Acessibilidade / Reportar erro

Reviewing physical exercise in non-obese diabetic Goto-Kakizaki rats

Abstract

There is a high incidence of non-obese type 2 diabetes mellitus (non-obese-T2DM) cases, particularly in Asian countries, for which the pathogenesis remains mainly unclear. Interestingly, Goto-Kakizaki (GK) rats spontaneously develop insulin resistance (IR) and non-obese-T2DM, making them a lean diabetes model. Physical exercise is a non-pharmacological therapeutic approach to reduce adipose tissue mass, improving peripheral IR, glycemic control, and quality of life in obese animals or humans with T2DM. In this narrative review, we selected and analyzed the published literature on the effects of physical exercise on the metabolic features associated with non-obese-T2DM. Only randomized controlled trials with regular physical exercise training, freely executed physical activity, or skeletal muscle stimulation protocols in GK rats published after 2008 were included. The results indicated that exercise reduces plasma insulin levels, increases skeletal muscle glycogen content, improves exercise tolerance, protects renal and myocardial function, and enhances blood oxygen flow in GK rats.

Exercise intensity; Insulin resistance; Glycemic control; Type 2 diabetes mellitus; Skeletal muscle; Metabolism


Introduction

Type 2 diabetes mellitus: lean vs obese patients

Excessive accumulation of adipose tissue has been considered a public health problem for the last 50 years and today has reached epidemic proportions globally. This condition results from a positive energy balance, in which energy intake exceeds energy expenditure. Indeed, it has been shown that excessive food intake and insufficient physical activity reduce the ability of peripheral tissues to respond to insulin, a process known as insulin resistance (IR) (11. Ioannidis I. The road from obesity to type 2 diabetes. Angiology 2008; 59: 39S-43S, doi: 10.1177/0003319708318583.
https://doi.org/10.1177/0003319708318583...
).

Obesity is a multifactorial disease associated with various genetic and environmental factors. Obese individuals have an increased risk of developing chronic disorders such as hypertension, cardiovascular disease, IR, metabolic syndrome, and type 2 diabetes mellitus (T2DM). Notably, individuals with obesity-related diseases display more severe symptoms and suffer from higher mortality rates when infected with viruses such as COVID-19 (22. Mohammad S, Aziz R, Al Mahri S, Malik SS, Haji E, Khan AH, et al. Obesity and COVID-19: what makes obese host so vulnerable? Immun Ageing 2021; 18: 1, doi: 10.1186/s12979-020-00212-x.
https://doi.org/10.1186/s12979-020-00212...
).

It has been known for decades that obesity leads to T2DM. This disease is a public health problem characterized by high blood sugar concentrations (hyperglycemia) associated with impaired insulin sensitivity in the liver, skeletal muscle, and adipose tissue and reduced insulin secretion (33. Kuwabara WMT, Panveloski-Costa AC, Yokota CNF, Pereira JNB, Filho JM, Torres RP, et al. Comparison of Goto-Kakizaki rats and high fat diet-induced obese rats: are they reliable models to study Type 2 Diabetes mellitus? PLoS One 2017; 12: e0189622, doi: 10.1371/journal.pone.0189622.
https://doi.org/10.1371/journal.pone.018...
,44. Kuwabara WMT, Yokota CNF, Curi R, Alba-Loureiro TC. Obesity and type 2 Diabetes mellitus induce lipopolysaccharide tolerance in rat neutrophils. Sci Rep 2018; 8: 17534, doi: 10.1038/s41598-018-35809-2.
https://doi.org/10.1038/s41598-018-35809...
). It is estimated that T2DM corresponds to 90-95% of all diabetes cases (55. WHO (World Health Organization). Classification of Diabetes mellitus. World Health Organization; 2019. Report No.: 978-92-4-151570-2.). Furthermore, obese T2DM patients typically present a low-grade and chronic inflammation (66. Barazzoni R, Gortan Cappellari G, Ragni M, Nisoli E. Insulin resistance in obesity: an overview of fundamental alterations. Eat Weight Disord 2018; 23: 149-157, doi: 10.1007/s40519-018-0481-6.
https://doi.org/10.1007/s40519-018-0481-...
,77. Kuang J, Chen L, Tang Q, Zhang J, Li Y, He J. The role of Sirt6 in obesity and diabetes. Front Physiol 2018; 9: 135, doi: 10.3389/fphys.2018.00135.
https://doi.org/10.3389/fphys.2018.00135...
), including pro-inflammatory cytokines (TNF-α, IL-6, IL-8, and IFN-α) and C-reactive protein (CRP).

For example, several lines of evidence indicate that adipsin is a fat inflammation-related and visceral fat accumulation biomarker (88. Kyohara M, Shirakawa J, Okuyama T, Togashi Y, Inoue R, Li J, et al. Soluble EGFR, a hepatokine, and adipsin, an adipokine, are biomarkers correlated with distinct aspects of insulin resistance in type 2 diabetes subjects. Diabetol Metab Syndr 2020; 12: 83, doi: 10.1186/s13098-020-00591-7.
https://doi.org/10.1186/s13098-020-00591...
,99. Wang JS, Lee WJ, Lee IT, Lin SY, Lee WL, Liang KW, et al. Association between serum adipsin levels and insulin resistance in subjects with various degrees of glucose intolerance. J Endocr Soc 2019; 3: 403-410, doi: 10.1210/js.2018-00359.
https://doi.org/10.1210/js.2018-00359...
). Other biomarkers, including plasma leptin, resistin, tumor necrosis factor-alpha (TNF-α), plasminogen activator inhibitor 1(PAI-1), interleukins (IL) IL-1β, IL-6, and IL-8, insulin-like growth factor 1 (IGF-1), monocyte chemoattractant protein 1 (MCP-1), and visfatin, have also been reported. The plasma IL-6 concentration was proposed to be a T2DM and cardiovascular disease marker associated with an inflammatory state, and increased systemic IL-6 concentrations have been correlated with increased fat mass in both rodent models and obese humans (44. Kuwabara WMT, Yokota CNF, Curi R, Alba-Loureiro TC. Obesity and type 2 Diabetes mellitus induce lipopolysaccharide tolerance in rat neutrophils. Sci Rep 2018; 8: 17534, doi: 10.1038/s41598-018-35809-2.
https://doi.org/10.1038/s41598-018-35809...
,1010. Wernstedt I, Olsson B, Jernås M, Paglialunga S, Carlsson LM, Smith U, et al. Increased levels of acylation-stimulating protein in interleukin-6-deficient (IL-6(-/-)) mice. Endocrinology 2006; 147: 2690-2695, doi: 10.1210/en.2005-1133.
https://doi.org/10.1210/en.2005-1133...
) . Moreover, reduced serum adiponectin levels in obese individuals were associated with T2DM and attenuated cardiovascular function (1111. Achari AE, Jain SK. Adiponectin, a therapeutic target for obesity, diabetes, and endothelial dysfunction. Int J Mol Sci 2017; 18: 1321, doi: 10.3390/ijms18061321.
https://doi.org/10.3390/ijms18061321...
).

It is well known that chronic inflammation reduces the peripheral insulin response, promoting resistance to the hormone, and, in TNF-α knockout rats, improved insulin sensitivity has been reported. Additionally, obese TNF-α knockout mice have reduced fat pad weights and enhanced responses to exogenous glucose and insulin than obese mice with unaltered peripheral insulin sensitivity. These results are due to high tissue TNF-α levels promoting insulin receptor substrate-1 (IRS-1) serine phosphorylation rather than the tyrosine kinase-dependent phosphorylation, ultimately inhibiting insulin signaling (1212. Furman D, Campisi J, Verdin E, Carrera-Bastos P, Targ S, Franceschi C, et al. Chronic inflammation in the etiology of disease across the life span. Nat Med 2019; 25: 1822-1832, doi: 10.1038/s41591-019-0675-0.
https://doi.org/10.1038/s41591-019-0675-...
).

In the last 25 years, the prevalence of obese T2DM has increased from 13 to 31% in eastern populations (55. WHO (World Health Organization). Classification of Diabetes mellitus. World Health Organization; 2019. Report No.: 978-92-4-151570-2.,1313. Ramachandran A, Snehalatha C, Ma RC. Diabetes in South-East Asia: an update. Diabetes Res Clin Pract 2014; 103: 231-237, doi: 10.1016/j.diabres.2013.11.011.
https://doi.org/10.1016/j.diabres.2013.1...
). However, T2DM also occurs in non-obese individuals (1414. Al-Goblan AS, Al-Alfi MA, Khan MZ. Mechanism linking diabetes mellitus and obesity. Diabetes Metab Syndr Obes 2014; 7: 587-591, doi: 10.2147/DMSO.S67400.
https://doi.org/10.2147/DMSO.S67400...
). For example, in Asia, 70% of T2DM patients are non-obese with body mass index (BMI) values of <19 kg/m2 (1515. Goto Y, Kakizaki M, Masaki N. Production of spontaneous diabetic rats by repetition of selective breeding. Tohoku J Exp Med 1976; 119: 85-90, doi: 10.1620/tjem.119.85.
https://doi.org/10.1620/tjem.119.85...
,1616. Anoop S, Misra A, Bhatt SP, Gulati S, Mahajan H. High fasting C-peptide levels and insulin resistance in non-lean & non-obese (BMI >19 to <25kg/m. Diabetes Metab Syndr 2019; 13: 708-715, doi: 10.1016/j.dsx.2018.11.041.
https://doi.org/10.1016/j.dsx.2018.11.04...
) and present the ketosis-resistant diabetes young (KRDY) phenotype, which is associated with a high propensity for beta-cell function failure. Notably, KRDY patients have a higher mortality rate compared to obese subjects.

A similar disease feature was reported in a comparative study of lean and obese diabetic patients in the United States (1717. Coleman NJ, Miernik J, Philipson L, Fogelfeld L. Lean versus obese diabetes mellitus patients in the United States minority population. J Diabetes Complications 2014; 28: 500-505, doi: 10.1016/j.jdiacomp.2013.11.010.
https://doi.org/10.1016/j.jdiacomp.2013....
). The lean patients from the US study exhibited an increased propensity for pancreatic beta-cell failure, a low total triglycerides/high-density lipoprotein ratio (TG/HDL) (an indirect marker of hepatic IR), and central obesity (i.e., hip circumference) (1717. Coleman NJ, Miernik J, Philipson L, Fogelfeld L. Lean versus obese diabetes mellitus patients in the United States minority population. J Diabetes Complications 2014; 28: 500-505, doi: 10.1016/j.jdiacomp.2013.11.010.
https://doi.org/10.1016/j.jdiacomp.2013....
). Moreover, in a study with non-obese Filipino T2DM patients, Bautista et al. (1818. Bautista FP, Jasul Jr G, Dampil OA. Insulin resistance and β-cell function of lean versus overweight or obese Filipino patients with newly diagnosed type 2 Diabetes mellitus. J ASEAN Fed Endocr Soc 2019; 34: 164-170, doi: 10.15605/jafes.034.02.07.
https://doi.org/10.15605/jafes.034.02.07...
) detected reduced pancreatic beta-cell function using the Homeostasis Model Assessment (HOMA) index compared to overweight/obese patients. In non-obese diabetic patients, the impaired pancreatic beta-cell function is associated with genetic factors, including different alleles in Japanese and Chinese populations (1919. Goto A, Noda M, Goto M, Yasuda K, Mizoue T, Yamaji T, et al. Predictive performance of a genetic risk score using 11 susceptibility alleles for the incidence of type 2 diabetes in a general Japanese population: a nested case-control study. Diabet Med 2018; 35: 602-611, doi: 10.1111/dme.13602.
https://doi.org/10.1111/dme.13602...
,2020. Galli J, Li LS, Glaser A, Ostenson CG, Jiao H, Fakhrai-Rad H, et al. Genetic analysis of non-insulin dependent diabetes mellitus in the GK rat. Nat Genet 1996; 12: 31-37, doi: 10.1038/ng0196-31.
https://doi.org/10.1038/ng0196-31...
), and acquired autoimmunity, requiring exogenous insulin administration for glycemic control (1717. Coleman NJ, Miernik J, Philipson L, Fogelfeld L. Lean versus obese diabetes mellitus patients in the United States minority population. J Diabetes Complications 2014; 28: 500-505, doi: 10.1016/j.jdiacomp.2013.11.010.
https://doi.org/10.1016/j.jdiacomp.2013....
) . Since environmental and genetic factors are associated with non-obese T2DM development, future studies will need to be conducted to understand the underlying pathophysiological mechanisms associated with this disease.

Physical exercise in T2DM subjects

Sedentarism is one of the main factors for developing metabolic syndrome, cardiovascular diseases, and T2DM. Studies have shown that regular physical activity reduces the prevalence and helps manage these obesity-related diseases (2121. Hawley JA, Lessard SJ. Exercise training-induced improvements in insulin action. Acta Physiol (Oxf) 2008; 192: 127-135, doi: 10.1111/j.1748-1716.2007.01783.x.
https://doi.org/10.1111/j.1748-1716.2007...
,2222. Zhao RR, O'Sullivan AJ, Singh MAF. Exercise or physical activity and cognitive function in adults with type 2 diabetes, insulin resistance or impaired glucose tolerance: a systematic review. Eur Rev Aging Phys Act 2018; 15: 1, doi: 10.1186/s11556-018-0190-1.
https://doi.org/10.1186/s11556-018-0190-...
). For example, regular physical activity improved glucose tolerance, blood lipid level profiles, and reduced risk factors for developing cardiovascular diseases in T2DM patients (2323. Ivy JL, Zderic TW, Fogt DL. Prevention and treatment of non-insulin-dependent Diabetes mellitus. Exerc Sport Sci Rev 1999; 27: 1-35, doi: 10.1249/00003677-199900270-00003.
https://doi.org/10.1249/00003677-1999002...
). Additionally, physical exercise programs have been shown to ameliorate glucose uptake via insulin-independent translocation and glucose transporter 4 (GLUT-4) expression, potentiate insulin signaling during the post-exercise period, increase skeletal muscle oxidative capacity, attenuate lipid metabolite generation, up-regulate the expression of genes associated with mitochondrial biogenesis and metabolism, decrease inflammatory markers, and restore immune function (2424. Dos Santos JM, Moreli ML, Tewari S, Benite-Ribeiro SA. The effect of exercise on skeletal muscle glucose uptake in type 2 diabetes: an epigenetic perspective. Metabolism 2015; 64: 1619-1628, doi: 10.1016/j.metabol.2015.09.013.
https://doi.org/10.1016/j.metabol.2015.0...

25. de Matos MA, Duarte TC, Ottone VeO, Sampaio PF, Costa KB, de Oliveira MF, et al. The effect of insulin resistance and exercise on the percentage of CD16(+) monocyte subset in obese individuals. Cell Biochem Funct 2016; 34: 209-216, doi: 10.1002/cbf.3178.
https://doi.org/10.1002/cbf.3178...

