Skip to content
Publicly Available Published by De Gruyter June 10, 2016

The regulation of FGF21 gene expression by metabolic factors and nutrients

  • Anjeza Erickson and Régis Moreau EMAIL logo

Abstract

Fibroblast growth factor 21 (FGF21) gene expression is altered by a wide array of physiological, metabolic, and environmental factors. Among dietary factors, high dextrose, low protein, methionine restriction, short-chain fatty acids (butyric acid and lipoic acid), and all-trans-retinoic acid were repeatedly shown to induce FGF21 expression and circulating levels. These effects are usually more pronounced in liver or isolated hepatocytes than in adipose tissue or isolated fat cells. Although peroxisome proliferator-activated receptor α (PPARα) is a key mediator of hepatic FGF21 expression and function, including the regulation of gluconeogenesis, ketogenesis, torpor, and growth inhibition, there is increasing evidence of PPARα-independent transactivation of the FGF21 gene by dietary molecules. FGF21 expression is believed to follow the circadian rhythm and be placed under the control of first order clock-controlled transcription factors, retinoic acid receptor-related orphan receptors (RORs) and nuclear receptors subfamily 1 group D (REV-ERBs), with FGF21 rhythm being anti-phase to REV-ERBs. Key metabolic hormones such as glucagon, insulin, and thyroid hormone have presumed or clearly demonstrated roles in regulating FGF21 transcription and secretion. The control of the FGF21 gene by glucagon and insulin appears more complex than first anticipated. Some discrepancies are noted and will need continued studies. The complexity in assessing the significance of FGF21 gene expression resides in the difficulty to ascertain (i) when transcription results in local or systemic increase of FGF21 protein; (ii) if FGF21 is among the first or second order genes upregulated by physiological, metabolic, and environmental stimuli, or merely an epiphenomenon; and (iii) whether FGF21 may have some adverse effects alongside beneficial outcomes.

Introduction

The importance of diet and nutrition in the etiology of a number of diseases affecting morbidity and mortality is well recognized. However, the exact nature of how diet impacts health and disease is complex and not fully understood. Nutrients and dietary molecules alter gene expression, modulate protein and metabolite levels in blood and tissues, modify cellular and metabolic pathways, affect epigenetic phenomena, and modify response to drugs. These nutrient-gene interactions thus influence an individual’s response to the environment – including diet – and to therapy. Nutrients act directly, indirectly, and cooperatively with hormones to influence the rate and extent of transcription. Among nutrients, glucose, fatty acids, sterols, vitamin A, vitamin D, and their metabolites are prominent regulators of gene expression. Chief among hormones, insulin, glucagon, glucocorticoids, and growth and thyroid hormones, are essential regulators of metabolic homeostasis.

Since fibroblast growth factor 21 (FGF21) was discovered nearly two decades ago [1], its physiological and pharmacological effects have been extensively studied whereas, comparatively, the regulation of FGF21 gene expression by diet, nutrients, and dietary bioactive compounds is not well characterized. FGF21 is expressed across several tissues, predominantly in liver and adipocyte tissues, and, to a lower extent, in the pancreas, skeletal muscle, heart, kidneys and testes [13]. Unlike most members of the FGF family, FGF21 lacks the FGF heparin-binding domain, allowing FGF21 to be secreted in the bloodstream and act peripherally as well as centrally [4]. Circulating levels of FGF21 are increased in response to fasting [57], macronutrient diet composition [6, 8], and physiological or environmental stress [9] in individuals with obesity, type 2 diabetes, and non-alcoholic fatty liver disease [10]. The broad range responsiveness of FGF21 expression to metabolic variations suggests that FGF21 contributes to metabolic homeostasis in health and disease. Indeed, the pharmacological administration of recombinant FGF21 resulted in a plethora of metabolic outcomes including the increase of fat utilization and energy expenditure, the promotion of fat browning in white and brown adipose tissue, the improvement of thermogenesis, the stimulation of fatty acid oxidation, the stimulation of peroxisome proliferator-activated receptor α (PPARα) and PPARγ transactivation, glucose tolerance and insulin sensitivity, decreased body weight and lowered cholesterol along with serum and hepatic triglyceride levels [5, 6, 1121]. This review focuses on the nutritional factors and metabolic hormones that modulate FGF21 gene expression in rodents and humans, and discusses the underlying transcriptional mechanisms.

Body of review

Regulation by starvation and overnutrition

Animal studies have shown that FGF21 expression is upregulated by drastically different nutritional states, such as in starvation and overnutrition. In contrast, the FGF21 serum levels of healthy subjects remain stable over the course of a day despite wide interindividual variations (21–5300 pg/mL; mean, 450 pg/mL) [22]. Moreover, serum FGF21 is largely unchanged in humans who fasted for 2 days [22], suggesting that secreted FGF21 remains within the tissue of production and acts locally. It is after 7 days of fasting that serum FGF21 notably increases in human subjects [22, 23]. Plasma FGF21 levels are reportedly higher in the obese, in individuals with type 2 diabetes, and in subjects with non-alcoholic liver disease, which correlate with elevated levels of triacylglycerols in the liver [2429]. Overnutrition and refeeding also upregulate FGF21 gene expression. This response may be induced by dietary carbohydrates as it was shown that carbohydrates induce FGF21 gene expression through the transcription factor carbohydrate response element binding protein (ChREBP).

In the liver and isolated hepatocytes, FGF21 expression is induced by starvation in PPARα-dependent and independent manners [57, 30, 31]. As a transcription factor activated by fatty acids, phosphatidylcholine, oleoylethanolamine, and hypolipidemic fibrate drugs, PPARα binds to PPRE in the 5′-flanking region of the FGF21 gene and induces transcription [5, 7] (Figure 1). Liver FGF21 is also induced by a high-fat low-carbohydrate diet [6], a condition that, similar to starvation, stimulates the breakdown of stored fat, the catabolism of fatty acids, ketogenesis, and gluconeogenesis. The FGF21 gene is also upregulated by fasting signals conveyed by glucagon. The actions of the glucagon receptor on glucose and lipid metabolism and on the regulation of body weight and fat mass are mediated in part by FGF21 [36]. The glucagon/FGF21 axis is further discussed below.

Figure 1: Mapping of the response elements and specific transcription factors documented to interact within the 5′-flanking region of the human, mouse, and rat FGF21 gene.(A) Overall layout of the response elements within the 4600 bp upstream of the transcription start site (TSS). (B) Alignment of human, mouse, and rat FGF21 5′-regulatory sequences shown in red or blue boxes the positions of known transcription factor binding sites. The binding sites were retrieved from the literature and confirmed using MatInspector (Genomatix Software GmbH, Munich, Germany). Segments of the 5′-flanking region that do not harbor any documented transcription binding sites were omitted. The 5′ genomic sequences for human, mouse, and rat FGF21 were retrieved from NCBI [32, 33] and aligned using Clustal Omega [34]. The TSSs were positioned based on the following transcript sequences retrieved from ENSEMBL [35]: ENST00000222157 for hFGF21, ENSMUST00000033099 for mFGF21, and ENSRNOT00000028490 for rFGF21.
Figure 1:

Mapping of the response elements and specific transcription factors documented to interact within the 5′-flanking region of the human, mouse, and rat FGF21 gene.

(A) Overall layout of the response elements within the 4600 bp upstream of the transcription start site (TSS). (B) Alignment of human, mouse, and rat FGF21 5′-regulatory sequences shown in red or blue boxes the positions of known transcription factor binding sites. The binding sites were retrieved from the literature and confirmed using MatInspector (Genomatix Software GmbH, Munich, Germany). Segments of the 5′-flanking region that do not harbor any documented transcription binding sites were omitted. The 5′ genomic sequences for human, mouse, and rat FGF21 were retrieved from NCBI [32, 33] and aligned using Clustal Omega [34]. The TSSs were positioned based on the following transcript sequences retrieved from ENSEMBL [35]: ENST00000222157 for hFGF21, ENSMUST00000033099 for mFGF21, and ENSRNOT00000028490 for rFGF21.

Fat cell FGF21 has been shown to be upregulated during adipocyte differentiation [28] and to stimulate glucose uptake in adipocytes [11, 13]. ChREBP is upregulated during adipocyte differentiation and, thus, may be involved in the induction of FGF21 in these cells [37]. In adipocytes, FGF21 is induced by feeding in a PPARγ-dependent manner [38]. PPARγ agonists, such as the thiazolidinedione drugs, also induce FGF21 gene expression in white adipose tissue and isolated adipocytes [28, 39, 40]. Supporting the mediating role of FGF21 in the therapeutic effects of thiazolidinedione is the observation that FGF21-null mice are refractory to both the beneficial and adverse effects of this family of drugs [17].

There is evidence to indicate that the FGF21 gene is autoregulated by circulating FGF21. The administration of recombinant FGF21 to streptozotocin-treated mice dose-dependently repressed FGF21 gene expression in the liver to a greater extent than streptozotocin alone [41]. In a separate study, FGF21 transcript levels were substantially lowered in the liver following FGF21 administration [42]. These results suggest the presence of a negative feedback by FGF21 directed at its own expression.

Regulation by hormones

Glucagon and insulin

A physiological link was established between hepatic glucagon signaling, 5′ adenosine monophosphate-activated protein kinase (AMPK), PPARα, and the expression of FGF21 in mice lacking the glucagon receptor [43]. In these mice, glucagon failed to induce liver AMPK phosphorylation, PPARα and FGF21 gene expression. A lipid infusion composed of PPARα natural ligands also failed to induce liver AMPK phosphorylation, and PPARα and FGF21 gene expression in glucagon receptor-null mice, suggesting that a functional glucagon signaling is required for the PPARα-mediated upregulation of the FGF21 gene. The use of a glucagon receptor agonist raised hepatic expression and blood levels of FGF21 in wild-type mice [36], supporting the notion that chronic glucagon action is mediated through FGF21 expression. Cyphert et al. [44] found that glucagon stimulates FGF21 secretion via a translational and/or post-translational mechanism mediated by the cyclic adenosine monophosphate (cAMP)/protein kinase A (PKA) axis as well as a PKA-independent mechanism. The latter mechanism involves the cAMP activation of exchange protein directly activated by cAMP (EPAC), a guanine nucleotide exchange factor that activates the small guanosine-5′-phosphatase (GTPase) Ras-proximate-1 (Rap1) [45, 46]. Downstream of cAMP, the glucagon/cAMP signaling pathway implicates AMPK and p38 mitogen-activated protein kinase (MAPK).

