Submitted Successfully!
To reward your contribution, here is a gift for you: A free trial for our video production service.
Thank you for your contribution! You can also upload a video entry or images related to this topic.
Version Summary Created by Modification Content Size Created at Operation
1 -- 2777 2023-06-06 03:05:02 |
2 references update Meta information modification 2777 2023-06-06 11:24:55 |

Video Upload Options

Do you have a full video?

Confirm

Are you sure to Delete?
Cite
If you have any further questions, please contact Encyclopedia Editorial Office.
Heinle, J.W.; Dijoseph, K.; Sabag, A.; Oh, S.; Kimball, S.R.; Keating, S.; Stine, J.G. Exercise Is Medicine for Nonalcoholic Fatty Liver Disease. Encyclopedia. Available online: https://encyclopedia.pub/entry/45211 (accessed on 27 July 2024).
Heinle JW, Dijoseph K, Sabag A, Oh S, Kimball SR, Keating S, et al. Exercise Is Medicine for Nonalcoholic Fatty Liver Disease. Encyclopedia. Available at: https://encyclopedia.pub/entry/45211. Accessed July 27, 2024.
Heinle, James Westley, Kara Dijoseph, Angelo Sabag, Sechang Oh, Scot R. Kimball, Shelley Keating, Jonathan G. Stine. "Exercise Is Medicine for Nonalcoholic Fatty Liver Disease" Encyclopedia, https://encyclopedia.pub/entry/45211 (accessed July 27, 2024).
Heinle, J.W., Dijoseph, K., Sabag, A., Oh, S., Kimball, S.R., Keating, S., & Stine, J.G. (2023, June 06). Exercise Is Medicine for Nonalcoholic Fatty Liver Disease. In Encyclopedia. https://encyclopedia.pub/entry/45211
Heinle, James Westley, et al. "Exercise Is Medicine for Nonalcoholic Fatty Liver Disease." Encyclopedia. Web. 06 June, 2023.
Exercise Is Medicine for Nonalcoholic Fatty Liver Disease
Edit

Upwards of 30% of the world’s population has nonalcoholic fatty liver disease (NAFLD). There is no regulatory-agency-approved effective drug therapy or cure, and lifestyle modification with dietary change and increased physical activity remains crucial in the clinical management of all types of NAFLD, leads to many benefits within and outside the liver in patients with NAFLD. These benefits include an improvement in liver fat, histologic NASH activity, change in body composition, gain in physical fitness, reduction in markers of cardiovascular risk, improvement in health-related quality of life and possibly a reduction in oncologic risk. 

nonalcoholic fatty liver disease steatosis physical activity

1. Introduction

Upwards of 30% of the world’s population has nonalcoholic fatty liver disease (NAFLD) [1]. At this point in time, there is no regulatory-agency-approved effective drug therapy or cure, and lifestyle modification with dietary change and increased physical activity remains crucial in the clinical management of all types of NAFLD, including nonalcoholic steatohepatitis (NASH). Even when a regulatory-agency-approved drug therapy becomes widely available, lifestyle modification will always play a key role in the prevention and treatment of NAFLD and lessen the burden of associated extrahepatic disease from cardiovascular disease events and cancer. Following years of research, it is widely accepted that regular physical activity and, in particular, exercise training, which is a type of physical activity that is planned, structured, repetitive and with a specific goal in mind [2], leads to many benefits within and outside the liver in patients with NAFLD [3][4]. These benefits include an improvement in liver fat, histologic NASH activity, change in body composition, gain in physical fitness, reduction in markers of cardiovascular risk, improvement in health-related quality of life and possibly a reduction in oncologic risk [4]. Importantly, many of these improvements, including the reduction in magnetic resonance imaging (MRI)-measured liver fat, may occur without clinically significant body weight loss [5].

2. Exercise Training and Mechanistic Pathways Involved in Hepatic Steatosis

2.1. AMP-Activated Protein Kinase (AMPK)

AMPK is a fuel-sensing enzyme that is activated by energy stress [6] and is composed of a trimeric complex with a catalytic subunit (α) and two regulatory subunits (β and γ). AMPK also has a liver-specific role in hepatic de novo lipogenesis, fatty acid oxidation, glycogenolysis and gluconeogenesis. AMPK activity is abnormally low in patients with NAFLD, leading to excessive accumulation of liver fat [7]. For this reason, AMPK remains a drug target of interest, and in fact, the recent Phase 2a STAMP-NAFLD study enrolled 120 patients who were randomized to treatment with PXL770, a direct AMPK activator, or a placebo [8]. Unfortunately, this study did not meet its primary endpoint of statistically significant MRI-determined liver fat reduction; however, subgroup analysis limited only to patients with type 2 diabetes demonstrated a significant reduction in liver fat and corresponding metabolic parameters, including glycemic control.
Animal models of NAFLD demonstrate exercise changes the AMPK pathway, leading to less liver fat accumulation by reducing lipogenesis and increasing fatty acid oxidation [9][10][11][12][13]. Importantly, exercise-induced AMPK activation appears to be dose-related. As exercise intensity and duration increase, ATP usage increases to the point where it cannot be regenerated quickly enough, increasing the AMP/ADP:ATP ratio and activating AMPK [14]. Further, in order to generate additional ATP during exercise, glycogen, which is the main energy substrate used during exercise at higher intensities (typically greater than 70% of VO2max), dissociates from AMPK, leading to AMPK activation (glycogen-bound AMPK is inactive) [6][15][16]. Sustained moderate–vigorous intensity exercise seems to be required to deplete glycogen enough to activate AMPK [16][17][18][19]. While several small studies have demonstrated that exercise can favorably impact targets downstream of AMPK, including FGF-21 [20] and also ribosomal protein s6 [21], in patients with NASH, researchers await a definitive study showing that exercise can directly activate AMPK in this patient population.
There is a clear and consistent body of evidence to support the role of the AMPK pathway as a key pathway modulated by exercise training that appears to be related to both intensity and exercise volume across animal and human studies.

