IMR Press / FBL / Volume 28 / Issue 12 / DOI: 10.31083/j.fbl2812351
Open Access Review
FGF21: A Sharp Weapon in the Process of Exercise to Improve NAFLD
Show Less
1 School of Sports Science, Beijing Sport University, 100080 Beijing, China
2 Key Laboratory of the Ministry of Education of Exercise and Physical Fitness, Beijing Sport University, 100080 Beijing, China
*Correspondence: ymzhangno1@163.com (Yimin Zhang)
Front. Biosci. (Landmark Ed) 2023, 28(12), 351; https://doi.org/10.31083/j.fbl2812351
Submitted: 18 July 2023 | Revised: 9 September 2023 | Accepted: 18 September 2023 | Published: 27 December 2023
Copyright: © 2023 The Author(s). Published by IMR Press.
This is an open access article under the CC BY 4.0 license.
Abstract

Nonalcoholic fatty liver disease (NAFLD) is a pathological syndrome characterized by excessive fat deposition in hepatocytes. A sedentary lifestyle is a major risk factor for NAFLD, and regular exercise is considered a cornerstone of NAFLD treatment independent of weight loss. Even low-intensity activity could have beneficial effects on NAFLD. Fibroblast growth factor 21 (FGF21), a cytokine mainly secreted by the liver, improves glucolipid metabolism, reduces inflammation and oxidative stress, increases insulin sensitivity, and acts on multiple organs through autocrine, paracrine, and endocrine actions. Both clinical trials and animal experiments have shown a high correlation between liver fat content and circulating blood FGF21 levels, and abnormal FGF21 signaling appears to be an important mechanism for the development of NAFLD. FGF21 is an exerkine that responds to exercise; therefore, it may be a key target in exercise to improve NAFLD. This review provides an overview of NAFLD and its pathogenesis, and summarizes the effects of exercise intervention on NAFLD, as well as the role of FGF21 in NAFLD. Emphasis is placed on possible mechanisms for improving NAFLD by targeting FGF21 during exercise.

Keywords
NAFLD
FGF21
exercise intervention
1. Introduction

Non-alcoholic fatty liver disease (NAFLD) is the most prevalent chronic liver disease globally, affecting about 25.24% of the population worldwide [1, 2]. It causes liver inflammation, fibrosis, cirrhosis and even liver cancer, and lead to cardiovascular and metabolic diseases, making it a major public health burden [3]. Obesity, lack of exercise and unhealthy dietary patterns are important risk factors for NAFLD [4]. Although the dangers of NAFLD are well known, no drugs have been approved specifically for the treatment of this disease [5]. Several promising targets for the treatment of NAFLD such as peroxisome proliferator-activated receptor (PPAR), sodium-glucose transport protein 2, glucagon-like peptide 1, farnesoid X receptor, fibroblast growth factor 19 (FGF19), and FGF21 are being actively explored, and related drugs such as pioglitazone, canagliflozin, liraglutid, obeticholic acid, NGM282, and pegbelfermin (BMS-986036) are undergoing clinical studies [6]. Lifestyle and dietary interventions, as well as possible bariatric surgery, are generally recognized as the cornerstones of NAFLD treatment [7].

Long-term sedentary lifestyle and inactivity are important health hazards, and the risk of multiple chronic diseases is directly related to a lack of physical activity [8]. Only recently has the biomedical scientific community clearly acknowledged the significant benefits of regular exercise on the prevention and improvement of several illnesses, even showing exercise to be more effective than some low-performing medicines [9]. Regular exercise reduces liver fat content, increases insulin sensitivity and improves lipid metabolism, thereby relieving NAFLD and other metabolic diseases, independent of weight loss [10]. During exercise, organs such as skeletal muscle, liver, and adipose tissue secrete a series of cytokines such as irisin, interleukin 6 (IL-6), brain-derived neurotrophic factor and FGF21, called exerkines, which have important health benefits [9].

FGF21 is an atypical member of the FGF family with multiple biological roles [11]. It is mainly derived from the liver, although it can also be secreted from other organs such as skeletal muscle, adipose tissue and heart [12]. FGF21 acts via autocrine, paracrine, and endocrine functions, or is released into the circulatory system to exert metabolic regulation on multiple target organs such as the hypothalamus, adipose tissue and pancreas [13]. FGF21 has various important biological roles such as ameliorating oxidative stress [14], exerting anti-inflammatory [15] and anti-apoptotic effects [16], and alleviating endoplasmic reticulum (ER) stress [17], and is a potential drug target for the treatment of various metabolic diseases such as obesity [18, 19], type 2 diabetes mellitus (T2DM) [20], and NAFLD [21].

This review provides an overview of NAFLD and its pathogenesis, and summarizes the effects of exercise intervention on NAFLD, as well as the role of FGF21 in NAFLD and its response to exercise. Emphasis is placed on the effects of FGF21 on exercise intervention in NAFLD and its possible mechanisms.

2. NAFLD Overview
2.1 Definition of NAFLD

NAFLD is the accumulation of triglycerides (TGs) in hepatocytes exceeding at least 5% of the liver weight of individuals with little or no alcohol consumption and no other liver disease [22]. The progression of liver disease usually includes the following stages: NAFL (without hepatocellular injury), non-alcoholic steatohepatitis (NASH, inflammation and fibrosis), cirrhosis and Hepatocellular carcinoma (HCC) (Fig. 1) [23, 24]. Liver biopsy is one way to histologically diagnose NAFLD despite its invasive nature. Transient elastography based on ultrasound techniques [25] and magnetic resonance elastography [6] are important noninvasive methods for the histologic diagnosis of NAFLD. In addition, some predictive models of NAFLD, such as fatty liver index [26], hepatic steatosis index [27], fibrosis 4 score [28], Framingham steatosis index [29], percentage of hepatic fat [30], and NAFLD Screening Score [31]; and blood biochemical markers such as aspartate aminotransferase (AST) and alanine aminotransferase (ALT) are also diagnostic for NAFLD.

Fig. 1.

Liver disease progression. Healthy liver to NAFL, NASH, cirrhosis and HCC. Abbreviations: NAFL, non-alcoholic fatty liver; NASH, nonalcoholic steatohepatitis; HCC, hepatocellular carcinoma; TGs, triglycerides. Fig. 1 created with BioRender.com.

2.2 Pathogenesis of NAFLD

As scientific research continues, the possible pathogenesis of NAFLD is gradually being refined (Fig. 2). The “two-hit hypothesis” was originally proposed as one of the hypotheses to explain the pathogenesis of NAFLD [32]. The hypothesis suggested that the “first-hit” was due to a metabolic disturbance caused by insulin resistance (IR), which allowed excess free fatty acids (FFAs) to flow into the liver, promoting the production and accumulation of TGs and causing hepatic steatosis. According to the “second-hit” hypothesis, an increase in pro-inflammatory cytokines and oxidative stress due to hepatic steatosis caused by TG accumulation, results in NASH and fibrosis [33]. However, as the influencing factors of NAFLD were revealed, it was gradually found that the “two-hit hypothesis” was insufficient to explain the process of NAFLD. Therefore, the “multiple-hit hypothesis” was proposed [34]. This hypothesis considers that the pathogenesis of NAFLD may be due to a combination of factors including increased pro-inflammatory cytokines, increased oxidative stress, IR, nutritional factors, alterations in gut microbes, and genetic limitations [34]. Although the cellular and molecular mechanisms of NAFLD have been extensively studied in recent years, the exact mechanism of action of disease development is still not fully understood.

Fig. 2.

Two-Hit Hypothesis and Multiple-Hit Hypothesis of NAFLD pathogenesis. Abbreviations: ROS, reactive oxygen species. Fig. 2 created with BioRender.com.

3. Exercise Intervention for NAFLD
3.1 NAFLD Improvement by Exercise

Exercise not only improves NAFLD but also serves as an effective preventive tool for several metabolic diseases, such as diabetes [35] and hypertension [36]. The effects of exercise on improving NAFLD include, but are not limited to, reducing intrahepatic lipid content, improving insulin sensitivity, increasing caloric consumption, and promoting weight loss [37]. A variety of regular exercise modalities including aerobic exercise, resistance exercise, and flexibility training could improve NAFLD independent of weight loss [10].

Aerobic exercise is one of the most commonly used forms of exercise to improve NAFLD, as it reduces blood levels of AST and ALT, which are biomarkers of liver damage [38, 39]. Meanwhile, low-intensity aerobic exercises such as brisk walking [40] and cycling [41] are able to reduce liver fat content in patients with NAFLD or obesity, despite no significant change in body weight. In addition, studies have shown that high-intensity aerobic exercise may improve liver fat content and stiffness in patients with NAFLD [42].

Resistance exercise, also known as strength training, is effective in improving muscle strength and mass and is one of the most commonly used exercise methods for NAFLD patients [43]. It has been shown that 8 weeks of resistance exercise is able to reduce liver fat content and increase insulin sensitivity independent of weight loss [44]. In addition, other modalities including high-intensity interval training (HIIT) [45] and concurrent exercise [46] are also exercise methods to improve NAFLD.