The cAMP-stimulated transcription factor cyclic AMP-responsive element-binding protein 3-like protein 3 (CREBH) is upregulated during fasting [47, 48] and possibly by glucagon. Fasting and glucagon increases CREBH acetylation, which is required for the formation of the CREBH-PPARα transcriptional complex [49]. In addition to regulating each other’s transcription in fasted liver [47], CREBH and PPARα synergize to activate FGF21 gene expression through the binding of the CREBH-PPARα complex to a highly conserved CRE-peroxisome proliferator-activated response element (PPRE) site in the FGF21 proximal promoter [50]. Glucocorticoid signaling is involved in the regulation of CREBH [48]. In the fasting state, the adrenal gland secretes glucocorticoids, which contribute to upregulate gluconeogenesis and stimulate hepatic glucose output [51].

Glucagon and insulin synergized in primary rat hepatocytes to induce FGF21 mRNA and FGF21 secretion, whereas glucagon alone in the absence of insulin lowered FGF21 mRNAs [44]. In contrast, Uebanso et al. [52] found that glucagon and insulin function in an opposite manner in rat primary hepatocytes: glucagon induced FGF21 mRNA whereas insulin inhibited gene expression. Insulin, through protein kinase B (Akt), reportedly induced FGF21 expression in cultured skeletal muscle cells [53]. Of note, glucagon has been shown to enhance the action of insulin on Akt in hepatocytes [54]. Hence, although glucagon and insulin have opposite broad effects on metabolism, Cyphert et al. [44] showed that these hormones stimulate FGF21 secretion either alone [53] or in combination [44]. Overall, the regulation of FGF21 mRNA levels by glucagon and insulin appears more complex than first anticipated. Either glucagon or insulin has the potential to upregulate FGF21 gene expression.

Thyroid hormone T3

The intraperitoneal injection of thyroid hormone tri-iodothyronine (T3) in mice has been shown to induce hepatic FGF21 gene expression in a dose-dependent manner [55]. The induction was abolished in PPARα-null mice, suggesting that PPARα mediates the effects of T3. A direct interaction among thyroid hormone receptor β, PPARα, and retinoid X receptor (RXR) in hepatocytes was suggested based on the rapid induction of the FGF21 gene by T3 [55]. A distal PPRE site in the 5′ regulatory region of the FGF21 gene was required for T3 action. T3 did not induce FGF21 mRNA in the white adipose tissue of wild-type mice, indicating that tissue specificity may result from a distinct expression profile of the thyroid hormone receptor subtypes. As abnormal blood lipids can be improved with thyroid hormone receptor β agonists, similarly with recombinant FGF21, FGF21 may be a physiologically relevant mediator of T3’s lipid-ameliorating properties.

Regulation by miRNA

Recent studies have found evidence linking microRNAs (miRNAs) to the regulation of metabolism and metabolic disorders [56]. miRNAs are circulating, small, non-coding RNAs of about 19–23 nucleotides long, which regulate gene expression post-transcriptionally by binding primarily to the 3′-untranslated region (3′UTR) of mRNAs and targeting mRNAs for translational repression and degradation. Blood-circulating miRNAs function as signaling molecules by altering gene expression not only in cells and tissues where they are produced but also in distant organs. Their critical role in gene regulation suggests that miRNAs can be markers of health and disease, and potential therapeutic targets. The discovery of miRNAs able to regulate FGF21 expression and impact the metabolic effects of FGF21 has been of special interest in recent years.

FGF21 is a well-known regulator of pancreatic insulin production. Treatment of diabetic mice and rats with FGF21 increased insulin secretion and content in pancreatic islets, improved glucose clearance, and promoted the survival of islets and insulin-secreting INS-1E pancreatic β-cells against glucolipotoxicity and cytokine-induced apoptosis [57]. A recent study investigated miR-577 as a novel mechanism of improving pancreatic β-cell survival and function in diabetic patients [58], in part because miR-577 was predicted to target FGF21 [59]. Chen et al. [58] observed that the blood of diabetic children contained elevated levels of miR-577. They showed that FGF21 3′UTR was a direct target of miR-577 in mouse embryonic pancreatic β-cells as the suppression of miR-577 with anti-miR-577 increased FGF21 mRNA and protein expression in these β-cells. Additional evidence that miR-577 targets FGF21 was provided when the activation of the extracellular signal-regulated kinase 1/2 (ERK1/2) and Akt signaling pathways was blocked by miR-577, and when the glucose-stimulated FGF21-induced production of insulin was repressed by miR-577. Hence, their work points to anti-miR-577 as a means to protect and restore the insulinotropic action of FGF21. There are other predicted targets of miR-577 that belong to the FGF family, including FGF5 and FGF23 [59].

Another miRNA that has been shown to play an important role in metabolic disorders is microRNA-212 (miR-212). miR-212 modulates alcoholic liver disease [60] and cardiac insulin resistance during overnutrition [61]. This miRNA is primarily expressed in the central nervous system and, to a lower extent, in adipose, liver, and many other tissues. Xiao et al. [62] reported on the beneficial effects of miR-212 downregulation through exercise in the prevention of non-alcoholic fatty liver disease (NAFLD). The evidence that miR-212 targets FGF21 derives primarily from the observation that synthetic miR-212 mimics decreased FGF21 expression whereas the miR-212 inhibitor increased FGF21 protein levels and decreased lipid synthesis in HepG2 cells. The silencing of FGF21 increased intracellular lipid droplets in HepG2 cells and hindered the anti-lipogenic effect of the miR-212 inhibitor in these cells. In animals, hepatic miR-212 was increased in mice fed a high-fat diet, and negatively regulated FGF21. Collectively, it was concluded that FGF21 is a target of miR-212 and anti-miR-212 therapeutic approaches could be useful for NAFLD patients [62]. Other predicted targets of miR-212 that belong to the FGF family of proteins include FGF1 [63], FGF2, FGF5, FGF9, FGF11, FGF12, and FGF23 [64].

Regulation by endoplasmic reticulum (ER) stress

The ER plays a prominent role in cellular homeostasis by orchestrating the synthesis, folding, maturation, and distribution of over a third of all proteins in the cell [65, 66]. A plethora of studies have linked ER stress with metabolic disturbances and diseases such as insulin resistance, type 2 diabetes, and obesity [6769]. New evidence has indicated that FGF21 gene expression is induced under conditions causing cellular stress, and the upregulation is mediated by activating transcription factor 4 (ATF4) and CCAAT enhancer binding-protein homologous protein (CHOP) [9, 7074]. For the mechanism, the phosphorylation of the α subunit of the eukaryotic initiation factor (eIF2) by protein kinase R-like endoplasmic reticulum kinase (PERK) prompts ATF4 mRNA to be translated. ATF4 translocates to the nucleus and induces transcription of the unfolded protein response (UPR) target genes. These genes include CHOP, a proapoptotic transcription factor that promotes cell death when stress conditions persist [75]. While conserved ATF4-binding sites are apparent in the FGF21 promoter [70], current evidence indicates an absence of a conserved CHOP binding site in the 5′ flanking region (–1497/+5), therefore suggesting that CHOP stabilizes FGF21 transcripts rather than induces FGF21 gene expression [74]. Alternatively, the inositol-requiring enzyme 1 α (IRE1α)/X-box binding protein 1 (XBP1) branch of the UPR was implicated in FGF21 upregulation by ER stressors tunicamycin and thapsigargin [76]. ER stress response elements (ERSE, 5′-CCATT…N(n)…CCACG) to which XBP1 can bind were identified in the human (–150/–128), mouse (–142/–108), and rat (–319/–285) FGF21 promoters [76]. The upregulation of FGF21 expression by the UPR program is viewed as a countermeasure to ER stress as FGF21 acts subsequently to suppress the eukaryotic initiation factor 2α (eIF2α)/ATF4/CHOP pathway [76].

Regulation by nutrients

Carbohydrates

FGF21 hepatic expression and circulating FGF21 levels are influenced by dietary macronutrients. High-carbohydrate diets have been shown to induce FGF21 expression. Liver FGF21 mRNA levels were upregulated in mice fed a lipogenic high-carbohydrate liquid diet rich in dextrose, but not by a high-fat obesogenic diet, although both diets induced hepatosteatosis [77]. The stimulatory role of dextrose (27.5 mM) on FGF21 gene expression had been shown earlier in isolated rat hepatocytes [78]. Hepatic FGF21 gene expression is induced not only by glucose but also by fructose, sucrose, and to a lesser extent, saccharin (an artificial non-caloric sweetener) when provided ad libitum to mice in the drinking water for 24 h [79]. Glucose, fructose, and sucrose, but not saccharin, raised plasma FGF21 levels in these mice. Similar results were observed in human subjects infused with dextrose to reproduce a state of hyperglycemia [79]. ChREBP was implicated in the upregulation of hepatic FGF21 gene expression in response to sucrose intake in mice. ChREBP binds to the ChoRE site in the human FGF21 promoter [80]. A typical ChoRE comprising two E-boxes separated by a 5-bp space exists in the mouse FGF21 promoter [81]. In humans, the ChoRE comprises two imperfect E-boxes separated by 8 bp at –380 to –366 [80]. A negative feedback loop of liver-derived FGF21 acting centrally in the hypothalamus to repress sweet-seeking behavior and reduce meal size has been proposed [79]. Counteracting the stimulation by sugars, the addition of a soybean oil-based emulsion to a high-carbohydrate dextrose-rich diet led to the suppression of FGF21 gene expression in mouse tissues and a lowering of plasma FGF21 levels [77].