2.2. Fibroblast Growth Factor (FGF)-19 and -21

FGF is a complex family of peptide hormones that has a crucial implication on regulating energy homeostasis and metabolism [22]. Multiple isoforms of FGF are involved in the cascade, but FGF-19 and FGF-21 are closely related to fat metabolism [23] and are the signaling pathways for both hormones involved in NAFLD and NASH development and the focus of drug discovery [24][25]. Both the FALCON and BALANCED trials investigated the efficacy of FGF-21 analogues pegbelfermin and efruxifermin in patients with NASH [25][26][27].
FGF-19 is expressed in the ileum and is released in response to bile acid stimuli. FGF-19 is activated postprandially, regulating the transcription of hepatic protein and glycogen synthesis and inhibiting hepatic gluconeogenesis. FGF-19 appears to largely act locally on the hepatocytes, whereas FGF-21 tends to be expressed systemically and, of particular interest, in the skeletal muscle, adipose tissue and in the liver tissue [28][29]. Importantly, FGF-21 relies heavily on the coreceptor β-klotho [30]. If there is a lower expression of β-klotho, resistance to FGF-21 has been observed, resulting in impaired fatty acid oxidation [31]. In fact, NAFLD is felt to be an FGF-21-resistant state [32][33].
Because FGF-21 is widely and variably expressed in the human body, it has been challenging to identify the relationship between exercise and FGF-21 expression. Despite this, there is a robust and consistent body of evidence describing this relationship in pre-clinical animal models of NAFLD and in clinical studies involving patients who are inactive, overweight, obese or diabetic [34][35][36]. Importantly, the relationship between exercise and FGF-21 expression appears to exist across differing intensities, where both moderate and vigorous intensity exercise can change FGF-21 expression [37]. In terms of patients with NAFLD, a recent study by Takahashi et al. [38] found that after 12 weeks of resistance training in which participants performed push-ups and squats three times a week on non-consecutive days, serum FGF-21 level was significantly reduced, confirming the results seen with aerobic exercise training in animal models of NAFLD. Furthermore, a study of 24 patients with biopsy-proven NASH reported that 20 weeks of aerobic exercise training significantly reduced serum FGF-21 in parallel with gains in cardiorespiratory fitness [20]. Despite these differences in methodology, it is generally agreed upon that while acute exercise tends to increase plasma levels of FGF-21, perhaps due to increased production by skeletal muscle, chronic exercise training programs of four weeks or more duration lead to a reduction in serum FGF-21 level while simultaneously increasing expression of FGF receptors and β-klotho in not only liver tissue but also in adipose tissue and skeletal muscle [39].
Although closely related to FGF-21, exercise may play a different role in FGF-19 expression. There have been observed correlations between resistance training and upregulation of FGF-19, but further studies are needed to ensure no other external factors are contributing, such as fasting states. These potentially confounding factors may explain the conflicting results of FGF-19 downregulation following an acute one-hour bout of aerobic exercise [40] as well as multiple negative studies, which are limited by small sample sizes. 
To date, the scientific literature suggests that exercise training across different exercise intensities and volumes can activate FGF-21. 

2.3. Glucagon-like Peptide-1 (GLP-1)

The liver plays a central role in insulin metabolism and is impacted by multiple gut hormones. One such hormone is GLP-1, an incretin, which helps to regulate satiety and lipid metabolism in both the fasting and glucose-stimulated states [41]. GLP-1 has recently emerged at the forefront of drug development in NASH given the recent promising results of early-phase studies using both semaglutide [42] as well as liraglutide [43] and the fact that several GLP-1 receptor agonists are regulatory-agency-approved for the medical treatment of overweight and obesity as well as type two diabetes [44][45].
Exercise can impact serum levels of GLP-1, and in fact, exercise can increase GLP-1 levels in healthy individuals and in persons with obesity and suppress appetite [46][47]. In patients with NAFLD, Kullman et al. [48] measured the impact of a short-term, one-week high-intensity exercise program and found that while GLP-1 level remained unchanged, exercise instead reversed the GLP-1 resistant state of NAFLD by restoring the normal physiologic response of GLP-1 to glucose stimulation. This raises the question of the importance of increasing serum GLP-1 level versus ameliorating GLP-1 resistance, building on previous work showing short-term exercise programs to improve hepatic insulin extraction in patients with NAFLD [49]. To our knowledge, the effects of a long-term exercise training program on GLP-1 have not yet been investigated in patients with NAFLD or NASH.
While GLP-1 and other gut hormones present intriguing avenues of research, the existing scientific data prevent strong conclusions regarding the impact of exercise training programs on this therapeutic target.

2.4. Mitochondrial Function and Beta Oxidation

The liver plays a principal role in lipid metabolism as the primary site of de novo lipogenesis and fatty acid oxidation [50]. In fact, lipid-derived energy production in the liver occurs through the β-oxidation of fatty acids [50]. However, mitochondrial defects, which are related to both physical inactivity and obesity [51], reduce the oxidative capacity of the mitochondria, which results in incomplete β-oxidation and the accumulation of metabolic by-products, such as ceramides and diacylglycerides [52]. While intrahepatic triglycerides themselves do not cause hepatic insulin resistance, it is thought that the accumulation of the aforementioned by-products impairs insulin receptor signaling through various mechanisms and has been identified as critical in the pathogenesis of hepatic insulin resistance [53].
Regular exercise has been shown to improve mitochondrial oxidative capacity and increase mitochondrial content, which are related to increases in cardiorespiratory fitness [54][55]. In fact, cardiorespiratory fitness is inversely related to hepatic steatosis [56], and improvements in cardiorespiratory fitness are independently associated with improvements in steatosis [57][58]. As cardiorespiratory fitness has been shown to improve to a similar degree with both high-intensity interval training (HIIT) and more traditional moderate-intensity continuous training [59], this may, in part, explain why HIIT leads to similar improvements in hepatic steatosis to more moderate-intensity continuous exercise despite expending less energy [60].
Although most work in this space has been conducted in animal models, the available data from human studies support the role of improving cardiorespiratory fitness as a key therapeutic target in the management of NAFLD.

2.5. Mitochondrial Uncoupling Proteins (UCP)

Mitochondria are vital organelles that are at the forefront of cellular metabolism, especially in the liver, which is the primary metabolic organ in the human body. UCPs are a key component of mitochondrial metabolism and are mitochondrial inner-membrane proteins which mediate proton leak across the inner membrane through anion transport and uncouple substrate oxidation from ATP synthesis [61]. Five key mitochondrial UCPs have been discovered. UCP-1 is found largely in brown adipose tissue and plays a role in thermogenesis and energy expenditure; UCP-2, while fairly ubiquitous, is found in high concentrations in the liver and regulates insulin secretion from pancreatic β-cells as well as fatty acid metabolism; UCP-3 is expressed largely in brown adipose tissue and also skeletal muscle and influences fatty acid metabolism and insulin sensitivity [62][63][64][65]. UCP-4 and UCP-5 are found in the brain. The dysfunction of these transporters has been correlated with various metabolic disorders, such as obesity and diabetes [66][67], and also NAFLD [68][69]. Genetic variation in UCP polymorphisms is also important. In patients with type 2 diabetes, the CC genotype of the UCP-1 rs3811791 polymorphism blunts the response to regular physical activity in terms of insulin resistance and also lipid control, even at guideline-based amounts of 150 min/wk. of moderate-intensity activity [70]. Moreover, the INDOGENIC cohort study demonstrated that even in healthy individuals, the GG genotype for the UCP-2 G-866A polymorphism changed the physiologic response to energy intake, making these individuals more prone to weight gain and overeating over a two year follow-up period [71]. Consequently, targeting UCPs to enhance energy utilization remains an attractive treatment option in NASH. However, when medications with this mechanism of action have been used to induce body weight loss, they have been significantly limited by a strong side effect profile, and this target must be approached with caution [72]. A recent phase 2a trial for a novel mitochondrial uncoupler, HU6, appears to have addressed some of these concerns by demonstrating a favorable side effect profile while inducing significant body weight loss and MRI-measured liver fat reduction in patients with obesity and NASH [73].
Exercise is well known to upregulate the expression of various mitochondrial UCPs. Animal models of aerobic exercise have demonstrated that UCP-1 can be upregulated in brown adipose tissue, white adipose tissue as it browns in response to exercise and skeletal muscle [74][75][76][77]. Animal models have also demonstrated that UCP-2 is modulated by exercise in the vascular endothelium, myocardium, adipose tissue and skeletal muscle [74][78], as is UCP-3 in skeletal muscle [79]. When limited to animal models of NASH, aerobic training reverses dysfunction in UCP-2 in the liver [80]; however, researchers are unaware of any studies to date in patients with NAFLD or NASH confirming these findings.
In summary, animal models suggest a role for exercise training in upregulating UCPs.

