benchaouir R, Joanne P, Peat RA, Mougenot N, Agbulut O, et al. Effect of voluntary physical activity initiated at age 7 months on skeletal hindlimb and cardiac muscle function in mdx mice of both genders. Muscle Nerve 2015;52:788-94.

https://doi.org/10.1002/mus.24604

Van Erp C, Loch D, Laws N, Trebbin A, Hoey AJ. Timeline of cardiac dystrophy in 3-18-month-old mdx mice. Muscle Nerve 2010;42:504-13. https://doi.org/10.1002/mus.21716

Schill KE, Altenberger AR, Lowe J, Periasamy M, Villamena FA, Rafael-Fortney JiA, et al. Muscle damage, metabolism, and oxidative stress in mdx mice: Impact of aerobic running. Muscle Nerve 2016;54:110-7. https://doi.org/10.1002/mus.25015

Deconinck AE, Rafael JA, Skinner JA, Brown SC, Potter AC, Metzinger L, et al. Utrophin-dystrophin-deficient mice as a model for Duchenne muscular dystrophy. Cell 1997;90:717-27.

https://doi.org/10.1016/s0092-8674(00)80532-2

Willmann R, Possekel S, Dubach-Powell J, Meier T, Ruegg MA. Mammalian animal models for Duchenne muscular dystrophy. Neuromuscul Disord 2009;19:241-9.

http://dx.doi.org/10.1016/j.nmd.2008.11.015

Ishizaki M, Suga T, Kimura E, Shiota T, Kawano R, Uchida Y, et al. Mdx respiratory impairment following fibrosis of the diaphragm. Neuromuscul Disord 2008;18:342-8.

https://doi.org/10.1016/j.nmd.2008.02.002

Dupont-Versteegden EE, McCarter RJ, Katz MS. Voluntary exercise decreases progression of muscular dystrophy in diaphragm of mdx mice. J Appl Physiol 1994;77:1736-41.

https://doi.org/10.1152/jappl.1994.77.4.1736

Barbin ICC, Pereira JA, Bersan Rovere M, Oliveira Moreira D, Marques MJ, Santo Neto H. Diaphragm degeneration and cardiac structure in mdx mouse: Potential clinical implications for Duchenne muscular dystrophy. J Anat 2016;228:784-91.

https://doi.org/10.1111/joa.12443

Frinchi M, MacAluso F, Licciardi A, Perciavalle V, Coco M, Belluardo N, et al. Recovery of damaged skeletal muscle in mdx mice through low-intensity endurance exercise. Int J Sports Med 2014;35:19-27.

https://doi.org/10.1055/s-0033-1343405

Morici G, Frinchi M, Pitruzzella A, Di Liberto V, Barone R, Pace A, et al. Mild Aerobic Exercise Training Hardly Affects the Diaphragm of mdx Mice. J Cell Physiol 2017;232:2044-52.

https://doi.org/10.1002/jcp.25573

Downloads

Publicado

2022-11-22

Como Citar

Santos, A. F., Peixoto Gaid Machado, T. ., Alves, L. A., Santos, A. P., & Dias Machado, A. S. (2022). Treino de baixa intensidade mantem a funcionalidade e fibrose em camundongos mdx. Revista Neurociências, 30, 1–28. https://doi.org/10.34024/rnc.2022.v30.13541

Edição

Seção

Artigos Originais
Recebido: 2022-03-03
Aceito: 2022-06-23
Publicado: 2022-11-22

Artigos mais lidos pelo mesmo(s) autor(es)