Proteins (low-protein diet, methionine- or leucine-restricted diet)

FGF21 hepatic expression and circulating FGF21 are influenced by the level of dietary protein. Serum FGF21 increases on low-protein diets that typically are high in carbohydrates. Singly restricted amino acids, such as methionine [8285] or leucine [70], have been shown to increase hepatic expression and circulating levels of FGF21. Serum FGF21 also increases in the context of ketogenic diets that are high in fat, very low in carbohydrate, and moderate to low in protein. Since FGF21 is induced by both high and low carbohydrate diets, the view is that FGF21 is not regulated by carbohydrates alone but also by the reduction in protein [86]. Feeding a low-protein (5% casein) isoenergetic diet (vs. 15%–20% casein in the control diet) either short-term (8 h) in rats or longer-term (5–11 days) in mice led to an upregulation of FGF21 gene expression in the liver and an increase of FGF21 levels in plasma [87]. In humans, restricting dietary protein for 28 days caused plasma FGF21 to rise [86]. Mechanistically, the PERK/eIF2α/ATF4 branch of the UPR was implicated in FGF21 upregulation by a low-protein diet [86] and methionine restriction [84]. Dietary methionine restriction resulted in the upregulation of hepatic ATF4 expression. Subsequently, ATF4 bound to cAMP response element (CRE). The 5′ flanking region of human FGF21 has two functional ATF4 binding sites associated with amino acid response elements (AARE) [9, 70]. Disruption of these sites abolished ER stress-mediated induction of FGF21. Since leucine deprivation induces the kinase general control non-derepressible protein 2 (GCN2), the involvement of GCN2 (an upstream regulator of ATF4) in FGF21 expression has been proposed. Evidence shows that hepatic FGF21 mRNA levels are markedly decreased in GCN2-null mice fed a low-protein diet [86]; however, low dietary protein is still capable of FGF21 induction, suggesting that there might be alternative mechanisms independent of GCN2 that link low protein and FGF21 upregulation. These alternative mechanisms include PPARα, which when knocked out in mouse liver elicited low serum FGF21 concentrations and weak FGF21 induction by a low-protein diet [86].

Lipids (high-fat diet, fatty acids, retinoic acid)

High-fat diet and polyunsaturated fatty acids

Hepatic FGF21 gene expression and plasma FGF21 levels were induced in mice fed a corn oil-based high-fat diet compared to control mice fed a high-carbohydrate low-fat diet for 5 or 8 weeks [88]. In this study, mice on the high-fat diet gained more weight, consumed more food, had higher epididymal white adipose fat and blood triacylglycerols than control mice. The sustained feeding of the high-fat diet for 8 weeks led to an upregulation of FGF21 mRNA in the liver and white and brown adipose tissue. In contrast, Hao et al. [77] did not observe a significant change in hepatic FGF21 mRNA levels in mice fed a high-fat vs. low-fat diet for 16 weeks. The apparent inconsistencies suggest that high dietary fat is only one factor implicated in FGF21 upregulation and warrant continued investigation. The upregulation of FGF21 gene expression in the adipose tissue by feeding was consistent with the study of Dutchak et al. [15] that showed a meal-dependent increase in FGF21 mRNA in white adipose tissue 4 h after the initiation of a meal during the dark phase. These results illustrate the upregulation of FGF21 in liver and adipose tissue in the context of overnutrition and obesity. Supplementation of the high-fat diet with long-chain polyunsaturated n-3 fatty acids (PUFA), among which are eicosapentaenoic acid (EPA) and docosahexaenoic acid (DHA), caused a comparatively modest induction of hepatic and adipose FGF21 gene expression above that of mice fed the control high-carbohydrate low-fat diet [88]. Mice fed a PUFA-supplemented high-fat diet gained less weight than mice fed the high-fat diet, and stored less fat in the epididymal pads and interscapular depot. Their blood triacylglycerols were also lower than in mice fed the high-fat diet devoid of PUFA. From these studies, it is concluded that although PUFA ameliorated the lipid profile, the mechanism did not overtly implicate FGF21.

Short-chain fatty acids

Butyric acid is a short-chain fatty acid produced in large quantities by bacterial fermentation of dietary fiber in the large intestine. Many of butyrate’s mechanisms of action implicate the regulation of gene expression since butyric acid has histone deacetylase inhibitor (HDAC) properties. Li et al. [89] showed that butyrate induced FGF21 gene expression in the liver of dietary obese mice and in human hepatocellular carcinoma HepG2 cells. The peritoneal injection of butyrate (500 mg/kg body weight) in mice after an overnight fast induced plasma levels of FGF21 and β-hydroxybutyrate, indicating butyrate-enhanced ketogenesis in mice [89]. Other exogenously supplied short-chain fatty acids, such as β-hydroxybutyrate and hexanoate, did not induce FGF21 mRNA or FGF21 protein levels, suggesting the specific role of butyrate. Functional analysis of the FGF21 gene promoter (–1497/+5) revealed the putative involvement of a bezafibrate-responsive distal PPRE site and HDAC3 in the mechanism of butyrate. The use of small interfering RNA (siRNA) to block HDAC3 enhanced the activity of the promoter, suggesting that butyrate induced FGF21 gene expression through the inhibition of HDAC3, but not because of the short-chain fatty acid structure of butyrate. Li et al. [89] examined the regulatory role of butyrate on FGF21 in mouse epididymal fat and did not observe any effect.

α-Lipoic acid (α-LA) is a naturally occurring enzyme cofactor synthesized from octanoic acid in most prokaryotic and eukaryotic microorganisms as well as plant and animal mitochondria and plant plastids [90, 91]. α-LA can also be absorbed from foods (leafy green vegetables and red meats) and dietary supplements. Animal studies indicated that dietary supplementation of (R)-α-LA induced hepatic FGF21 expression and raised FGF21 blood levels above the levels induced by fasting alone [9294]. Moreover, feeding (R)-α-LA to rats reproduced the metabolic effects of FGF21, including the lowering of liver and blood triacylglycerol concentrations, and the decreased expression of lipogenic genes including acetyl-CoA carboxylase 1 (ACACA), fatty acid synthase (FASN), acyl-CoA desaturase 1 (SCD1), elongation of very long chain fatty acids protein 6 (ELOV6), and glycerol-3-phosphate acyltransferase 1 (GPAM) [9294]. (R)-α-LA (50 μM) stimulated the transcription and secretion of FGF21 from human hepatocellular carcinoma HepG2 cells [31]. A comparable outcome was observed in mouse hepatocyte AML12 cells treated with 1 mM α-LA [95]. In these cells, Bae et al. [95] found that α-LA increased FGF21 promoter activity by upregulating CREBH expression and increasing CREBH binding to the FGF21 promoter. The knockdown of CREBH with siRNA attenuated the α-LA-driven induction of the FGF21 gene and protein expression. In the same study, the peritoneal injection of α-LA (100 mg/kg body weight per day for 7 days) in mice induced hepatic CREBH and FGF21 transcript and protein levels, and raised serum FGF21 levels in the fed and fasted states [95]. In HepG2 cells, α-LA altered the chromatin structure of a distal 5′-flanking 400-bp region (–1524/–1124 bp) of the FGF21 gene [31]. The 400-bp fragment was enriched in the acetylated histone H3, which indicated its transcriptional competency to bind transcriptional factors. In silico analysis of putative transcription factors ranked the testis-specific bHLH-Zip transcription factor (SPZ1) as a lead candidate with three mixed E/G-boxes located at –1524/–1513, –1273/–1283, and –1161/–1151 bp in the 5′-flanking region. Reports have indicated that α-LA stimulates the cAMP/PKA signaling [96] and other protein kinases downstream of cAMP, including ERK1/2 [97, 98] and p38MAPK [99, 100]. As SPZ1 has been shown to act downstream of MAPK [101], the cAMP/p38MAPK/SPZ1 axis may play a central role in the mechanism of α-LA.

α-LA is known to induce Phase II detoxification enzymes in a nuclear factor E2-related factor 2 (Nrf2)-dependent manner by eliciting Nrf2 nuclear translocation through the enhancement of NRF2 mRNA translation and the increase of Nrf2 protein half-life [102, 103]. Nrf2 plays a central role in recruiting cellular defense systems against oxidative and electrophilic stress and in maintaining redox, lipid, and energy homeostasis. Cellular Nfr2 is sequestered in a cytosolic complex with the negative regulator Kelch-like ECH-associated protein 1 (Keap1). In one study, Nrf2 induction by the genetic knockdown of Keap1 increased hepatic FGF21 expression and plasma FGF21 levels in diabetic db/db mice and diet-induced obese mice [104]. A synthetic inducer of Nrf2 reproduced these effects in these mice. As a transcription factor, Nrf2 regulates antioxidant/electrophile responsive element (ARE/EpRE)-containing genes. Several ARE/EpREs were identified in the 5′-regulatory region of the mouse FGF21 gene [104]. However, chromatin immunoprecipitation (ChIP) analysis did not reveal significant Nrf2 binding [104].

Retinoic acid

Li et al. [30] identified FGF21 as a novel target gene of all-trans-retinoic acid (t-RA) through the action of the retinoic acid receptor (RAR). They showed that adenoviral overexpression of RARβ in mouse liver (i) stimulated hepatic mRNA and protein levels of FGF21, (ii) induced hepatic mRNA levels of carnitine palmitoyltransferase 1 α (CPT1α) (fatty acid catabolism), medium-chain acyl-CoA dehydrogenase (MCAD, a β-oxidation enzyme), 3-hydroxy-3-methylglutaryl-CoA synthase 2 (HMGCS2, a ketogenic enzyme), and 3-hydroxy-3-methylglutaryl-CoA lyase (HMGCL, also a ketogenic enzyme), (iii) increased plasma levels of ketone body and β-hydroxybutyrate, and iv) raised whole-body energy expenditure. These effects were consistent with the physiological response to fasting observed upon FGF21 administration, except that in the study of Li et al. [30], the mice were sacrificed while having free access to food. Li et al. [30] further identified a putative RAR response element (RARE) separated by one base (DR1) in the human FGF21 gene located at –645/–632, and two putative RARE sites spaced by five nucleotides (DR5) located at –537/–521 and –602/–586 in the regulatory region of the mouse FGF21 gene. The functional relevance of this RARE-like site was established when point mutations in the RARE abolished the transactivity of t-RA/RAR for the FGF21 gene. Moreover, the induction of FGF21 by the PPARα agonist, GW7647, combined with t-RA appeared to be synergistic. The knockdown of PPARα abolished GW7647-mediated induction of FGF21 gene expression, whereas the knockdown of RARβ did not, indicating that FGF21 transcription is independently upregulated by PPARα and RARβ [30].

The retinoic acid receptor-related receptors (RORs) also play an important regulatory role in lipid metabolism. RORs regulate gene expression by binding DNA as monomers to ROR response elements (ROREs) consisting of an AGGTCA half-site preceded by a 5′ AT-rich sequence. Wang et al. [105] identified a conserved classical RORE site positioned at –1563/–1554 in the 5′ regulatory region of the human FGF21 gene and –1223/–1213 upstream the mouse gene. This RORE site locates >1000-bp upstream of two additional putative proximally located RORE sites (–105/–96 and –88/–82 in the human FGF21 promoter, and –87/–78 and –69/–64 in the mouse FGF21 promoter). These proximal sites mediated REV-ERBα repression of the FGF21 gene in mouse primary hepatocytes [106].