2.6. Peroxisome Proliferator-Activated Receptor (PPAR)-α/γ

PPARα is a nuclear receptor that plays a key role in regulating lipid metabolism. It is specifically activated by fatty acids and their derivatives. The receptor can be found in many key organs, including the liver, and has three subtypes, PPAR-α, PPAR-γ and PPAR-β/δ [81]. When expressed in the liver, PPAR-α is responsible for fatty acid catabolism and energy homeostasis [82][83]. PPAR-γ is heavily involved in glucagon signaling and insulin sensitivity and, thus, is closely related to adiposity-related disorders [84][85]. The PPAR pathway remains one of many targets for drug discovery of anti-steatogenic medications, where late-phase studies have demonstrated PPAR-γ agonists, such as pioglitazone [86], and dual PPAR-α/γ agonists, such as saroglitazar [87], improve insulin resistance and lipid metabolism through regulation of fatty acid metabolism and modulation of inflammatory adipocytokines and adiponectin secretion. Moreover, the PPAR pathway is intricately linked to several other signaling pathways involved in NAFLD and NASH pathogenesis, including FGF-21, AMPK, and uncoupling proteins (UCP) [88][89][90].
The PPAR-α pathway is known to be strongly influenced by exercise training in non-NAFLD populations [91][92][93], including patients who are physically inactive and are overweight or obese [94]. In animal models of NAFLD, multiple studies have shown exercise-induced PPAR-α activation to mediate liver fat reduction [12][85][90][95]. Importantly, the impact of exercise training on the PPAR-α pathway appears to be independent of exercise type, in that PPAR activation has been observed in animals who performed either moderate-intensity aerobic exercise training with either swimming or running [85][90], HIIT [95][96] or resistance training [97]. While exercise per se can influence the regulation of PPAR activity, epigenetic factors may also play a role. For example, maternal exercise during pregnancy may confer protection against the development of NAFLD early in the life of offspring exposed to a high-fat diet. Bae-Gartz et al. [98] demonstrated that male offspring of exercised mouse dams were protected from adult-onset NAFLD mediated through greater activation of PPAR-α. The PPAR-γ receptor may also be affected by exercise training. Batatinha et al. [99] demonstrated that after eight weeks of treadmill training, PPAR-α knockout mice that were fed a high-fat diet to induce NAFLD still experienced a decrease in fat accumulation, perhaps owing to changes in PPAR-γ activity and fatty acid oxidation in the skeletal muscle.
In summary, the current available evidence indicates that exercise-mediated activation of PPAR appears to be mediated by exercise intensity and volume.

2.7. Thyroid Receptor (THR)-β

The thyroid gland and NAFLD are intricately linked in that thyroid hormones, including triiodothyronine (T3) and thyroxine (T4), are not only involved in lipid metabolism regulation, glucose uptake and increased size and number of mitochondria [100][101] but also that hypothyroidism is associated with increased NAFLD risk [102][103]. As a result, thyroid-hormone-based treatments, including resmetirom [104], are an attractive therapeutic target in patients with NAFLD and NASH. Two THR subtypes are found in the human body: THR-α, which is predominantly expressed in cardiac tissue, and THR-β, which is most commonly found in the liver and is responsible for the intrahepatic response to T3 [105]. THR-β is a critical receptor in the regulation of cholesterol metabolism and fatty acid oxidation, as exhibited in mouse models [69][106]. Importantly, THR-β is intricately linked to other pathways of interest in NAFLD, including PPAR-α, FGF-21 and UCP1 [69][107][108]. Since THR-β is interrelated to many metabolic pathways, it has been the focus of both pharmacologic and non-pharmacologic therapies. Resmetirom is an emerging therapy in NASH clinical trials that has the potential to significantly reduce hepatic fat, making it a potential therapy for patients with NASH [104].
Regular physical activity, including exercise training, is also well known to significantly impact circulating levels of thyroid hormone at the population level [109], in smaller groups of healthy adults who perform regular resistance training [110] and also in post-menopausal women with metabolic syndrome [111]. Specifically, exercise training leads to increased turnover of T3 and T4 at the same work rate, effectively lowering resting concentrations of thyroid hormone. The intensity of exercise is an important consideration as it appears to cause differential effects on thyroid hormones, with vigorous intensity leading to the greatest change [112]. When limiting solely to NAFLD and NASH, the data are more sparse. 