3.2 Mechanisms Underlying the Effects of Exercise on NAFLD Improvement

Exercise improves IR, and improvement of IR is one of the prominent mechanisms by which exercise alleviates NAFLD [47]. IR in peripheral tissues such as skeletal muscle and adipose tissue increases lipolysis and the release of FFAs, which is subsequently taken up by the liver, resulting in disruption of hepatic fatty acid metabolism [48]. Exercise improves IR in peripheral tissues, increases insulin sensitivity, and may reduce excess FFAs flow to the liver [49]. Increased insulin sensitivity increases glycogen synthesis in the liver and glucose uptake capacity in skeletal muscle [50]. In addition, exercise also increases the expression of AMP-activated protein kinase (AMPK) [51] and PPARγ [46], and inhibits the expression of sterol regulatory element binding protein 1 (SREBP-1) [52], which is a major regulator of hepatic fatty acid metabolism, thereby reducing the accumulation of fat in the liver and ameliorating disorders of hepatic fatty acid metabolism, thus positively affecting NAFLD.

Exercise improves oxidative stress and inflammatory responses. Oxidative stress reflects an imbalance between oxidative and antioxidant systems, leading to the excessive production of reactive oxygen species (ROS), which severely impairs cell function and leads to cell death [53]. The overproduction of ROS and the occurrence of oxidative stress are key mechanisms in the development of NAFLD, leading to impaired antioxidant systems as well as lipid peroxidation and possibly to the activation of inflammation to the point where liver fibrosis and hepatocyte cell death occur [54]. Regular exercise is considered an effective antioxidant [55]. As early as 1983, it was shown that long-term exercise training in rodents could increase the level of antioxidant enzymes, such as glutathione peroxidase (GPX) and superoxide dismutase (SOD), to prevent liver damage caused by oxidative stress [56]. Until recently, studies related to the antioxidant effects of exercise were still being reported. Zhang et al. [57] showed that exercise training alleviated hepatic lipid accumulation and ameliorated lipotoxicity-induced apoptosis in db/db mice via the AMPK/nuclear factor erythroid 2-related factor 2 (Nrf2)/heme oxygenase 1 (HO-1) signaling pathway. Zou et al. [58] reported that swimming intervention attenuated hepatic pathological changes in NAFLD zebrafish through silent information regulator 1 (SIRT1)/AMPK/Nrf2 signaling.

The hepatic inflammatory response is a key mechanism for the progression of NAFLD to more severe stages, including NASH and fibrosis [59]. During NAFLD, hepatic Kupffer cells are activated, leading to the massive release of pro-inflammatory mediators such as tumor necrosis factor alpha (TNF-α), IL-1β, and IL-6, which affects the normal function of hepatocytes [60]. Studies have shown that exercise training improves the hepatic inflammatory response by downregulating levels of pro-inflammatory mediators [55]. Exercise training also inhibits the activation of the NOD-like receptor thermal protein domain associated protein 3 (NLRP3) inflammasome in mice, leading to an improvement in NASH [61]. Voluntary exercise has been shown to improve fatty acid and glucose homeostasis and reduce macrophage-related hepatic inflammatory responses in NAFLD mice [62]. In addition, high-intensity exercise is reportedly more effective than low-intensity exercise in improving liver inflammation and NASH progression [63].

Exercise promotes organ crosstalk (Fig. 3). Skeletal muscle is one of the main effector organs in response to movement and is also recognized as an endocrine organ [64]. During exercise, skeletal muscle is able to release exercise-induced proteins, peptides and cytokines that exert autocrine, paracrine, and endocrine effects, collectively known as myokines, to enable crosstalk with other organs. Interestingly, other tissues such as the liver and adipose also respond to exercise by releasing hepatokines and adipokines; myokines, hepatokines and adipokines released during exercise are collectively known as exerkines [65].

Fig. 3.

Organ Crosstalk during exercise. During exercise, Myokines, Hepatokines, and Adipokines released from skeletal muscle, liver, and adipose tissue interact through blood circulation. Abbreviations: SeP, selenoprotein P; FGF21, fibroblast growth factor 21; ANGPTL4, angiopoietin-like 4; IL-6, interleukin 6; IL-15, interleukin 15; BAIBA, beta-aminoisobutyric acid; TGF-β2, transforming growth factor beta 2. Fig. 3 created with BioRender.com.

The exerkines released during exercise may be an important pathway to organ crosstalk. For example, FGF21 is thought to be an exerkine that improves glucolipid metabolism, reduces hepatic steatosis, and improves insulin sensitivity [66]. Although FGF21 is less expressed in skeletal muscle under normal conditions, during exercise, FGF21 may be released by skeletal muscle as a myokine and enter the circulation to interact with other organs such as the liver and adipose tissue [64]. Therefore, organ crosstalk may be one of the mechanisms by which exercise improves NAFLD.

4. The Role of FGF21 in NAFLD
4.1 FGF Family

The FGF family is a group of growth factors closely related to cell growth and differentiation, proliferation, wound healing and repair, angiogenesis, embryonic development and other processes. Currently, 23 members of the FGF family have been identified, mainly including endocrine and paracrine types. The FGF family has seven subfamilies. The paracrine FGF1 subfamily, comprising FGF1 and FGF2, play important roles in tissue and organ formation, neural cell regeneration and angiogenesis [67]. The paracrine FGF4 subfamily, comprising FGF4, FGF5, and FGF6, regulates cell proliferation and differentiation, the hair follicle growth cycle, and muscle repair and myocardial function [68]. The paracrine FGF7 subfamily, comprising FGF3, FGF7, FGF10, and FGF22, promotes bone formation and restores trauma repair processes [69, 70]. The paracrine FGF8 subfamily, comprising FGF8, FGF17, and FGF18, is involved in tissue and organ formation, embryonic development, and skeletal developmental processes [71, 72]. The paracrine FGF9 subfamily, comprising FGF9, FGF16, and FGF20, regulates white fat browning and cardiac function [73, 74]. The intracellular FGF11 subfamily, comprising FGF11, FGF12, FGF13, and FGF14, regulates bone resorption and synaptic plasticity [75, 76]. The endocrine FGF15/19 subfamily, comprising FGF15/19, FGF21, FGF23, regulates bile acid homeostasis and glucolipid metabolism, and maintenance of homeostasis [77].

4.2 FGF21 Overview

FGF21 is an effective regulator of glucose and lipid metabolism, exerting its biological effects through FGF receptors (FGFRs) and β-klotho [78]. FGF21 is normally expressed in a variety of tissues, including liver, adipose tissue, skeletal muscle, and pancreas, in response to different physiological states such as fasting, feeding, exercise, and cold exposure [13]. Liver-derived FGF21 has been widely studied and several signaling mechanisms regulating FGF21 expression in the liver have been described. Fasting increases the level of FGF21 in the blood of rodents, primates, and humans, which is mainly regulated by PPARα, and the liver is the main source of FGF21 during fasting [79]. High-sugar consumption is able to increase hepatic FGF21 mRNA expression and blood levels of FGF21, mainly dependent on carbohydrate response element-binding protein [80]. Furthermore, evidence suggests that hepatocytes increase FGF21 expression and secretion in an activating transcription factor 4 (ATF4)-dependent way in response to ER stress [81]. The brain [82], adipose tissue [83], heart [13] and the liver itself [84] are the main effector organs for hepatic FGF21.

Adipose tissue, skeletal muscle, pancreas, and the heart are also capable of secreting FGF21 [13]. Nutritional excess [85] and cold exposure [86] lead to the increased expression of FGF21 in adipose tissue, which acts in association with PPARγ [87], uncoupling protein 1 (UCP-1) [88], and ATF2 [89], mainly in an autocrine manner [90]. During exercise, increased FGF21 expression is associated with skeletal muscle [91]. Although most studies suggest that the exercise-induced increase in FGF21 is mainly of hepatic origin, emerging evidence suggests that skeletal muscle may also be a source of FGF21 in the circulation during exercise and is regulated by AMPK [92] and protein kinase B (PKB/AKT) [93]. In addition, FGF21 secretion in the pancreas is regulated by phospholipase C gamma [94], whereas in the heart it is associated with extracellular signal-regulated kinase 1/2 [95].

4.3 FGF21 Actions in Target Organs

FGF21 functions in complex with FGFRs and β-klotho. FGFRs, which include FGFR1, FGFR2, FGFR3, and FGFR4 are transmembrane proteins. FGF21 is required to increase its affinity to the receptor in the presence of β-klotho [96]. FGFRs are widely expressed in multiple organs, and β-klotho is predominantly expressed in adipose tissue, brain, the liver, and pancreas. Thus, the selective expression of β-klotho in tissues determines the tissue specificity of FGF21 to exert its biological functions [97]. Circulating FGF21 is able to cross the blood-brain barrier and act on the central nervous system to regulate water and alcohol preference and food intake, reduce energy expenditure, and adjust circadian rhythms via the hypothalamic-pituitary-adrenal axis [98, 99]. In addition, FGF21 increases insulin sensitivity and reduces lipid accumulation in the liver and skeletal muscle [100]. Also, FGF21 decreases the inflammatory response in the heart, liver, kidney and pancreas; enhances the survival of pancreatic islet β-cells; and reduces cardiomyocyte apoptosis and renal fibrosis [13, 101, 102]. Furthermore, FGF21 increases lipocalin production in white adipose tissue, decreases lipolysis, and promotes browning [83]. Meanwhile, FGF21 increases insulin sensitivity in brown adipose tissue, increases glucose uptake, and enhances thermogenesis [66] (Fig. 4).

Fig. 4.

Metabolic effects of FGF21 on different types of tissues. Abbreviations: FGF21, fibroblast growth factor 21; BAT, brown adipose tissue; WAT, white adipose tissue. Fig. 4 created with BioRender.com.