Through their inductive and repressive activities, the first order clock-controlled genes RORs and REV-ERBs coordinate the circadian expression of FGF21 directly [105, 106] as well as indirectly through PPARα/FGF21 [107] (Figure 2). RORs and REV-ERBs are activated by the core clock genes aryl hydrocarbon receptor nuclear translocator-like protein 1 (BMAL1) and circadian locomotor output cycles kaput (CLOCK) and, in turn, provide feedback to the core clock genes BMAL1/CLOCK and period circadian protein homologue 1 (PER)/cryptochrome 1 (CRY) [108]. While the expression of FGF21 in the mouse liver exhibited a circadian rhythm, the oscillation pattern remains ill defined. Wild-type male Jcl:ICR mice had to be fed bezafibrate (a PPARα agonist) to exhibit temporal oscillation in the transcription of hepatic FGF21, which peaked at the time of the dark-to-light transition (ZT0, ZT is Zeitgeber time within a 24-h cycle composed of 12 h of light and 12 h of darkness). For comparison, the 24-h profile of FGF21 mRNA was flat in control Jcl:ICR mice not receiving bezafibrate [107]. In contrast, Tong et al. [109] found that, in C57BL6 mice, FGF21 mRNA peaked around circadian time CT6 in the first 24-h cycle and at CT12 in the second 24-h cycle. In the latter study, mice were maintained on a 12 h/12 h light/dark cycle with free access to food and water for 2 weeks prior to being transferred to the dark for 48 h. Under these conditions, mice were sacrificed every 4 h for two 24-h cycles. Hence, CT6 represents the 6-h time point after lights were turned on during the 2-week feeding. In this study, the oscillation of FGF21 mRNA was clearly anti-phase to that of E4-binding protein 4 gene (E4BP4), which encodes a bHLH-Zip transcription factor also called nuclear factor interleukin 3 regulated (NFIL3). E4BP4 has a role in the regulation of circadian rhythm by repressing the expression of a core clock gene periodic circadian protein homologue 1 (PER). E4BP4 is regulated by RORs and REV-ERBs, explaining why its circadian expression profile is in phase with that of BMAL1 and CLOCK [108, 110]. E4BP4 suppressed FGF21 gene expression by direct binding to a D-box element in the distal promoter (–1033 to –1015) [109]. In mouse liver, temporal FGF21 expression was low during the light phase and steadily increased during the dark phase (ZT12–ZT24) when the animals actively fed [111]. This profile was exacerbated in mice (TSC1lox/lox Alb-CreTg/0) in which liver mammalian target of rapamycin complex 1 (mTORC1) activity was induced by the knockdown of its repressor hamartin, also known as tuberous sclerosis 1 protein (TSC1) [111]. FGF21 expression was induced secondarily to PPARγ coactivator-1α (PGC-1α) activation in response to mTORC1-mediated depletion of glutamine in the liver. In these mice, RORα and RORγ expression displayed a circadian rhythm in phase with FGF21, while that of REV-ERBα and REV-ERBβ were anti-phase to FGF21.

Figure 2: Schematic representation of the role of first order clock-controlled genes in the circadian regulation of FGF21 gene expression.The model depicts the activating and repressing transcriptional activity of core clock protein complexes BMAL1/CLOCK and PER/CRY onto their own genes (the core circadian feedback loop) and first order clock-controlled genes (ROR, E4BP4, REV-ERB, PPARα) mediated by E-boxes. The model also depicts the activating and repressing transcriptional activity of first order clock-controlled genes onto the core clock genes and onto FGF21, a second order clock-controlled gene, mediated by RORE, D-box, and PPRE.
Figure 2:

Schematic representation of the role of first order clock-controlled genes in the circadian regulation of FGF21 gene expression.

The model depicts the activating and repressing transcriptional activity of core clock protein complexes BMAL1/CLOCK and PER/CRY onto their own genes (the core circadian feedback loop) and first order clock-controlled genes (ROR, E4BP4, REV-ERB, PPARα) mediated by E-boxes. The model also depicts the activating and repressing transcriptional activity of first order clock-controlled genes onto the core clock genes and onto FGF21, a second order clock-controlled gene, mediated by RORE, D-box, and PPRE.

Polyphenol (curcumin)

Curcumin is the major polyphenol pigment isolated from the plant Curcuma longa, commonly known as turmeric. Curcumin has a variety of pharmacologic properties. It has been demonstrated to have systemic anti-inflammatory activity and beneficial effects in several types of cancers both in animals and humans [112117], and in individuals with dermatitis [118], Crohn’s disease [119], and ulcerative colitis [120]. In addition, curcumin has been shown to improve metabolic disorders associated with hyperlipidemia and hyperglycemia [121125]. Curcumin gavage to mice stimulated FGF21 expression in liver [126]. A small dose of curcumin (2 μM) also increased FGF21 protein abundance in mouse primary hepatocytes and human hepatocellular carcinoma HepG2 cells [126]. The underlying transcriptional mechanism is currently unknown and should be a matter of further investigation.

Expert opinion

Recombinant FGF21 (rFGF21) administration is an experimental polypeptide therapy against type 2 diabetes and lipid anomalies. However, the high costs of producing rFGF21 and the mode of delivery by injection are important limitations to the wide therapeutic use of engineered FGF21. The stimulation of endogenous FGF21 production through diet should be explored as a practical and cost-effective alternative approach. Among dietary factors, high dextrose, low protein, methionine restriction, short-chain fatty acids (butyric acid and lipoic acid), and all-trans-retinoic acid have been shown to induce FGF21 expression and circulating levels. These effects, though, are usually more pronounced in liver tissue and isolated hepatocytes than they are in adipose tissue and isolated fat cells. This observation calls for further investigation of the regulation of the FGF21 gene in non-hepatic tissues by dietary and non-dietary compounds. If new approaches were to emerge from this research, they could be combined with liver-effective strategies to assess broader efficacy.

Outlook

Given that all macronutrients and a growing list of structurally diverse dietary molecules have been shown to regulate FGF21 transcription in recent years, it is likely that other dietary compounds will be shown to alter FGF21 gene expression in the future. Regulation of FGF21 transcripts by non-coding RNAs is expected to develop from its current rudimentary beginnings. It is expected that further research will be devoted to elucidate the cause and significance of interindividual variations in serum FGF21 levels among human subjects.

Highlights

  • Anti-miR-577-based approaches may boost the insulinotropic action of FGF21.

  • CREBH and PPARα synergize to activate FGF21 gene expression through the binding of the CREBH-PPARα-RXR complex.

  • Glucagon and insulin can cooperate to induce FGF21 expression in liver cells.

  • FGF21 gene expression is upregulated by the eIF2α/ATF4/CHOP and the IRE1α/XBP1 branches of the UPR.

  • PPARα and RARβ can transactivate the FGF21 gene synergistically.

  • E4BP4 (also known as NFIL3) represses FGF21 gene expression and regulates its circadian oscillation.

  • The nutritional and physiological factors influencing the response of the FGF21 gene to high-fat diet need further studies.

  • The transcriptional mechanism of butyric acid on FGF21 expression requires further investigation.

Award Identifier / Grant number: 2013-403

Funding statement: Funding was provided in part by the University of Nebraska-Lincoln ARD Hatch Act, Allen Foundation (2013-403), and USDA-NIFA (2015-05502). The authors have declared no conflict of interest.

Acknowledgments:

Funding was provided in part by the University of Nebraska-Lincoln ARD Hatch Act, Allen Foundation (2013-403), and USDA-NIFA (2015-05502). The authors have declared no conflict of interest.

References

1. Nishimura T, Nakatake Y, Konishi M, Itoh N. Identification of a novel FGF, FGF-21, preferentially expressed in the liver. Biochim Biophys Acta 2000;1492:203–6.10.1016/S0167-4781(00)00067-1Search in Google Scholar

2. Kharitonenkov A, Shanafelt AB. FGF21: a novel prospect for the treatment of metabolic diseases. Curr Opin Investig Drugs 2009;10:359–64.Search in Google Scholar

3. Fon Tacer K, Bookout AL, Ding X, Kurosu H, John GB, Wang L, Goetz R, Mohammadi M, Kuro-o M, Mangelsdorf DJ, Kliewer SA. Research resource: comprehensive expression atlas of the fibroblast growth factor system in adult mouse. Mol Endocrinol 2010;24:2050–64.10.1210/me.2010-0142Search in Google Scholar PubMed PubMed Central

4. Owen BM, Ding X, Morgan DA, Coate KC, Bookout AL, Rahmouni K, Kliewer SA, Mangelsdorf DJ. FGF21 acts centrally to induce sympathetic nerve activity, energy expenditure, and weight loss. Cell Metab 2014;20:670–7.10.1016/j.cmet.2014.07.012Search in Google Scholar PubMed PubMed Central

5. Inagaki T, Dutchak P, Zhao G, Ding X, Gautron L, Parameswara V, Li Y, Goetz R, Mohammadi M, Esser V, Elmquist JK, Gerard RD, Burgess SC, Hammer RE, Mangelsdorf DJ, Kliewer SA. Endocrine regulation of the fasting response by PPARalpha-mediated induction of fibroblast growth factor 21. Cell Metab 2007;5:415–25.10.1016/j.cmet.2007.05.003Search in Google Scholar PubMed

6. Badman MK, Pissios P, Kennedy AR, Koukos G, Flier JS, Maratos-Flier E. Hepatic fibroblast growth factor 21 is regulated by PPARalpha and is a key mediator of hepatic lipid metabolism in ketotic states. Cell Metab 2007;5y:426–37.10.1016/j.cmet.2007.05.002Search in Google Scholar PubMed

7. Lundasen T, Hunt MC, Nilsson LM, Sanyal S, Angelin B, Alexson SE, Rudling M. PPARalpha is a key regulator of hepatic FGF21. Biochem Bioph Res Co 2007;360:437–40.10.1016/j.bbrc.2007.06.068Search in Google Scholar PubMed

8. Murata Y, Nishio K, Mochiyama T, Konishi M, Shimada M, Ohta H, Itoh N. Fgf21 impairs adipocyte insulin sensitivity in mice fed a low-carbohydrate, high-fat ketogenic diet. PLoS One 2013;8:e69330.10.1371/journal.pone.0069330Search in Google Scholar PubMed PubMed Central