26. Mazur-Bialy AI, Pocheć E, Zarawski M. Anti-inflammatory properties of irisin, mediator of physical activity, are connected with TLR4/MyD88 signaling pathway activation. Int J Mol Sci 2017; 18: 701, doi: 10.3390/ijms18040701.
https://doi.org/10.3390/ijms18040701...
-2727. Passos MEP, Borges L, Dos Santos-Oliveira LC, Alecrim-Zeza AL, Lobato TB, de Oliveira HH, et al. Recreational dance practice modulates lymphocyte profile and function in diabetic women. Int J Sports Med 2021; 42: 749-759, doi: 10.1055/a-1309-2037.
https://doi.org/10.1055/a-1309-2037...
). Interestingly, improved mitochondrial function and biogenesis have also been observed in the skeletal muscle of diabetic rats and humans following exercise training (2828. Raza H, John A, Shafarin J, Howarth FC. Exercise-induced alterations in pancreatic oxidative stress and mitochondrial function in type 2 diabetic Goto-Kakizaki rats. Physiol Rep 2016; 4: e12751, doi: 10.14814/phy2.12751.
https://doi.org/10.14814/phy2.12751...
,2929. Bishop DJ, Botella J, Genders AJ, Lee MJC, Saner NJ, Kuang J, et al. High-intensity exercise and mitochondrial biogenesis: current controversies and future research directions. Physiology (Bethesda) 2019; 34: 56-70, doi: 10.1152/physiol.00038.2018.
https://doi.org/10.1152/physiol.00038.20...
).

Furthermore, physical exercise reduces cell hypertrophy and hyperplasia in adipose tissue and increases GLUT-4 expression and insulin sensitivity (3030. Gollisch KS, Brandauer J, Jessen N, Toyoda T, Nayer A, Hirshman MF, et al. Effects of exercise training on subcutaneous and visceral adipose tissue in normal- and high-fat diet-fed rats. Am J Physiol Endocrinol Metab 2009; 297: E495-E504, doi: 10.1152/ajpendo.90424.2008.
https://doi.org/10.1152/ajpendo.90424.20...
). It has also been shown to increase IL-6, IL-1ra, and IL-10 and decrease TNF-α plasma levels, with an inflammation modulatory effect (3131. McFarlin BK, Flynn MG, Campbell WW, Craig BA, Robinson JP, Stewart LK, et al. Physical activity status, but not age, influences inflammatory biomarkers and toll-like receptor 4. J Gerontol A Biol Sci Med Sci 2006; 61: 388-393, doi: 10.1093/gerona/61.4.388.
https://doi.org/10.1093/gerona/61.4.388...
,3232. Galan BS, Carvalho FG, Santos PC, Gobbi RB, Kalva-Filho CA, Papoti M, et al. Effects of taurine on markers of muscle damage, inflammatory response and physical performance in triathletes. J Sports Med Phys Fitness 2018; 58: 1318-1324, doi: 10.23736/S0022-4707.17.07497-7.
https://doi.org/10.23736/S0022-4707.17.0...
). Additionally, the activation of signaling pathways due to muscle contraction increases glucose and fatty acid (FA) metabolism through gene expression regulation. For example, during physical exercise, AMP-activated protein kinase (AMPK) and calcium-calmodulin-dependent protein kinase (CaMK) are activated (3333. Reznick RM, Shulman GI. The role of AMP-activated protein kinase in mitochondrial biogenesis. J Physiol 2006; 574: 33-39, doi: 10.1113/jphysiol.2006.109512.
https://doi.org/10.1113/jphysiol.2006.10...
).

Exercise-induced AMPK activation has been shown to lead to 1) reductions in malonyl-CoA generation, which inhibits carnitine palmitoyltransferase 1 (CPT-1), by phosphorylating and inhibiting acetyl-CoA carboxylase, consequently increasing FA oxidation acutely; 2) increased insulin-independent glucose uptake due to GLUT-4 translocation to the plasma membrane; and 3) up-regulated expression of genes related to mitochondrial metabolism and biogenesis, resulting in persistently increased FA oxidation (3434. Imierska M, Kurianiuk A, Błachnio-Zabielska A. The influence of physical activity on the bioactive lipids metabolism in obesity-induced muscle insulin resistance. Biomolecules 2020; 10: 1665, doi: 10.3390/biom10121665.
https://doi.org/10.3390/biom10121665...
). CaMK regulates genes associated with mitochondrial biogenesis induced by muscle contraction (3333. Reznick RM, Shulman GI. The role of AMP-activated protein kinase in mitochondrial biogenesis. J Physiol 2006; 574: 33-39, doi: 10.1113/jphysiol.2006.109512.
https://doi.org/10.1113/jphysiol.2006.10...
,3535. Smith JAH, Collins M, Grobler LA, Magee CJ, Ojuka EO. Exercise and CaMK activation both increase the binding of MEF2A to the Glut4 promoter in skeletal muscle in vivo. Am J Physiol Endocrinol Metab 2007; 292: E413-E420, doi: 10.1152/ajpendo.00142.2006.
https://doi.org/10.1152/ajpendo.00142.20...
).

GK rats: a lean model of type 2 diabetes mellitus

The Goto-Kakizaki (GK) rat, a spontaneous non-obese-T2DM animal model, was generated by selective inbreeding using glucose-intolerant Wistar rats with a hyperglycemic phenotype (1515. Goto Y, Kakizaki M, Masaki N. Production of spontaneous diabetic rats by repetition of selective breeding. Tohoku J Exp Med 1976; 119: 85-90, doi: 10.1620/tjem.119.85.
https://doi.org/10.1620/tjem.119.85...
). Many studies have utilized GK rats to evaluate non-obese T2DM features, development, and complications (33. Kuwabara WMT, Panveloski-Costa AC, Yokota CNF, Pereira JNB, Filho JM, Torres RP, et al. Comparison of Goto-Kakizaki rats and high fat diet-induced obese rats: are they reliable models to study Type 2 Diabetes mellitus? PLoS One 2017; 12: e0189622, doi: 10.1371/journal.pone.0189622.
https://doi.org/10.1371/journal.pone.018...
,44. Kuwabara WMT, Yokota CNF, Curi R, Alba-Loureiro TC. Obesity and type 2 Diabetes mellitus induce lipopolysaccharide tolerance in rat neutrophils. Sci Rep 2018; 8: 17534, doi: 10.1038/s41598-018-35809-2.
https://doi.org/10.1038/s41598-018-35809...
,3636. Portha B. Programmed disorders of beta-cell development and function as one cause for type 2 diabetes? The GK rat paradigm. Diabetes Metab Res Rev 2005; 21: 495-504, doi: 10.1002/dmrr.566.
https://doi.org/10.1002/dmrr.566...

37. Portha B, Giroix MH, Serradas P, Gangnerau MN, Movassat J, Rajas F, et al. Beta-cell function and viability in the spontaneously diabetic GK rat: information from the GK/Par colony. Diabetes 2001; 50: S89-S93, doi: 10.2337/diabetes.50.2007.S89.
https://doi.org/10.2337/diabetes.50.2007...

38. Panveloski-Costa AC, Kuwabara WMT, Munhoz AC, Lucena CF, Curi R, Carpinelli AR, et al. The insulin resistance is reversed by exogenous 3,5,3'triiodothyronine in type 2 diabetic Goto-Kakizaki rats by an inflammatory-independent pathway. Endocrine 2020; 68: 287-295, doi: 10.1007/s12020-020-02208-5.
https://doi.org/10.1007/s12020-020-02208...
-3939. Pereira JNB, Murata GM, Sato FT, Marosti AR, Carvalho CRO, Curi R. Small intestine remodeling in male Goto-Kakizaki rats. Physiol Rep 2021; 9: e14755, doi: 10.14814/phy2.14755.
https://doi.org/10.14814/phy2.14755...
). These rats display insulin sensitivity impairment and T2DM similar to high-caloric diet-induced obese animals. Interestingly, in contrast to the changes in rats submitted to diet-induced obesity, the increased expression of proinflammatory cytokines is not observed in the skeletal muscle and retroperitoneal adipose tissue depots of 16-week-old GK rats (33. Kuwabara WMT, Panveloski-Costa AC, Yokota CNF, Pereira JNB, Filho JM, Torres RP, et al. Comparison of Goto-Kakizaki rats and high fat diet-induced obese rats: are they reliable models to study Type 2 Diabetes mellitus? PLoS One 2017; 12: e0189622, doi: 10.1371/journal.pone.0189622.
https://doi.org/10.1371/journal.pone.018...
,3838. Panveloski-Costa AC, Kuwabara WMT, Munhoz AC, Lucena CF, Curi R, Carpinelli AR, et al. The insulin resistance is reversed by exogenous 3,5,3'triiodothyronine in type 2 diabetic Goto-Kakizaki rats by an inflammatory-independent pathway. Endocrine 2020; 68: 287-295, doi: 10.1007/s12020-020-02208-5.
https://doi.org/10.1007/s12020-020-02208...
).

GK rats develop T2DM earlier in life, exhibiting a reduced pancreatic beta-cell mass at 16.5 days of fetal age. At 28 days of age, basal hyperglycemia, IR, and impaired insulin secretion by pancreatic beta-cells are observed. However, GK rats present a preserved beta-cell response to other secretagogues, increased hepatic glucose production, liver inflammation and liver glucotoxicity, and late-stage diabetes complications (33. Kuwabara WMT, Panveloski-Costa AC, Yokota CNF, Pereira JNB, Filho JM, Torres RP, et al. Comparison of Goto-Kakizaki rats and high fat diet-induced obese rats: are they reliable models to study Type 2 Diabetes mellitus? PLoS One 2017; 12: e0189622, doi: 10.1371/journal.pone.0189622.
https://doi.org/10.1371/journal.pone.018...
,1515. Goto Y, Kakizaki M, Masaki N. Production of spontaneous diabetic rats by repetition of selective breeding. Tohoku J Exp Med 1976; 119: 85-90, doi: 10.1620/tjem.119.85.
https://doi.org/10.1620/tjem.119.85...
,3737. Portha B, Giroix MH, Serradas P, Gangnerau MN, Movassat J, Rajas F, et al. Beta-cell function and viability in the spontaneously diabetic GK rat: information from the GK/Par colony. Diabetes 2001; 50: S89-S93, doi: 10.2337/diabetes.50.2007.S89.
https://doi.org/10.2337/diabetes.50.2007...
,4040. Guest PC. Characterization of the Goto-Kakizaki (GK) rat model of type 2 diabetes. Methods Mol Biol 2019; 1916: 203-211, doi: 10.1007/978-1-4939-8994-2.
https://doi.org/10.1007/978-1-4939-8994-...
).

GK rats also exhibit typical T2DM hallmarks such as hyperinsulinemia, increased gluconeogenesis, and elevated plasma lipid levels (33. Kuwabara WMT, Panveloski-Costa AC, Yokota CNF, Pereira JNB, Filho JM, Torres RP, et al. Comparison of Goto-Kakizaki rats and high fat diet-induced obese rats: are they reliable models to study Type 2 Diabetes mellitus? PLoS One 2017; 12: e0189622, doi: 10.1371/journal.pone.0189622.
https://doi.org/10.1371/journal.pone.018...
,3636. Portha B. Programmed disorders of beta-cell development and function as one cause for type 2 diabetes? The GK rat paradigm. Diabetes Metab Res Rev 2005; 21: 495-504, doi: 10.1002/dmrr.566.
https://doi.org/10.1002/dmrr.566...
). Indeed, impaired skeletal muscle oxidative capacity has been associated with T2DM pathogenesis in non-obese individuals. It has been suggested that the lower skeletal muscle oxidative capacity and attenuated mRNA expression of the nuclear receptor coactivator PGC1-α is related to impaired mitochondrial function (4141. Nagatomo F, Fujino H, Kondo H, Gu N, Takeda I, Ishioka N, et al. PGC-1α mRNA level and oxidative capacity of the plantaris muscle in rats with metabolic syndrome, hypertension, and type 2 diabetes. Acta Histochem Cytochem 2011; 44: 73-80, doi: 10.1267/ahc.10041.
https://doi.org/10.1267/ahc.10041...
,4242. Caria ACI, Nonaka CKV, Pereira CS, Soares MBP, Macambira SG, Souza BSF. Exercise training-induced changes in microRNAs: beneficial regulatory effects in hypertension, type 2 diabetes, and obesity. Int J Mol Sci 2018; 19: 3608, doi: 10.3390/ijms19113608.
https://doi.org/10.3390/ijms19113608...
).

Xue et al. (4343. Xue B, Sukumaran S, Nie J, Jusko WJ, Dubois DC, Almon RR. Adipose tissue deficiency and chronic inflammation in diabetic Goto-Kakizaki rats. PLoS One 2011; 6: e17386, doi: 10.1371/journal.pone.0017386.
https://doi.org/10.1371/journal.pone.001...
) studied GK rats from 4-20 weeks of age and reported that 412 genes were differentially expressed in white adipose tissue (WAT). This result is not entirely surprising, given that a progressive failure of body fat accumulation was associated with animal age. The authors also detected down-regulated gene expression of fatty acid synthase (Fasn) and signal transducer and activator of transcription-3 (Stat-3), a transcription factor activated by leptin, and up-regulated expression of inflammatory response and interferon-regulated genes. Overall, the GK rats exhibit increased caloric intake, less body fat accumulation, and chronic inflammation in the absence of obesity.

In pancreatic beta-cells, ATP-sensitive K+ (KATP) channel closure, due to plasma membrane depolarization and voltage-dependent calcium channel opening, is crucial for glucose-stimulated insulin secretion (GSIS) (4444. Haber EP, Procópio J, Carvalho CRO, Carpinelli AR, Newsholme P, Curi R. New insights into fatty acid modulation of pancreatic beta-cell function. Int Rev Cytol 2006; 248: 1-41, doi: 10.1016/S0074-7696(06)48001-3.
https://doi.org/10.1016/S0074-7696(06)48...
,4545. Newsholme P, Haber EP, Hirabara SM, Rebelato EL, Procopio J, Morgan D, et al. Diabetes associated cell stress and dysfunction: role of mitochondrial and non-mitochondrial ROS production and activity. J Physiol 2007; 583: 9-24, doi: 10.1113/jphysiol.2007.135871.
https://doi.org/10.1113/jphysiol.2007.13...
). Alterations in KATP channel activity can lead to changes in plasma membrane polarization, impaired insulin secretion, and diabetes mellitus. In GK rats, insufficient KATP channel closure caused by reduced glucose metabolism in pancreatic beta-cells culminates in GSIS impairment (4646. Tsuura Y, Ishida H, Hayashi S, Sakamoto K, Horie M, Seino Y. Nitric oxide opens ATP-sensitive K+ channels through suppression of phosphofructokinase activity and inhibits glucose-induced insulin release in pancreatic beta cells. J Gen Physiol 1994; 104: 1079-1098, doi: 10.1085/jgp.104.6.1079.
https://doi.org/10.1085/jgp.104.6.1079...
).

Our group recently described marked changes in the three small intestine segments (duodenum, jejunum, and ileum) of four-month-old male GK rats (3939. Pereira JNB, Murata GM, Sato FT, Marosti AR, Carvalho CRO, Curi R. Small intestine remodeling in male Goto-Kakizaki rats. Physiol Rep 2021; 9: e14755, doi: 10.14814/phy2.14755.
https://doi.org/10.14814/phy2.14755...
). Compared to Wistar rats, GK rats have a reduced small intestine, diminished crypt depth in the duodenum and ileum, increased crypt depth in the jejunum, and longer and thicker villi in the jejunum and ileum. The three intestine segments also had thicker muscular layers. At the molecular level, GK rats display elevated IL-1β levels in the duodenum and jejunum and elevated NF-κB p65 in all three segments. These changes are associated with a high IL-1β reactivity in the muscle layer, myenteric neurons, and glial cells. Moreover, a significant reduction in submucosal neuron density in the jejunum and ileum, ganglionic hypertrophy in all three intestinal segments, and a slower intestinal transit were observed. It was concluded that IR and T2DM development in GK rats is associated with small intestine morphology changes, tissue inflammation, and decreased intestinal transit.