References

  1. Younossi, Z.M.; Golabi, P.; Paik, J.M.; Henry, A.; Van Dongen, C.; Henry, L. The global epidemiology of nonalcoholic fatty liver disease (NAFLD) and nonalcoholic steatohepatitis (NASH): A systematic review. Hepatology 2023, 77, 1335–1347.
  2. Caspersen, C.J.; Powell, K.E.; Christenson, G.M. Physical activity, exercise, and physical fitness: Definitions and distinctions for health-related research. Public Health Rep. 1985, 100, 126–131.
  3. Thorp, A.; Stine, J.G. Exercise as Medicine: The Impact of Exercise Training on Nonalcoholic Fatty Liver Disease. Curr. Hepatol. Rep. 2020, 19, 402–411.
  4. Stine, J.G.; Long, M.T.; Corey, K.E.; Sallis, R.E.; Allen, A.M.; Armstrong, M.J.; Conroy, D.E.; Cuthbertson, D.J.; Duarte-Rojo, A.; Hallsworth, K.; et al. American College of Sports Medicine (ACSM) International Multidisciplinary Roundtable Report on Physical Activity and Nonalcoholic Fatty Liver Disease. Hepatol. Commun. 2023, 7, e0108.
  5. Stine, J.G.; DiJoseph, K.; Pattison, Z.; Harrington, A.; Chinchilli, V.M.; Schmitz, K.H.; Loomba, R. Exercise Training Is Associated with Treatment Response in Liver Fat Content by Magnetic Resonance Imaging Independent of Clinically Significant Body Weight Loss in Patients with Nonalcoholic Fatty Liver Disease: A Systematic Review and Meta-Analysis. Am. J. Gastroenterol. 2023. Online ahead of print.
  6. Richter, E.A.; Ruderman, N.B. AMPK and the biochemistry of exercise: Implications for human health and disease. Biochem. J. 2009, 418, 261–275.
  7. Smith, B.K.; Marcinko, K.; Desjardins, E.M.; Lally, J.S.; Ford, R.J.; Steinberg, G.R. Treatment of nonalcoholic fatty liver disease: Role of AMPK. Am. J. Physiol. Endocrinol. Metab. 2016, 311, E730–E740.
  8. Cusi, K.; Alkhouri, N.; Harrison, S.A.; Fouqueray, P.; Moller, D.E.; Hallakou-Bozec, S.; Bolze, S.; Grouin, J.M.; Megnien, S.J.; Dubourg, J.; et al. Efficacy and safety of PXL770, a direct AMP kinase activator, for the treatment of non-alcoholic fatty liver disease (STAMP-NAFLD): A randomised, double-blind, placebo-controlled, phase 2a study. Lancet Gastroenterol. Hepatol. 2021, 6, 889–902.
  9. Gehrke, N.; Biedenbach, J.; Huber, Y.; Straub, B.K.; Galle, P.R.; Simon, P.; Schattenberg, J.M. Voluntary exercise in mice fed an obesogenic diet alters the hepatic immune phenotype and improves metabolic parameters—An animal model of life style intervention in NAFLD. Sci. Rep. 2019, 9, 4007.
  10. Alex, S.; Boss, A.; Heerschap, A.; Kersten, S. Exercise training improves liver steatosis in mice. Nutr. Metab. 2015, 12, 29.
  11. Rector, R.S.; Uptergrove, G.M.; Morris, E.M.; Borengasser, S.J.; Laughlin, M.H.; Booth, F.W.; Thyfault, J.P.; Ibdah, J.A. Daily exercise vs. caloric restriction for prevention of nonalcoholic fatty liver disease in the OLETF rat model. Am. J. Physiol. Gastrointest. Liver Physiol. 2011, 300, G874–G883.
  12. Cho, J.; Lee, I.; Kim, D.; Koh, Y.; Kong, J.; Lee, S.; Kang, H. Effect of aerobic exercise training on non-alcoholic fatty liver disease induced by a high fat diet in C57BL/6 mice. J. Exerc. Nutr. Biochem. 2014, 18, 339–346.
  13. Cintra, D.E.; Ropelle, E.R.; Vitto, M.F.; Luciano, T.F.; Souza, D.R.; Engelmann, J.; Marques, S.O.; Lira, F.S.; de Pinho, R.A.; Pauli, J.R.; et al. RETRACTED: Reversion of hepatic steatosis by exercise training in obese mice: The role of sterol regulatory element-binding protein-1c. Life Sci. 2012, 91, 395–401.
  14. Hardie, D.G. AMP-activated protein kinase: An energy sensor that regulates all aspects of cell function. Genes Dev. 2011, 25, 1895–1908.
  15. Apro, W.; Wang, L.; Ponten, M.; Blomstrand, E.; Sahlin, K. Resistance exercise induced mTORC1 signaling is not impaired by subsequent endurance exercise in human skeletal muscle. Am. J. Physiol. Endocrinol. Metab. 2013, 305, E22–E32.
  16. Chen, Z.P.; Stephens, T.J.; Murthy, S.; Canny, B.J.; Hargreaves, M.; Witters, L.A.; Kemp, B.E.; McConell, G.K. Effect of exercise intensity on skeletal muscle AMPK signaling in humans. Diabetes 2003, 52, 2205–2212.
  17. Steinberg, G.R.; Watt, M.J.; McGee, S.L.; Chan, S.; Hargreaves, M.; Febbraio, M.A.; Stapleton, D.; Kemp, B.E. Reduced glycogen availability is associated with increased AMPKalpha2 activity, nuclear AMPKalpha2 protein abundance, and GLUT4 mRNA expression in contracting human skeletal muscle. Appl. Physiol. Nutr. Metab. Physiol. Appl. Nutr. Metab. 2006, 31, 302–312.
  18. Jensen, J.; Rustad, P.I.; Kolnes, A.J.; Lai, Y.-C. The role of skeletal muscle glycogen breakdown for regulation of insulin sensitivity by exercise. Front. Physiol. 2011, 2, 112.
  19. Casuso, R.A.; Plaza-Díaz, J.; Ruiz-Ojeda, F.J.; Aragón-Vela, J.; Robles-Sanchez, C.; Nordsborg, N.B.; Hebberecht, M.; Salmeron, L.M.; Huertas, J.R. High-intensity high-volume swimming induces more robust signaling through PGC-1α and AMPK activation than sprint interval swimming in m. triceps brachii. PLoS ONE 2017, 12, e0185494.
  20. Stine, J.G.; Welles, J.E.; Keating, S.; Hussaini, Z.; Soriano, C.; Heinle, J.W.; Geyer, N.; Chinchilli, V.M.; Loomba, R.; Kimball, S.R. Serum Fibroblast Growth Factor 21 Is Markedly Decreased following Exercise Training in Patients with Biopsy-Proven Nonalcoholic Steatohepatitis. Nutrients 2023, 15, 1481.
  21. Stine, J.G.; Xu, D.; Schmitz, K.; Sciamanna, C.; Kimball, S.R. Exercise Attenuates Ribosomal Protein Six Phosphorylation in Fatty Liver Disease. Dig. Dis. Sci. 2020, 65, 3238–3243.
  22. Yun, Y.R.; Won, J.E.; Jeon, E.; Lee, S.; Kang, W.; Jo, H.; Jang, J.H.; Shin, U.S.; Kim, H.W. Fibroblast growth factors: Biology, function, and application for tissue regeneration. J. Tissue Eng. 2010, 2010, 218142.