9. Schaap FG, Kremer AE, Lamers WH, Jansen PL, Gaemers IC. Fibroblast growth factor 21 is induced by endoplasmic reticulum stress. Biochimie 2013;95:692–9.10.1016/j.biochi.2012.10.019Search in Google Scholar PubMed

10. Iglesias P, Selgas R, Romero S, Diez JJ. Biological role, clinical significance, and therapeutic possibilities of the recently discovered metabolic hormone fibroblastic growth factor 21. Eur J Endocrinol 2012;167:301–9.10.1530/EJE-12-0357Search in Google Scholar PubMed

11. Kharitonenkov A, Shiyanova TL, Koester A, Ford AM, Micanovic R, Galbreath EJ, Sandusky GE, Hammond LJ, Moyers JS, Owens RA, Gromada J, Brozinick JT, Hawkins ED, Wroblewski VJ, Li DS, Mehrbod F, Jaskunas SR, Shanafelt AB. FGF-21 as a novel metabolic regulator. J Clin Invest 2005;115:1627–35.10.1172/JCI23606Search in Google Scholar PubMed PubMed Central

12. Kharitonenkov A, Wroblewski VJ, Koester A, Chen YF, Clutinger CK, Tigno XT, Hansen BC, Shanafelt AB, Etgen GJ. The metabolic state of diabetic monkeys is regulated by fibroblast growth factor-21. Endocrinology 2007;148:774–81.10.1210/en.2006-1168Search in Google Scholar PubMed

13. Moyers JS, Shiyanova TL, Mehrbod F, Dunbar JD, Noblitt TW, Otto KA, Reifel-Miller A, Kharitonenkov A. Molecular determinants of FGF-21 activity-synergy and cross-talk with PPARgamma signaling. J Cell Physiol 2007;210:1–6.10.1002/jcp.20847Search in Google Scholar PubMed

14. Fisher FM, Estall JL, Adams AC, Antonellis PJ, Bina HA, Flier JS, Kharitonenkov A, Spiegelman BM, Maratos-Flier E. Integrated regulation of hepatic metabolism by fibroblast growth factor 21 (FGF21) in vivo. Endocrinology 2011;152:2996–3004.10.1210/en.2011-0281Search in Google Scholar PubMed PubMed Central

15. Dutchak PA, Katafuchi T, Bookout AL, Choi JH, Yu RT, Mangelsdorf DJ, Kliewer SA. Fibroblast growth factor-21 regulates PPARgamma activity and the antidiabetic actions of thiazolidinediones. Cell 2012;148:556–67.10.1016/j.cell.2011.11.062Search in Google Scholar PubMed PubMed Central

16. Fisher FM, Kleiner S, Douris N, Fox EC, Mepani RJ, Verdeguer F, Wu J, Kharitonenkov A, Flier JS, Maratos-Flier E, Spiegelman BM. FGF21 regulates PGC-1alpha and browning of white adipose tissues in adaptive thermogenesis. Genes Dev 2012;26:271–81.10.1101/gad.177857.111Search in Google Scholar PubMed PubMed Central

17. Potthoff MJ, Kliewer SA, Mangelsdorf DJ. Endocrine fibroblast growth factors 15/19 and 21: from feast to famine. Genes Dev 2012;26:312–24.10.1101/gad.184788.111Search in Google Scholar PubMed PubMed Central

18. Kharitonenkov A, Beals JM, Micanovic R, Strifler BA, Rathnachalam R, Wroblewski VJ, Li S, Koester A, Ford AM, Coskun T, Dunbar JD, Cheng CC, Frye CC, Bumol TF, Moller DE. Rational design of a fibroblast growth factor 21-based clinical candidate, LY2405319. PLoS One 2013;8:e58575.10.1371/journal.pone.0058575Search in Google Scholar PubMed PubMed Central

19. Adams AC, Halstead CA, Hansen BC, Irizarry AR, Martin JA, Myers SR, Reynolds VL, Smith HW, Wroblewski VJ, Kharitonenkov A. LY2405319, an Engineered FGF21 Variant, Improves the Metabolic Status of Diabetic Monkeys. PLoS One 2013;8:e65763.10.1371/journal.pone.0065763Search in Google Scholar PubMed PubMed Central

20. Gaich G, Chien JY, Fu H, Glass LC, Deeg MA, Holland WL, Kharitonenkov A, Bumol T, Schilske HK, Moller DE. The effects of LY2405319, an FGF21 analog, in obese human subjects with type 2 diabetes. Cell Metab 2013;18:333–40.10.1016/j.cmet.2013.08.005Search in Google Scholar PubMed

21. Bae KH, Kim JG, Park KG. Transcriptional regulation of fibroblast growth factor 21 expression. Endocrinol Metab (Seoul) 2014;29:105–11.10.3803/EnM.2014.29.2.105Search in Google Scholar PubMed PubMed Central

22. Galman C, Lundasen T, Kharitonenkov A, Bina HA, Eriksson M, Hafstrom I, Dahlin M, Amark P, Angelin B, Rudling M. The circulating metabolic regulator FGF21 is induced by prolonged fasting and PPARalpha activation in man. Cell Metab 2008;8:169–74.10.1016/j.cmet.2008.06.014Search in Google Scholar PubMed

23. Hafstrom I, Ringertz B, Gyllenhammar H, Palmblad J, Harms-Ringdahl M. Effects of fasting on disease activity, neutrophil function, fatty acid composition, and leukotriene biosynthesis in patients with rheumatoid arthritis. Arthritis Rheum 1988;31:585–92.10.1002/art.1780310502Search in Google Scholar PubMed

24. Mraz M, Bartlova M, Lacinova Z, Michalsky D, Kasalicky M, Haluzikova D, Matoulek M, Dostalova I, Humenanska V, Haluzik M. Serum concentrations and tissue expression of a novel endocrine regulator fibroblast growth factor-21 in patients with type 2 diabetes and obesity. Clin Endocrinol (Oxf) 2009;71:369–75.10.1111/j.1365-2265.2008.03502.xSearch in Google Scholar PubMed

25. Dushay J, Chui PC, Gopalakrishnan GS, Varela-Rey M, Crawley M, Fisher FM, Badman MK, Martinez-Chantar ML, Maratos-Flier E. Increased fibroblast growth factor 21 in obesity and nonalcoholic fatty liver disease. Gastroenterology 2010;139:456–63.10.1053/j.gastro.2010.04.054Search in Google Scholar PubMed PubMed Central

26. Li H, Fang Q, Gao F, Fan J, Zhou J, Wang X, Zhang H, Pan X, Bao Y, Xiang K, Xu A, Jia W. Fibroblast growth factor 21 levels are increased in nonalcoholic fatty liver disease patients and are correlated with hepatic triglyceride. J Hepatol 2010;53:934–40.10.1016/j.jhep.2010.05.018Search in Google Scholar PubMed

27. Yilmaz Y, Eren F, Yonal O, Kurt R, Aktas B, Celikel CA, Ozdogan O, Imeryuz N, Kalayci C, Avsar E. Increased serum FGF21 levels in patients with nonalcoholic fatty liver disease. Eur J Clin Invest 2010;40:887–92.10.1111/j.1365-2362.2010.02338.xSearch in Google Scholar PubMed

28. Zhang X, Yeung DC, Karpisek M, Stejskal D, Zhou ZG, Liu F, Wong RL, Chow WS, Tso AW, Lam KS, Xu A. Serum FGF21 levels are increased in obesity and are independently associated with the metabolic syndrome in humans. Diabetes 2008;57:1246–53.10.2337/db07-1476Search in Google Scholar PubMed

29. 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;32:1542–6.10.2337/dc09-0684Search in Google Scholar PubMed PubMed Central

30. Li Y, Wong K, Walsh K, Gao B, Zang M. Retinoic acid receptor beta stimulates hepatic induction of fibroblast growth factor 21 to promote fatty acid oxidation and control whole-body energy homeostasis in mice. J Biol Chem 2013;288:10490–504.10.1074/jbc.M112.429852Search in Google Scholar PubMed PubMed Central

31. Xia M, Erickson A, Yi X, Moreau R. Mapping the response of human fibroblast growth factor 21 (FGF21) promoter to serum availability and lipoic acid in HepG2 hepatoma cells. Biochim Biophys Acta 2016;1860:498–507.10.1016/j.bbagen.2015.12.004Search in Google Scholar PubMed

32. Benson DA, Cavanaugh M, Clark K, Karsch-Mizrachi I, Lipman DJ, Ostell J, Sayers EW. GenBank. Nucleic Acids Res 2013;41(Database issue):D36–42.10.1093/nar/gks1195Search in Google Scholar PubMed PubMed Central

33. Pruitt KD, Brown GR, Hiatt SM, Thibaud-Nissen F, Astashyn A, Ermolaeva O, Farrell CM, Hart J, Landrum MJ, McGarvey KM, Murphy MR, O’Leary NA, Pujar S, Rajput B, Rangwala SH, Riddick LD, Shkeda A, Sun H, Tamez P, Tully RE, Wallin C, Webb D, Weber J, Wu W, DiCuccio M, Kitts P, Maglott DR, Murphy TD, Ostell JM. RefSeq: an update on mammalian reference sequences. Nucleic Acids Res 2014;42(Database issue):D756–63.10.1093/nar/gkt1114Search in Google Scholar PubMed PubMed Central

34. Sievers F, Wilm A, Dineen D, Gibson TJ, Karplus K, Li W, Lopez R, McWilliam H, Remmert M, Soding J, Thompson JD, Higgins DG. Fast, scalable generation of high-quality protein multiple sequence alignments using Clustal Omega. Mol Syst Biol 2011;7:539.10.1038/msb.2011.75Search in Google Scholar PubMed PubMed Central

35. Herrero J, Muffato M, Beal K, Fitzgerald S, Gordon L, Pignatelli M, Vilella AJ, Searle SM, Amode R, Brent S, Spooner W, Kulesha E, Yates A, Flicek P. Ensembl comparative genomics resources. Database (Oxford) 2016;2016:1–7.10.1093/database/baw053Search in Google Scholar PubMed PubMed Central