Considering the evidence discussed above, along with the fact that regular physical activity combined with weight loss improves glucose homeostasis by raising glucose disposal and insulin action in obese T2DM patients (4747. Magkos F, Hjorth MF, Astrup A. Diet and exercise in the prevention and treatment of type 2 Diabetes mellitus. Nat Rev Endocrinol 2020; 16: 545-555, doi: 10.1038/s41574-020-0381-5.
https://doi.org/10.1038/s41574-020-0381-...
), this review sought to evaluate the literature reporting the effects of physical exercise on diabetic features of GK rats.

Methodology

Physical activity/exercise practice in GK rats

Eligibility criteria and key points for selecting the related studies

The PICO (Patient, Intervention, Comparison and Outcome) strategy was used to develop and write this narrative review. This approach reduces the possible risks of bias in the article selection process. This study reviewed and analyzed the published literature on chronic physical exercise interventions in GK rats, focusing on metabolic and physiological alterations. The study results are relevant to understanding how physical exercise can specifically benefit non-obese T2DM patients.

Two review team researchers independently searched electronic databases [PubMed, MedLine, Cochrane, Web of Science, and Scopus] from January to November of 2021 for studies employing T2DM GK rats of any age OR sex AND involving voluntary OR non-voluntary exercise training OR other types of physical activity OR exercise OR exercise training AND non-obese type 2 diabetes mellitus OR muscle electrostimulation from 2008 to 2021. We limited the search to include randomized controlled trials published only in English in international indexed journals.

The articles used in this review were selected by two independent researchers and based on the titles, objectives, methodologies, and results of each article. Discrepancies between the two researchers were resolved by discussion with a third researcher. At the end of the selection process, 16 studies were identified as being associated with the evaluation of the effects of physical activity or exercise training intervention on non-obese T2DM GK rats.

A flow chart of the search strategy is presented in Figure 1, and a summary of the risk of bias is presented in Figure 2. The topics in this review, including obesity, insulin resistance, physical exercise, type 2 diabetes, and metabolism, are supported by solid research and vast literature. Thus, the selected articles facilitate the discussion of the central theme.

Figure 1
Flow chart of the study selection process for review preparation. GK: Goto-Kakizaki.
Figure 2
Risk of bias assessments for review studies of exercise intervention in Goto-Kakizaki rats. Low risk of bias: if present, is unlikely to alter the results seriously; unclear risk of bias: a risk of bias that raises some doubt about results; high risk of bias: bias may alter the results seriously.

Main effects of physical exercise in GK rats

Effects on metabolic and diabetic features

As shown in Supplementary Table S1, the exercise training protocols included moderate-intensity swimming (n=1), moderate-intensity treadmill running (n=9), low-intensity treadmill running (n=3), low-intensity voluntary wheel activity (n=2), and blood restriction and muscle stimulation (n=1).

In general, moderate physical activity markedly improved the diabetic features of GK rats. Tufescu et al. (4848. Tufescu A, Kanazawa M, Ishida A, Lu H, Sasaki Y, Ootaka T, et al. Combination of exercise and losartan enhances renoprotective and peripheral effects in spontaneously type 2 diabetes mellitus rats with nephropathy. J Hypertens 2008; 26: 312-321, doi: 10.1097/HJH.0b013e3282f2450b.
https://doi.org/10.1097/HJH.0b013e3282f2...
) reported that treadmill exercise reduced urinary protein excretion, indicating renoprotective effects, and enhanced type I fiber capillarization and proportion in extensor digitorum longus (EDL) muscle. Type I fibers exhibit a high capacity for oxidative metabolism and are resistant to fatigue. Qi et al. (4949. Qi Z, He J, Zhang Y, Shao Y, Ding S. Exercise training attenuates oxidative stress and decreases p53 protein content in skeletal muscle of type 2 diabetic Goto-Kakizaki rats. Free Radic Biol Med 2011; 50: 794-800, doi: 10.1016/j.freeradbiomed.2010.12.022.
https://doi.org/10.1016/j.freeradbiomed....
) reported that 10-12 week-old GK rats, submitted to 30-60 min of moderate-intensity treadmill exercise six days per week for eight weeks, displayed no differences in body mass or blood glucose levels but had increased serum adiponectin and insulin levels. Moreover, in skeletal muscle, the authors observed enhanced cytochrome c oxidase (COX) activity and mitochondrial DNA (mtDNA) markers, such as COXII protein content, and attenuated p53 protein and tumor p53-induced glycolysis and apoptosis regulator (TIGAR) expression levels.

It is well known that p53 protein regulates aerobic metabolism and is involved in various steps of glycolysis, inhibiting glucose transport through down-regulated GLUT-1 and GLUT-4 gene expression (4949. Qi Z, He J, Zhang Y, Shao Y, Ding S. Exercise training attenuates oxidative stress and decreases p53 protein content in skeletal muscle of type 2 diabetic Goto-Kakizaki rats. Free Radic Biol Med 2011; 50: 794-800, doi: 10.1016/j.freeradbiomed.2010.12.022.
https://doi.org/10.1016/j.freeradbiomed....
,5050. Itahana Y, Itahana K. Emerging roles of p53 family members in glucose metabolism. Int J Mol Sci 2018; 19: 776, doi: 10.3390/ijms19030776.
https://doi.org/10.3390/ijms19030776...
). Additionally, p53 is involved in mitochondrial function, mtDNA content, and biogenesis. The lack of this protein has been associated with low oxygen consumption and high lactate production, favoring substrate flux through the glycolytic pathway to maintain the required ATP production (5151. Olovnikov IA, Kravchenko JE, Chumakov PM. Homeostatic functions of the p53 tumor suppressor: regulation of energy metabolism and antioxidant defense. Semin Cancer Biol 2009; 19: 32-41, doi: 10.1016/j.semcancer.2008.11.005.
https://doi.org/10.1016/j.semcancer.2008...
).

In skeletal muscle of 46-52-week-old GK rats, treadmill training increased the protein levels of phosphorylated AMPK, peroxisome proliferator-activated receptor coactivator 1 alpha (PGC-1α), GLUT-4, lactate dehydrogenase (LDH), monocarboxylate transporters 1-4 (MCT-1 and -4), and COX-IV (5252. Kim SS, Koo JH, Kwon IS, Oh YS, Lee SJ, Kim EJ, et al. Exercise training and selenium or a combined treatment ameliorates aberrant expression of glucose and lactate metabolic proteins in skeletal muscle in a rodent model of diabetes. Nutr Res Pract 2011; 5: 205-213, doi: 10.4162/nrp.2011.5.3.205.
https://doi.org/10.4162/nrp.2011.5.3.205...
). The same study also reported increased glycogen content and citrate synthase activity in the liver and skeletal muscle of rats subjected to exercise training (5252. Kim SS, Koo JH, Kwon IS, Oh YS, Lee SJ, Kim EJ, et al. Exercise training and selenium or a combined treatment ameliorates aberrant expression of glucose and lactate metabolic proteins in skeletal muscle in a rodent model of diabetes. Nutr Res Pract 2011; 5: 205-213, doi: 10.4162/nrp.2011.5.3.205.
https://doi.org/10.4162/nrp.2011.5.3.205...
). These results corroborated reports that exercise-induced glucose uptake (5353. Richter EA, Hargreaves M. Exercise, GLUT4, and skeletal muscle glucose uptake. Physiol Rev 2013; 93: 993-1017, doi: 10.1152/physrev.00038.2012.
https://doi.org/10.1152/physrev.00038.20...
) correlates with skeletal muscle LDH and MCT-1 activities, oxidative capacity, and lactate oxidation. It has been reported that the up-regulation of LDH, MCT-1, and COX-IV protein levels reduces hyperlactatemia in diabetic animals after an exercise session. PGC-1α is involved in mitochondrial biogenesis, insulin sensitivity, and lipid metabolism, playing a central regulator of phenotypic adaptation and substrate utilization induced by physical exercise (5454. Lira VA, Benton CR, Yan Z, Bonen A. PGC-1alpha regulation by exercise training and its influences on muscle function and insulin sensitivity. Am J Physiol Endocrinol Metab 2010; 299: E145-E161, doi: 10.1152/ajpendo.00755.2009.
https://doi.org/10.1152/ajpendo.00755.20...
). AMPK is involved in the activation of mitophagy, autophagy, and lipolysis. AMPK also inhibits other metabolic pathways, such as lipogenesis, protein synthesis, and gluconeogenesis (5555. Vieira R, Junqueira R, Gaspar R, Munoz V. Exercise ativates AMPK signaling: Impact on glucose uptake in the skeletal muscle in aging. J Rehab Ther 2020.,5656. Pinto AP, da Rocha AL, Marafon BB, Rovina RL, Muãoz VR, da Silva LECM, et al. Impact of different physical exercises on the expression of autophagy markers in mice. Int J Mol Sci 2021; 22: 2635, doi: 10.3390/ijms22052635.
https://doi.org/10.3390/ijms22052635...
). Notably, Kim et al. (5252. Kim SS, Koo JH, Kwon IS, Oh YS, Lee SJ, Kim EJ, et al. Exercise training and selenium or a combined treatment ameliorates aberrant expression of glucose and lactate metabolic proteins in skeletal muscle in a rodent model of diabetes. Nutr Res Pract 2011; 5: 205-213, doi: 10.4162/nrp.2011.5.3.205.
https://doi.org/10.4162/nrp.2011.5.3.205...
) reported improved glucose homeostasis, oxidative metabolism, IR, and up-regulated AMPK, PGC-1α, and GLUT-4 protein expression in GK rats after exercise training.

Nitric oxide synthase (NOS) produces nitric oxide (NO), which plays a crucial role in nervous system function and vascular tone (5757. Ding Q, Cecarini V, Keller JN. Interplay between protein synthesis and degradation in the CNS: physiological and pathological implications. Trends Neurosci 2007; 30: 31-36, doi: 10.1016/j.tins.2006.11.003.
https://doi.org/10.1016/j.tins.2006.11.0...
). Increased blood glucose levels are associated with impaired NO production and altered endothelial NOS (eNOS) and nicotinamide adenine nucleotide phosphate (NADPH) oxidase activities (5858. Patti ME, Corvera S. The role of mitochondria in the pathogenesis of type 2 diabetes. Endocr Rev 2010; 31: 364-395, doi: 10.1210/er.2009-0027.
https://doi.org/10.1210/er.2009-0027...
). Indeed, it has been demonstrated that humans and animals with T2DM present impaired mitochondrial oxidative capacity (5858. Patti ME, Corvera S. The role of mitochondria in the pathogenesis of type 2 diabetes. Endocr Rev 2010; 31: 364-395, doi: 10.1210/er.2009-0027.
https://doi.org/10.1210/er.2009-0027...
). Thus, it is plausible that the benefits of moderate exercise on the mitochondrial oxidative capacity of T2DM subjects are mediated by increased NO production (5959. Grijalva J, Hicks S, Zhao X, Medikayala S, Kaminski PM, Wolin MS, et al. Exercise training enhanced myocardial endothelial nitric oxide synthase (eNOS) function in diabetic Goto-Kakizaki (GK) rats. Cardiovasc Diabetol 2008; 7: 34, doi: 10.1186/1475-2840-7-34.
https://doi.org/10.1186/1475-2840-7-34...
). Interestingly, a study performed with plantaris muscle and left cardiac ventricle of 9-week-old GK rats submitted to 9 weeks of running exercise reported enhanced insulin sensitivity and eNOS expression (5959. Grijalva J, Hicks S, Zhao X, Medikayala S, Kaminski PM, Wolin MS, et al. Exercise training enhanced myocardial endothelial nitric oxide synthase (eNOS) function in diabetic Goto-Kakizaki (GK) rats. Cardiovasc Diabetol 2008; 7: 34, doi: 10.1186/1475-2840-7-34.
https://doi.org/10.1186/1475-2840-7-34...
). The same authors showed improved mitochondrial biogenesis and function through increased eNOS, PCG1-α, and UCP3 (uncoupling protein 3) protein expression in the thoracic aorta of GK rats subjected to moderate-intensity exercise (5959. Grijalva J, Hicks S, Zhao X, Medikayala S, Kaminski PM, Wolin MS, et al. Exercise training enhanced myocardial endothelial nitric oxide synthase (eNOS) function in diabetic Goto-Kakizaki (GK) rats. Cardiovasc Diabetol 2008; 7: 34, doi: 10.1186/1475-2840-7-34.
https://doi.org/10.1186/1475-2840-7-34...
).

Another study performed with 18-week-old T2DM rats submitted to 8 and 14 days of physical exercise with or without saxagliptin, an inhibitor of glucagon-like peptide 1 (GLP-1) degradation, investigated eNOS activity stimulation (6060. Keller AC, Knaub LA, Miller MW, Birdsey N, Klemm DJ, Reusch JE. Saxagliptin restores vascular mitochondrial exercise response in the Goto-Kakizaki rat. J Cardiovasc Pharmacol 2015; 65: 137-147, doi: 10.1097/FJC.0000000000000170.
https://doi.org/10.1097/FJC.000000000000...
). The authors found that exercise training improved mitochondrial function and markers in older T2DM rats. Raza et al. (2828. Raza H, John A, Shafarin J, Howarth FC. Exercise-induced alterations in pancreatic oxidative stress and mitochondrial function in type 2 diabetic Goto-Kakizaki rats. Physiol Rep 2016; 4: e12751, doi: 10.14814/phy2.12751.
https://doi.org/10.14814/phy2.12751...
) reported that eight weeks of moderate-intensity exercise inhibited NADPH oxidase activity, reactive oxygen species (ROS) production, and superoxide dismutase (SOD) activity in the pancreas of 11-month-old GK rats. These results suggested improved mitochondrial function due to enhanced oxygen utilization via the modulation of mitochondrial complex IV activity and PPAR-γ levels.

Seven-month-old T2DM rats previously submitted to 8 weeks of treadmill running presented improved insulin sensitivity and citrate synthase activity in the gastrocnemius muscle (6161. Macia M, Pecchi E, Desrois M, Lan C, Vilmen C, Portha B, et al. Exercise training impacts exercise tolerance and bioenergetics in gastrocnemius muscle of non-obese type-2 diabetic Goto-Kakizaki rat in vivo. Biochimie 2018; 148: 36-45, doi: 10.1016/j.biochi.2018.02.014.
https://doi.org/10.1016/j.biochi.2018.02...
). Additionally, the ventricular myocyte of 11-month-old GK rats submitted to 2-3 months of treadmill exercise exhibited increased intercellular Gap junctional (Gja1) expression, a gene involved in heart contraction. It should be pointed out that the increased Gja1 expression was not followed by altered calcium (Ca2+) transport (6262. Salem KA, Qureshi MA, Sydorenko V, Parekh K, Jayaprakash P, Iqbal T, et al. Effects of exercise training on excitation-contraction coupling and related mRNA expression in hearts of Goto-Kakizaki type 2 diabetic rats. Mol Cell Biochem 2013; 380: 83-96, doi: 10.1007/s11010-013-1662-2.
https://doi.org/10.1007/s11010-013-1662-...
), indicating that exercise intervention preserves the ventricular shortening of myocytes and Ca2+ transport (6262. Salem KA, Qureshi MA, Sydorenko V, Parekh K, Jayaprakash P, Iqbal T, et al. Effects of exercise training on excitation-contraction coupling and related mRNA expression in hearts of Goto-Kakizaki type 2 diabetic rats. Mol Cell Biochem 2013; 380: 83-96, doi: 10.1007/s11010-013-1662-2.
https://doi.org/10.1007/s11010-013-1662-...
).