4.4 FGF21 as a Biomarker of NAFLD

FGF21 levels are elevated in the circulation of NAFLD patients [103], and the same phenomenon has been observed in rodents [104]. Therefore, the notion of “FGF21 resistance” [105] was proposed to explain this change, similar to IR [106], as a compensatory change in organisms. High serum FGF21 are highly correlated with the development of NAFLD [107]. Increased serum FGF21 levels are positively correlated with the degree of hepatic steatosis, as well as serum TG and insulin levels in humans [108], and are negatively associated with high density lipoprotein and the body mass index [109]. In a prospective cohort study of 808 subjects over a 3-year period, elevated serum FGF21 levels were found to predict the development of hepatic steatosis [110]. Another study showed that serum FGF21 level was closely related to hepatic lipid content in a dose-dependent manner in NAFLD patients with central obesity [111]. In addition, it is thought that a reduction in serum FGF21 levels due to gastric bypass surgery may lead to an improvement in NAFLD [112]. In addition, some analogues of FGF21 such as BFKB8488A [113] and efruxifermin [114] have emerged as drug candidates for NAFLD and NASH and entered clinical trial studies. Therefore, FGF21 may serve as a biological marker of NAFLD.

4.5 FGF21 in the Development of NAFLD

FGF21 reduces steatosis in the development of NAFLD. Hepatic steatosis is the accumulation of excess TGs in hepatocytes, which is one of the main characteristics of NAFLD [115]. Although the excessive accumulation of TGs in hepatocytes has no direct side effects, its continuous release of intermediates, such as fatty acids and ceramides, has toxic effects on the liver [116, 117]. In addition, these intermediates are able to destabilize lysosomes and enhance the inflammatory response and ER stress, thus causing tissue damage [118, 119]. Therefore, hepatic steatosis is an important factor in the development of NAFLD. The role of FGF21 in improving hepatic steatosis in NAFLD has been reported in several studies: First, FGF21 inhibits lipolysis and prevents the flow of excess FFAs into the liver, thus reducing fat accumulation in the liver [120, 121]. Second, mammalian target of rapamycin (mTOR) plays an important role in lipid metabolism and FGF21 is able to inhibit mTOR expression in the liver thereby exerting hypolipidemic effects [122]. In addition, FGF21 inhibits SREBP1c transcription [123] and increases insulin sensitivity [124] to exert lipid regulation.

FGF21 ameliorates oxidative stress in the development of NAFLD. Oxidative stress is a state of imbalance between oxidation and antioxidation in the body, leading to the high production of ROS, which in turn have harmful effects on the organism [53]. Oxidative stress is related to the pathophysiology of metabolic diseases such as NAFLD. For example, excess ROS leads to disorders of hepatic lipid metabolism and IR, triggering liver fibrosis and hepatitis [53, 54]. Elevated ROS levels disrupt ER homeostasis and activate the unfolding protein response signal transduction pathway, which increases FGF21 expression through various pathways including inositol-requiring enzyme 1 alpha/X box binding protein 1 [125], ATF4 [126] and CCAAT enhancer binding protein homologous protein [127]. FGF21 is an important protective factor against oxidative stress in vivo, and is capable of activating a variety of signals such as Nrf2 [128] and AMPK [129]. Exogenous FGF21 reduces ROS production in HepG2 HCC cells [130], and FGF21 analogs alleviate hepatic steatosis, hepatitis, and liver fibrosis in patients with NASH [18]. Therefore, FGF21 is considered an effective regulator of NAFLD [11].

FGF21 ameliorates inflammatory responses in the development of NAFLD. Inflammation is an immune protective reaction of the organism in response to various endogenous or exogenous stimuli; moderate inflammation is beneficial to the organism, while excessive inflammation is harmful [131]. The pathogenesis of NAFLD is associated with a chronic inflammatory state. Sustained upregulation of signals including Toll-like receptor 4 (TLR4) [132], nuclear factor kappa-B (NF-κB) [133], and NLRP3 [134] activates hepatic Kupffer cells and elevates inflammatory factor concentrations in the liver, resulting in hepatocyte injury and apoptosis, ultimately leading to loss of liver function [60]. FGF21 exerts direct anti-inflammatory effects on NALFD and other models of inflammatory diseases [135]. FGF21 suppresses macrophage-mediated inflammation in RAW 264.7 cells through activation of Nrf2 and inhibition of the NF-κB signaling pathway [136]. Exogenous FGF21 analogs reduce inflammatory cytokine levels in NAFLD mice [137]. Similarly, administration of FGF21 analogs to ob/ob mice reduce c-Jun N-terminal kinase enzyme 1/2 and NF-κB signaling in the liver, inhibit TNF-α expression, and enhance hepatic mitochondrial function [138]. By contrast, a lack of FGF21 results in the significant upregulation of TLR4 and NF-κB signaling in the liver of NASH mice [139]. In addition, FGF21 inhibits NLRP3 inflammasome-mediated cell pyroptosis to improve atherosclerosis in mice [140].

5. The Role of FGF21 in the Improvement of NAFLD by Exercise
5.1 Effects of Exercise on FGF21

Studies have shown that different exercise intensities have different effects on serum levels of FGF21. Experiments in 10 healthy men with moderate-intensity exercise (55% maximal oxygen uptake) and high-intensity exercise (75% maximal oxygen uptake) found that both exercise intensities increased their serum FGF21 levels, and that exercise-induced changes in FGF21 were exercise-intensity dependent, with high-intensity exercise providing greater metabolic benefits than low-intensity exercise [141]. FGF21 was also found to be regulated by glucagon. High-intensity exercise has a greater effect on glucagon compared to moderate intensity; therefore FGF21 changes more significantly during high-intensity exercise [141]. In addition, there are significant individualized features of the effect of exercise on serum FGF21. He et al. [142] showed that either HIIT or resistance training increased serum FGF21 levels in untrained male subjects. However, in subjects with moderate levels of training, serum FGF21 levels did not significantly increase even with greater intensity training [143]. Different views have also been reported. Takahashi et al. [144] reported that 12 weeks of resistance exercise reduced serum FGF21 levels in NAFLD patients. Motahari et al. [145] found no significant change in serum FGF21 levels in male T2DM patients with concurrent exercise.

It was also found that long-term metabolic dysregulation such as obesity can cause a decrease in expression of the FGF21/FGFRs/β-klotho receptor complex, leading to FGF21 resistance and thus a compensatory increase in circulating levels of FGF21 [105]. Although the effect of exercise on FGF21 stimulation was significantly diminished in the FGF21-resistant state, exercise restored FGF21 sensitivity by increasing the expression of FGF21/FGFRs/β-klotho receptor complexes [146]. Fletcher et al. [147] reported that while 36 weeks of exercise intervention decreased FGF21 mRNA expression in rat liver, its receptor FGFR2 mRNA transcript levels were significantly upregulated, suggesting that exercise increases FGF21 sensitivity by increasing FGF21 receptor expression to improve FGF21 resistance status. It has also been reported [146] that 12 weeks of exercise intervention increased the mRNA expression of β-klotho, FGFR1, and FGFR2 in adipose tissue. This study also showed that even a 3-day exercise intervention was able to significantly increase the expression of β-klotho and FGFR1 in the adipose tissue of obese mice, suggesting that exercise positively enhances the actual biological effects of FGF21 on mice.

5.2 Possible Mechanism by which Exercise Targets FGF21 to Improve NAFLD

As an important cytokine in vivo, FGF21 is secreted by multiple organs and is able to enter the circulation to exert multi-organ interactions [13]. At the same time, FGF21 is an exerkine that responds to exercise [9]. Therefore, it is highly likely that one mechanism of action by which exercise improves NAFLD is by stimulating the secretion of FGF21 from multiple organs, acting on the liver in an autocrine, endocrine, and paracrine manners.

During exercise, organs such as the liver and skeletal muscle are able to secrete FGF21 and enter the bloodstream for action. In skeletal muscle, exercise induces the sustained activation of AMPK [92] and AKT [93] signaling, significantly upregulates FGF21 expression in skeletal muscle, and induces the secretion of myogenic FGF21. In the liver, exercise stimulates a large increase in FFAs and depletion of hepatic glycogen, which induces the large secretion of hepatogenic FGF21 via PPARα [148] and ATF4 [149]. At the same time, exercise-induced upregulation of glucagon and decrease in insulin levels can enable the pancreas to secrete FGF21 into the bloodstream [150]. In addition, it has also been suggested that activation of cardiac SIRT1 pathway during exercise causes cardiogenic FGF21 secretion [151] (Fig. 5).

Fig. 5.

The mechanism of action of FGF21 in the improvement of NAFLD by exercise. FGF21 is secreted by organs such as skeletal muscle and pancreas during exercise and acts on the liver through the blood circulation. At the same time, the liver itself is able to secrete FGF21 during exercise, acting in an autocrine, paracrine or endocrine manner. Abbreviations: FGFR, fibroblast growth factor receptor; ER, endoplasmic reticulum; AMPK, AMP-activated protein kinase; PKB/AKT, protein kinase B; Nrf2, nuclear factor erythroid 2-related factor 2; SOD, superoxide dismutase; GSH, glutathione; HO-1, heme oxygenase 1; NF-κB, nuclear factor kappa-B; TNF-α, tumor necrosis factor alpha; NLRP3, NOD-like receptor thermal protein domain associated protein 3; IL-1β, interleukin 1 beta; PGC1-α, peroxisome proliferator-activated receptor γ coactivator l alpha; PI3K, phosphoinositide 3-kinase; GSK3β, glycogen synthase kinase-3 beta; GS, glycogen synthase; ATF4, activating transcription factor 4; PPARα, peroxisome proliferator-activated receptor alpha. Fig. 5 created with BioRender.com.