36. Habegger KM, Stemmer K, Cheng C, Muller TD, Heppner KM, Ottaway N, Holland J, Hembree JL, Smiley D, Gelfanov V, Krishna R, Arafat AM, Konkar A, Belli S, Kapps M, Woods SC, Hofmann SM, D’Alessio D, Pfluger PT, Perez-Tilve D, Seeley RJ, Konishi M, Itoh N, Kharitonenkov A, Spranger J, DiMarchi RD, Tschop MH. Fibroblast growth factor 21 mediates specific glucagon actions. Diabetes 2013;62:1453–63.10.2337/db12-1116Search in Google Scholar PubMed PubMed Central

37. He Z, Jiang T, Wang Z, Levi M, Li J. Modulation of carbohydrate response element-binding protein gene expression in 3T3-L1 adipocytes and rat adipose tissue. Am J Physiol Endocrinol Metab 2004;287:E424–30.10.1152/ajpendo.00568.2003Search in Google Scholar PubMed

38. Zhang F, Yu L, Lin X, Cheng P, He L, Li X, Lu X, Tan Y, Yang H, Cai L, Zhang C. Minireview: Roles of Fibroblast Growth Factors 19 and 21 in Metabolic Regulation and Chronic Diseases. Mol Endocrinol 2015;29:1400–13.10.1210/me.2015-1155Search in Google Scholar PubMed PubMed Central

39. Muise ES, Azzolina B, Kuo DW, El-Sherbeini M, Tan Y, Yuan X, Mu J, Thompson JR, Berger JP, Wong KK. Adipose fibroblast growth factor 21 is up-regulated by peroxisome proliferator-activated receptor gamma and altered metabolic states. Mol Pharmacol 2008;74:403–12.10.1124/mol.108.044826Search in Google Scholar PubMed

40. Wang H, Qiang L, Farmer SR. Identification of a domain within peroxisome proliferator-activated receptor gamma regulating expression of a group of genes containing fibroblast growth factor 21 that are selectively repressed by SIRT1 in adipocytes. Mol Cell Biol 2008;28:188–200.10.1128/MCB.00992-07Search in Google Scholar PubMed PubMed Central

41. Andersen B, Omar BA, Rakipovski G, Raun K, Ahren B. Fibroblast growth factor 21 prevents glycemic deterioration in insulin deficient mouse models of diabetes. Eur J Pharmacol 2015;764:189–94.10.1016/j.ejphar.2015.07.003Search in Google Scholar PubMed

42. Wang WF, Li SM, Ren GP, Zheng W, Lu YJ, Yu YH, Xu WJ, Li TH, Zhou LH, Liu Y, Li DS. Recombinant murine fibroblast growth factor 21 ameliorates obesity-related inflammation in monosodium glutamate-induced obesity rats. Endocrine 2015;49:119–29.10.1007/s12020-014-0433-5Search in Google Scholar PubMed

43. Berglund ED, Kang L, Lee-Young RS, Hasenour CM, Lustig DG, Lynes SE, Donahue EP, Swift LL, Charron MJ, Wasserman DH. Glucagon and lipid interactions in the regulation of hepatic AMPK signaling and expression of PPARalpha and FGF21 transcripts in vivo. Am J Physiol Endocrinol Metab 2010;299:E607–14.10.1152/ajpendo.00263.2010Search in Google Scholar PubMed PubMed Central

44. Cyphert HA, Alonge KM, Ippagunta SM, Hillgartner FB. Glucagon stimulates hepatic FGF21 secretion through a PKA- and EPAC-dependent posttranscriptional mechanism. PLoS One 2014;9:e94996.10.1371/journal.pone.0094996Search in Google Scholar PubMed PubMed Central

45. Cullen KA, McCool J, Anwer MS, Webster CR. Activation of cAMP-guanine exchange factor confers PKA-independent protection from hepatocyte apoptosis. Am J Physiol Gastrointest Liver Physiol 2004;287:G334–43.10.1152/ajpgi.00517.2003Search in Google Scholar PubMed

46. Gloerich M, Bos JL. Epac: defining a new mechanism for cAMP action. Annu Rev Pharmacol Toxicol 2010;50:355–75.10.1146/annurev.pharmtox.010909.105714Search in Google Scholar PubMed

47. Danno H, Ishii KA, Nakagawa Y, Mikami M, Yamamoto T, Yabe S, Furusawa M, Kumadaki S, Watanabe K, Shimizu H, Matsuzaka T, Kobayashi K, Takahashi A, Yatoh S, Suzuki H, Yamada N, Shimano H. The liver-enriched transcription factor CREBH is nutritionally regulated and activated by fatty acids and PPARalpha. Biochem Biophys Res Commun 2010;391:1222–7.10.1016/j.bbrc.2009.12.046Search in Google Scholar PubMed

48. Lee MW, Chanda D, Yang J, Oh H, Kim SS, Yoon YS, Hong S, Park KG, Lee IK, Choi CS, Hanson RW, Choi HS, Koo SH. Regulation of hepatic gluconeogenesis by an ER-bound transcription factor, CREBH. Cell Metab 2010;11:331–9.10.1016/j.cmet.2010.02.016Search in Google Scholar PubMed

49. Kim H, Mendez R, Chen X, Fang D, Zhang K. Lysine Acetylation of CREBH Regulates Fasting-Induced Hepatic Lipid Metabolism. Mol Cell Biol 2015;35:4121–34.10.1128/MCB.00665-15Search in Google Scholar PubMed PubMed Central

50. Kim H, Mendez R, Zheng Z, Chang L, Cai J, Zhang R, Zhang K. Liver-enriched transcription factor CREBH interacts with peroxisome proliferator-activated receptor alpha to regulate metabolic hormone FGF21. Endocrinology 2014;155:769–82.10.1210/en.2013-1490Search in Google Scholar PubMed PubMed Central

51. Pilkis SJ, el-Maghrabi MR, Claus TH. Hormonal regulation of hepatic gluconeogenesis and glycolysis. Annu Rev Biochem 1988;57:755–83.10.1146/annurev.bi.57.070188.003543Search in Google Scholar PubMed

52. Uebanso T, Taketani Y, Fukaya M, Sato K, Takei Y, Sato T, Sawada N, Amo K, Harada N, Arai H, Yamamoto H, Takeda E. Hypocaloric high-protein diet improves fatty liver and hypertriglyceridemia in sucrose-fed obese rats via two pathways. Am J Physiol Endocrinol Metab 2009;297:E76–84.10.1152/ajpendo.00014.2009Search in Google Scholar PubMed

53. Izumiya Y, Bina HA, Ouchi N, Akasaki Y, Kharitonenkov A, Walsh K. FGF21 is an Akt-regulated myokine. FEBS Lett 2008;582:3805–10.10.1016/j.febslet.2008.10.021Search in Google Scholar PubMed PubMed Central

54. Zhao AZ, Shinohara MM, Huang D, Shimizu M, Eldar-Finkelman H, Krebs EG, Beavo JA, Bornfeldt KE. Leptin induces insulin-like signaling that antagonizes cAMP elevation by glucagon in hepatocytes. J Biol Chem 2000;275:11348–54.10.1074/jbc.275.15.11348Search in Google Scholar PubMed

55. Adams AC, Astapova I, Fisher FM, Badman MK, Kurgansky KE, Flier JS, Hollenberg AN, Maratos-Flier E. Thyroid hormone regulates hepatic expression of fibroblast growth factor 21 in a PPARalpha-dependent manner. J Biol Chem 2010;285:14078–82.10.1074/jbc.C110.107375Search in Google Scholar PubMed PubMed Central

56. Rottiers V, Naar AM. MicroRNAs in metabolism and metabolic disorders. Nat Rev Mol Cell Biol 2012;13:239–50.10.1038/nrm3313Search in Google Scholar PubMed PubMed Central

57. Wente W, Efanov AM, Brenner M, Kharitonenkov A, Koster A, Sandusky GE, Sewing S, Treinies I, Zitzer H, Gromada J. Fibroblast growth factor-21 improves pancreatic beta-cell function and survival by activation of extracellular signal-regulated kinase 1/2 and Akt signaling pathways. Diabetes 2006;55:2470–8.10.2337/db05-1435Search in Google Scholar PubMed

58. Chen XY, Li GM, Dong Q, Peng H. miR-577 inhibits pancreatic beta-cell function and survival by targeting fibroblast growth factor 21 (FGF-21) in pediatric diabetes. Genet Mol Res 2015;14:15462–70.10.4238/2015.November.30.24Search in Google Scholar PubMed

59. Agarwal V, Bell GW, Nam JW, Bartel DP. Predicting effective microRNA target sites in mammalian mRNAs. eLife 2015;4:e05005.10.7554/eLife.05005Search in Google Scholar PubMed PubMed Central

60. Tang Y, Banan A, Forsyth CB, Fields JZ, Lau CK, Zhang LJ, Keshavarzian A. Effect of alcohol on miR-212 expression in intestinal epithelial cells and its potential role in alcoholic liver disease. Alcohol Clin Exp Res 2008;32:355–64.10.1111/j.1530-0277.2007.00584.xSearch in Google Scholar PubMed

61. Pulakat L, Aroor AR, Gul R, Sowers JR. Cardiac insulin resistance and microRNA modulators. Exp Diabetes Res 2012;2012:654904.10.1155/2012/654904Search in Google Scholar PubMed PubMed Central

62. Xiao J, Bei Y, Liu J, Dimitrova-Shumkovska J, Kuang D, Zhou Q, Li J, Yang Y, Xiang Y, Wang F, Yang C, Yang W. miR-212 downregulation contributes to the protective effect of exercise against non-alcoholic fatty liver via targeting FGF-21. J Cell Mol Med 2016;20:204–16.10.1111/jcmm.12733Search in Google Scholar PubMed PubMed Central

63. Wong N, Wang X. miRDB: an online resource for microRNA target prediction and functional annotations. Nucleic Acids Res 2015;43(Database issue):D146–52.10.1093/nar/gku1104Search in Google Scholar PubMed PubMed Central

64. Kozomara A, Griffiths-Jones S. miRBase: annotating high confidence microRNAs using deep sequencing data. Nucleic Acids Res 2014;42(Database issue):D68–73.10.1093/nar/gkt1181Search in Google Scholar PubMed PubMed Central

65. Anelli T, Sitia R. Protein quality control in the early secretory pathway. EMBO J 2008;27:315–27.10.1038/sj.emboj.7601974Search in Google Scholar PubMed PubMed Central

66. Oakes SA, Papa FR. The role of endoplasmic reticulum stress in human pathology. Annu Rev Pathol 2015;10:173–94.10.1146/annurev-pathol-012513-104649Search in Google Scholar PubMed PubMed Central