Several authors described the beneficial effect of physical exercise intensity in obese-associated T2DM rat models (6161. Macia M, Pecchi E, Desrois M, Lan C, Vilmen C, Portha B, et al. Exercise training impacts exercise tolerance and bioenergetics in gastrocnemius muscle of non-obese type-2 diabetic Goto-Kakizaki rat in vivo. Biochimie 2018; 148: 36-45, doi: 10.1016/j.biochi.2018.02.014.
https://doi.org/10.1016/j.biochi.2018.02...
,6262. Salem KA, Qureshi MA, Sydorenko V, Parekh K, Jayaprakash P, Iqbal T, et al. Effects of exercise training on excitation-contraction coupling and related mRNA expression in hearts of Goto-Kakizaki type 2 diabetic rats. Mol Cell Biochem 2013; 380: 83-96, doi: 10.1007/s11010-013-1662-2.
https://doi.org/10.1007/s11010-013-1662-...
). However, only three studies evaluated the efficacy of low-intensity exercise in GK rats (6363. Kondo H, Fujino H, Murakami S, Tanaka M, Kanazashi M, Nagatomo F, et al. Low-intensity running exercise enhances the capillary volume and pro-angiogenic factors in the soleus muscle of type 2 diabetic rats. Muscle Nerve 2015; 51: 391-399, doi: 10.1002/mus.24316.
https://doi.org/10.1002/mus.24316...

64. Kupai K, Szabó R, Veszelka M, Awar AA, Török S, Csonka A, et al. Consequences of exercising on ischemia-reperfusion injury in type 2 diabetic Goto-Kakizaki rat hearts: role of the HO/NOS system. Diabetol Metab Syndr 2015; 7: 85, doi: 10.1186/s13098-015-0080-x.
https://doi.org/10.1186/s13098-015-0080-...
-6565. Nakamoto I, Ishihara A. Effects of voluntary running exercise on skeletal muscle properties in nonobese rats with type 2 diabetes. Physiol Res 2020; 69: 73-84, doi: 10.33549/physiolres.934178.
https://doi.org/10.33549/physiolres.9341...
). Kupai et al. (6464. Kupai K, Szabó R, Veszelka M, Awar AA, Török S, Csonka A, et al. Consequences of exercising on ischemia-reperfusion injury in type 2 diabetic Goto-Kakizaki rat hearts: role of the HO/NOS system. Diabetol Metab Syndr 2015; 7: 85, doi: 10.1186/s13098-015-0080-x.
https://doi.org/10.1186/s13098-015-0080-...
) observed cardio-protective effects in GK rats after six weeks of voluntary exercise, which could be associated with increased NOS activity in cardiac and aortic tissues. After three weeks of treadmill running, Kondo et al. (6363. Kondo H, Fujino H, Murakami S, Tanaka M, Kanazashi M, Nagatomo F, et al. Low-intensity running exercise enhances the capillary volume and pro-angiogenic factors in the soleus muscle of type 2 diabetic rats. Muscle Nerve 2015; 51: 391-399, doi: 10.1002/mus.24316.
https://doi.org/10.1002/mus.24316...
) observed a PGC-1α-induced increase in mitochondrial function in GK rat muscle. Moreover, after six weeks of treadmill running, GK rats exhibited enhanced mitochondrial oxidative capacity due to the increased succinate dehydrogenase (SHD) activity, an enzyme associated with microangiopathy prevention (6666. Morifuji T, Murakami S, Fujita N, Kondo H, Fujino H. Exercise training prevents decrease in luminal capillary diameter of skeletal muscles in rats with type 2 diabetes. ScientificWorldJournal 2012; 2012: 645891, doi: 10.1100/2012/645891.
https://doi.org/10.1100/2012/645891...
). Nakamoto et al. (6565. Nakamoto I, Ishihara A. Effects of voluntary running exercise on skeletal muscle properties in nonobese rats with type 2 diabetes. Physiol Res 2020; 69: 73-84, doi: 10.33549/physiolres.934178.
https://doi.org/10.33549/physiolres.9341...
) also reported increased SHD activity and detected up-regulated PGC1-α expression in the soleus muscle of six-week-old GK rats after six weeks of voluntary wheel running. Exercise training increased this animal model's oxidative activity and oxygen uptake in skeletal and cardiac muscles (6767. Ventura-Clapier R. Exercise training, energy metabolism, and heart failure. Appl Physiol Nutr Metab 2009; 34: 336-339, doi: 10.1139/H09-013.
https://doi.org/10.1139/H09-013...
). These results are consistent with these tissues exhibiting high energy metabolism levels (6767. Ventura-Clapier R. Exercise training, energy metabolism, and heart failure. Appl Physiol Nutr Metab 2009; 34: 336-339, doi: 10.1139/H09-013.
https://doi.org/10.1139/H09-013...
,6868. De Sousa E, Lechêne P, Fortin D, N'Guessan B, Belmadani S, Bigard X, et al. Cardiac and skeletal muscle energy metabolism in heart failure: beneficial effects of voluntary activity. Cardiovasc Res 2002; 56: 260-268, doi: 10.1016/S0008-6363(02)00540-0.
https://doi.org/10.1016/S0008-6363(02)00...
) and suggest that low-intensity exercise enhances mitochondrial function and oxidative activity in skeletal and cardiac muscle of lean T2DM subjects.

Previous studies have also shown that hyperglycemia generates advanced glycation end products (AGEs), which have been shown to accumulate in tissues (6969. Rasheed Z, Akhtar N, Haqqi TM. Advanced glycation end products induce the expression of interleukin-6 and interleukin-8 by receptor for advanced glycation end product-mediated activation of mitogen-activated protein kinases and nuclear factor-κB in human osteoarthritis chondrocytes. Rheumatology (Oxford) 2011; 50: 838-851, doi: 10.1093/rheumatology/keq380.
https://doi.org/10.1093/rheumatology/keq...
) and are associated with the down-regulation of skeletal muscle protein synthesis and subsequent skeletal muscle atrophy (7070. Chiu CY, Yang RS, Sheu ML, Chan DC, Yang TH, Tsai KS, et al. Advanced glycation end-products induce skeletal muscle atrophy and dysfunction in diabetic mice via a RAGE-mediated, AMPK-down-regulated, Akt pathway. J Pathol 2016; 238: 470-482, doi: 10.1002/path.4674.
https://doi.org/10.1002/path.4674...
). Using diabetic subjects, Goldin et al. (7171. Goldin A, Beckman JA, Schmidt AM, Creager MA. Advanced glycation end products: sparking the development of diabetic vascular injury. Circulation 2006; 114: 597-605, doi: 10.1161/CIRCULATIONAHA.106.621854.
https://doi.org/10.1161/CIRCULATIONAHA.1...
) demonstrated that AGEs inhibit NO activity, increase ROS production, and impair vasculature. A study performed with GK rats submitted to a combined electro-stimulation treatment and blood flow restriction reported attenuated AGE production and skeletal muscle protein synthesis activation (7272. Tanaka M, Morifuji T, Yoshikawa M, Nakanishi R, Fujino H. Effects of combined treatment with blood flow restriction and low-intensity electrical stimulation on diabetes mellitus-associated muscle atrophy in rats. J Diabetes 2019; 11: 326-334, doi: 10.1111/1753-0407.12857.
https://doi.org/10.1111/1753-0407.12857...
). Based on these findings, it appears that even low-intensity physical exercise reduces AGE levels and could prevent skeletal muscle atrophy in lean T2DM individuals. The exercise training protocols reported in the reviewed articles are summarized in Figure 3.

Figure 3
Main findings reported in the reviewed articles on physical exercise in Goto-Kakizaki (GK) rats.

Varying beneficial effects according to exercise intensity in GK rats

Exercise intensity refers to the amount of energy required for the performance of physical activity per unit of time. Exercise intensity can be measured directly using respiratory gas analysis to quantify oxygen uptake during exercise or through standard regression models to estimate energy expenditure per a given work rate. Another way to express exercise intensity is as multiples of resting oxygen requirement [metabolic equivalents (METs)]. In this approach, one MET corresponds to the amount of oxygen consumed by a resting, awake individual and is equivalent to 3.5 mL O2 per kg body weight per minute. According to previous studies, low-intensity exercise corresponds to activities requiring <3 METs, moderate-intensity requires 3-6 METs, and high-intensity requires >6 METs (7373. Awtry EH, Balady GJ. Exercise and the heart. Cardiology Secrets; 2010. p 311-315.). The American College of Sports Medicine describes moderate-intensity exercise ranges between 40 and 60% of the maximal capacity, whereas high-intensity exercise is above 64% of the maximal capacity. Most studies describe high-intensity exercise as above 80-85% of peak power output or maximal velocity. In general, activities requiring <80% of peak output are not considered high-intensity training (7474. American College of Sports Medicine and American Diabetes Association joint position statement. Diabetes mellitus and exercise. Med Sci Sports Exerc 1997; 29: i-vi.,7575. De Feo P. Is high-intensity exercise better than moderate-intensity exercise for weight loss? Nutr Metab Cardiovasc Dis 2013; 23: 1037-1042, doi: 10.1016/j.numecd.2013.06.002.
https://doi.org/10.1016/j.numecd.2013.06...
).

However, in rats, the exercise intensity tolerance varies depending on the rat strain and associated disease, reflecting a different work intensity and probably altering the final metabolic and physiologic assessments if the correct intensity for each rat strain and comorbidity is not utilized. It is also necessary to consider other variables, such as treadmill inclination, speed, oxygen consumption (VO2), and/or intensity tracker. Despite these potential challenges, all protocols included in this review demonstrate that low- to moderate-intensity exercise (i.e., treadmill, swimming, and voluntary exercise) produced beneficial effects in young and adult GK rats. Studies seeking to determine the exercise intensity tolerance of GK rats still need to be performed.

Maximum VO2 (VO2 max) reflects an adaptation of the cardiorespiratory system and has been employed for validating clinical studies and calculating endurance work capacity (7676. Leandro CG, Levada AC, Hirabara SM, Manhães-de-Castro R, De-Castro CB, Curi R, et al. A program of moderate physical training for Wistar rats based on maximal oxygen consumption. J Strength Cond Res 2007; 21: 751-756, doi: 10.1519/R-20155.1.
https://doi.org/10.1519/R-20155.1...
). It has been reported that VO2 max rates between 50-70% correspond to moderate-intensity exercise (7777. Boutcher YN, Boutcher SH. Exercise intensity and hypertension: what's new? J Hum Hypertens 2017; 31: 157-164, doi: 10.1038/jhh.2016.62.
https://doi.org/10.1038/jhh.2016.62...
,7878. Gomes-Neto M, Durães AR, Reis HFCD, Neves VR, Martinez BP, Carvalho VO. High-intensity interval training versus moderate-intensity continuous training on exercise capacity and quality of life in patients with coronary artery disease: A systematic review and meta-analysis. Eur J Prev Cardiol 2017; 24: 1696-1707, doi: 10.1177/2047487317728370.
https://doi.org/10.1177/2047487317728370...
). In this context, only two studies [Grijalva et al. (5959. Grijalva J, Hicks S, Zhao X, Medikayala S, Kaminski PM, Wolin MS, et al. Exercise training enhanced myocardial endothelial nitric oxide synthase (eNOS) function in diabetic Goto-Kakizaki (GK) rats. Cardiovasc Diabetol 2008; 7: 34, doi: 10.1186/1475-2840-7-34.
https://doi.org/10.1186/1475-2840-7-34...
) and Keller et al. (6060. Keller AC, Knaub LA, Miller MW, Birdsey N, Klemm DJ, Reusch JE. Saxagliptin restores vascular mitochondrial exercise response in the Goto-Kakizaki rat. J Cardiovasc Pharmacol 2015; 65: 137-147, doi: 10.1097/FJC.0000000000000170.
https://doi.org/10.1097/FJC.000000000000...
)] mentioned the use of 50% VO2 max, which was based on a previous study (7979. Criswell D, Powers S, Dodd S, Lawler J, Edwards W, Renshler K, et al. High intensity training-induced changes in skeletal muscle antioxidant enzyme activity. Med Sci Sports Exerc 1993; 25: 1135-1140, doi: 10.1249/00005768-199310000-00009.
https://doi.org/10.1249/00005768-1993100...
). However, it is important to mention that the exercise intensity was not directly assessed in these studies, which can result in significant differences in relation to the markers evaluated in GK rats submitted to moderate physical exercise, as described in Supplementary Table S1.

Three studies used blood lactate analyses before and after exercise to quantify the exercise intensity (6363. Kondo H, Fujino H, Murakami S, Tanaka M, Kanazashi M, Nagatomo F, et al. Low-intensity running exercise enhances the capillary volume and pro-angiogenic factors in the soleus muscle of type 2 diabetic rats. Muscle Nerve 2015; 51: 391-399, doi: 10.1002/mus.24316.
https://doi.org/10.1002/mus.24316...
,6666. Morifuji T, Murakami S, Fujita N, Kondo H, Fujino H. Exercise training prevents decrease in luminal capillary diameter of skeletal muscles in rats with type 2 diabetes. ScientificWorldJournal 2012; 2012: 645891, doi: 10.1100/2012/645891.
https://doi.org/10.1100/2012/645891...
,8080. Tsutsumi E, Murata Y, Sakamoto M, Horikawa E. Effects of exercise on the nephron of Goto-Kakizaki rats: morphological, and advanced glycation end-products and inducible nitric oxide synthase immunohistochemical analyses. J Diabetes Complications 2015; 29: 472-478, doi: 10.1016/j.jdiacomp.2015.03.002.
https://doi.org/10.1016/j.jdiacomp.2015....
). This analysis can detect the thresholds of sub-lactate (∼10 m/min, low-intensity) and supra-lactate (∼25 m/min, moderate-intensity). It is important to mention that sub-lactate threshold intensities have been suggested to promote no significant physiological changes associated with stress responses (8181. Soya H, Mukai A, Deocaris CC, Ohiwa N, Chang H, Nishijima T, et al. Threshold-like pattern of neuronal activation in the hypothalamus during treadmill running: establishment of a minimum running stress (MRS) rat model. Neurosci Res 2007; 58: 341-348, doi: 10.1016/j.neures.2007.04.004.
https://doi.org/10.1016/j.neures.2007.04...
).