  23. Kurosu, H.; Choi, M.; Ogawa, Y.; Dickson, A.S.; Goetz, R.; Eliseenkova, A.V.; Mohammadi, M.; Rosenblatt, K.P.; Kliewer, S.A.; Kuro, O.M. Tissue-specific expression of betaKlotho and fibroblast growth factor (FGF) receptor isoforms determines metabolic activity of FGF19 and FGF21. J. Biol. Chem. 2007, 282, 26687–26695.
  24. Harrison, S.A.; Abdelmalek, M.F.; Neff, G.; Gunn, N.; Guy, C.D.; Alkhouri, N.; Bashir, M.R.; Freilich, B.; Kohli, A.; Khazanchi, A.; et al. Aldafermin in patients with non-alcoholic steatohepatitis (ALPINE 2/3): A randomised, double-blind, placebo-controlled, phase 2b trial. Lancet Gastroenterol. Hepatol. 2022, 7, 603–616.
  25. Abdelmalek, M.F.; Charles, E.D.; Sanyal, A.J.; Harrison, S.A.; Neuschwander-Tetri, B.A.; Goodman, Z.; Ehman, R.A.; Karsdal, M.; Nakajima, A.; Du, S.; et al. The FALCON program: Two phase 2b randomized, double-blind, placebo-controlled studies to assess the efficacy and safety of pegbelfermin in the treatment of patients with nonalcoholic steatohepatitis and bridging fibrosis or compensated cirrhosis. Contemp. Clin. Trials 2021, 104, 106335.
  26. Harrison, S.A.; Ruane, P.J.; Freilich, B.L.; Neff, G.; Patil, R.; Behling, C.A.; Hu, C.; Fong, E.; de Temple, B.; Tillman, E.J.; et al. Efruxifermin in non-alcoholic steatohepatitis: A randomized, double-blind, placebo-controlled, phase 2a trial. Nat. Med. 2021, 27, 1262–1271.
  27. Brown, E.A.; Minnich, A.; Sanyal, A.J.; Loomba, R.; Du, S.; Schwarz, J.; Ehman, R.L.; Karsdal, M.; Leeming, D.J.; Cizza, G.; et al. Effect of pegbelfermin on NASH and fibrosis-related biomarkers and correlation with histological response in the FALCON 1 trial. JHEP Rep. Innov. Hepatol. 2023, 5, 100661.
  28. Fukumoto, S. Actions and mode of actions of FGF19 subfamily members. Endocr. J. 2008, 55, 23–31.
  29. Lin, B.C.; Wang, M.; Blackmore, C.; Desnoyers, L.R. Liver-specific activities of FGF19 require Klotho beta. J. Biol. Chem. 2007, 282, 27277–27284.
  30. Suzuki, M.; Uehara, Y.; Motomura-Matsuzaka, K.; Oki, J.; Koyama, Y.; Kimura, M.; Asada, M.; Komi-Kuramochi, A.; Oka, S.; Imamura, T. betaKlotho is required for fibroblast growth factor (FGF) 21 signaling through FGF receptor (FGFR) 1c and FGFR3c. Mol. Endocrinol. 2008, 22, 1006–1014.
  31. Moure, R.; Cairó, M.; Morón-Ros, S.; Quesada-López, T.; Campderrós, L.; Cereijo, R.; Hernáez, A.; Villarroya, F.; Giralt, M. Levels of β-klotho determine the thermogenic responsiveness of adipose tissues: Involvement of the autocrine action of FGF21. Am. J. Physiol. Endocrinol. Metab. 2021, 320, E822–E834.
  32. Liu, J.; Xu, Y.; Hu, Y.; Wang, G. The role of fibroblast growth factor 21 in the pathogenesis of non-alcoholic fatty liver disease and implications for therapy. Metabolism 2015, 64, 380–390.
  33. Falamarzi, K.; Malekpour, M.; Tafti, M.F.; Azarpira, N.; Behboodi, M.; Zarei, M. The role of FGF21 and its analogs on liver associated diseases. Front. Med. 2022, 9, 967375.
  34. Slusher, A.L.; Whitehurst, M.; Zoeller, R.F.; Mock, J.T.; Maharaj, M.; Huang, C.J. Attenuated fibroblast growth factor 21 response to acute aerobic exercise in obese individuals. Nutr. Metab. Cardiovasc. Dis. NMCD 2015, 25, 839–845.
  35. Banitalebi, E.; Kazemi, A.; Faramarzi, M.; Nasiri, S.; Haghighi, M.M. Effects of sprint interval or combined aerobic and resistance training on myokines in overweight women with type 2 diabetes: A randomized controlled trial. Life Sci. 2019, 217, 101–109.
  36. Rad, M.M.; Bijeh, N.; Hosseini, S.R.A.; Saeb, A.R. The effect of two concurrent exercise modalities on serum concentrations of FGF21, irisin, follistatin, and myostatin in men with type 2 diabetes mellitus. Arch. Physiol. Biochem. 2020, 129, 424–433.
  37. Kong, Z.; Sun, S.; Liu, M.; Shi, Q. Short-Term High-Intensity Interval Training on Body Composition and Blood Glucose in Overweight and Obese Young Women. J. Diabetes Res. 2016, 2016, 4073618.
  38. Takahashi, A.; Abe, K.; Fujita, M.; Hayashi, M.; Okai, K.; Ohira, H. Simple resistance exercise decreases cytokeratin 18 and fibroblast growth factor 21 levels in patients with nonalcoholic fatty liver disease: A retrospective clinical study. Medicine 2020, 99, e20399.
  39. Porflitt-Rodríguez, M.; Guzmán-Arriagada, V.; Sandoval-Valderrama, R.; Tam, C.S.; Pavicic, F.; Ehrenfeld, P.; Martínez-Huenchullán, S. Effects of aerobic exercise on fibroblast growth factor 21 in overweight and obesity. A systematic review. Metabolism 2022, 129, 155137.
  40. Morville, T.; Sahl, R.E.; Trammell, S.A.; Svenningsen, J.S.; Gillum, M.P.; Helge, J.W.; Clemmensen, C. Divergent effects of resistance and endurance exercise on plasma bile acids, FGF19, and FGF21 in humans. JCI Insight 2018, 3, e122737.
  41. Baggio, L.L.; Drucker, D.J. Biology of incretins: GLP-1 and GIP. Gastroenterology 2007, 132, 2131–2157.
  42. Newsome, P.N.; Buchholtz, K.; Cusi, K.; Linder, M.; Okanoue, T.; Ratziu, V.; Sanyal, A.J.; Sejling, A.S.; Harrison, S.A. A Placebo-Controlled Trial of Subcutaneous Semaglutide in Nonalcoholic Steatohepatitis. N. Engl. J. Med. 2021, 384, 1113–1124.
  43. Armstrong, M.J.; Gaunt, P.; Aithal, G.P.; Barton, D.; Hull, D.; Parker, R.; Hazlehurst, J.M.; Guo, K.; Abouda, G.; Aldersley, M.A.; et al. Liraglutide safety and efficacy in patients with non-alcoholic steatohepatitis (LEAN): A multicentre, double-blind, randomised, placebo-controlled phase 2 study. Lancet 2016, 387, 679–690.
  44. Wilding, J.P.H.; Batterham, R.L.; Calanna, S.; Davies, M.; Van Gaal, L.F.; Lingvay, I.; McGowan, B.M.; Rosenstock, J.; Tran, M.T.D.; Wadden, T.A.; et al. Once-Weekly Semaglutide in Adults with Overweight or Obesity. N. Engl. J. Med. 2021, 384, 989.