67. Ozcan U, Cao Q, Yilmaz E, Lee AH, Iwakoshi NN, Ozdelen E, Tuncman G, Gorgun C, Glimcher LH, Hotamisligil GS. Endoplasmic reticulum stress links obesity, insulin action, and type 2 diabetes. Science 2004;306:457–61.10.1126/science.1103160Search in Google Scholar PubMed

68. Hotamisligil GS. Endoplasmic reticulum stress and the inflammatory basis of metabolic disease. Cell 2010;140:900–17.10.1016/j.cell.2010.02.034Search in Google Scholar PubMed PubMed Central

69. Ozcan L, Tabas I. Role of endoplasmic reticulum stress in metabolic disease and other disorders. Annu Rev Med 2012;63:317–28.10.1146/annurev-med-043010-144749Search in Google Scholar PubMed PubMed Central

70. De Sousa-Coelho AL, Marrero PF, Haro D. Activating transcription factor 4-dependent induction of FGF21 during amino acid deprivation. Biochem J 2012;443:165–71.10.1042/BJ20111748Search in Google Scholar PubMed

71. Kim KH, Jeong YT, Oh H, Kim SH, Cho JM, Kim YN, Kim SS, Kim do H, Hur KY, Kim HK, Ko T, Han J, Kim HL, Kim J, Back SH, Komatsu M, Chen H, Chan DC, Konishi M, Itoh N, Choi CS, Lee MS. Autophagy deficiency leads to protection from obesity and insulin resistance by inducing Fgf21 as a mitokine. Nat Med 2013;19:83–92.10.1038/nm.3014Search in Google Scholar PubMed

72. Jiang X, Zhang C, Xin Y, Huang Z, Tan Y, Huang Y, Wang Y, Feng W, Li X, Li W, Qu Y, Cai L. Protective effect of FGF21 on type 1 diabetes-induced testicular apoptotic cell death probably via both mitochondrial- and endoplasmic reticulum stress-dependent pathways in the mouse model. Toxicol Lett 2013;219:65–76.10.1016/j.toxlet.2013.02.022Search in Google Scholar PubMed

73. Luo Y, McKeehan WL. Stressed Liver and Muscle Call on Adipocytes with FGF21. Front Endocrinol (Lausanne) 2013;4:194.10.3389/fendo.2013.00194Search in Google Scholar PubMed PubMed Central

74. Wan XS, Lu XH, Xiao YC, Lin Y, Zhu H, Ding T, Yang Y, Huang Y, Zhang Y, Liu YL, Xu ZM, Xiao J, Li XK. ATF4- and CHOP-dependent induction of FGF21 through endoplasmic reticulum stress. Biomed Res Int 2014;2014:807874.10.1155/2014/807874Search in Google Scholar PubMed PubMed Central

75. Basseri S, Austin RC. Endoplasmic reticulum stress and lipid metabolism: mechanisms and therapeutic potential. Biochem Res Int 2012;2012:841362.10.1155/2012/841362Search in Google Scholar PubMed PubMed Central

76. Jiang S, Yan C, Fang QC, Shao ML, Zhang YL, Liu Y, Deng YP, Shan B, Liu JQ, Li HT, Yang L, Zhou J, Dai Z, Jia WP. Fibroblast growth factor 21 is regulated by the IRE1alpha-XBP1 branch of the unfolded protein response and counteracts endoplasmic reticulum stress-induced hepatic steatosis. J Biol Chem 2014;289:29751–65.10.1074/jbc.M114.565960Search in Google Scholar PubMed PubMed Central

77. Hao L, Huang KH, Ito K, Sae-Tan S, Lambert JD, Ross AC. Fibroblast Growth Factor 21 (Fgf21) Gene Expression Is Elevated in the Liver of Mice Fed a High-Carbohydrate Liquid Diet and Attenuated by a Lipid Emulsion but Is Not Upregulated in the Liver of Mice Fed a High-Fat Obesogenic Diet. J Nutr 2016;146:184–90.10.3945/jn.115.216572Search in Google Scholar PubMed PubMed Central

78. Ma L, Robinson LN, Towle HC. ChREBP*Mlx is the principal mediator of glucose-induced gene expression in the liver. J Biol Chem 2006;281:28721–30.10.1074/jbc.M601576200Search in Google Scholar PubMed

79. von Holstein-Rathlou S, BonDurant LD, Peltekian L, Naber MC, Yin TC, Claflin KE, Urizar AI, Madsen AN, Ratner C, Holst B, Karstoft K, Vandenbeuch A, Anderson CB, Cassell MD, Thompson AP, Solomon TP, Rahmouni K, Kinnamon SC, Pieper AA, Gillum MP, Potthoff MJ. FGF21 Mediates Endocrine Control of Simple Sugar Intake and Sweet Taste Preference by the Liver. Cell Metab 2016;23:335–43.10.1016/j.cmet.2015.12.003Search in Google Scholar PubMed PubMed Central

80. Uebanso T, Taketani Y, Yamamoto H, Amo K, Ominami H, Arai H, Takei Y, Masuda M, Tanimura A, Harada N, Yamanaka-Okumura H, Takeda E. Paradoxical regulation of human FGF21 by both fasting and feeding signals: is FGF21 a nutritional adaptation factor? PLoS One 2011;6:e22976.10.1371/journal.pone.0022976Search in Google Scholar PubMed PubMed Central

81. Iizuka K, Takeda J, Horikawa Y. Glucose induces FGF21 mRNA expression through ChREBP activation in rat hepatocytes. FEBS Lett 2009;583:2882–6.10.1016/j.febslet.2009.07.053Search in Google Scholar PubMed

82. Ables GP, Perrone CE, Orentreich D, Orentreich N. Methionine-restricted C57BL/6J mice are resistant to diet-induced obesity and insulin resistance but have low bone density. PLoS One 2012;7:e51357.10.1371/journal.pone.0051357Search in Google Scholar

83. Perrone CE, Mattocks DA, Plummer JD, Chittur SV, Mohney R, Vignola K, Orentreich DS, Orentreich N. Genomic and metabolic responses to methionine-restricted and methionine-restricted, cysteine-supplemented diets in Fischer 344 rat inguinal adipose tissue, liver and quadriceps muscle. J Nutrigenet Nutrigenomics 2012;5:132–57.10.1159/000339347Search in Google Scholar

84. Lees EK, Krol E, Grant L, Shearer K, Wyse C, Moncur E, Bykowska AS, Mody N, Gettys TW, Delibegovic M. Methionine restriction restores a younger metabolic phenotype in adult mice with alterations in fibroblast growth factor 21. Aging Cell 2014;13:817–27.10.1111/acel.12238Search in Google Scholar

85. Stone KP, Wanders D, Orgeron M, Cortez CC, Gettys TW. Mechanisms of increased in vivo insulin sensitivity by dietary methionine restriction in mice. Diabetes 2014;63:3721–33.10.2337/db14-0464Search in Google Scholar

86. Laeger T, Henagan TM, Albarado DC, Redman LM, Bray GA, Noland RC, Munzberg H, Hutson SM, Gettys TW, Schwartz MW, Morrison CD. FGF21 is an endocrine signal of protein restriction. J Clin Invest 2014;124:3913–22.10.1172/JCI74915Search in Google Scholar

87. Ozaki Y, Saito K, Nakazawa K, Konishi M, Itoh N, Hakuno F, Takahashi S, Kato H, Takenaka A. Rapid increase in fibroblast growth factor 21 in protein malnutrition and its impact on growth and lipid metabolism. Br J Nutr 2015;114:1410–8.10.1017/S0007114515002846Search in Google Scholar

88. Villarroya J, Flachs P, Redondo-Angulo I, Giralt M, Medrikova D, Villarroya F, Kopecky J, Planavila A. Fibroblast growth factor-21 and the beneficial effects of long-chain n-3 polyunsaturated fatty acids. Lipids 2014;49:1081–9.10.1007/s11745-014-3948-xSearch in Google Scholar

89. Li H, Gao Z, Zhang J, Ye X, Xu A, Ye J, Jia W. Sodium Butyrate Stimulates Expression of Fibroblast Growth Factor 21 in Liver by Inhibition of Histone Deacetylase 3. Diabetes 2012;61:797–806.10.2337/db11-0846Search in Google Scholar

90. Herbert AA, Guest JR. Lipoic acid content of Escherichia coli and other microorganisms. Arch Microbiol 1975;106:259–66.10.1007/BF00446532Search in Google Scholar

91. Yasuno R, Wada H. The biosynthetic pathway for lipoic acid is present in plastids and mitochondria in Arabidopsis thaliana. FEBS Lett 2002;517:110–4.10.1016/S0014-5793(02)02589-9Search in Google Scholar

92. Finlay LA, Michels AJ, Butler JA, Smith EJ, Monette JS, Moreau RF, Petersen SK, Frei B, Hagen TM. R-alpha-lipoic acid does not reverse hepatic inflammation of aging, but lowers lipid anabolism, while accentuating circadian rhythm transcript profiles. Am J Physiol Regul Integr Comp Physiol 2012;302:R587–97.10.1152/ajpregu.00393.2011Search in Google Scholar PubMed PubMed Central

93. Pashaj A, Yi X, Xia M, Canny S, Riethoven JJ, Moreau R. Characterization of genome-wide transcriptional changes in liver and adipose tissues of ZDF (fa/fa) rats fed R-alpha-lipoic acid by next-generation sequencing. Physiol Genomics 2013;45:1136–43.10.1152/physiolgenomics.00138.2013Search in Google Scholar PubMed

94. Yi X, Pashaj A, Xia M, Moreau R. Reversal of obesity-induced hypertriglyceridemia by (R)-α-lipoic acid in ZDF (fa/fa) rats. Biochem Bioph Res Co 2013;439:390–95.10.1016/j.bbrc.2013.08.063Search in Google Scholar PubMed

95. Bae KH, Min AK, Kim JG, Lee IK, Park KG. Alpha lipoic acid induces hepatic fibroblast growth factor 21 expression via up-regulation of CREBH. Biochem Biophys Res Commun 2014;455:212–7.10.1016/j.bbrc.2014.10.147Search in Google Scholar PubMed

96. Salinthone S, Schillace RV, Marracci GH, Bourdette DN, Carr DW. Lipoic acid stimulates cAMP production via the EP2 and EP4 prostanoid receptors and inhibits IFN gamma synthesis and cellular cytotoxicity in NK cells. J Neuroimmunol 2008;199:46–55.10.1016/j.jneuroim.2008.05.003Search in Google Scholar PubMed PubMed Central

