Skip to main content
Erschienen in: Journal of Translational Medicine 1/2016

Open Access 01.12.2016 | Review

Regulation of fibroblast growth factor 15/19 and 21 on metabolism: in the fed or fasted state

Erschienen in: Journal of Translational Medicine | Ausgabe 1/2016

Abstract

Fibroblast growth factor (FGF) 15/19 and FGF21 are two atypical members of FGF19 subfamily that function as hormones. Exogenous FGF15/19 and FGF21 have pharmacological effects, and endogenous FGF15/19 and FGF21 play vital roles in the maintenance of energy homeostasis. Recent reports have expanded the effects of FGF15/19 and FGF21 on carbohydrate and lipid metabolism. However, the regulations of FGF15/19 and FGF21 on metabolism are different. FGF15/19 is mainly secreted from the small intestine in response to feeding, and FGF21 is secreted from the liver in response to extended fasting and from the liver and adipose tissue in response to feeding. In this work, we reviewed the regulatory effects of FGF15/19 and FGF21 on metabolism in the fast and fed states. This information may provide some insight into the metabolic regulation of FGF15/19 and FGF21 in different physiological condition.
Hinweise
Dandan Guan and Lidan Zhao contributed equally to this work
Abkürzungen
FGF
fibroblast growth factor
GFGR
fibroblast growth factor receptor
KL
Klotho
KLB
βKlotho
GS
glycogen synthase
GSK
glycogen synthase kinase
CYP7A1
cholesterol 7a-hydroxylase
Akt
protein kinase B
PI3K
phosphatidylinositol 3-kinase
CREB
cAMP response element-binding protein
PGC-1α
peroxisome proliferator-activated receptor-1α
FXR
farnesoid X receptor
Ig
immunoglogulin
ERK
extracellular regulated protein kinases
CCK
cholecystokinin
STAT 5
signal transducer and activator of transcription 5
SHP
small heterodimer partner
PPARα
peroxisome proliferator-activated receptor α
GH
growth hormone
WAT
white adipose tissue
CPT-1a
carnitine palmitoyl transferase-1a
HMGCS2
hydroxymethyl glutaryl-CoA synthase-2

Background

Fibroblast growth factors (FGFs) are a group of structurally related polypeptides, involved in various biological processes such as neuronal functions, development, differentiation, and metabolism [13]. There have been 22 FGFs, FGF1–FGF23, identified in mouse and/or human, among which human FGF19 is the orthologous gene of mouse FGF15. The FGFs can be divided into seven subfamilies according to their gene locus, phylogenetic analyses and action modes [4].
These mammalian subfamilies are also classified into three groups based on their action mechanisms, including the intracellular FGFs, the hormone-like FGFs and the canonical FGFs [5, 6] (Table 1). The intracellular subfamily, function as nonsecreted signaling molecules and mainly plays a role in neuronal functions [7, 8]. The hormone-like subfamily, functions over long distances in an endocrine manner and mainly plays a role in metabolism [6]. Canonical FGFs, function as autocrine and/or paracrine in multiple developmental processes [2, 9, 10]. Most FGFs have a high affinity for heparin sulfate in the extracellular matrix except the endocrine FGFs, which include FGF15/19, FGF21, and FGF23, have little or no affinity for heparin sulfate [11].
Table 1
FGFs super family
FGFs
Intracellular subfamily
FGF11, FGF12, FGF13, FGF14
Endocrine subfamily
FGF15/19, FGF21, FGF23
Canonical subfamily
FGF1/2/5, FGF3/4/6, FGF7/10/22, FGF8/17/18, FGF9/16/20
Fibroblast growth factors (FGFs) are a group of proteins. Currently, the FGF family consists of 22 members that can be classified into three groups and can also be divided into 7 subfamilies
FGF15/19 is mostly secreted from the small intestine in response to feeding. The expression of FGF21 is induced in multiple organs in response to diverse nutrition stressors, such as fasting and amino acid deprivation [12]. FGF15/19 is secreted from the ileum in response to feeding, it acts as endocrine hormones and takes part in the regulation of glucose and lipid metabolism [13]. After entering the portal venous circulation, FGF15/19 represses bile acid synthesis and gluconeogenesis, promotes glycogen synthesis [14], and stimulates gallbladder filling [15]. Unlike other members in the FGF family, FGF21 is a newly discovered factor for metabolism [5], it lacks heparin-binding domain, and has no effect on promoting mitosis and proliferative activity [16, 17]. In response to fasting, FGF21 expression is induced in the liver [18, 19]. Secreted FGF21 acts as an endocrine hormone to induce ketogenesis and gluconeogenesis. In response to feeding, FGF21 expression is induced in WAT and liver [2023]. In WAT, FGF21 acts through an autocrine mechanism to stimulate PPARγ activity and glucose uptake, and via an endocrine mechanism to repress lipolysis in liver [14, 18, 24]. Therefore, we reviewed the regulatory effects of FGF15/19 and FGF21 on nutrient metabolism in the fast and fed states in the present work.