  45. Tchang, B.G.; Aras, M.; Kumar, R.B.; Aronne, L.J. Pharmacologic Treatment of Overweight and Obesity in Adults. In Endotext; Feingold, K.R., Anawalt, B., Blackman, M.R., Boyce, A., Chrousos, G., Corpas, E., de Herder, W.W., Dhatariya, K., Dungan, K., Hofland, J., et al., Eds.; MDText.com, Inc.: South Dartmouth, MA, USA, 2000.
  46. Ueda, S.Y.; Yoshikawa, T.; Katsura, Y.; Usui, T.; Fujimoto, S. Comparable effects of moderate intensity exercise on changes in anorectic gut hormone levels and energy intake to high intensity exercise. J. Endocrinol. 2009, 203, 357–364.
  47. Martins, C.; Stensvold, D.; Finlayson, G.; Holst, J.; Wisloff, U.; Kulseng, B.; Morgan, L.; King, N.A. Effect of moderate- and high-intensity acute exercise on appetite in obese individuals. Med. Sci. Sport. Exerc. 2015, 47, 40–48.
  48. Kullman, E.L.; Kelly, K.R.; Haus, J.M.; Fealy, C.E.; Scelsi, A.R.; Pagadala, M.R.; Flask, C.A.; McCullough, A.J.; Kirwan, J.P. Short-term aerobic exercise training improves gut peptide regulation in nonalcoholic fatty liver disease. J. Appl. Physiol. 2016, 120, 1159–1164.
  49. Hari, A.; Fealy, C.E.; Axelrod, C.L.; Haus, J.M.; Flask, C.A.; McCullough, A.J.; Kirwan, J.P. Exercise Training Rapidly Increases Hepatic Insulin Extraction in NAFLD. Med. Sci. Sport. Exerc. 2020, 52, 1449–1455.
  50. Alves-Bezerra, M.; Cohen, D.E. Triglyceride Metabolism in the Liver. Compr. Physiol. 2017, 8, 1–8.
  51. Stevanović, J.; Beleza, J.; Coxito, P.; Ascensão, A.; Magalhães, J. Physical exercise and liver “fitness”: Role of mitochondrial function and epigenetics-related mechanisms in non-alcoholic fatty liver disease. Mol. Metab. 2020, 32, 1–14.
  52. Selen, E.S.; Choi, J.; Wolfgang, M.J. Discordant hepatic fatty acid oxidation and triglyceride hydrolysis leads to liver disease. JCI Insight 2021, 6, e135626.
  53. Petersen, M.C.; Shulman, G.I. Roles of Diacylglycerols and Ceramides in Hepatic Insulin Resistance. Trends Pharmacol. Sci. 2017, 38, 649–665.
  54. Morris, E.M.; McCoin, C.S.; Allen, J.A.; Gastecki, M.L.; Koch, L.G.; Britton, S.L.; Fletcher, J.A.; Fu, X.; Ding, W.X.; Burgess, S.C.; et al. Aerobic capacity mediates susceptibility for the transition from steatosis to steatohepatitis. J. Physiol. 2017, 595, 4909–4926.
  55. Fletcher, J.A.; Meers, G.M.; Linden, M.A.; Kearney, M.L.; Morris, E.M.; Thyfault, J.P.; Rector, R.S. Impact of various exercise modalities on hepatic mitochondrial function. Med. Sci. Sport. Exerc. 2014, 46, 1089–1097.
  56. Sabag, A.; Keating, S.E.; Way, K.L.; Sultana, R.N.; Lanting, S.M.; Twigg, S.M.; Johnson, N.A. The association between cardiorespiratory fitness, liver fat and insulin resistance in adults with or without type 2 diabetes: A cross-sectional analysis. BMC Sport. Sci. Med. Rehabil. 2021, 13, 40.
  57. Dahmus, J.; Hummer, B.; Rivas, G.; Schmitz, K.; Caldwell, S.H.; Argo, C.K.; Schreibman, I.; Stine, J.G. Patients with Nonalcoholic Steatohepatitis and Advanced Liver Disease Have the Lowest Cardiorespiratory Fitness. Dig. Dis. Sci. 2023, 68, 2695–2703.
  58. Sabag, A.; Way, K.L.; Sultana, R.N.; Keating, S.E.; Gerofi, J.A.; Chuter, V.H.; Byrne, N.M.; Baker, M.K.; George, J.; Caterson, I.D.; et al. The Effect of a Novel Low-Volume Aerobic Exercise Intervention on Liver Fat in Type 2 Diabetes: A Randomized Controlled Trial. Diabetes Care 2020, 43, 2371–2378.
  59. Sultana, R.N.; Sabag, A.; Keating, S.E.; Johnson, N.A. The Effect of Low-Volume High-Intensity Interval Training on Body Composition and Cardiorespiratory Fitness: A Systematic Review and Meta-Analysis. Sport. Med. 2019, 49, 1687–1721.
  60. Sabag, A.; Barr, L.; Armour, M.; Armstrong, A.; Baker, C.J.; Twigg, S.M.; Chang, D.; Hackett, D.A.; Keating, S.E.; George, J.; et al. The Effect of High-intensity Interval Training vs Moderate-intensity Continuous Training on Liver Fat: A Systematic Review and Meta-Analysis. J. Clin. Endocrinol. Metab. 2022, 107, 862–881.
  61. Ledesma, A.; de Lacoba, M.G.; Rial, E. The mitochondrial uncoupling proteins. Genome Biol. 2002, 3, 1–9.
  62. Nedergaard, J.; Cannon, B. The ‘novel’ ‘uncoupling’ proteins UCP2 and UCP3: What do they really do? Pros and cons for suggested functions. Exp. Physiol. 2003, 88, 65–84.
  63. Brand, M.D.; Esteves, T.C. Physiological functions of the mitochondrial uncoupling proteins UCP2 and UCP3. Cell Metab. 2005, 2, 85–93.
  64. Brand, M.D.; Affourtit, C.; Esteves, T.C.; Green, K.; Lambert, A.J.; Miwa, S.; Pakay, J.L.; Parker, N. Mitochondrial superoxide: Production, biological effects, and activation of uncoupling proteins. Free Radic. Biol. Med. 2004, 37, 755–767.
  65. Echtay, K.S.; Liu, Q.; Caskey, T.; Winkler, E.; Frischmuth, K.; Bienengräber, M.; Klingenberg, M. Regulation of UCP3 by nucleotides is different from regulation of UCP1. FEBS Lett. 1999, 450, 8–12.
  66. Liu, J.; Li, J.; Li, W.J.; Wang, C.M. The role of uncoupling proteins in diabetes mellitus. J. Diabetes Res. 2013, 2013, 585897.
  67. Dalgaard, L.T.; Pedersen, O. Uncoupling proteins: Functional characteristics and role in the pathogenesis of obesity and Type II diabetes. Diabetologia 2001, 44, 946–965.
  68. Chavin, K.D.; Yang, S.; Lin, H.Z.; Chatham, J.; Chacko, V.P.; Hoek, J.B.; Walajtys-Rode, E.; Rashid, A.; Chen, C.H.; Huang, C.C.; et al. Obesity induces expression of uncoupling protein-2 in hepatocytes and promotes liver ATP depletion. J. Biol. Chem. 1999, 274, 5692–5700.
  69. Ribeiro, M.O.; Bianco, S.D.; Kaneshige, M.; Schultz, J.J.; Cheng, S.Y.; Bianco, A.C.; Brent, G.A. Expression of uncoupling protein 1 in mouse brown adipose tissue is thyroid hormone receptor-beta isoform specific and required for adaptive thermogenesis. Endocrinology 2010, 151, 432–440.