The main protocols that analyzed exercise intensities were defined by analyzing the VO2 max and lactate threshold. The primary intensities used were based on references from other rat strains with different diseases, consequently generating differences in metabolic responses in GK rats. As previously described, exercise intensity modulates several physiological parameters, including VO2 and cytochrome c levels (8282. Bedford TG, Tipton CM, Wilson NC, Oppliger RA, Gisolfi CV. Maximum oxygen consumption of rats and its changes with various experimental procedures. J Appl Physiol Respir Environ Exerc Physiol 1979; 47: 1278-1283, doi: 10.1152/jappl.1979.47.6.1278.
https://doi.org/10.1152/jappl.1979.47.6....
). Moreover, regular physical activity or training at low to moderate intensity improves IR by increasing skeletal muscle fatty acid oxidation, augmenting hormonal responses (5252. Kim SS, Koo JH, Kwon IS, Oh YS, Lee SJ, Kim EJ, et al. Exercise training and selenium or a combined treatment ameliorates aberrant expression of glucose and lactate metabolic proteins in skeletal muscle in a rodent model of diabetes. Nutr Res Pract 2011; 5: 205-213, doi: 10.4162/nrp.2011.5.3.205.
https://doi.org/10.4162/nrp.2011.5.3.205...
,8383. Corcoran MP, Lamon-Fava S, Fielding RA. Skeletal muscle lipid deposition and insulin resistance: effect of dietary fatty acids and exercise. Am J Clin Nutr 2007; 85: 662-677, doi: 10.1093/ajcn/85.3.662.
https://doi.org/10.1093/ajcn/85.3.662...
) and modulating gene expression, protein signaling, and anti-inflammatory effects in GK rats (6565. Nakamoto I, Ishihara A. Effects of voluntary running exercise on skeletal muscle properties in nonobese rats with type 2 diabetes. Physiol Res 2020; 69: 73-84, doi: 10.33549/physiolres.934178.
https://doi.org/10.33549/physiolres.9341...
). In this sense, extensive investigations evaluating the effect of exercise intensity on GK rat physiology and health are crucial.

As described in Supplementary Table S1 and Table 1, low to submaximal exercise intensity positively affects GK-T2DM metabolism. Positive effects are reported at intensities of 50-70% of the VO2 max, which corresponds to 15-21 m/min for one hour on the treadmill during each training session. The exercise programs occurred 3-5 times per week and ranged from 8 days to 15 weeks of training. However, it is important to note that exercise intensity varies depending on oxidative stress and other metabolic parameters (8484. Schöler CM, Marques CV, da Silva GS, Heck TG, de Oliveira Junior LP, de Bittencourt Jr PIH. Modulation of rat monocyte/macrophage innate functions by increasing intensities of swimming exercise is associated with heat shock protein status. Mol Cell Biochem 2016; 421: 111-125, doi: 10.1007/s11010-016-2791-1.
https://doi.org/10.1007/s11010-016-2791-...
).

Table 1
Exercise protocols used in each study and the physical intensity mentioned.

Moderate-intensity training was achieved when GK rats ran at 20 m/min (∼75% of the VO2 max), suggesting that this rat model, when trained, can perform moderate-intensity running for one hour (4848. Tufescu A, Kanazawa M, Ishida A, Lu H, Sasaki Y, Ootaka T, et al. Combination of exercise and losartan enhances renoprotective and peripheral effects in spontaneously type 2 diabetes mellitus rats with nephropathy. J Hypertens 2008; 26: 312-321, doi: 10.1097/HJH.0b013e3282f2450b.
https://doi.org/10.1097/HJH.0b013e3282f2...
). Notably, even low-intensity exercise protocols, such as walking or running at around 50% of the VO2 max until the submaximal effort is achieved enhance aerobic and anaerobic functions. This observation is due to exercise training-induced increases in eNOS activity, NO bioavailability, and subsequent vasodilation of the diabetic heart (5959. Grijalva J, Hicks S, Zhao X, Medikayala S, Kaminski PM, Wolin MS, et al. Exercise training enhanced myocardial endothelial nitric oxide synthase (eNOS) function in diabetic Goto-Kakizaki (GK) rats. Cardiovasc Diabetol 2008; 7: 34, doi: 10.1186/1475-2840-7-34.
https://doi.org/10.1186/1475-2840-7-34...
).

Brooks and White (8585. Brooks GA, White TP. Determination of metabolic and heart rate responses of rats to treadmill exercise. J Appl Physiol Respir Environ Exer Physiol 1978; 45: 1009-1015, doi: 10.1152/jappl.1978.45.6.1009.
https://doi.org/10.1152/jappl.1978.45.6....
) reported differential effects in GK rats exposed to three conditions: a) rest; b) 14 m/min, at 1% inclination, easy exercise; and c) 28.7 m/min, at 15% inclination, heavy exercise. All exercise protocols lasted 90 minutes on a treadmill, where VO2, VCO2, and respiratory exchange ratio (RER = VCO2,/VO2) were determined to assess the metabolic responses. Interestingly, the classification and extrapolation of the proposed protocols (light, moderate, and heavy) did not directly mention the VO2 max intensity measurement.

Dudley et al. (8686. Dudley GA, Abraham WM, Terjung RL. Influence of exercise intensity and duration on biochemical adaptations in skeletal muscle. J Appl Physiol Respir Environ Exerc Physiol 1982; 53: 844-850, doi: 10.1152/jappl.1982.53.4.844.
https://doi.org/10.1152/jappl.1982.53.4....
) used a training protocol with 15% inclination for 30, 60, and 90 min at speeds of 10, 20, 30, 40, 50, and 60 m/min. Exercise intensity-related differences were observed in the volumes of fast-twitch white (FTW), fast-twitch red (FTR), and slow-twitch red (STR) fibers and cytochrome c concentrations. For FTR, the best response was detected at 83% VO2 max (∼30 m/min) and between 50-75% VO2 max (10-20 m/min). The best response for STR was observed at 80% VO2 max (30-40 m/min). Moreover, exercise at VO2 max values of <50% did not generate significant mitochondrial adaptations and the responses between 60 and 90 min did not differ statistically.

One crucial point is that there are currently no studies utilizing the GK rat model in an incremental test, which would allow correlations to be made with the VO2 max in the same protocol. Most studies are based on Wistar, Wistar-Kyoto, Sprague-Dawley, and Okamoto-Aoki rat strains (7676. Leandro CG, Levada AC, Hirabara SM, Manhães-de-Castro R, De-Castro CB, Curi R, et al. A program of moderate physical training for Wistar rats based on maximal oxygen consumption. J Strength Cond Res 2007; 21: 751-756, doi: 10.1519/R-20155.1.
https://doi.org/10.1519/R-20155.1...
,8282. Bedford TG, Tipton CM, Wilson NC, Oppliger RA, Gisolfi CV. Maximum oxygen consumption of rats and its changes with various experimental procedures. J Appl Physiol Respir Environ Exerc Physiol 1979; 47: 1278-1283, doi: 10.1152/jappl.1979.47.6.1278.
https://doi.org/10.1152/jappl.1979.47.6....
). It is also important to consider that, as analyzed by Bedford et al. (8282. Bedford TG, Tipton CM, Wilson NC, Oppliger RA, Gisolfi CV. Maximum oxygen consumption of rats and its changes with various experimental procedures. J Appl Physiol Respir Environ Exerc Physiol 1979; 47: 1278-1283, doi: 10.1152/jappl.1979.47.6.1278.
https://doi.org/10.1152/jappl.1979.47.6....
), different VO2 max values vary among animal strain and gender, with male Sprague-Dawley exhibiting values of 85.2 mL·kg-1·min-1, female Sprague-Dawley, 81.1 mL·kg-1·min-1, male Wistar-Kyoto, 65.3 mL·kg-1·min-1, and male Okamoto-Aoki 72.3 mL·kg-1·min-1. Thus, studies assessing and evaluating the experimental approaches employed in exercise studies with the GK rat model are lacking.

Three articles mentioned using low-intensity exercise protocols, as determined by blood lactate levels (6363. Kondo H, Fujino H, Murakami S, Tanaka M, Kanazashi M, Nagatomo F, et al. Low-intensity running exercise enhances the capillary volume and pro-angiogenic factors in the soleus muscle of type 2 diabetic rats. Muscle Nerve 2015; 51: 391-399, doi: 10.1002/mus.24316.
https://doi.org/10.1002/mus.24316...
,6666. Morifuji T, Murakami S, Fujita N, Kondo H, Fujino H. Exercise training prevents decrease in luminal capillary diameter of skeletal muscles in rats with type 2 diabetes. ScientificWorldJournal 2012; 2012: 645891, doi: 10.1100/2012/645891.
https://doi.org/10.1100/2012/645891...
,8080. Tsutsumi E, Murata Y, Sakamoto M, Horikawa E. Effects of exercise on the nephron of Goto-Kakizaki rats: morphological, and advanced glycation end-products and inducible nitric oxide synthase immunohistochemical analyses. J Diabetes Complications 2015; 29: 472-478, doi: 10.1016/j.jdiacomp.2015.03.002.
https://doi.org/10.1016/j.jdiacomp.2015....
). The protocol used by Tsutsumi et al. (8080. Tsutsumi E, Murata Y, Sakamoto M, Horikawa E. Effects of exercise on the nephron of Goto-Kakizaki rats: morphological, and advanced glycation end-products and inducible nitric oxide synthase immunohistochemical analyses. J Diabetes Complications 2015; 29: 472-478, doi: 10.1016/j.jdiacomp.2015.03.002.
https://doi.org/10.1016/j.jdiacomp.2015....
) demonstrated renoprotective action through the enhanced kidney-mediated AGE uptake. Additionally, Morifuji et al. (6666. Morifuji T, Murakami S, Fujita N, Kondo H, Fujino H. Exercise training prevents decrease in luminal capillary diameter of skeletal muscles in rats with type 2 diabetes. ScientificWorldJournal 2012; 2012: 645891, doi: 10.1100/2012/645891.
https://doi.org/10.1100/2012/645891...
) and Kondo et al. (6363. Kondo H, Fujino H, Murakami S, Tanaka M, Kanazashi M, Nagatomo F, et al. Low-intensity running exercise enhances the capillary volume and pro-angiogenic factors in the soleus muscle of type 2 diabetic rats. Muscle Nerve 2015; 51: 391-399, doi: 10.1002/mus.24316.
https://doi.org/10.1002/mus.24316...
) reported that low-intensity exercise led to augmented SDH activity and stimulated pro-angiogenic factors [e.g., vascular endothelial growth factor (VEGF) and fetal liver kinase 1 (FLK-1)], consequently preventing microangiopathy and improving capillary communication. However, the protocols did not reduce blood glucose levels.

Macia et al. (6161. Macia M, Pecchi E, Desrois M, Lan C, Vilmen C, Portha B, et al. Exercise training impacts exercise tolerance and bioenergetics in gastrocnemius muscle of non-obese type-2 diabetic Goto-Kakizaki rat in vivo. Biochimie 2018; 148: 36-45, doi: 10.1016/j.biochi.2018.02.014.
https://doi.org/10.1016/j.biochi.2018.02...
) described a reduction in plasma insulin levels promoted by low-intensity exercise. Along with this finding, increased citrate synthase activity, improved microcirculation, and up-regulated lactate and glucose metabolism-related protein expression (enzymes and transporters) were observed in the skeletal muscle.

Blood flow restriction combined with low-intensity electrical stimulation to induce muscle atrophy in GK rats prevented muscle mass loss, reduced AGE production and down-regulated the expression of receptor for AGE (RAGE), with no significant changes in blood glucose levels (7272. Tanaka M, Morifuji T, Yoshikawa M, Nakanishi R, Fujino H. Effects of combined treatment with blood flow restriction and low-intensity electrical stimulation on diabetes mellitus-associated muscle atrophy in rats. J Diabetes 2019; 11: 326-334, doi: 10.1111/1753-0407.12857.
https://doi.org/10.1111/1753-0407.12857...
).

Using voluntary activity (i.e., low-intensity), Nakamoto et al. (6565. Nakamoto I, Ishihara A. Effects of voluntary running exercise on skeletal muscle properties in nonobese rats with type 2 diabetes. Physiol Res 2020; 69: 73-84, doi: 10.33549/physiolres.934178.
https://doi.org/10.33549/physiolres.9341...
) and Kupai et al. (6464. Kupai K, Szabó R, Veszelka M, Awar AA, Török S, Csonka A, et al. Consequences of exercising on ischemia-reperfusion injury in type 2 diabetic Goto-Kakizaki rat hearts: role of the HO/NOS system. Diabetol Metab Syndr 2015; 7: 85, doi: 10.1186/s13098-015-0080-x.
https://doi.org/10.1186/s13098-015-0080-...
) reported significant SDH activity in the soleus muscle and, unlike previous articles, lower blood glucose and HbA1c levels in exercised GK rats compared to rats not subjected to exercise. The total distance covered in the voluntary exercise could account for these discrepancies.

Kim et al. (8787. Kim MS, Goo JS, Kim JE, Nam SH, Choi SI, Lee HR, et al. Overexpression of insulin degrading enzyme could greatly contribute to insulin down-regulation induced by short-term swimming exercise. Lab Anim Res 2011; 27: 29-36, doi: 10.5625/lar.2011.27.1.29.
https://doi.org/10.5625/lar.2011.27.1.29...
) used a swimming protocol to investigate the effects of physical exercise on GK rats but did not mention the exercise intensity level. The authors reported improved peripheral glucose utilization and insulin sensitivity. Tufescu et al. (4848. Tufescu A, Kanazawa M, Ishida A, Lu H, Sasaki Y, Ootaka T, et al. Combination of exercise and losartan enhances renoprotective and peripheral effects in spontaneously type 2 diabetes mellitus rats with nephropathy. J Hypertens 2008; 26: 312-321, doi: 10.1097/HJH.0b013e3282f2450b.
https://doi.org/10.1097/HJH.0b013e3282f2...
) demonstrated that exercise corresponding to 75% of the VO2 max (i.e., moderate-intensity) did not affect blood glucose levels but had a renoprotective effect that was enhanced by losartan.

Qi et al. (4949. Qi Z, He J, Zhang Y, Shao Y, Ding S. Exercise training attenuates oxidative stress and decreases p53 protein content in skeletal muscle of type 2 diabetic Goto-Kakizaki rats. Free Radic Biol Med 2011; 50: 794-800, doi: 10.1016/j.freeradbiomed.2010.12.022.
https://doi.org/10.1016/j.freeradbiomed....
) and Raza et al. (2828. Raza H, John A, Shafarin J, Howarth FC. Exercise-induced alterations in pancreatic oxidative stress and mitochondrial function in type 2 diabetic Goto-Kakizaki rats. Physiol Rep 2016; 4: e12751, doi: 10.14814/phy2.12751.
https://doi.org/10.14814/phy2.12751...
) reported that moderate-intensity exercise contributes to resolving oxidative stress and improving mitochondrial function, as determined by cyclooxygenase 2 (COXII), glutathione reductase (GSH), p53 (a tumor-suppressor protein), TIGAR (p53 inducible gene), ATPase6, cytochrome B (CytB), and nuclear factor-kappaB (NF-κB) levels. These results suggested that moderate-intensity exercise improves energy metabolism and mitochondrial respiratory function, induces antioxidant responses, and restores insulin sensitivity.

Many of the processes involving eNOS are linked to mitochondrial biogenesis, which, in the case of diabetes, can lead to improved metabolic function in the face of moderate exercise intervention. Some of these points are related to basal mitochondrial activity maintenance, redox balance regulation, and vascular restoration, mediated by glucagon-like peptide-1 (GLP-1) through cAMP, which improves myocardial contractility via Akt. The eNOS activity is increased by enhancing the bioavailability of tetrahydrobiopterin (BH4), a cofactor for NO synthesis (5959. Grijalva J, Hicks S, Zhao X, Medikayala S, Kaminski PM, Wolin MS, et al. Exercise training enhanced myocardial endothelial nitric oxide synthase (eNOS) function in diabetic Goto-Kakizaki (GK) rats. Cardiovasc Diabetol 2008; 7: 34, doi: 10.1186/1475-2840-7-34.
https://doi.org/10.1186/1475-2840-7-34...
,6060. Keller AC, Knaub LA, Miller MW, Birdsey N, Klemm DJ, Reusch JE. Saxagliptin restores vascular mitochondrial exercise response in the Goto-Kakizaki rat. J Cardiovasc Pharmacol 2015; 65: 137-147, doi: 10.1097/FJC.0000000000000170.
https://doi.org/10.1097/FJC.000000000000...
).