FGF21 secreted during exercise is able to act on the liver through blood circulation. First, FGF21 entering the circulation can activate Nrf2; upregulate the levels of antioxidant factors such as SOD, glutathione (GSH) and HO-1; and reduce the excessive accumulation of ROS, thus decreasing the level of oxidative stress in the liver [152]. Second, these cytokines suppress over-activation of NFκB signaling pathway; reduce the levels of pro-inflammatory cytokines such as TNF-α, NLRP3, and IL-1β; and inhibit the liver inflammatory response [136, 140]. Third, circulating FGF21 is able to inhibit apoptosis and NFκB-mediated inflammatory responses through AMPK signaling pathway on the one hand [153], and enhance liver mitochondrial function through activation of PPAR gamma coactivator l alpha on the other [154]. In addition, the secreted FGF21 promotes hepatic glycogen synthesis and inhibits IR through phosphoinositide 3-kinase/AKT signaling [155], and is also able to inhibit hepatic ER stress [156]. In parallel, circulating FGF21 also acts on other organs to alleviate NAFLD, such as skeletal muscle, where it increases mitochondrial activity via AMPK to improve skeletal muscle function [157]; and adipose tissue, where it regulates lipolysis via UCP-1 to inhibit ectopic fat deposition and induce browning [158].

6. Conclusions and Future Perspectives

Exercise interventions are increasingly being recognized as an important non-pharmacological treatment for the prevention and improvement of NAFLD. FGF21, one of the important indicators for improving NAFLD, serves as an exerkine in response to exercise, and multiple exercise modalities have beneficial effects on FGF21. FGF21 is increasingly becoming a crucial target in the development of exercise-improved NAFLD.

FGF21 is an important metabolic regulator that plays an important role in improving glucolipid metabolism, IR, inflammation, and oxidative stress. Changes in serum FGF21 are positively correlated with the development of several metabolic diseases and have become a predictor and biological marker for the early diagnosis of diseases such as NAFLD, T2DM and obesity. Various FGF21 analogs such as LY2405319 [159] and PF-05231023 [160] have entered phase I clinical trials. Although these FGF21 analogs have not achieved great results in glucose metabolism, they have had beneficial effects on improving lipid metabolism in patients with NAFLD and T2DM. Meanwhile, FGF21 is an exerkine that responds to exercise and enables crosstalk between multiple organs through autocrine, endocrine and paracrine approaches. Exercise is also able to improve FGF21 resistance status under pathological conditions. The combination of FGF21 analogs with exercise intervention may bring new hope for the improvement of metabolic diseases such as NAFLD and T2DM. It is worth noting that the short half-life, poor stability and solubility, as well as some potential side effects such as promotion of bone loss in mice [161] and effects on female reproduction [162], have led to concerns about the clinical application of FGF21-based therapies. Meanwhile, a number of drugs that have been approved for the treatment of T2DM have similarly shown benefit in ameliorating NAFLD and may pose a challenge to FGF21 as a therapeutic target for NAFLD. Despite these concerns, FGF21 still has significant potential as a powerful target for improving NAFLD. Thus, more in-depth studies are needed to elucidate the physiological and pharmacological roles of FGF21 and to provide new insights into the development of FGF21 as a therapeutic agent for NAFLD.

Last but not least, prevention rather than cure of NAFLD should be the common goal. How to encourage more patients to participate in exercise should be a key consideration for policy makers. Future research should focus on the precise molecular mechanisms by which FGF21 plays a multi-organ role during exercise, rather than just studying the actions of a single organ. A comprehensive understanding of the role of FGF21 in NAFLD and its changes during exercise are needed to provide novel targets for the establishment of exercise intervention strategies for patients with NAFLD.

Abbreviations

NAFLD, Non-alcoholic fatty liver disease; FGF21, Fibroblast growth factor 21; FGF, fibroblast growth factor; T2DM, type 2 diabetes mellitus; NASH, non-alcoholic steatohepatitis; AST, aspartate aminotransferase; ALT, alanine aminotransferase; FFA, free fatty acids; TGs, triglycerides; HIIT, High-intensity Interval Training; FGFR, fibroblast growth factor receptor; ER, endoplasmic reticulum; AMPK, AMP-activated protein kinase; PKB/AKT, protein kinase B; Nrf2, nuclear factor erythroid 2-related factor 2; SOD, superoxide dismutase; GSH, glutathione; HO-1, heme oxygenase 1; NF-κB, nuclear factor kappa-B; TNF-α, tumor necrosis factor alpha; NLRP3, NOD-like receptor thermal protein domain associated protein 3; IL-1β, interleukin 1 beta; PGC1-α, peroxisome proliferator-activated receptor γ coactivator l alpha; PI3K, phosphoinositide 3-kinase; GSK3β, glycogen synthase kinase-3 beta; GS, glycogen synthase; ATF4, activating transcription factor 4; PPARα, peroxisome proliferator-activated receptor alpha.

Author Contributions

ZW designed the research study. ZW, TS, JY, and SL performed the research. ZW, LG and YZ provided help and advice on conceptualization, data curation, methodology and formal analysis. All authors contributed to editorial changes in the manuscript. All authors read and approved the final manuscript.

Ethics Approval and Consent to Participate

Not applicable.

Acknowledgment

We thank the authors of the studies included in this review, and thanks to the biorender for the resources, all figures were created with https://www.biorender.com.

Funding

This research was funded by Special Fund for Basic Scientific Research of the Central Universities (grant number 20221016) and the Independent research project of Beijing Sport University (grant number 2021TD012).

Conflict of Interest

The authors declare no conflict of interest.