  70. Dong, C.; Lv, Y.; Xie, L.; Yang, R.; Chen, L.; Zhang, L.; Long, T.; Yang, H.; Mao, X.; Fan, Q.; et al. Association of UCP1 polymorphisms with type 2 diabetes mellitus and their interaction with physical activity and sedentary behavior. Gene 2020, 739, 144497.
  71. Muhammad, H.F.L.; Sulistyoningrum, D.C.; Huriyati, E.; Lee, Y.Y.; Muda, W. The interaction between energy intake, physical activity and UCP2 -866G/A gene variation on weight gain and changes in adiposity: An Indonesian Nutrigenetic Cohort (INDOGENIC). Br. J. Nutr. 2021, 125, 611–617.
  72. Grundlingh, J.; Dargan, P.I.; El-Zanfaly, M.; Wood, D.M. 2,4-dinitrophenol (DNP): A weight loss agent with significant acute toxicity and risk of death. J. Med. Toxicol. 2011, 7, 205–212.
  73. Noureddin, M.; Khan, S.; Portell, F.; Jorkasky, D.; Dennis, J.; Khan, O.; Sanyal, A.J. HU6 reduces liver fat in subjects with high BMI NAFLD: Top-line results from a phase 2a trial. Hepatology 2022, 76, S94–S95.
  74. Kim, H.J.; Kim, Y.J.; Seong, J.K. AMP-activated protein kinase activation in skeletal muscle modulates exercise-induced uncoupled protein 1 expression in brown adipocyte in mouse model. J. Physiol. 2022, 600, 2359–2376.
  75. Yin, R.; Ma, Y.; Zhang, N.; Yang, L.; Zhao, D. Combined effects of voluntary running and liraglutide on glucose homeostasis, fatty acid composition of brown adipose tissue phospholipids, and white adipose tissue browning in db/db mice. Chin. J. Physiol. 2022, 65, 117–124.
  76. Tanimura, R.; Kobayashi, L.; Shirai, T.; Takemasa, T. Effects of exercise intensity on white adipose tissue browning and its regulatory signals in mice. Physiol. Rep. 2022, 10, e15205.
  77. Liu, Y.; Guo, C.; Liu, S.; Zhang, S.; Mao, Y.; Fang, L. Eight Weeks of High-Intensity Interval Static Strength Training Improves Skeletal Muscle Atrophy and Motor Function in Aged Rats via the PGC-1α/FNDC5/UCP1 Pathway. Clin. Interv. Aging 2021, 16, 811–821.
  78. Hong, J.; Park, E.; Lee, J.; Lee, Y.; Rooney, B.V.; Park, Y. Exercise training mitigates ER stress and UCP2 deficiency-associated coronary vascular dysfunction in atherosclerosis. Sci. Rep. 2021, 11, 15449.
  79. Zhou, M.; Lin, B.Z.; Coughlin, S.; Vallega, G.; Pilch, P.F. UCP-3 expression in skeletal muscle: Effects of exercise, hypoxia, and AMP-activated protein kinase. Am. J. Physiol. Endocrinol. Metab. 2000, 279, E622–E629.
  80. Gonçalves, I.O.; Passos, E.; Rocha-Rodrigues, S.; Diogo, C.V.; Torrella, J.R.; Rizo, D.; Viscor, G.; Santos-Alves, E.; Marques-Aleixo, I.; Oliveira, P.J.; et al. Physical exercise prevents and mitigates non-alcoholic steatohepatitis-induced liver mitochondrial structural and bioenergetics impairments. Mitochondrion 2014, 15, 40–51.
  81. Tyagi, S.; Gupta, P.; Saini, A.S.; Kaushal, C.; Sharma, S. The peroxisome proliferator-activated receptor: A family of nuclear receptors role in various diseases. J. Adv. Pharm. Technol. Res. 2011, 2, 236–240.
  82. Poulsen, L.; Siersbæk, M.; Mandrup, S. PPARs: Fatty acid sensors controlling metabolism. Semin. Cell Dev. Biol. 2012, 23, 631–639.
  83. Amorim, R.; Simões, I.C.M.; Teixeira, J.; Cagide, F.; Potes, Y.; Soares, P.; Carvalho, A.; Tavares, L.C.; Benfeito, S.; Pereira, S.P.; et al. Mitochondria-targeted anti-oxidant AntiOxCIN(4) improved liver steatosis in Western diet-fed mice by preventing lipid accumulation due to upregulation of fatty acid oxidation, quality control mechanism and antioxidant defense systems. Redox Biol. 2022, 55, 102400.
  84. Berglund, E.D.; Lustig, D.G.; Baheza, R.A.; Hasenour, C.M.; Lee-Young, R.S.; Donahue, E.P.; Lynes, S.E.; Swift, L.L.; Charron, M.J.; Damon, B.M.; et al. Hepatic glucagon action is essential for exercise-induced reversal of mouse fatty liver. Diabetes 2011, 60, 2720–2729.
  85. Diniz, T.A.; de Lima Junior, E.A.; Teixeira, A.A.; Biondo, L.A.; da Rocha, L.A.F.; Valadão, I.C.; Silveira, L.S.; Cabral-Santos, C.; de Souza, C.O.; Neto, J.C.R. Aerobic training improves NAFLD markers and insulin resistance through AMPK-PPAR-α signaling in obese mice. Life Sci. 2021, 266, 118868.
  86. Sanyal, A.J.; Chalasani, N.; Kowdley, K.V.; McCullough, A.; Diehl, A.M.; Bass, N.M.; Neuschwander-Tetri, B.A.; Lavine, J.E.; Tonascia, J.; Unalp, A.; et al. Pioglitazone, vitamin E, or placebo for nonalcoholic steatohepatitis. N. Engl. J. Med. 2010, 362, 1675–1685.
  87. Gawrieh, S.; Noureddin, M.; Loo, N.; Mohseni, R.; Awasty, V.; Cusi, K.; Kowdley, K.V.; Lai, M.; Schiff, E.; Parmar, D.; et al. Saroglitazar, a PPAR-α/γ Agonist, for Treatment of NAFLD: A Randomized Controlled Double-Blind Phase 2 Trial. Hepatology 2021, 74, 1809–1824.
  88. Ricquier, D.; Bouillaud, F. Mitochondrial uncoupling proteins: From mitochondria to the regulation of energy balance. J. Physiol. 2000, 529 Pt 1, 3–10.
  89. Vernia, S.; Cavanagh-Kyros, J.; Garcia-Haro, L.; Sabio, G.; Barrett, T.; Jung, D.Y.; Kim, J.K.; Xu, J.; Shulha, H.P.; Garber, M.; et al. The PPARα-FGF21 hormone axis contributes to metabolic regulation by the hepatic JNK signaling pathway. Cell Metab. 2014, 20, 512–525.
  90. Li, D.D.; Ma, J.M.; Li, M.J.; Gao, L.L.; Fan, Y.N.; Zhang, Y.N.; Tao, X.J.; Yang, J.J. Supplementation of Lycium barbarum Polysaccharide Combined with Aerobic Exercise Ameliorates High-Fat-Induced Nonalcoholic Steatohepatitis via AMPK/PPARα/PGC-1α Pathway. Nutrients 2022, 14, 3247.
  91. Maciejewska, A.; Sawczuk, M.; Cięszczyk, P. Variation in the PPARα gene in Polish rowers. J. Sci. Med. Sport 2011, 14, 58–64.