Of all the articles selected for this narrative review, around 27% mentioned the use of low or moderate intensity exercise, while the remaining studies (∼73%) did not mention the exercise intensity in the protocols used. Because direct markers of exercise training were not assessed in these studies, including VO2 max, lactate threshold, and enzyme activities, it is difficult to accurately define the exercise intensity and control loads used during training sessions in GK rat models (Table 1). Thus, we tried to address and discuss the main relevant results and differences from these previous studies, which is fundamental to guide and design further strategies to corroborate or refute the observed findings.

Beneficial effects of exercise in GK rats of different ages

A summary of the effects of physical exercise on GK rats of different ages is presented in Figure 4. It shows that a 3-6-week exercise program with young 6-12-week-old GK rats prevented microangiopathy, counteracting the capillary regression in skeletal and cardiac muscle and increased SDH and PGC-1α expression. Voluntary activity or low-intensity exercise attenuates infarct size by stimulating heme-oxygenase (HO) activity and effectively delaying the onset and progression of TDM2 in GK rats (6363. Kondo H, Fujino H, Murakami S, Tanaka M, Kanazashi M, Nagatomo F, et al. Low-intensity running exercise enhances the capillary volume and pro-angiogenic factors in the soleus muscle of type 2 diabetic rats. Muscle Nerve 2015; 51: 391-399, doi: 10.1002/mus.24316.
https://doi.org/10.1002/mus.24316...

64. Kupai K, Szabó R, Veszelka M, Awar AA, Török S, Csonka A, et al. Consequences of exercising on ischemia-reperfusion injury in type 2 diabetic Goto-Kakizaki rat hearts: role of the HO/NOS system. Diabetol Metab Syndr 2015; 7: 85, doi: 10.1186/s13098-015-0080-x.
https://doi.org/10.1186/s13098-015-0080-...

65. Nakamoto I, Ishihara A. Effects of voluntary running exercise on skeletal muscle properties in nonobese rats with type 2 diabetes. Physiol Res 2020; 69: 73-84, doi: 10.33549/physiolres.934178.
https://doi.org/10.33549/physiolres.9341...
-6666. Morifuji T, Murakami S, Fujita N, Kondo H, Fujino H. Exercise training prevents decrease in luminal capillary diameter of skeletal muscles in rats with type 2 diabetes. ScientificWorldJournal 2012; 2012: 645891, doi: 10.1100/2012/645891.
https://doi.org/10.1100/2012/645891...
).

Figure 4
Summary of the physical exercise effects in Goto-Kakizaki (GK) rats of different ages.

Similarly, training programs or physical muscle stimulation for 1, 2, 8, 9, 12, and 15 weeks at low- or moderate-intensity increased eNOS and PGC-1α expression and NO production, consequently eliciting positive kidney function effects. Exercise programs have also been shown to inhibit diabetes-associated muscle atrophy and enhance muscle protein synthesis in 6-30-week-old GK rats (4848. Tufescu A, Kanazawa M, Ishida A, Lu H, Sasaki Y, Ootaka T, et al. Combination of exercise and losartan enhances renoprotective and peripheral effects in spontaneously type 2 diabetes mellitus rats with nephropathy. J Hypertens 2008; 26: 312-321, doi: 10.1097/HJH.0b013e3282f2450b.
https://doi.org/10.1097/HJH.0b013e3282f2...
,4949. Qi Z, He J, Zhang Y, Shao Y, Ding S. Exercise training attenuates oxidative stress and decreases p53 protein content in skeletal muscle of type 2 diabetic Goto-Kakizaki rats. Free Radic Biol Med 2011; 50: 794-800, doi: 10.1016/j.freeradbiomed.2010.12.022.
https://doi.org/10.1016/j.freeradbiomed....
,5959. Grijalva J, Hicks S, Zhao X, Medikayala S, Kaminski PM, Wolin MS, et al. Exercise training enhanced myocardial endothelial nitric oxide synthase (eNOS) function in diabetic Goto-Kakizaki (GK) rats. Cardiovasc Diabetol 2008; 7: 34, doi: 10.1186/1475-2840-7-34.
https://doi.org/10.1186/1475-2840-7-34...
,6060. Keller AC, Knaub LA, Miller MW, Birdsey N, Klemm DJ, Reusch JE. Saxagliptin restores vascular mitochondrial exercise response in the Goto-Kakizaki rat. J Cardiovasc Pharmacol 2015; 65: 137-147, doi: 10.1097/FJC.0000000000000170.
https://doi.org/10.1097/FJC.000000000000...
,7272. Tanaka M, Morifuji T, Yoshikawa M, Nakanishi R, Fujino H. Effects of combined treatment with blood flow restriction and low-intensity electrical stimulation on diabetes mellitus-associated muscle atrophy in rats. J Diabetes 2019; 11: 326-334, doi: 10.1111/1753-0407.12857.
https://doi.org/10.1111/1753-0407.12857...
,8080. Tsutsumi E, Murata Y, Sakamoto M, Horikawa E. Effects of exercise on the nephron of Goto-Kakizaki rats: morphological, and advanced glycation end-products and inducible nitric oxide synthase immunohistochemical analyses. J Diabetes Complications 2015; 29: 472-478, doi: 10.1016/j.jdiacomp.2015.03.002.
https://doi.org/10.1016/j.jdiacomp.2015....
).

The effect of moderate-intensity exercise training for 4, 6, and 8 weeks was investigated in aged (>30 weeks of age) GK rats. The authors reported increased muscle and liver glycogen content, improved glucose homeostasis, oxidative metabolism, and IR, and up-regulated AMPK, PGC-1α, and GLUT-4 expression (2828. Raza H, John A, Shafarin J, Howarth FC. Exercise-induced alterations in pancreatic oxidative stress and mitochondrial function in type 2 diabetic Goto-Kakizaki rats. Physiol Rep 2016; 4: e12751, doi: 10.14814/phy2.12751.
https://doi.org/10.14814/phy2.12751...
,5252. Kim SS, Koo JH, Kwon IS, Oh YS, Lee SJ, Kim EJ, et al. Exercise training and selenium or a combined treatment ameliorates aberrant expression of glucose and lactate metabolic proteins in skeletal muscle in a rodent model of diabetes. Nutr Res Pract 2011; 5: 205-213, doi: 10.4162/nrp.2011.5.3.205.
https://doi.org/10.4162/nrp.2011.5.3.205...
,6161. Macia M, Pecchi E, Desrois M, Lan C, Vilmen C, Portha B, et al. Exercise training impacts exercise tolerance and bioenergetics in gastrocnemius muscle of non-obese type-2 diabetic Goto-Kakizaki rat in vivo. Biochimie 2018; 148: 36-45, doi: 10.1016/j.biochi.2018.02.014.
https://doi.org/10.1016/j.biochi.2018.02...
,6262. Salem KA, Qureshi MA, Sydorenko V, Parekh K, Jayaprakash P, Iqbal T, et al. Effects of exercise training on excitation-contraction coupling and related mRNA expression in hearts of Goto-Kakizaki type 2 diabetic rats. Mol Cell Biochem 2013; 380: 83-96, doi: 10.1007/s11010-013-1662-2.
https://doi.org/10.1007/s11010-013-1662-...
). The authors also observed improvements in ventricular myocyte Ca2+ handling despite variable muscle protein expression in GK rats. Exercise improved mitochondrial respiratory function and energy expenditure, induced an antioxidant response mechanism, reduced plasma insulin levels, and increased ATP production in exercising muscle. Glucose and lactate metabolism-regulating proteins were also up-regulated in the muscle and improved the recovery time of well-trained GK rats (2828. Raza H, John A, Shafarin J, Howarth FC. Exercise-induced alterations in pancreatic oxidative stress and mitochondrial function in type 2 diabetic Goto-Kakizaki rats. Physiol Rep 2016; 4: e12751, doi: 10.14814/phy2.12751.
https://doi.org/10.14814/phy2.12751...
,5252. Kim SS, Koo JH, Kwon IS, Oh YS, Lee SJ, Kim EJ, et al. Exercise training and selenium or a combined treatment ameliorates aberrant expression of glucose and lactate metabolic proteins in skeletal muscle in a rodent model of diabetes. Nutr Res Pract 2011; 5: 205-213, doi: 10.4162/nrp.2011.5.3.205.
https://doi.org/10.4162/nrp.2011.5.3.205...
,6161. Macia M, Pecchi E, Desrois M, Lan C, Vilmen C, Portha B, et al. Exercise training impacts exercise tolerance and bioenergetics in gastrocnemius muscle of non-obese type-2 diabetic Goto-Kakizaki rat in vivo. Biochimie 2018; 148: 36-45, doi: 10.1016/j.biochi.2018.02.014.
https://doi.org/10.1016/j.biochi.2018.02...
,6262. Salem KA, Qureshi MA, Sydorenko V, Parekh K, Jayaprakash P, Iqbal T, et al. Effects of exercise training on excitation-contraction coupling and related mRNA expression in hearts of Goto-Kakizaki type 2 diabetic rats. Mol Cell Biochem 2013; 380: 83-96, doi: 10.1007/s11010-013-1662-2.
https://doi.org/10.1007/s11010-013-1662-...
,8787. Kim MS, Goo JS, Kim JE, Nam SH, Choi SI, Lee HR, et al. Overexpression of insulin degrading enzyme could greatly contribute to insulin down-regulation induced by short-term swimming exercise. Lab Anim Res 2011; 27: 29-36, doi: 10.5625/lar.2011.27.1.29.
https://doi.org/10.5625/lar.2011.27.1.29...
,8888. Sinon CG, Ottensmeyer A, Slone AN, Li DC, Allen RS, Pardue MT, et al. Prehabilitative exercise hastens recovery from isoflurane in diabetic and non-diabetic rats. Neurosci Lett 2021; 751: 135808, doi: 10.1016/j.neulet.2021.135808.
https://doi.org/10.1016/j.neulet.2021.13...
).

It is important to highlight that muscle function changes during life; the natural tendency during the aging process is the reduction in muscle protein balance, resulting in the reduction in muscle mass (8989. Clemens Z, Sivakumar S, Pius A, Sahu A, Shinde S, Mamiya H, et al. The biphasic and age-dependent impact of klotho on hallmarks of aging and skeletal muscle function. Elife 2021; 10: e61138, doi: 10.7554/eLife.61138.
https://doi.org/10.7554/eLife.61138...
). Some strategies have been proposed to reduce or prevent muscle mass loss during aging, including nutritional and physical exercise programs, especially in people with comorbidities, such as obesity, type 2 diabetes mellitus, cardiovascular diseases, metabolic syndrome, among others, because these comorbidities exacerbate the process of muscle mass loss (33. Kuwabara WMT, Panveloski-Costa AC, Yokota CNF, Pereira JNB, Filho JM, Torres RP, et al. Comparison of Goto-Kakizaki rats and high fat diet-induced obese rats: are they reliable models to study Type 2 Diabetes mellitus? PLoS One 2017; 12: e0189622, doi: 10.1371/journal.pone.0189622.
https://doi.org/10.1371/journal.pone.018...
,6565. Nakamoto I, Ishihara A. Effects of voluntary running exercise on skeletal muscle properties in nonobese rats with type 2 diabetes. Physiol Res 2020; 69: 73-84, doi: 10.33549/physiolres.934178.
https://doi.org/10.33549/physiolres.9341...
,6767. Ventura-Clapier R. Exercise training, energy metabolism, and heart failure. Appl Physiol Nutr Metab 2009; 34: 336-339, doi: 10.1139/H09-013.
https://doi.org/10.1139/H09-013...
).

In this review, it was observed that physical activity and exercise training programs are potential strategies to generate important metabolic and physiological adaptations, leading to the improvement of several disturbances associated with type 2 diabetes mellitus in GK rats of all ages. Further studies are required to elucidate the mechanisms involved in these processes, as well as the long-term effect of physical exercise in preventing muscle mass loss during aging.

Concluding Remarks

The exercise training protocols using GK rats cited in this review had beneficial effects on several T2DM features. Similar to obesity-related T2DM, these results indicated that moderate-intensity exercise training can attenuate or prevent complications in non-obese T2DM individuals. Furthermore, similar responses to exercise training were reported in the muscle, liver, adipose tissue, pancreas, and blood of obese and non-obese T2DM rats (Figure 5).

Figure 5
The effects of physical exercise on different tissues and organs of obese and non-obese (GK: Goto-Kakizaki) type 2 diabetes mellitus rats.

As discussed in this review, the diabetic characteristics and the protocols used to examine the regression of the T2DM condition confirmed that non-invasive treatments, such as physical exercise, promoted improvements in the physiometabolic response in GK and Wistar rats. However, it remains unclear if there is real health improvement or if physical exercise is an effective treatment. Further studies investigating how exercise intensity affects the responses and adaptation in non-obese T2DM animals are necessary. Filling in these knowledge gaps is essential for understanding the effects of exercise intensity (i.e., low, moderate, or high) and modality in different animal strains. Furthermore, cognitive and functional abilities need to be evaluated following exercise training (9090. Kirwan JP, Sacks J, Nieuwoudt S. The essential role of exercise in the management of type 2 diabetes. Cleve Clin J Med 2017; 84: S15-S21, doi: 10.3949/ccjm.84.s1.03.
https://doi.org/10.3949/ccjm.84.s1.03...
). In conclusion, proper interventions focusing on a healthy lifestyle, including appropriate exercise protocols, are necessary to preserve the quality of life of both obese and non-obese T2DM patients.

Acknowledgments

The work of our group on GK rats is supported by research funding from the National Council for Scientific and Technological Development (CNPq), Coordination for the Improvement of Higher Education Personnel (CAPES, 88881.068515/201401), and São Paulo State Research Foundation (FAPESP, 2018/09868-7, 2018/07283-1, and 2019/25892-8). B.S.M. Galán and R. Manoel are recipients of FAPESP scholarships (2020/06641-1 and 2020/06642-8), R. Curi and R. Gorjão are recipients of CNPq scholarships (308955/2020-0 and 310711/2020-7), and L.E. Rodrigues is the recipient of a CAPES scholarship (8888.7598043/2021-00).