References
[1]
Younossi Z, Anstee QM, Marietti M, Hardy T, Henry L, Eslam M, et al. Global burden of NAFLD and NASH: trends, predictions, risk factors and prevention. Nature Reviews. Gastroenterology & Hepatology. 2018; 15: 11–20.
[2]
Yang L, Zhou MT, Lu XC, Zhu HY, Han HM. Research Progress on the Mechanism of Vitamin D3 in the Occurrence and Development of Nonalcoholic Fatty Liver Disease. Journal of Biological Regulators and Homeostatic Agents. 2022; 36: 797–805.
[3]
Raza S, Rajak S, Upadhyay A, Tewari A, Anthony Sinha R. Current treatment paradigms and emerging therapies for NAFLD/NASH. Frontiers in Bioscience (Landmark Edition). 2021; 26: 206–237.
[4]
Zhang X, Goh GBB, Chan WK, Wong GLH, Fan JG, Seto WK, et al. Unhealthy lifestyle habits and physical inactivity among Asian patients with non-alcoholic fatty liver disease. Liver International: Official Journal of the International Association for the Study of the Liver. 2020; 40: 2719–2731.
[5]
Neuschwander-Tetri BA. Therapeutic Landscape for NAFLD in 2020. Gastroenterology. 2020; 158: 1984–1998.e3.
[6]
Romero FA, Jones CT, Xu Y, Fenaux M, Halcomb RL. The Race to Bash NASH: Emerging Targets and Drug Development in a Complex Liver Disease. Journal of Medicinal Chemistry. 2020; 63: 5031–5073.
[7]
Younossi ZM, Loomba R, Rinella ME, Bugianesi E, Marchesini G, Neuschwander-Tetri BA, et al. Current and future therapeutic regimens for nonalcoholic fatty liver disease and nonalcoholic steatohepatitis. Hepatology (Baltimore, Md.). 2018; 68: 361–371.
[8]
Booth FW, Roberts CK, Thyfault JP, Ruegsegger GN, Toedebusch RG. Role of Inactivity in Chronic Diseases: Evolutionary Insight and Pathophysiological Mechanisms. Physiological Reviews. 2017; 97: 1351–1402.
[9]
Thyfault JP, Bergouignan A. Exercise and metabolic health: beyond skeletal muscle. Diabetologia. 2020; 63: 1464–1474.
[10]
Sargeant JA, Gray LJ, Bodicoat DH, Willis SA, Stensel DJ, Nimmo MA, et al. The effect of exercise training on intrahepatic triglyceride and hepatic insulin sensitivity: a systematic review and meta-analysis. Obesity Reviews: an Official Journal of the International Association for the Study of Obesity. 2018; 19: 1446–1459.
[11]
Falamarzi K, Malekpour M, Tafti MF, Azarpira N, Behboodi M, Zarei M. The role of FGF21 and its analogs on liver associated diseases. Frontiers in Medicine. 2022; 9: 967375.
[12]
Lin X, Liu YB, Hu H. Metabolic role of fibroblast growth factor 21 in liver, adipose and nervous system tissues. Biomedical Reports. 2017; 6: 495–502.
[13]
Spann RA, Morrison CD, den Hartigh LJ. The Nuanced Metabolic Functions of Endogenous FGF21 Depend on the Nature of the Stimulus, Tissue Source, and Experimental Model. Frontiers in Endocrinology. 2022; 12: 802541.
[14]
Gómez-Sámano MÁ, Grajales-Gómez M, Zuarth-Vázquez JM, Navarro-Flores MF, Martínez-Saavedra M, Juárez-León ÓA, et al. Fibroblast growth factor 21 and its novel association with oxidative stress. Redox Biology. 2017; 11: 335–341.
[15]
Yu Y, Shen Y, Zhang S, Wang N, Luo L, Zhu X, et al. Suppression of Cutibacterium acnes-Mediated Inflammatory Reactions by Fibroblast Growth Factor 21 in Skin. International Journal of Molecular Sciences. 2022; 23: 3589.
[16]
Lee KM, Hwang YJ, Jung GS. Gemigliptin exerts protective effects against doxorubicin-induced hepatotoxicity by inhibiting apoptosis via the regulation of fibroblast growth factor 21 expression. Biochemical and Biophysical Research Communications. 2022; 626: 135–141.
[17]
Shimizu M, Sato R. Endocrine Fibroblast Growth Factors in Relation to Stress Signaling. Cells. 2022; 11: 505.
[18]
Jin L, Yang R, Geng L, Xu A. Fibroblast Growth Factor-Based Pharmacotherapies for the Treatment of Obesity-Related Metabolic Complications. Annual Review of Pharmacology and Toxicology. 2023; 63: 359–382.
[19]
Kozłowska-Flis M, Rodziewicz-Flis E, Micielska K, Kortas J, Jaworska J, Borkowska A, et al. Short and long-term effects of high-intensity interval training applied alone or with whole-body cryostimulation on glucose homeostasis and myokine levels in overweight to obese subjects. Frontiers in Bioscience-Landmark. 2021; 26: 1132–1146.
[20]
Guo C, Zhao L, Li Y, Deng X, Yuan G. Relationship between FGF21 and drug or nondrug therapy of type 2 diabetes mellitus. Journal of Cellular Physiology. 2021; 236: 55–67.
[21]
Talukdar S, Kharitonenkov A. FGF19 and FGF21: In NASH we trust. Molecular Metabolism. 2021; 46: 101152.
[22]
Kabarra K, Golabi P, Younossi ZM. Nonalcoholic steatohepatitis: global impact and clinical consequences. Endocrine Connections. 2021; 10: R240–R247.
[23]
Roeb E. Non-alcoholic fatty liver diseases: current challenges and future directions. Annals of Translational Medicine. 2021; 9: 726.
[24]
Ahmed IA, Mikail MA, Mustafa MR, Ibrahim M, Othman R. Lifestyle interventions for non-alcoholic fatty liver disease. Saudi Journal of Biological Sciences. 2019; 26: 1519–1524.
[25]
Rinaldi L, Giorgione C, Mormone A, Esposito F, Rinaldi M, Berretta M, et al. Non-Invasive Measurement of Hepatic Fibrosis by Transient Elastography: A Narrative Review. Viruses. 2023; 15: 1730.
[26]
Bedogni G, Bellentani S, Miglioli L, Masutti F, Passalacqua M, Castiglione A, et al. The Fatty Liver Index: a simple and accurate predictor of hepatic steatosis in the general population. BMC Gastroenterology. 2006; 6: 33.
[27]
Lee JH, Kim D, Kim HJ, Lee CH, Yang JI, Kim W, et al. Hepatic steatosis index: a simple screening tool reflecting nonalcoholic fatty liver disease. Digestive and Liver Disease: Official Journal of the Italian Society of Gastroenterology and the Italian Association for the Study of the Liver. 2010; 42: 503–508.
[28]
Park H, Yoon EL, Ito T, Jo AJ, Kim M, Lee J, et al. Diagnostic Performance of the Fibrosis-4 Index and Nonalcoholic Fatty Liver Disease Fibrosis Score in Lean Adults With Nonalcoholic Fatty Liver Disease. JAMA Network Open. 2023; 6: e2329568.
[29]
Cho YK, Kim M, Kim YJ, Jung CH, Lee WJ, Park JY. Predictive value of the Framingham steatosis index for cardiovascular risk: a nationwide population-based cohort study. Frontiers in Cardiovascular Medicine. 2023; 10: 1163052.
[30]
Kotronen A, Peltonen M, Hakkarainen A, Sevastianova K, Bergholm R, Johansson LM, et al. Prediction of non-alcoholic fatty liver disease and liver fat using metabolic and genetic factors. Gastroenterology. 2009; 137: 865–872.
[31]
Zhou YJ, Zhou YF, Zheng JN, Liu WY, Van Poucke S, Zou TT, et al. NAFL screening score: A basic score identifying ultrasound-diagnosed non-alcoholic fatty liver. Clinica Chimica Acta; International Journal of Clinical Chemistry. 2017; 475: 44–50.
[32]
Perdomo CM, Frühbeck G, Escalada J. Impact of Nutritional Changes on Nonalcoholic Fatty Liver Disease. Nutrients. 2019; 11: 677.
[33]
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. Molecular Metabolism. 2020; 32: 1–14.
[34]
Buzzetti E, Pinzani M, Tsochatzis EA. The multiple-hit pathogenesis of non-alcoholic fatty liver disease (NAFLD). Metabolism: Clinical and Experimental. 2016; 65: 1038–1048.
[35]
Balducci S, Sacchetti M, Haxhi J, Orlando G, D’Errico V, Fallucca S, et al. Physical exercise as therapy for type 2 diabetes mellitus. Diabetes/metabolism Research and Reviews. 2014; 30: 13–23.
[36]
Pedersen BK, Saltin B. Exercise as medicine - evidence for prescribing exercise as therapy in 26 different chronic diseases. Scandinavian Journal of Medicine & Science in Sports. 2015; 25: 1–72.
[37]
Ratziu V, Ghabril M, Romero-Gomez M, Svegliati-Baroni G. Recommendations for Management and Treatment of Nonalcoholic Steatohepatitis. Transplantation. 2019; 103: 28–38.
[38]
Xiong Y, Peng Q, Cao C, Xu Z, Zhang B. Effect of Different Exercise Methods on Non-Alcoholic Fatty Liver Disease: A Meta-Analysis and Meta-Regression. International Journal of Environmental Research and Public Health. 2021; 18: 3242.
[39]
Babu AF, Csader S, Lok J, Gómez-Gallego C, Hanhineva K, El-Nezami H, et al. Positive Effects of Exercise Intervention without Weight Loss and Dietary Changes in NAFLD-Related Clinical Parameters: A Systematic Review and Meta-Analysis. Nutrients. 2021; 13: 3135.
[40]
Sullivan S, Kirk EP, Mittendorfer B, Patterson BW, Klein S. Randomized trial of exercise effect on intrahepatic triglyceride content and lipid kinetics in nonalcoholic fatty liver disease. Hepatology (Baltimore, Md.). 2012; 55: 1738–1745.
[41]
Johnson NA, Sachinwalla T, Walton DW, Smith K, Armstrong A, Thompson MW, et al. Aerobic exercise training reduces hepatic and visceral lipids in obese individuals without weight loss. Hepatology (Baltimore, Md.). 2009; 50: 1105–1112.