Receptors of FGF15/19 and FGF21

FGFs exert their function by binding to their tyrosine kinase receptors, FGF receptors (FGFRs). FGF receptors consist of three extracellular immunoglogulin (Ig)-like domains and a single transmembrane domain [25]. Four FGF receptors, FGFR-1 through FGFR-4, have been identified so far [26]. There are many types of FGFs, which require the diversity of FGFR. However, by alternated splicing, the same FGFR genes could generate a variety of different isoforms [27]. The most variant parts are the extracellular Ig domains. FGFR may lack one Ig domain or use different exon for the same Ig-like domains. There are three types of third Ig domains (IIIa, IIIb, and IIIc). The binding of FGF to its receptor requires proteoglycans, namely heparin or heparin sulfate [28], which protects FGF from degradation and creates a local reservoir of FGF.
In addition to FGFRs, the FGF19 subfamily members need Klotho (KL) or βKlotho (KLB) protein as a co-receptor to activate their signaling pathway [3] (Fig. 1). KL is a single-pass trans-membrane protein, which has two homologous domains in the extracellular domain and a short intracellular tail [29]. KLB is expressed in liver, adipose tissue, pancreas and muscle, whereas KL is expressed in kidney and intestine [30]. KL was first identified in mice as an age suppressor gene. A defect in KL resulted in multiple ageing-like phenotypes, and KL overexpression extends life span in mice [31]. The study with global KLB-knockout mice showed that KLB is essential for most of the physiological functions of FGF15/19 and FGF21 [30].
FGF19 has low affinity for heparin. KLB is essential for FGF15/19 interaction with FGFRs 1c, 2c, 3c and 4c [30, 32]. KLB appears to stabilize the interaction of this ligand with its receptor, perhaps acting as a surrogate for heparin [33]. FGF15/19 is also able to interact directly with FGFR4 in the absence of KLB in a heparin-dependent manner [32, 34]. Therefore, FGF19 can activate FGFR4 in a KLB-dependent or heparin-dependent manner [33]. A recent study noted that FGF15/19 binds both FGFR1 and FGFR4 in the presence of KLB with comparable affinity, but not to FGFR1 alone although there is 10 % binding to FGFR4 alone. Like FGF15/19, FGF21 binds to KLB in complex with FGFR1c, 2c, or 3c. FGF21 has much higher affinity to FGFR1 than FGFR4 in the presence of KLB [29].
It has been believed that FGF21 forms complexes with FGFR and KLB to activate downstream signaling pathways [3537]. However, in vitro experiments show that KLB is indispensable for FGF21 [35, 36]. What causes these controversial results is still unclear. Possible explanations proposed by researches were the specific characteristics of cultured cells and an artificial abundance of KLB or FGF21 by adding them into the medium [38].
Typically, FGF binding to FGFR requires heparin sulfate cofactor that limits the diffusion of FGFs from their site of release, so FGF acts as a paracrine or autocrine factor [39]. However, FGF19 subfamily members have low affinity to heparin sulfate, which allow them to enter the circulation and function as hormones [16]. They have several effects that are similar to those of insulin, including stimulation of glycogen synthesis and suppression of gluconeogenesis [13].