References

  • 1
    Ioannidis I. The road from obesity to type 2 diabetes. Angiology 2008; 59: 39S-43S, doi: 10.1177/0003319708318583.
    » https://doi.org/10.1177/0003319708318583
  • 2
    Mohammad S, Aziz R, Al Mahri S, Malik SS, Haji E, Khan AH, et al. Obesity and COVID-19: what makes obese host so vulnerable? Immun Ageing 2021; 18: 1, doi: 10.1186/s12979-020-00212-x.
    » https://doi.org/10.1186/s12979-020-00212-x
  • 3
    Kuwabara WMT, Panveloski-Costa AC, Yokota CNF, Pereira JNB, Filho JM, Torres RP, et al. Comparison of Goto-Kakizaki rats and high fat diet-induced obese rats: are they reliable models to study Type 2 Diabetes mellitus? PLoS One 2017; 12: e0189622, doi: 10.1371/journal.pone.0189622.
    » https://doi.org/10.1371/journal.pone.0189622
  • 4
    Kuwabara WMT, Yokota CNF, Curi R, Alba-Loureiro TC. Obesity and type 2 Diabetes mellitus induce lipopolysaccharide tolerance in rat neutrophils. Sci Rep 2018; 8: 17534, doi: 10.1038/s41598-018-35809-2.
    » https://doi.org/10.1038/s41598-018-35809-2
  • 5
    WHO (World Health Organization). Classification of Diabetes mellitus. World Health Organization; 2019. Report No.: 978-92-4-151570-2.
  • 6
    Barazzoni R, Gortan Cappellari G, Ragni M, Nisoli E. Insulin resistance in obesity: an overview of fundamental alterations. Eat Weight Disord 2018; 23: 149-157, doi: 10.1007/s40519-018-0481-6.
    » https://doi.org/10.1007/s40519-018-0481-6
  • 7
    Kuang J, Chen L, Tang Q, Zhang J, Li Y, He J. The role of Sirt6 in obesity and diabetes. Front Physiol 2018; 9: 135, doi: 10.3389/fphys.2018.00135.
    » https://doi.org/10.3389/fphys.2018.00135
  • 8
    Kyohara M, Shirakawa J, Okuyama T, Togashi Y, Inoue R, Li J, et al. Soluble EGFR, a hepatokine, and adipsin, an adipokine, are biomarkers correlated with distinct aspects of insulin resistance in type 2 diabetes subjects. Diabetol Metab Syndr 2020; 12: 83, doi: 10.1186/s13098-020-00591-7.
    » https://doi.org/10.1186/s13098-020-00591-7
  • 9
    Wang JS, Lee WJ, Lee IT, Lin SY, Lee WL, Liang KW, et al. Association between serum adipsin levels and insulin resistance in subjects with various degrees of glucose intolerance. J Endocr Soc 2019; 3: 403-410, doi: 10.1210/js.2018-00359.
    » https://doi.org/10.1210/js.2018-00359
  • 10
    Wernstedt I, Olsson B, Jernås M, Paglialunga S, Carlsson LM, Smith U, et al. Increased levels of acylation-stimulating protein in interleukin-6-deficient (IL-6(-/-)) mice. Endocrinology 2006; 147: 2690-2695, doi: 10.1210/en.2005-1133.
    » https://doi.org/10.1210/en.2005-1133
  • 11
    Achari AE, Jain SK. Adiponectin, a therapeutic target for obesity, diabetes, and endothelial dysfunction. Int J Mol Sci 2017; 18: 1321, doi: 10.3390/ijms18061321.
    » https://doi.org/10.3390/ijms18061321
  • 12
    Furman D, Campisi J, Verdin E, Carrera-Bastos P, Targ S, Franceschi C, et al. Chronic inflammation in the etiology of disease across the life span. Nat Med 2019; 25: 1822-1832, doi: 10.1038/s41591-019-0675-0.
    » https://doi.org/10.1038/s41591-019-0675-0
  • 13
    Ramachandran A, Snehalatha C, Ma RC. Diabetes in South-East Asia: an update. Diabetes Res Clin Pract 2014; 103: 231-237, doi: 10.1016/j.diabres.2013.11.011.
    » https://doi.org/10.1016/j.diabres.2013.11.011
  • 14
    Al-Goblan AS, Al-Alfi MA, Khan MZ. Mechanism linking diabetes mellitus and obesity. Diabetes Metab Syndr Obes 2014; 7: 587-591, doi: 10.2147/DMSO.S67400.
    » https://doi.org/10.2147/DMSO.S67400
  • 15
    Goto Y, Kakizaki M, Masaki N. Production of spontaneous diabetic rats by repetition of selective breeding. Tohoku J Exp Med 1976; 119: 85-90, doi: 10.1620/tjem.119.85.
    » https://doi.org/10.1620/tjem.119.85
  • 16
    Anoop S, Misra A, Bhatt SP, Gulati S, Mahajan H. High fasting C-peptide levels and insulin resistance in non-lean & non-obese (BMI >19 to <25kg/m. Diabetes Metab Syndr 2019; 13: 708-715, doi: 10.1016/j.dsx.2018.11.041.
    » https://doi.org/10.1016/j.dsx.2018.11.041
  • 17
    Coleman NJ, Miernik J, Philipson L, Fogelfeld L. Lean versus obese diabetes mellitus patients in the United States minority population. J Diabetes Complications 2014; 28: 500-505, doi: 10.1016/j.jdiacomp.2013.11.010.
    » https://doi.org/10.1016/j.jdiacomp.2013.11.010
  • 18
    Bautista FP, Jasul Jr G, Dampil OA. Insulin resistance and β-cell function of lean versus overweight or obese Filipino patients with newly diagnosed type 2 Diabetes mellitus. J ASEAN Fed Endocr Soc 2019; 34: 164-170, doi: 10.15605/jafes.034.02.07.
    » https://doi.org/10.15605/jafes.034.02.07
  • 19
    Goto A, Noda M, Goto M, Yasuda K, Mizoue T, Yamaji T, et al. Predictive performance of a genetic risk score using 11 susceptibility alleles for the incidence of type 2 diabetes in a general Japanese population: a nested case-control study. Diabet Med 2018; 35: 602-611, doi: 10.1111/dme.13602.
    » https://doi.org/10.1111/dme.13602
  • 20
    Galli J, Li LS, Glaser A, Ostenson CG, Jiao H, Fakhrai-Rad H, et al. Genetic analysis of non-insulin dependent diabetes mellitus in the GK rat. Nat Genet 1996; 12: 31-37, doi: 10.1038/ng0196-31.
    » https://doi.org/10.1038/ng0196-31
  • 21
    Hawley JA, Lessard SJ. Exercise training-induced improvements in insulin action. Acta Physiol (Oxf) 2008; 192: 127-135, doi: 10.1111/j.1748-1716.2007.01783.x.
    » https://doi.org/10.1111/j.1748-1716.2007.01783.x
  • 22
    Zhao RR, O'Sullivan AJ, Singh MAF. Exercise or physical activity and cognitive function in adults with type 2 diabetes, insulin resistance or impaired glucose tolerance: a systematic review. Eur Rev Aging Phys Act 2018; 15: 1, doi: 10.1186/s11556-018-0190-1.
    » https://doi.org/10.1186/s11556-018-0190-1
  • 23
    Ivy JL, Zderic TW, Fogt DL. Prevention and treatment of non-insulin-dependent Diabetes mellitus. Exerc Sport Sci Rev 1999; 27: 1-35, doi: 10.1249/00003677-199900270-00003.
    » https://doi.org/10.1249/00003677-199900270-00003
  • 24
    Dos Santos JM, Moreli ML, Tewari S, Benite-Ribeiro SA. The effect of exercise on skeletal muscle glucose uptake in type 2 diabetes: an epigenetic perspective. Metabolism 2015; 64: 1619-1628, doi: 10.1016/j.metabol.2015.09.013.
    » https://doi.org/10.1016/j.metabol.2015.09.013
  • 25
    de Matos MA, Duarte TC, Ottone VeO, Sampaio PF, Costa KB, de Oliveira MF, et al. The effect of insulin resistance and exercise on the percentage of CD16(+) monocyte subset in obese individuals. Cell Biochem Funct 2016; 34: 209-216, doi: 10.1002/cbf.3178.
    » https://doi.org/10.1002/cbf.3178
  • 26
    Mazur-Bialy AI, Pocheć E, Zarawski M. Anti-inflammatory properties of irisin, mediator of physical activity, are connected with TLR4/MyD88 signaling pathway activation. Int J Mol Sci 2017; 18: 701, doi: 10.3390/ijms18040701.
    » https://doi.org/10.3390/ijms18040701
  • 27
    Passos MEP, Borges L, Dos Santos-Oliveira LC, Alecrim-Zeza AL, Lobato TB, de Oliveira HH, et al. Recreational dance practice modulates lymphocyte profile and function in diabetic women. Int J Sports Med 2021; 42: 749-759, doi: 10.1055/a-1309-2037.
    » https://doi.org/10.1055/a-1309-2037
  • 28
    Raza H, John A, Shafarin J, Howarth FC. Exercise-induced alterations in pancreatic oxidative stress and mitochondrial function in type 2 diabetic Goto-Kakizaki rats. Physiol Rep 2016; 4: e12751, doi: 10.14814/phy2.12751.
    » https://doi.org/10.14814/phy2.12751
  • 29
    Bishop DJ, Botella J, Genders AJ, Lee MJC, Saner NJ, Kuang J, et al. High-intensity exercise and mitochondrial biogenesis: current controversies and future research directions. Physiology (Bethesda) 2019; 34: 56-70, doi: 10.1152/physiol.00038.2018.
    » https://doi.org/10.1152/physiol.00038.2018
  • 30
    Gollisch KS, Brandauer J, Jessen N, Toyoda T, Nayer A, Hirshman MF, et al. Effects of exercise training on subcutaneous and visceral adipose tissue in normal- and high-fat diet-fed rats. Am J Physiol Endocrinol Metab 2009; 297: E495-E504, doi: 10.1152/ajpendo.90424.2008.
    » https://doi.org/10.1152/ajpendo.90424.2008
  • 31
    McFarlin BK, Flynn MG, Campbell WW, Craig BA, Robinson JP, Stewart LK, et al. Physical activity status, but not age, influences inflammatory biomarkers and toll-like receptor 4. J Gerontol A Biol Sci Med Sci 2006; 61: 388-393, doi: 10.1093/gerona/61.4.388.
    » https://doi.org/10.1093/gerona/61.4.388
  • 32
    Galan BS, Carvalho FG, Santos PC, Gobbi RB, Kalva-Filho CA, Papoti M, et al. Effects of taurine on markers of muscle damage, inflammatory response and physical performance in triathletes. J Sports Med Phys Fitness 2018; 58: 1318-1324, doi: 10.23736/S0022-4707.17.07497-7.
    » https://doi.org/10.23736/S0022-4707.17.07497-7
  • 33
    Reznick RM, Shulman GI. The role of AMP-activated protein kinase in mitochondrial biogenesis. J Physiol 2006; 574: 33-39, doi: 10.1113/jphysiol.2006.109512.
    » https://doi.org/10.1113/jphysiol.2006.109512
  • 34
    Imierska M, Kurianiuk A, Błachnio-Zabielska A. The influence of physical activity on the bioactive lipids metabolism in obesity-induced muscle insulin resistance. Biomolecules 2020; 10: 1665, doi: 10.3390/biom10121665.
    » https://doi.org/10.3390/biom10121665
  • 35
    Smith JAH, Collins M, Grobler LA, Magee CJ, Ojuka EO. Exercise and CaMK activation both increase the binding of MEF2A to the Glut4 promoter in skeletal muscle in vivo Am J Physiol Endocrinol Metab 2007; 292: E413-E420, doi: 10.1152/ajpendo.00142.2006.
    » https://doi.org/10.1152/ajpendo.00142.2006
  • 36
    Portha B. Programmed disorders of beta-cell development and function as one cause for type 2 diabetes? The GK rat paradigm. Diabetes Metab Res Rev 2005; 21: 495-504, doi: 10.1002/dmrr.566.
    » https://doi.org/10.1002/dmrr.566
  • 37
    Portha B, Giroix MH, Serradas P, Gangnerau MN, Movassat J, Rajas F, et al. Beta-cell function and viability in the spontaneously diabetic GK rat: information from the GK/Par colony. Diabetes 2001; 50: S89-S93, doi: 10.2337/diabetes.50.2007.S89.
    » https://doi.org/10.2337/diabetes.50.2007.S89
  • 38
    Panveloski-Costa AC, Kuwabara WMT, Munhoz AC, Lucena CF, Curi R, Carpinelli AR, et al. The insulin resistance is reversed by exogenous 3,5,3'triiodothyronine in type 2 diabetic Goto-Kakizaki rats by an inflammatory-independent pathway. Endocrine 2020; 68: 287-295, doi: 10.1007/s12020-020-02208-5.
    » https://doi.org/10.1007/s12020-020-02208-5
  • 39
    Pereira JNB, Murata GM, Sato FT, Marosti AR, Carvalho CRO, Curi R. Small intestine remodeling in male Goto-Kakizaki rats. Physiol Rep 2021; 9: e14755, doi: 10.14814/phy2.14755.
    » https://doi.org/10.14814/phy2.14755
  • 40
    Guest PC. Characterization of the Goto-Kakizaki (GK) rat model of type 2 diabetes. Methods Mol Biol 2019; 1916: 203-211, doi: 10.1007/978-1-4939-8994-2.
    » https://doi.org/10.1007/978-1-4939-8994-2
  • 41
    Nagatomo F, Fujino H, Kondo H, Gu N, Takeda I, Ishioka N, et al. PGC-1α mRNA level and oxidative capacity of the plantaris muscle in rats with metabolic syndrome, hypertension, and type 2 diabetes. Acta Histochem Cytochem 2011; 44: 73-80, doi: 10.1267/ahc.10041.
    » https://doi.org/10.1267/ahc.10041
  • 42
    Caria ACI, Nonaka CKV, Pereira CS, Soares MBP, Macambira SG, Souza BSF. Exercise training-induced changes in microRNAs: beneficial regulatory effects in hypertension, type 2 diabetes, and obesity. Int J Mol Sci 2018; 19: 3608, doi: 10.3390/ijms19113608.
    » https://doi.org/10.3390/ijms19113608
  • 43
    Xue B, Sukumaran S, Nie J, Jusko WJ, Dubois DC, Almon RR. Adipose tissue deficiency and chronic inflammation in diabetic Goto-Kakizaki rats. PLoS One 2011; 6: e17386, doi: 10.1371/journal.pone.0017386.
    » https://doi.org/10.1371/journal.pone.0017386
  • 44
    Haber EP, Procópio J, Carvalho CRO, Carpinelli AR, Newsholme P, Curi R. New insights into fatty acid modulation of pancreatic beta-cell function. Int Rev Cytol 2006; 248: 1-41, doi: 10.1016/S0074-7696(06)48001-3.
    » https://doi.org/10.1016/S0074-7696(06)48001-3
  • 45
    Newsholme P, Haber EP, Hirabara SM, Rebelato EL, Procopio J, Morgan D, et al. Diabetes associated cell stress and dysfunction: role of mitochondrial and non-mitochondrial ROS production and activity. J Physiol 2007; 583: 9-24, doi: 10.1113/jphysiol.2007.135871.
    » https://doi.org/10.1113/jphysiol.2007.135871
  • 46
    Tsuura Y, Ishida H, Hayashi S, Sakamoto K, Horie M, Seino Y. Nitric oxide opens ATP-sensitive K+ channels through suppression of phosphofructokinase activity and inhibits glucose-induced insulin release in pancreatic beta cells. J Gen Physiol 1994; 104: 1079-1098, doi: 10.1085/jgp.104.6.1079.
    » https://doi.org/10.1085/jgp.104.6.1079
  • 47