[42]
Oh S, So R, Shida T, Matsuo T, Kim B, Akiyama K, et al. High-Intensity Aerobic Exercise Improves Both Hepatic Fat Content and Stiffness in Sedentary Obese Men with Nonalcoholic Fatty Liver Disease. Scientific Reports. 2017; 7: 43029.
[43]
Hallsworth K, Adams LA. Lifestyle modification in NAFLD/NASH: Facts and figures. JHEP Reports: Innovation in Hepatology. 2019; 1: 468–479.
[44]
Hallsworth K, Fattakhova G, Hollingsworth KG, Thoma C, Moore S, Taylor R, et al. Resistance exercise reduces liver fat and its mediators in non-alcoholic fatty liver disease independent of weight loss. Gut. 2011; 60: 1278–1283.
[45]
Babu AF, Csader S, Männistö V, Tauriainen MM, Pentikäinen H, Savonen K, et al. Effects of exercise on NAFLD using non-targeted metabolomics in adipose tissue, plasma, urine, and stool. Scientific Reports. 2022; 12: 6485.
[46]
Zheng F, Cai Y. Concurrent exercise improves insulin resistance and nonalcoholic fatty liver disease by upregulating PPAR-γ and genes involved in the beta-oxidation of fatty acids in ApoE-KO mice fed a high-fat diet. Lipids in Health and Disease. 2019; 18: 6.
[47]
Cigrovski Berkovic M, Bilic-Curcic I, Mrzljak A, Cigrovski V. NAFLD and Physical Exercise: Ready, Steady, Go!. Frontiers in Nutrition. 2021; 8: 734859.
[48]
Lavoie JM, Gauthier MS. Regulation of fat metabolism in the liver: link to non-alcoholic hepatic steatosis and impact of physical exercise. Cellular and Molecular Life Sciences: CMLS. 2006; 63: 1393–1409.
[49]
Diniz TA, de Lima Junior EA, Teixeira AA, Biondo LA, da Rocha LAF, Valadão IC, et al. Aerobic training improves NAFLD markers and insulin resistance through AMPK-PPAR-α signaling in obese mice. Life Sciences. 2021; 266: 118868.
[50]
Houttu V, Boulund U, Grefhorst A, Soeters MR, Pinto-Sietsma SJ, Nieuwdorp M, et al. The role of the gut microbiome and exercise in non-alcoholic fatty liver disease. Therapeutic Advances in Gastroenterology. 2020; 13: 1756284820941745.
[51]
Li H, Dun Y, Zhang W, You B, Liu Y, Fu S, et al. Exercise improves lipid droplet metabolism disorder through activation of AMPK-mediated lipophagy in NAFLD. Life Sciences. 2021; 273: 119314.
[52]
Bae JY. Resistance Exercise Regulates Hepatic Lipolytic Factors as Effective as Aerobic Exercise in Obese Mice. International Journal of Environmental Research and Public Health. 2020; 17: 8307.
[53]
Chen Z, Tian R, She Z, Cai J, Li H. Role of oxidative stress in the pathogenesis of nonalcoholic fatty liver disease. Free Radical Biology & Medicine. 2020; 152: 116–141.
[54]
Clare K, Dillon JF, Brennan PN. Reactive Oxygen Species and Oxidative Stress in the Pathogenesis of MAFLD. Journal of Clinical and Translational Hepatology. 2022; 10: 939–946.
[55]
Farzanegi P, Dana A, Ebrahimpoor Z, Asadi M, Azarbayjani MA. Mechanisms of beneficial effects of exercise training on non-alcoholic fatty liver disease (NAFLD): Roles of oxidative stress and inflammation. European Journal of Sport Science. 2019; 19: 994–1003.
[56]
Salminen A, Vihko V. Lipid peroxidation in exercise myopathy. Experimental and Molecular Pathology. 1983; 38: 380–388.
[57]
Zhang Y, Liu Y, Liu X, Yuan X, Xiang M, Liu J, et al. Exercise and Metformin Intervention Prevents Lipotoxicity-Induced Hepatocyte Apoptosis by Alleviating Oxidative and ER Stress and Activating the AMPK/Nrf2/HO-1 Signaling Pathway in db/db Mice. Oxidative Medicine and Cellular Longevity. 2022; 2022: 2297268.
[58]
Zou Y, Chen Z, Sun C, Yang D, Zhou Z, Peng X, et al. Exercise Intervention Mitigates Pathological Liver Changes in NAFLD Zebrafish by Activating SIRT1/AMPK/NRF2 Signaling. International Journal of Molecular Sciences. 2021; 22: 10940.
[59]
Parthasarathy G, Revelo X, Malhi H. Pathogenesis of Nonalcoholic Steatohepatitis: An Overview. Hepatology Communications. 2020; 4: 478–492.
[60]
Diehl AM, Day C. Cause, Pathogenesis, and Treatment of Nonalcoholic Steatohepatitis. The New England Journal of Medicine. 2017; 377: 2063–2072.
[61]
Yang W, Liu L, Wei Y, Fang C, Liu S, Zhou F, et al. Exercise suppresses NLRP3 inflammasome activation in mice with diet-induced NASH: a plausible role of adropin. Laboratory Investigation; a Journal of Technical Methods and Pathology. 2021; 101: 369–380.
[62]
Gehrke N, Biedenbach J, Huber Y, Straub BK, Galle PR, Simon P, et al. 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. Scientific Reports. 2019; 9: 4007.
[63]
Fredrickson G, Barrow F, Dietsche K, Parthiban P, Khan S, Robert S, et al. Exercise of high intensity ameliorates hepatic inflammation and the progression of NASH. Molecular Metabolism. 2021; 53: 101270.
[64]
Sabaratnam R, Wojtaszewski JFP, Højlund K. Factors mediating exercise-induced organ crosstalk. Acta Physiologica (Oxford, England). 2022; 234: e13766.
[65]
Safdar A, Saleem A, Tarnopolsky MA. The potential of endurance exercise-derived exosomes to treat metabolic diseases. Nature Reviews. Endocrinology. 2016; 12: 504–517.
[66]
Geng L, Lam KSL, Xu A. The therapeutic potential of FGF21 in metabolic diseases: from bench to clinic. Nature Reviews. Endocrinology. 2020; 16: 654–667.
[67]
Suh JM, Jonker JW, Ahmadian M, Goetz R, Lackey D, Osborn O, et al. Endocrinization of FGF1 produces a neomorphic and potent insulin sensitizer. Nature. 2014; 513: 436–439.
[68]
Kosaka N, Sakamoto H, Terada M, Ochiya T. Pleiotropic function of FGF-4: its role in development and stem cells. Developmental Dynamics: an Official Publication of the American Association of Anatomists. 2009; 238: 265–276.
[69]
Poudel SB, Bhattarai G, Kim JH, Kook SH, Seo YK, Jeon YM, et al. Local delivery of recombinant human FGF7 enhances bone formation in rat mandible defects. Journal of Bone and Mineral Metabolism. 2017; 35: 485–496.
[70]
Prochazkova M, Prochazka J, Marangoni P, Klein OD. Bones, Glands, Ears and More: The Multiple Roles of FGF10 in Craniofacial Development. Frontiers in Genetics. 2018; 9: 542.
[71]
Atsuta Y, Takahashi Y. FGF8 coordinates tissue elongation and cell epithelialization during early kidney tubulogenesis. Development (Cambridge, England). 2015; 142: 2329–2337.
[72]
Maruoka Y, Ohbayashi N, Hoshikawa M, Itoh N, Hogan BL, Furuta Y. Comparison of the expression of three highly related genes, Fgf8, Fgf17 and Fgf18, in the mouse embryo. Mechanisms of Development. 1998; 74: 175–177.
[73]
Sun Y, Wang R, Zhao S, Li W, Liu W, Tang L, et al. FGF9 inhibits browning program of white adipocytes and associates with human obesity. Journal of Molecular Endocrinology. 2019; 62: 79–90.
[74]
Rulifson IC, Collins P, Miao L, Nojima D, Lee KJ, Hardy M, et al. In Vitro and in Vivo Analyses Reveal Profound Effects of Fibroblast Growth Factor 16 as a Metabolic Regulator. The Journal of Biological Chemistry. 2017; 292: 1951–1969.
[75]
Knowles HJ. Hypoxia-Induced Fibroblast Growth Factor 11 Stimulates Osteoclast-Mediated Resorption of Bone. Calcified Tissue International. 2017; 100: 382–391.
[76]
Alshammari MA, Alshammari TK, Nenov MN, Scala F, Laezza F. Fibroblast Growth Factor 14 Modulates the Neurogenesis of Granule Neurons in the Adult Dentate Gyrus. Molecular Neurobiology. 2016; 53: 7254–7270.
[77]
Owen BM, Mangelsdorf DJ, Kliewer SA. Tissue-specific actions of the metabolic hormones FGF15/19 and FGF21. Trends in Endocrinology and Metabolism: TEM. 2015; 26: 22–29.
[78]
Flippo KH, Potthoff MJ. Metabolic Messengers: FGF21. Nature Metabolism. 2021; 3: 309–317.
[79]
Nygaard EB, Ørskov C, Almdal T, Vestergaard H, Andersen B. Fasting decreases plasma FGF21 in obese subjects and the expression of FGF21 receptors in adipose tissue in both lean and obese subjects. The Journal of Endocrinology. 2018; 239: 73–80.
[80]
Iroz A, Montagner A, Benhamed F, Levavasseur F, Polizzi A, Anthony E, et al. A Specific ChREBP and PPARα Cross-Talk Is Required for the Glucose-Mediated FGF21 Response. Cell Reports. 2017; 21: 403–416.
[81]
Yamada Y, Saito H, Araki M, Tsuchimoto Y, Muroi SI, Suzuki K, et al. Wogonin, a Compound in Scutellaria baicalensis, Activates ATF4-FGF21 Signaling in Mouse Hepatocyte AML12 Cells. Nutrients. 2022; 14: 3920.
[82]
Lan T, Morgan DA, Rahmouni K, Sonoda J, Fu X, Burgess SC, et al. FGF19, FGF21, and an FGFR1/β-Klotho-Activating Antibody Act on the Nervous System to Regulate Body Weight and Glycemia. Cell Metabolism. 2017; 26: 709–718.e3.
[83]
Abu-Odeh M, Zhang Y, Reilly SM, Ebadat N, Keinan O, Valentine JM, et al. FGF21 promotes thermogenic gene expression as an autocrine factor in adipocytes. Cell Reports. 2021; 35: 109331.