  92. Ahmetov, I.I.; Mozhayskaya, I.A.; Flavell, D.M.; Astratenkova, I.V.; Komkova, A.I.; Lyubaeva, E.V.; Tarakin, P.P.; Shenkman, B.S.; Vdovina, A.B.; Netreba, A.I.; et al. PPARalpha gene variation and physical performance in Russian athletes. Eur. J. Appl. Physiol. 2006, 97, 103–108.
  93. Nishida, Y.; Iyadomi, M.; Tominaga, H.; Taniguchi, H.; Higaki, Y.; Tanaka, H.; Horita, M.; Shimanoe, C.; Hara, M.; Tanaka, K. Influence of Single-Nucleotide Polymorphisms in PPAR-δ, PPAR-γ, and PRKAA2 on the Changes in Anthropometric Indices and Blood Measurements through Exercise-Centered Lifestyle Intervention in Japanese Middle-Aged Men. Int. J. Mol. Sci. 2018, 19, 703.
  94. Blond, M.B.; Schnurr, T.M.; Rosenkilde, M.; Quist, J.S.; Gram, A.S.; Reichkendler, M.H.; Auerbach, P.L.; Nordby, P.; Skovgaard, L.T.; Ribel-Madsen, R.; et al. PPARG Pro12Ala Ala carriers exhibit greater improvements in peripheral insulin sensitivity in response to 12 weeks of aerobic exercise training. Physiol. Genom. 2019, 51, 254–260.
  95. Gu, X.; Ma, X.; Mo, L.; Wang, Q. The role of exercise intensity on fatty liver in rats. Chin. J. Physiol. 2022, 65, 301–310.
  96. Kapravelou, G.; Martínez, R.; Andrade, A.M.; Nebot, E.; Camiletti-Moirón, D.; Aparicio, V.A.; Lopez-Jurado, M.; Aranda, P.; Arrebola, F.; Fernandez-Segura, E.; et al. Aerobic interval exercise improves parameters of nonalcoholic fatty liver disease (NAFLD) and other alterations of metabolic syndrome in obese Zucker rats. Appl. Physiol. Nutr. Metab. Physiol. Appl. Nutr. Metab. 2015, 40, 1242–1252.
  97. Nikroo, H.; Hosseini, S.R.A.; Fathi, M.; Sardar, M.A.; Khazaei, M. The effect of aerobic, resistance, and combined training on PPAR-α, SIRT1 gene expression, and insulin resistance in high-fat diet-induced NAFLD male rats. Physiol. Behav. 2020, 227, 113149.
  98. Bae-Gartz, I.; Kasper, P.; Großmann, N.; Breuer, S.; Janoschek, R.; Kretschmer, T.; Appel, S.; Schmitz, L.; Vohlen, C.; Quaas, A.; et al. Maternal exercise conveys protection against NAFLD in the offspring via hepatic metabolic programming. Sci. Rep. 2020, 10, 15424.
  99. Batatinha, H.A.; Lima, E.A.; Teixeira, A.A.; Souza, C.O.; Biondo, L.A.; Silveira, L.S.; Lira, F.S.; Neto, J.C.R. Association between Aerobic Exercise and Rosiglitazone Avoided the NAFLD and Liver Inflammation Exacerbated in PPAR-α Knockout Mice. J. Cell. Physiol. 2017, 232, 1008–1019.
  100. Sinha, R.A.; Singh, B.K.; Yen, P.M. Direct effects of thyroid hormones on hepatic lipid metabolism. Nat. Rev. Endocrinol. 2018, 14, 259–269.
  101. Simonet, W.S.; Ness, G.C. Transcriptional and posttranscriptional regulation of rat hepatic 3-hydroxy-3-methylglutaryl-coenzyme A reductase by thyroid hormones. J. Biol. Chem. 1988, 263, 12448–12453.
  102. Ritter, M.J.; Amano, I.; Hollenberg, A.N. Thyroid Hormone Signaling and the Liver. Hepatology 2020, 72, 742–752.
  103. Qiu, S.; Cao, P.; Guo, Y.; Lu, H.; Hu, Y. Exploring the Causality between Hypothyroidism and Non-alcoholic Fatty Liver: A Mendelian Randomization Study. Front. Cell Dev. Biol. 2021, 9, 643582.
  104. Harrison, S.A.; Bashir, M.R.; Guy, C.D.; Zhou, R.; Moylan, C.A.; Frias, J.P.; Alkhouri, N.; Bansal, M.B.; Baum, S.; Neuschwander-Tetri, B.A.; et al. Resmetirom (MGL-3196) for the treatment of non-alcoholic steatohepatitis: A multicentre, randomised, double-blind, placebo-controlled, phase 2 trial. Lancet 2019, 394, 2012–2024.
  105. Li, L.M.; Song, Y.; Shi, Y.Q.; Sun, L.L. Thyroid hormone receptor-beta agonists in NAFLD therapy: Possibilities and challenges. J. Clin. Endocrinol. Metab. 2023. Online ahead of print.
  106. Ying, H.; Araki, O.; Furuya, F.; Kato, Y.; Cheng, S.Y. Impaired adipogenesis caused by a mutated thyroid hormone alpha1 receptor. Mol. Cell. Biol. 2007, 27, 2359–2371.
  107. Domouzoglou, E.M.; Fisher, F.M.; Astapova, I.; Fox, E.C.; Kharitonenkov, A.; Flier, J.S.; Hollenberg, A.N.; Maratos-Flier, E. Fibroblast growth factor 21 and thyroid hormone show mutual regulatory dependency but have independent actions in vivo. Endocrinology 2014, 155, 2031–2040.
  108. Videla, L.A.; Fernández, V.; Vargas, R.; Cornejo, P.; Tapia, G.; Varela, N.; Valenzuela, R.; Arenas, A.; Fernández, J.; Hernández-Rodas, M.C.; et al. Upregulation of rat liver PPARα-FGF21 signaling by a docosahexaenoic acid and thyroid hormone combined protocol. BioFactors 2016, 42, 638–646.
  109. Klasson, C.L.; Sadhir, S.; Pontzer, H. Daily physical activity is negatively associated with thyroid hormone levels, inflammation, and immune system markers among men and women in the NHANES dataset. PLoS ONE 2022, 17, e0270221.
  110. Aristizabal, J.C.; Freidenreich, D.J.; Volk, B.M.; Kupchak, B.R.; Saenz, C.; Maresh, C.M.; Kraemer, W.J.; Volek, J.S. Effect of resistance training on resting metabolic rate and its estimation by a dual-energy X-ray absorptiometry metabolic map. Eur. J. Clin. Nutr. 2015, 69, 831–836.
  111. Berahman, H.; Elmieh, A.; Chafy, M.R.F. The effect of water-based rhythmic exercise training on glucose homeostasis and thyroid hormones in postmenopausal women with metabolic syndrome. Horm. Mol. Biol. Clin. Investig. 2021, 42, 189–193.
  112. Ciloglu, F.; Peker, I.; Pehlivan, A.; Karacabey, K.; Ilhan, N.; Saygin, O.; Ozmerdivenli, R. Exercise intensity and its effects on thyroid hormones. Neuro Endocrinol. Lett. 2005, 26, 830–834.
More
Information
Contributors MDPI registered users' name will be linked to their SciProfiles pages. To register with us, please refer to https://encyclopedia.pub/register : , , , , , ,
View Times: 315
Revisions: 2 times (View History)
Update Date: 06 Jun 2023
1000/1000
Video Production Service