    Magkos F, Hjorth MF, Astrup A. Diet and exercise in the prevention and treatment of type 2 Diabetes mellitus. Nat Rev Endocrinol 2020; 16: 545-555, doi: 10.1038/s41574-020-0381-5.
    » https://doi.org/10.1038/s41574-020-0381-5
  • 48
    Tufescu A, Kanazawa M, Ishida A, Lu H, Sasaki Y, Ootaka T, et al. Combination of exercise and losartan enhances renoprotective and peripheral effects in spontaneously type 2 diabetes mellitus rats with nephropathy. J Hypertens 2008; 26: 312-321, doi: 10.1097/HJH.0b013e3282f2450b.
    » https://doi.org/10.1097/HJH.0b013e3282f2450b
  • 49
    Qi Z, He J, Zhang Y, Shao Y, Ding S. Exercise training attenuates oxidative stress and decreases p53 protein content in skeletal muscle of type 2 diabetic Goto-Kakizaki rats. Free Radic Biol Med 2011; 50: 794-800, doi: 10.1016/j.freeradbiomed.2010.12.022.
    » https://doi.org/10.1016/j.freeradbiomed.2010.12.022
  • 50
    Itahana Y, Itahana K. Emerging roles of p53 family members in glucose metabolism. Int J Mol Sci 2018; 19: 776, doi: 10.3390/ijms19030776.
    » https://doi.org/10.3390/ijms19030776
  • 51
    Olovnikov IA, Kravchenko JE, Chumakov PM. Homeostatic functions of the p53 tumor suppressor: regulation of energy metabolism and antioxidant defense. Semin Cancer Biol 2009; 19: 32-41, doi: 10.1016/j.semcancer.2008.11.005.
    » https://doi.org/10.1016/j.semcancer.2008.11.005
  • 52
    Kim SS, Koo JH, Kwon IS, Oh YS, Lee SJ, Kim EJ, et al. Exercise training and selenium or a combined treatment ameliorates aberrant expression of glucose and lactate metabolic proteins in skeletal muscle in a rodent model of diabetes. Nutr Res Pract 2011; 5: 205-213, doi: 10.4162/nrp.2011.5.3.205.
    » https://doi.org/10.4162/nrp.2011.5.3.205
  • 53
    Richter EA, Hargreaves M. Exercise, GLUT4, and skeletal muscle glucose uptake. Physiol Rev 2013; 93: 993-1017, doi: 10.1152/physrev.00038.2012.
    » https://doi.org/10.1152/physrev.00038.2012
  • 54
    Lira VA, Benton CR, Yan Z, Bonen A. PGC-1alpha regulation by exercise training and its influences on muscle function and insulin sensitivity. Am J Physiol Endocrinol Metab 2010; 299: E145-E161, doi: 10.1152/ajpendo.00755.2009.
    » https://doi.org/10.1152/ajpendo.00755.2009
  • 55
    Vieira R, Junqueira R, Gaspar R, Munoz V. Exercise ativates AMPK signaling: Impact on glucose uptake in the skeletal muscle in aging. J Rehab Ther 2020.
  • 56
    Pinto AP, da Rocha AL, Marafon BB, Rovina RL, Muãoz VR, da Silva LECM, et al. Impact of different physical exercises on the expression of autophagy markers in mice. Int J Mol Sci 2021; 22: 2635, doi: 10.3390/ijms22052635.
    » https://doi.org/10.3390/ijms22052635
  • 57
    Ding Q, Cecarini V, Keller JN. Interplay between protein synthesis and degradation in the CNS: physiological and pathological implications. Trends Neurosci 2007; 30: 31-36, doi: 10.1016/j.tins.2006.11.003.
    » https://doi.org/10.1016/j.tins.2006.11.003
  • 58
    Patti ME, Corvera S. The role of mitochondria in the pathogenesis of type 2 diabetes. Endocr Rev 2010; 31: 364-395, doi: 10.1210/er.2009-0027.
    » https://doi.org/10.1210/er.2009-0027
  • 59
    Grijalva J, Hicks S, Zhao X, Medikayala S, Kaminski PM, Wolin MS, et al. Exercise training enhanced myocardial endothelial nitric oxide synthase (eNOS) function in diabetic Goto-Kakizaki (GK) rats. Cardiovasc Diabetol 2008; 7: 34, doi: 10.1186/1475-2840-7-34.
    » https://doi.org/10.1186/1475-2840-7-34
  • 60
    Keller AC, Knaub LA, Miller MW, Birdsey N, Klemm DJ, Reusch JE. Saxagliptin restores vascular mitochondrial exercise response in the Goto-Kakizaki rat. J Cardiovasc Pharmacol 2015; 65: 137-147, doi: 10.1097/FJC.0000000000000170.
    » https://doi.org/10.1097/FJC.0000000000000170
  • 61
    Macia M, Pecchi E, Desrois M, Lan C, Vilmen C, Portha B, et al. Exercise training impacts exercise tolerance and bioenergetics in gastrocnemius muscle of non-obese type-2 diabetic Goto-Kakizaki rat in vivo Biochimie 2018; 148: 36-45, doi: 10.1016/j.biochi.2018.02.014.
    » https://doi.org/10.1016/j.biochi.2018.02.014
  • 62
    Salem KA, Qureshi MA, Sydorenko V, Parekh K, Jayaprakash P, Iqbal T, et al. Effects of exercise training on excitation-contraction coupling and related mRNA expression in hearts of Goto-Kakizaki type 2 diabetic rats. Mol Cell Biochem 2013; 380: 83-96, doi: 10.1007/s11010-013-1662-2.
    » https://doi.org/10.1007/s11010-013-1662-2
  • 63
    Kondo H, Fujino H, Murakami S, Tanaka M, Kanazashi M, Nagatomo F, et al. Low-intensity running exercise enhances the capillary volume and pro-angiogenic factors in the soleus muscle of type 2 diabetic rats. Muscle Nerve 2015; 51: 391-399, doi: 10.1002/mus.24316.
    » https://doi.org/10.1002/mus.24316
  • 64
    Kupai K, Szabó R, Veszelka M, Awar AA, Török S, Csonka A, et al. Consequences of exercising on ischemia-reperfusion injury in type 2 diabetic Goto-Kakizaki rat hearts: role of the HO/NOS system. Diabetol Metab Syndr 2015; 7: 85, doi: 10.1186/s13098-015-0080-x.
    » https://doi.org/10.1186/s13098-015-0080-x
  • 65
    Nakamoto I, Ishihara A. Effects of voluntary running exercise on skeletal muscle properties in nonobese rats with type 2 diabetes. Physiol Res 2020; 69: 73-84, doi: 10.33549/physiolres.934178.
    » https://doi.org/10.33549/physiolres.934178
  • 66
    Morifuji T, Murakami S, Fujita N, Kondo H, Fujino H. Exercise training prevents decrease in luminal capillary diameter of skeletal muscles in rats with type 2 diabetes. ScientificWorldJournal 2012; 2012: 645891, doi: 10.1100/2012/645891.
    » https://doi.org/10.1100/2012/645891
  • 67
    Ventura-Clapier R. Exercise training, energy metabolism, and heart failure. Appl Physiol Nutr Metab 2009; 34: 336-339, doi: 10.1139/H09-013.
    » https://doi.org/10.1139/H09-013
  • 68
    De Sousa E, Lechêne P, Fortin D, N'Guessan B, Belmadani S, Bigard X, et al. Cardiac and skeletal muscle energy metabolism in heart failure: beneficial effects of voluntary activity. Cardiovasc Res 2002; 56: 260-268, doi: 10.1016/S0008-6363(02)00540-0.
    » https://doi.org/10.1016/S0008-6363(02)00540-0
  • 69
    Rasheed Z, Akhtar N, Haqqi TM. Advanced glycation end products induce the expression of interleukin-6 and interleukin-8 by receptor for advanced glycation end product-mediated activation of mitogen-activated protein kinases and nuclear factor-κB in human osteoarthritis chondrocytes. Rheumatology (Oxford) 2011; 50: 838-851, doi: 10.1093/rheumatology/keq380.
    » https://doi.org/10.1093/rheumatology/keq380
  • 70
    Chiu CY, Yang RS, Sheu ML, Chan DC, Yang TH, Tsai KS, et al. Advanced glycation end-products induce skeletal muscle atrophy and dysfunction in diabetic mice via a RAGE-mediated, AMPK-down-regulated, Akt pathway. J Pathol 2016; 238: 470-482, doi: 10.1002/path.4674.
    » https://doi.org/10.1002/path.4674
  • 71
    Goldin A, Beckman JA, Schmidt AM, Creager MA. Advanced glycation end products: sparking the development of diabetic vascular injury. Circulation 2006; 114: 597-605, doi: 10.1161/CIRCULATIONAHA.106.621854.
    » https://doi.org/10.1161/CIRCULATIONAHA.106.621854
  • 72
    Tanaka M, Morifuji T, Yoshikawa M, Nakanishi R, Fujino H. Effects of combined treatment with blood flow restriction and low-intensity electrical stimulation on diabetes mellitus-associated muscle atrophy in rats. J Diabetes 2019; 11: 326-334, doi: 10.1111/1753-0407.12857.
    » https://doi.org/10.1111/1753-0407.12857
  • 73
    Awtry EH, Balady GJ. Exercise and the heart. Cardiology Secrets; 2010. p 311-315.
  • 74
    American College of Sports Medicine and American Diabetes Association joint position statement. Diabetes mellitus and exercise. Med Sci Sports Exerc 1997; 29: i-vi.
  • 75
    De Feo P. Is high-intensity exercise better than moderate-intensity exercise for weight loss? Nutr Metab Cardiovasc Dis 2013; 23: 1037-1042, doi: 10.1016/j.numecd.2013.06.002.
    » https://doi.org/10.1016/j.numecd.2013.06.002
  • 76
    Leandro CG, Levada AC, Hirabara SM, Manhães-de-Castro R, De-Castro CB, Curi R, et al. A program of moderate physical training for Wistar rats based on maximal oxygen consumption. J Strength Cond Res 2007; 21: 751-756, doi: 10.1519/R-20155.1.
    » https://doi.org/10.1519/R-20155.1
  • 77
    Boutcher YN, Boutcher SH. Exercise intensity and hypertension: what's new? J Hum Hypertens 2017; 31: 157-164, doi: 10.1038/jhh.2016.62.
    » https://doi.org/10.1038/jhh.2016.62
  • 78
    Gomes-Neto M, Durães AR, Reis HFCD, Neves VR, Martinez BP, Carvalho VO. High-intensity interval training versus moderate-intensity continuous training on exercise capacity and quality of life in patients with coronary artery disease: A systematic review and meta-analysis. Eur J Prev Cardiol 2017; 24: 1696-1707, doi: 10.1177/2047487317728370.
    » https://doi.org/10.1177/2047487317728370
  • 79
    Criswell D, Powers S, Dodd S, Lawler J, Edwards W, Renshler K, et al. High intensity training-induced changes in skeletal muscle antioxidant enzyme activity. Med Sci Sports Exerc 1993; 25: 1135-1140, doi: 10.1249/00005768-199310000-00009.
    » https://doi.org/10.1249/00005768-199310000-00009
  • 80
    Tsutsumi E, Murata Y, Sakamoto M, Horikawa E. Effects of exercise on the nephron of Goto-Kakizaki rats: morphological, and advanced glycation end-products and inducible nitric oxide synthase immunohistochemical analyses. J Diabetes Complications 2015; 29: 472-478, doi: 10.1016/j.jdiacomp.2015.03.002.
    » https://doi.org/10.1016/j.jdiacomp.2015.03.002
  • 81
    Soya H, Mukai A, Deocaris CC, Ohiwa N, Chang H, Nishijima T, et al. Threshold-like pattern of neuronal activation in the hypothalamus during treadmill running: establishment of a minimum running stress (MRS) rat model. Neurosci Res 2007; 58: 341-348, doi: 10.1016/j.neures.2007.04.004.
    » https://doi.org/10.1016/j.neures.2007.04.004
  • 82
    Bedford TG, Tipton CM, Wilson NC, Oppliger RA, Gisolfi CV. Maximum oxygen consumption of rats and its changes with various experimental procedures. J Appl Physiol Respir Environ Exerc Physiol 1979; 47: 1278-1283, doi: 10.1152/jappl.1979.47.6.1278.
    » https://doi.org/10.1152/jappl.1979.47.6.1278
  • 83
    Corcoran MP, Lamon-Fava S, Fielding RA. Skeletal muscle lipid deposition and insulin resistance: effect of dietary fatty acids and exercise. Am J Clin Nutr 2007; 85: 662-677, doi: 10.1093/ajcn/85.3.662.
    » https://doi.org/10.1093/ajcn/85.3.662
  • 84
    Schöler CM, Marques CV, da Silva GS, Heck TG, de Oliveira Junior LP, de Bittencourt Jr PIH. Modulation of rat monocyte/macrophage innate functions by increasing intensities of swimming exercise is associated with heat shock protein status. Mol Cell Biochem 2016; 421: 111-125, doi: 10.1007/s11010-016-2791-1.
    » https://doi.org/10.1007/s11010-016-2791-1
  • 85
    Brooks GA, White TP. Determination of metabolic and heart rate responses of rats to treadmill exercise. J Appl Physiol Respir Environ Exer Physiol 1978; 45: 1009-1015, doi: 10.1152/jappl.1978.45.6.1009.
    » https://doi.org/10.1152/jappl.1978.45.6.1009
  • 86
    Dudley GA, Abraham WM, Terjung RL. Influence of exercise intensity and duration on biochemical adaptations in skeletal muscle. J Appl Physiol Respir Environ Exerc Physiol 1982; 53: 844-850, doi: 10.1152/jappl.1982.53.4.844.
    » https://doi.org/10.1152/jappl.1982.53.4.844
  • 87
    Kim MS, Goo JS, Kim JE, Nam SH, Choi SI, Lee HR, et al. Overexpression of insulin degrading enzyme could greatly contribute to insulin down-regulation induced by short-term swimming exercise. Lab Anim Res 2011; 27: 29-36, doi: 10.5625/lar.2011.27.1.29.
    » https://doi.org/10.5625/lar.2011.27.1.29
  • 88
    Sinon CG, Ottensmeyer A, Slone AN, Li DC, Allen RS, Pardue MT, et al. Prehabilitative exercise hastens recovery from isoflurane in diabetic and non-diabetic rats. Neurosci Lett 2021; 751: 135808, doi: 10.1016/j.neulet.2021.135808.
    » https://doi.org/10.1016/j.neulet.2021.135808
  • 89
    Clemens Z, Sivakumar S, Pius A, Sahu A, Shinde S, Mamiya H, et al. The biphasic and age-dependent impact of klotho on hallmarks of aging and skeletal muscle function. Elife 2021; 10: e61138, doi: 10.7554/eLife.61138.
    » https://doi.org/10.7554/eLife.61138
  • 90
    Kirwan JP, Sacks J, Nieuwoudt S. The essential role of exercise in the management of type 2 diabetes. Cleve Clin J Med 2017; 84: S15-S21, doi: 10.3949/ccjm.84.s1.03.
    » https://doi.org/10.3949/ccjm.84.s1.03

Supplementary Material

Click to view [pdf].

Publication Dates

  • Publication in this collection
    27 May 2022
  • Date of issue
    2022

History

  • Received
    25 Sept 2021
  • Accepted
    9 Mar 2022
Associação Brasileira de Divulgação Científica Av. Bandeirantes, 3900, 14049-900 Ribeirão Preto SP Brazil, Tel. / Fax: +55 16 3315-9120 - Ribeirão Preto - SP - Brazil
E-mail: bjournal@terra.com.br