[84]
Li X. The FGF metabolic axis. Frontiers of Medicine. 2019; 13: 511–530.
[85]
Nygaard EB, Møller CL, Kievit P, Grove KL, Andersen B. Increased fibroblast growth factor 21 expression in high-fat diet-sensitive non-human primates (Macaca mulatta). International Journal of Obesity (2005). 2014; 38: 183–191.
[86]
Moure R, Cairó M, Morón-Ros S, Quesada-López T, Campderrós L, Cereijo R, et al. Levels of β-klotho determine the thermogenic responsiveness of adipose tissues: involvement of the autocrine action of FGF21. American Journal of Physiology. Endocrinology and Metabolism. 2021; 320: E822–E834.
[87]
Kroon T, Harms M, Maurer S, Bonnet L, Alexandersson I, Lindblom A, et al. PPARγ and PPARα synergize to induce robust browning of white fat in vivo. Molecular Metabolism. 2020; 36: 100964.
[88]
Liu X, Zhang H, Zhang S, Mao W, Liu L, Deng C, et al. Olanzapine-induced decreases of FGF21 in brown adipose tissue via histone modulations drive UCP1-dependent thermogenetic impairment. Progress in Neuro-psychopharmacology & Biological Psychiatry. 2023; 122: 110692.
[89]
Ng R, Hussain NA, Zhang Q, Chang C, Li H, Fu Y, et al. miRNA-32 Drives Brown Fat Thermogenesis and Trans-activates Subcutaneous White Fat Browning in Mice. Cell Reports. 2017; 19: 1229–1246.
[90]
Justesen S, Haugegaard KV, Hansen JB, Hansen HS, Andersen B. The autocrine role of FGF21 in cultured adipocytes. The Biochemical Journal. 2020; 477: 2477–2487.
[91]
Porflitt-Rodríguez M, Guzmán-Arriagada V, Sandoval-Valderrama R, Tam CS, Pavicic F, Ehrenfeld P, et al. Effects of aerobic exercise on fibroblast growth factor 21 in overweight and obesity. A systematic review. Metabolism: Clinical and Experimental. 2022; 129: 155137.
[92]
Park SY, Lee HJ, Song JH, Shin YK, Abd El-Aty AM, Ramadan A, et al. Dimethyl itaconate attenuates palmitate-induced insulin resistance in skeletal muscle cells through the AMPK/FGF21/PPARδ-mediated suppression of inflammation. Life Sciences. 2021; 287: 120129.
[93]
Izumiya Y, Bina HA, Ouchi N, Akasaki Y, Kharitonenkov A, Walsh K. FGF21 is an Akt-regulated myokine. FEBS Letters. 2008; 582: 3805–3810.
[94]
Coate KC, Hernandez G, Thorne CA, Sun S, Le TDV, Vale K, et al. FGF21 Is an Exocrine Pancreas Secretagogue. Cell Metabolism. 2017; 25: 472–480.
[95]
Xu Z, Sun J, Tong Q, Lin Q, Qian L, Park Y, et al. The Role of ERK1/2 in the Development of Diabetic Cardiomyopathy. International Journal of Molecular Sciences. 2016; 17: 2001.
[96]
Micanovic R, Raches DW, Dunbar JD, Driver DA, Bina HA, Dickinson CD, et al. Different roles of N- and C- termini in the functional activity of FGF21. Journal of Cellular Physiology. 2009; 219: 227–234.
[97]
Fon Tacer K, Bookout AL, Ding X, Kurosu H, John GB, Wang L, et al. Research resource: Comprehensive expression atlas of the fibroblast growth factor system in adult mouse. Molecular Endocrinology (Baltimore, Md.). 2010; 24: 2050–2064.
[98]
Kliewer SA, Mangelsdorf DJ. A Dozen Years of Discovery: Insights into the Physiology and Pharmacology of FGF21. Cell Metabolism. 2019; 29: 246–253.
[99]
Bookout AL, de Groot MHM, Owen BM, Lee S, Gautron L, Lawrence HL, et al. FGF21 regulates metabolism and circadian behavior by acting on the nervous system. Nature Medicine. 2013; 19: 1147–1152.
[100]
So WY, Leung PS. Fibroblast Growth Factor 21 As an Emerging Therapeutic Target for Type 2 Diabetes Mellitus. Medicinal Research Reviews. 2016; 36: 672–704.
[101]
Zhang Y, Liu D, Long XX, Fang QC, Jia WP, Li HT. The role of FGF21 in the pathogenesis of cardiovascular disease. Chinese Medical Journal. 2021; 134: 2931–2943.
[102]
Salgado JV, Goes MA, Salgado Filho N. FGF21 and Chronic Kidney Disease. Metabolism: Clinical and Experimental. 2021; 118: 154738.
[103]
He L, Deng L, Zhang Q, Guo J, Zhou J, Song W, et al. Diagnostic Value of CK-18, FGF-21, and Related Biomarker Panel in Nonalcoholic Fatty Liver Disease: A Systematic Review and Meta-Analysis. BioMed Research International. 2017; 2017: 9729107.
[104]
Kaelin BR, McKenzie CM, Hempel KW, Lang AL, Arteel GE, Beier JI. Adipose tissue-liver crosstalk during pathologic changes caused by vinyl chloride metabolites in mice. Toxicology and Applied Pharmacology. 2020; 399: 115068.
[105]
Fisher FM, Chui PC, Antonellis PJ, Bina HA, Kharitonenkov A, Flier JS, et al. Obesity is a fibroblast growth factor 21 (FGF21)-resistant state. Diabetes. 2010; 59: 2781–2789.
[106]
Könner AC, Brüning JC. Selective insulin and leptin resistance in metabolic disorders. Cell Metabolism. 2012; 16: 144–152.
[107]
Kim TH, Hong DG, Yang YM. Hepatokines and Non-Alcoholic Fatty Liver Disease: Linking Liver Pathophysiology to Metabolism. Biomedicines. 2021; 9: 1903.
[108]
Staiger H, Keuper M, Berti L, Hrabe de Angelis M, Häring HU. Fibroblast Growth Factor 21-Metabolic Role in Mice and Men. Endocrine Reviews. 2017; 38: 468–488.
[109]
Chavez AO, Molina-Carrion M, Abdul-Ghani MA, Folli F, Defronzo RA, Tripathy D. Circulating fibroblast growth factor-21 is elevated in impaired glucose tolerance and type 2 diabetes and correlates with muscle and hepatic insulin resistance. Diabetes Care. 2009; 32: 1542–1546.
[110]
Wu G, Li H, Fang Q, Zhang J, Zhang M, Zhang L, et al. Complementary Role of Fibroblast Growth Factor 21 and Cytokeratin 18 in Monitoring the Different Stages of Nonalcoholic Fatty Liver Disease. Scientific Reports. 2017; 7: 5095.
[111]
Xiao F, Shi X, Huang P, Zeng X, Wang L, Zeng J, et al. Dose-response relationship between serum fibroblast growth factor 21 and liver fat content in non-alcoholic fatty liver disease. Diabetes & Metabolism. 2021; 47: 101221.
[112]
Guo JY, Chen HH, Lee WJ, Chen SC, Lee SD, Chen CY. Fibroblast Growth Factor 19 and Fibroblast Growth Factor 21 Regulation in Obese Diabetics, and Non-Alcoholic Fatty Liver Disease after Gastric Bypass. Nutrients. 2022; 14: 645.
[113]
Wong C, Dash A, Fredrickson J, Lewin-Koh N, Chen S, Yoshida K, et al. Fibroblast growth factor receptor 1/Klothoβ agonist BFKB8488A improves lipids and liver health markers in patients with diabetes or NAFLD: A phase 1b randomized trial. Hepatology (Baltimore, Md.). 2023; 78: 847–862.
[114]
Harrison SA, Ruane PJ, Freilich BL, Neff G, Patil R, Behling CA, et al. Efruxifermin in non-alcoholic steatohepatitis: a randomized, double-blind, placebo-controlled, phase 2a trial. Nature Medicine. 2021; 27: 1262–1271.
[115]
Yin X, Guo X, Liu Z, Wang J. Advances in the Diagnosis and Treatment of Non-Alcoholic Fatty Liver Disease. International Journal of Molecular Sciences. 2023; 24: 2844.
[116]
Chaurasia B, Tippetts TS, Mayoral Monibas R, Liu J, Li Y, Wang L, et al. Targeting a ceramide double bond improves insulin resistance and hepatic steatosis. Science (New York, N.Y.). 2019; 365: 386–392.
[117]
Neuschwander-Tetri BA. Hepatic lipotoxicity and the pathogenesis of nonalcoholic steatohepatitis: the central role of nontriglyceride fatty acid metabolites. Hepatology (Baltimore, Md.). 2010; 52: 774–788.
[118]
Feldstein AE, Werneburg NW, Canbay A, Guicciardi ME, Bronk SF, Rydzewski R, et al. Free fatty acids promote hepatic lipotoxicity by stimulating TNF-alpha expression via a lysosomal pathway. Hepatology (Baltimore, Md.). 2004; 40: 185–194.
[119]
Piccolis M, Bond LM, Kampmann M, Pulimeno P, Chitraju C, Jayson CBK, et al. Probing the Global Cellular Responses to Lipotoxicity Caused by Saturated Fatty Acids. Molecular Cell. 2019; 74: 32–44.e8.
[120]
Henriksson E, Andersen B. FGF19 and FGF21 for the Treatment of NASH-Two Sides of the Same Coin? Differential and Overlapping Effects of FGF19 and FGF21 From Mice to Human. Frontiers in Endocrinology. 2020; 11: 601349.
[121]
Li X, Ge H, Weiszmann J, Hecht R, Li YS, Véniant MM, et al. Inhibition of lipolysis may contribute to the acute regulation of plasma FFA and glucose by FGF21 in ob/ob mice. FEBS Letters. 2009; 583: 3230–3234.
[122]
Gong Q, Hu Z, Zhang F, Cui A, Chen X, Jiang H, et al. Fibroblast growth factor 21 improves hepatic insulin sensitivity by inhibiting mammalian target of rapamycin complex 1 in mice. Hepatology (Baltimore, Md.). 2016; 64: 425–438.
[123]
Zhang Y, Lei T, Huang JF, Wang SB, Zhou LL, Yang ZQ, et al. The link between fibroblast growth factor 21 and sterol regulatory element binding protein 1c during lipogenesis in hepatocytes. Molecular and Cellular Endocrinology. 2011; 342: 41–47.
[124]
Jimenez V, Jambrina C, Casana E, Sacristan V, Muñoz S, Darriba S, et al. FGF21 gene therapy as treatment for obesity and insulin resistance. EMBO Molecular Medicine. 2018; 10: e8791.