97. Suzuki YJ, Shi SS, Day RM, Blumberg JB. Differential regulation of MAP kinase signaling by pro- and antioxidant biothiols. Ann NY Acad Sci 2000;899:159–67.10.1111/j.1749-6632.2000.tb06184.xSearch in Google Scholar PubMed

98. Shi SS, Day RM, Halpner AD, Blumberg JB, Suzuki YJ. Homocysteine and alpha-lipoic acid regulate p44/42 MAP kinase phosphorylation in NIH/3T3 cells. Antioxid Redox Sign 1999;1:123–8.10.1089/ars.1999.1.1-123Search in Google Scholar PubMed

99. Konrad D, Somwar R, Sweeney G, Yaworsky K, Hayashi M, Ramlal T, Klip A. The antihyperglycemic drug alpha-lipoic acid stimulates glucose uptake via both GLUT4 translocation and GLUT4 activation: potential role of p38 mitogen-activated protein kinase in GLUT4 activation. Diabetes 2001;50:1464–71.10.2337/diabetes.50.6.1464Search in Google Scholar PubMed

100. Delghandi MP, Johannessen M, Moens U. The cAMP signalling pathway activates CREB through PKA, p38 and MSK1 in NIH 3T3 cells. Cell Signal 2005;17:1343–51.10.1016/j.cellsig.2005.02.003Search in Google Scholar PubMed

101. Hsu SH, Hsieh-Li HM, Huang HY, Huang PH, Li H. bHLH-zip transcription factor Spz1 mediates mitogen-activated protein kinase cell proliferation, transformation, and tumorigenesis. Cancer Res 2005;65:4041–50.10.1158/0008-5472.CAN-04-3658Search in Google Scholar PubMed

102. Shay KP, Moreau RF, Smith EJ, Smith AR, Hagen TM. Alpha-lipoic acid as a dietary supplement: molecular mechanisms and therapeutic potential. Biochim Biophys Acta 2009;1790:1149–60.10.1016/j.bbagen.2009.07.026Search in Google Scholar PubMed PubMed Central

103. Shay KP, Michels AJ, Li W, Kong AN, Hagen TM. Cap-independent Nrf2 translation is part of a lipoic acid-stimulated detoxification stress response. Biochimica Biophy Acta 2012;1823:1102–9.10.1016/j.bbamcr.2012.04.002Search in Google Scholar PubMed PubMed Central

104. Furusawa Y, Uruno A, Yagishita Y, Higashi C, Yamamoto M. Nrf2 induces fibroblast growth factor 21 in diabetic mice. Genes Cells 2014;19:864–78.10.1111/gtc.12186Search in Google Scholar PubMed

105. Wang Y, Solt LA, Burris TP. Regulation of FGF21 expression and secretion by retinoic acid receptor-related orphan receptor alpha. J Biol Chem 2010;285:15668–73.10.1074/jbc.M110.102160Search in Google Scholar PubMed PubMed Central

106. Estall JL, Ruas JL, Choi CS, Laznik D, Badman M, Maratos-Flier E, Shulman GI, Spiegelman BM. PGC-1alpha negatively regulates hepatic FGF21 expression by modulating the heme/Rev-Erb(alpha) axis. Proc Natl Acad Sci USA 2009;106:22510–5.10.1073/pnas.0912533106Search in Google Scholar PubMed PubMed Central

107. Oishi K, Uchida D, Ishida N. Circadian expression of FGF21 is induced by PPARalpha activation in the mouse liver. FEBS Lett 2008;582:3639–42.10.1016/j.febslet.2008.09.046Search in Google Scholar PubMed

108. Liu AC, Tran HG, Zhang EE, Priest AA, Welsh DK, Kay SA. Redundant function of REV-ERBalpha and beta and non-essential role for Bmal1 cycling in transcriptional regulation of intracellular circadian rhythms. PLoS Genet 2008;4:e1000023.10.1371/journal.pgen.1000023Search in Google Scholar PubMed PubMed Central

109. Tong X, Muchnik M, Chen Z, Patel M, Wu N, Joshi S, Rui L, Lazar MA, Yin L. Transcriptional repressor E4-binding protein 4 (E4BP4) regulates metabolic hormone fibroblast growth factor 21 (FGF21) during circadian cycles and feeding. J Biol Chem 2010;285:36401–9.10.1074/jbc.M110.172866Search in Google Scholar PubMed PubMed Central

110. Cho H, Zhao X, Hatori M, Yu RT, Barish GD, Lam MT, Chong LW, DiTacchio L, Atkins AR, Glass CK, Liddle C, Auwerx J, Downes M, Panda S, Evans RM. Regulation of circadian behaviour and metabolism by REV-ERB-alpha and REV-ERB-beta. Nature 2012;485:123–7.10.1038/nature11048Search in Google Scholar PubMed PubMed Central

111. Cornu M, Oppliger W, Albert V, Robitaille AM, Trapani F, Quagliata L, Fuhrer T, Sauer U, Terracciano L, Hall MN. Hepatic mTORC1 controls locomotor activity, body temperature, and lipid metabolism through FGF21. Proc Natl Acad Sci USA 2014;111:11592–9.10.1073/pnas.1412047111Search in Google Scholar PubMed PubMed Central

112. Cheng KW, Wong CC, Mattheolabakis G, Xie G, Huang L, Rigas B. Curcumin enhances the lung cancer chemopreventive efficacy of phospho-sulindac by improving its pharmacokinetics. Int J Oncol 2013;43:895–902.10.3892/ijo.2013.1995Search in Google Scholar PubMed PubMed Central

113. Lin YC, Chen HW, Kuo YC, Chang YF, Lee YJ, Hwang JJ. Therapeutic efficacy evaluation of curcumin on human oral squamous cell carcinoma xenograft using multimodalities of molecular imaging. Am J Chin Med 2010;38:343–58.10.1142/S0192415X10007890Search in Google Scholar PubMed

114. Yoysungnoen P, Wirachwong P, Bhattarakosol P, Niimi H, Patumraj S. Effects of curcumin on tumor angiogenesis and biomarkers, COX-2 and VEGF, in hepatocellular carcinoma cell-implanted nude mice. Clin Hemorheol Microcirc 2006;34:109–15.Search in Google Scholar

115. Huang MT, Lou YR, Xie JG, Ma W, Lu YP, Yen P, Zhu BT, Newmark H, Ho CT. Effect of dietary curcumin and dibenzoylmethane on formation of 7,12-dimethylbenz[a]anthracene-induced mammary tumors and lymphomas/leukemias in Sencar mice. Carcinogenesis 1998;19:1697–700.10.1093/carcin/19.9.1697Search in Google Scholar PubMed

116. Perkins S, Verschoyle RD, Hill K, Parveen I, Threadgill MD, Sharma RA, Williams ML, Steward WP, Gescher AJ. Chemopreventive efficacy and pharmacokinetics of curcumin in the min/+ mouse, a model of familial adenomatous polyposis. Cancer Epidemiol Biomarkers Prev 2002;11:535–40.Search in Google Scholar

117. Pettan-Brewer C MJ, Mangalindan R, Ladiges W. Curcumin suppresses intestinal polyps in APC Min mice fed a high fat diet. Pathobiol Aging Age Relat Dis 2011;1:7013.10.3402/pba.v1i0.7013Search in Google Scholar PubMed PubMed Central

118. Ryan JL, Heckler CE, Ling M, Katz A, Williams JP, Pentland AP, Morrow GR. Curcumin for radiation dermatitis: a randomized, double-blind, placebo-controlled clinical trial of thirty breast cancer patients. Radiat Res 2013;180:34–43.10.1667/RR3255.1Search in Google Scholar PubMed PubMed Central

119. Holt PR, Katz S, Kirshoff R. Curcumin therapy in inflammatory bowel disease: a pilot study. Dig Dis Sci 2005;50:2191–3.10.1007/s10620-005-3032-8Search in Google Scholar PubMed

120. Hanai H, Sugimoto K. Curcumin has bright prospects for the treatment of inflammatory bowel disease. Curr Pharm Design 2009;15:2087–94.10.2174/138161209788489177Search in Google Scholar PubMed

121. Tang M, Larson-Meyer DE, Liebman M. Effect of cinnamon and turmeric on urinary oxalate excretion, plasma lipids, and plasma glucose in healthy subjects. Am J Clin Nutr 2008;87:1262–7.10.1093/ajcn/87.5.1262Search in Google Scholar PubMed

122. Weisberg SP, Leibel R, Tortoriello DV. Dietary curcumin significantly improves obesity-associated inflammation and diabetes in mouse models of diabesity. Endocrinology 2008;149:3549–58.10.1210/en.2008-0262Search in Google Scholar PubMed PubMed Central

123. Chuengsamarn S, Rattanamongkolgul S, Luechapudiporn R, Phisalaphong C, Jirawatnotai S. Curcumin extract for prevention of type 2 diabetes. Diabetes Care 2012;35:2121–7.10.2337/dc12-0116Search in Google Scholar PubMed PubMed Central

124. Shao W, Yu Z, Chiang Y, Yang Y, Chai T, Foltz W, Lu H, Fantus IG, Jin T. Curcumin prevents high fat diet induced insulin resistance and obesity via attenuating lipogenesis in liver and inflammatory pathway in adipocytes. PLoS One 2012;7:e28784.10.1371/journal.pone.0028784Search in Google Scholar PubMed PubMed Central

125. Yu S, Shen G, Khor TO, Kim JH, Kong AN. Curcumin inhibits Akt/mammalian target of rapamycin signaling through protein phosphatase-dependent mechanism. Mol Cancer Ther 2008;7:2609–20.10.1158/1535-7163.MCT-07-2400Search in Google Scholar PubMed PubMed Central

126. Tian L, Zeng K, Shao W, Yang BB, Fantus IG, Weng J, Jin T. Short-Term Curcumin Gavage Sensitizes Insulin Signaling in Dexamethasone-Treated C57BL/6 Mice J Nutr 2015;145:2300–7.10.3945/jn.115.216853Search in Google Scholar PubMed

Received: 2016-3-28
Accepted: 2016-5-8
Published Online: 2016-6-10

©2017 Walter de Gruyter GmbH, Berlin/Boston

Downloaded on 29.3.2024 from https://www.degruyter.com/document/doi/10.1515/hmbci-2016-0016/html
Scroll to top button