[125]
Jiang S, Yan C, Fang QC, Shao ML, Zhang YL, Liu Y, et al. Fibroblast growth factor 21 is regulated by the IRE1α-XBP1 branch of the unfolded protein response and counteracts endoplasmic reticulum stress-induced hepatic steatosis. The Journal of Biological Chemistry. 2014; 289: 29751–29765.
[126]
Dong K, Li H, Zhang M, Jiang S, Chen S, Zhou J, et al. Endoplasmic reticulum stress induces up-regulation of hepatic β-Klotho expression through ATF4 signaling pathway. Biochemical and Biophysical Research Communications. 2015; 459: 300–305.
[127]
Wan XS, Lu XH, Xiao YC, Lin Y, Zhu H, Ding T, et al. ATF4- and CHOP-dependent induction of FGF21 through endoplasmic reticulum stress. BioMed Research International. 2014; 2014: 807874.
[128]
Wu A, Feng B, Yu J, Yan L, Che L, Zhuo Y, et al. Fibroblast growth factor 21 attenuates iron overload-induced liver injury and fibrosis by inhibiting ferroptosis. Redox Biology. 2021; 46: 102131.
[129]
Zhang Y, Yin R, Lang J, Fu Y, Yang L, Zhao D. Epigallocatechin-3-gallate ameliorates hepatic damages by relieve FGF21 resistance and promotion of FGF21-AMPK pathway in mice fed a high fat diet. Diabetology & Metabolic Syndrome. 2022; 14: 53.
[130]
Zhu S, Ma L, Wu Y, Ye X, Zhang T, Zhang Q, et al. FGF21 treatment ameliorates alcoholic fatty liver through activation of AMPK-SIRT1 pathway. Acta Biochimica et Biophysica Sinica. 2014; 46: 1041–1048.
[131]
Medzhitov R. Origin and physiological roles of inflammation. Nature. 2008; 454: 428–435.
[132]
Khanmohammadi S, Kuchay MS. Toll-like receptors and metabolic (dysfunction)-associated fatty liver disease. Pharmacological Research. 2022; 185: 106507.
[133]
Wei W, Liu L, Liu X, Tao Y, Gong J, Wang Y, et al. Black ginseng protects against Western diet-induced nonalcoholic steatohepatitis by modulating the TLR4/NF-κB signaling pathway in mice. Journal of Food Biochemistry. 2022; 46: e14432.
[134]
Yan BF, Wang Y, Wang WB, Ding XJ, Wei B, Liu SJ, et al. Huangqin decoction mitigates hepatic inflammation in high-fat diet-challenged rats by inhibiting TLR4/NF-κB/NLRP3 pathway. Journal of Ethnopharmacology. 2023; 303: 115999.
[135]
Tillman EJ, Rolph T. FGF21: An Emerging Therapeutic Target for Non-Alcoholic Steatohepatitis and Related Metabolic Diseases. Frontiers in Endocrinology. 2020; 11: 601290.
[136]
Yu Y, He J, Li S, Song L, Guo X, Yao W, et al. Fibroblast growth factor 21 (FGF21) inhibits macrophage-mediated inflammation by activating Nrf2 and suppressing the NF-κB signaling pathway. International Immunopharmacology. 2016; 38: 144–152.
[137]
Chikamatsu M, Watanabe H, Shintani Y, Murata R, Miyahisa M, Nishinoiri A, et al. Albumin-fused long-acting FGF21 analogue for the treatment of non-alcoholic fatty liver disease. Journal of Controlled Release: Official Journal of the Controlled Release Society. 2023; 355: 42–53.
[138]
Lee JH, Kang YE, Chang JY, Park KC, Kim HW, Kim JT, et al. An engineered FGF21 variant, LY2405319, can prevent non-alcoholic steatohepatitis by enhancing hepatic mitochondrial function. American Journal of Translational Research. 2016; 8: 4750–4763.
[139]
Zheng Q, Martin RC, Shi X, Pandit H, Yu Y, Liu X, et al. Lack of FGF21 promotes NASH-HCC transition via hepatocyte-TLR4-IL-17A signaling. Theranostics. 2020; 10: 9923–9936.
[140]
Zeng Z, Zheng Q, Chen J, Tan X, Li Q, Ding L, et al. FGF21 mitigates atherosclerosis via inhibition of NLRP3 inflammasome-mediated vascular endothelial cells pyroptosis. Experimental Cell Research. 2020; 393: 112108.
[141]
Willis SA, Sargeant JA, Thackray AE, Yates T, Stensel DJ, Aithal GP, et al. Effect of exercise intensity on circulating hepatokine concentrations in healthy men. Applied Physiology, Nutrition, and Metabolism. 2019; 44: 1065–1072.
[142]
He Z, Tian Y, Valenzuela PL, Huang C, Zhao J, Hong P, et al. Myokine Response to High-Intensity Interval vs. Resistance Exercise: An Individual Approach. Frontiers in Physiology. 2018; 9: 1735.
[143]
Morville T, Sahl RE, Trammell SA, Svenningsen JS, Gillum MP, Helge JW, et al. Divergent effects of resistance and endurance exercise on plasma bile acids, FGF19, and FGF21 in humans. JCI Insight. 2018; 3: e122737.
[144]
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.
[145]
Motahari Rad M, Bijeh N, Attarzadeh Hosseini SR, Raouf Saeb A. The effect of two concurrent exercise modalities on serum concentrations of FGF21, irisin, follistatin, and myostatin in men with type 2 diabetes mellitus. Archives of Physiology and Biochemistry. 2023; 129: 424–433.
[146]
Geng L, Liao B, Jin L, Huang Z, Triggle CR, Ding H, et al. Exercise Alleviates Obesity-Induced Metabolic Dysfunction via Enhancing FGF21 Sensitivity in Adipose Tissues. Cell Reports. 2019; 26: 2738–2752.e4.
[147]
Fletcher JA, Meers GM, Laughlin MH, Ibdah JA, Thyfault JP, Rector RS. Modulating fibroblast growth factor 21 in hyperphagic OLETF rats with daily exercise and caloric restriction. Applied Physiology, Nutrition, and Metabolism. 2012; 37: 1054–1062.
[148]
Kim KH, Kim SH, Min YK, Yang HM, Lee JB, Lee MS. Acute exercise induces FGF21 expression in mice and in healthy humans. PLoS ONE. 2013; 8: e63517.
[149]
Kim KH, Jeong YT, Oh H, Kim SH, Cho JM, Kim YN, et al. Autophagy deficiency leads to protection from obesity and insulin resistance by inducing Fgf21 as a mitokine. Nature Medicine. 2013; 19: 83–92.
[150]
Ennequin G, Sirvent P, Whitham M. Role of exercise-induced hepatokines in metabolic disorders. American Journal of Physiology. Endocrinology and Metabolism. 2019; 317: E11–E24.
[151]
Guo Y, Liu Q, Gui Y, Liao C, Xu D. Exercise promotes cardiac-specific fibroblast growth factor 21 expression. International Journal of Cardiology. 2016; 203: 532–533.
[152]
Yu Y, Bai F, Liu Y, Yang Y, Yuan Q, Zou D, et al. Fibroblast growth factor (FGF21) protects mouse liver against D-galactose-induced oxidative stress and apoptosis via activating Nrf2 and PI3K/Akt pathways. Molecular and Cellular Biochemistry. 2015; 403: 287–299.
[153]
Kang K, Xia A, Meng F, Chunyu J, Sun X, Ren G, et al. FGF21 alleviates chronic inflammatory injury in the aging process through modulating polarization of macrophages. International Immunopharmacology. 2021; 96: 107634.
[154]
Wu L, Mo W, Feng J, Li J, Yu Q, Li S, et al. Astaxanthin attenuates hepatic damage and mitochondrial dysfunction in non-alcoholic fatty liver disease by up-regulating the FGF21/PGC-1α pathway. British Journal of Pharmacology. 2020; 177: 3760–3777.
[155]
Pan Y, Wang B, Zheng J, Xiong R, Fan Z, Ye Y, et al. Pancreatic fibroblast growth factor 21 protects against type 2 diabetes in mice by promoting insulin expression and secretion in a PI3K/Akt signaling-dependent manner. Journal of Cellular and Molecular Medicine. 2019; 23: 1059–1071.
[156]
Kim SH, Kim KH, Kim HK, Kim MJ, Back SH, Konishi M, et al. Fibroblast growth factor 21 participates in adaptation to endoplasmic reticulum stress and attenuates obesity-induced hepatic metabolic stress. Diabetologia. 2015; 58: 809–818.
[157]
Loyd C, Magrisso IJ, Haas M, Balusu S, Krishna R, Itoh N, et al. Fibroblast growth factor 21 is required for beneficial effects of exercise during chronic high-fat feeding. Journal of Applied Physiology (Bethesda, Md.: 1985). 2016; 121: 687–698.
[158]
Porter JW, Rowles JL, 3rd, Fletcher JA, Zidon TM, Winn NC, McCabe LT, et al. Anti-inflammatory effects of exercise training in adipose tissue do not require FGF21. The Journal of Endocrinology. 2017; 235: 97–109.
[159]
Gaich G, Chien JY, Fu H, Glass LC, Deeg MA, Holland WL, et al. The effects of LY2405319, an FGF21 analog, in obese human subjects with type 2 diabetes. Cell Metabolism. 2013; 18: 333–340.
[160]
Talukdar S, Zhou Y, Li D, Rossulek M, Dong J, Somayaji V, et al. A Long-Acting FGF21 Molecule, PF-05231023, Decreases Body Weight and Improves Lipid Profile in Non-human Primates and Type 2 Diabetic Subjects. Cell Metabolism. 2016; 23: 427–440.
[161]
Wei W, Dutchak PA, Wang X, Ding X, Wang X, Bookout AL, et al. Fibroblast growth factor 21 promotes bone loss by potentiating the effects of peroxisome proliferator-activated receptor γ. Proceedings of the National Academy of Sciences of the United States of America. 2012; 109: 3143–3148.
[162]
Owen BM, Bookout AL, Ding X, Lin VY, Atkin SD, Gautron L, et al. FGF21 contributes to neuroendocrine control of female reproduction. Nature Medicine. 2013; 19: 1153–1156.

Publisher’s Note: IMR Press stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share
Back to top