Role of FGF15/19 and FGF21 in metabolism

Regulation of FGF15/19 on metabolism

FGF15/19 is expressed in small intestine under the regulation of bile acid (BA) nuclear receptor farnesoid X receptor (FXR) [40, 41], and is a negative feedback regulator of BA synthesis and gallbladder filling (Fig. 2).
In response to fasting, BAs are stored in the gallbladder until they are needed for digestion normally [15]. In fed state, BAs are released from the gallbladder into the intestine, bind to and activate FXR, thereby induce expression of FGF19 [42]. In humans, serum FGF19 levels exhibited a rhythm with peaks occurring 90–120 min after the postprandial rise in serum BAs, and the FGF19 peaks in turn preceded the declining phase of BA synthesis [43].
In small intestine, BA induces FGF15/19 expression by activating FXR. FXR induces the expression of small heterodimer partner (SHP) in liver [44, 45]. However, unlike most nuclear receptors, SHP lacks a DNA-binding domain and binds indirectly to the CYP7A1 promoter [4446]. Knockout studies in FXR-KO and SHP-KO mice has been demonstrated the significance of the FXR–SHP pathway in bile acid homeostasis, both of which increase CYP7A1 expression [4749], the rate-limiting enzyme in the classical BA biosynthetic pathway [14]. FGF15/19 represses CYP7A1 by binding to the FGFR4/KLB receptor complex to activate downstream signaling cascade [43, 50]. A more recent study showed that an uncharacterized gene, Diet1, transcriptionally and post-transcriptionally influences FGF15/19 level as well as CYP7A1 level, and it co-localize with FGF19 in cultured intestinal cells [51]. This suggests that Diet1 plays a role in FGF15/19 intestine-liver axis involved in the BA synthesis. FGF15/19, a hormone made by the distal small intestine in response to BAs, also promotes relaxation and refilling of the gallbladder after a meal. Cholecystokinin (CCK) is a hormone secreted by duodenum causing gallbladder contraction to release bile, which facilitates lipid digestion. Bile acids travel to ileum, where they induce FGF15 synthesis. FGF15 in turn stimulates gallbladder filling by relaxing smooth muscle in gallbladder [15].
After a meal, besides regulation of BA synthesis and gallbladder filling, FGF15/19 has an effect on glycogen synthesis. FGF15/19 acts on FGFR/KLB receptor complexes to represses cholesterol 7a-hydroxylase (CYP7A1) through small heteromer partner [41, 52], and then increase hepatic glycogen synthase (GS) activity and glycogen synthesis in an insulin-independent manner by inducing the phosphorylation and inactivation of GSK3s [53]. Serum FGF19 levels peak approximately 3 h after a meal [43] and increase glycogen synthesis by activation of the Ras/ERK pathway; in contrast, serum insulin levels peak within 1 h after a meal and stimulate glycogen synthesis by the phosphoinositide 3-kinase (Akt) pathway [42].
In addition to glycogen synthesis, FGF15/19 also has an effect on gluconeogenesis. To date, gluconeogenesis inhibition is also differently mediated by FGF19 and insulin by dephosphorylation and inactivation of cAMP response element-binding protein (CREB) and Akt-dependent phosphorylation and FoxO1 degradation, respectively [54]. Unlike insulin, FGF15/19 represses gluconeogenesis gene expressions by promoting protein kinase B(Akt) dependent FOXO1 phosphorylation and dephosphorylation, FGF15/19 cannot activate the PI3K/Akt pathway [42]. The mechanism by which FGF15/19 blocks the expression of gluconeogenesis genes involves dephosphorylation and inactivation of the transcription factor CREB [13]. This in turn down-regulates peroxisome proliferator-activated receptor-1α (PGC1α) transcription, which subsequently decreases its binding to glucose-6-phosphatase catalytic subunit gene and phosphoenolpyruvate carborykinase gene promoters [13]. Therefore, FGF15/19 inhibits gluconeogenesis via regulating the expression of genes involved in gluconeogenesis.
In fasted status, FGF19 increased the phosphorylation of ERK1 and ERK2 in liver. FGF19 induced phosphorylation of both glycogen synthase kinase (GSK) 3α and GSK3β in animals fasted overnight, which correlated with decreased phosphorylation of Ser641 and Ser645 on glycogen synthase and increased glycogen synthase activity [53]. However, the effects of FGF15/19 and insulin on metabolism in fasted status are noticeable. Although both of them can stimulate glycogen and repress gluconeogenesis, there still are important differences as insulin acts through the insulin receptor-PI3K-Akt pathway, and FGF15/19 mediates its effects through the FGFR/KLB-ERK pathway. In addition, there are significant temporal differences. In humans, insulin is released minutes after a meal, and in rodent experiments, serum insulin concentrations and its downstream Akt phosphorylation in liver peak approximately 15 min after a high-carbohydrate or high-fat diet. In contrast, FGF15 mRNA levels in ileum and downstream hepatic ERK1/2 phosphorylation peak about 1 h after feeding [13]. Likewise, FGF19 serum levels peak near 2 h after a meal [43], and accordingly, circulating FGF19 levels in humans negatively correlate with fasting glucose levels and metabolic syndrome [5557]. Thus, FGF15/19 acts after insulin in the transition from the fed to the fasted state.

Regulation of FGF21 on metabolism

FGF21 is an important regulator of metabolism. A larger number of recent reports have expanded that FGF21 expression is induced in various tissues in response to fasting and feeding. The physiological function of FGF21 in the maintenance of nutritional homeostasis has been suggested (Fig. 3).
Emerging evidences have shown that fasting increases hepatic FGF21 mRNA expression and plasma FGF21 level in mice. Fasting mediated induction of FGF21 requires the peroxisome proliferator-activated receptor a (PPARα) [18, 58, 59]. PPARα can bind directly to the FGF21 gene promoter to induce its transcription [18]. It has been shown that fasting-induced FGF21 in liver increases gluconeogenesis, but does not increase glycogenolysis [60]. Gluconeogenesis is controlled by several key enzymes including fructose-1,6-bisphosphatase, glucose-6-phosphatase and phosphoenopyruvate carboxykinase. An acute FGF21 treatment leads to the gene expressions of hepatic glucose-6-phosphatase and phosphoenopyruvate carborykinase [61]. PGC-lα as a transcriptional coactivator regulating gluconeogenic gene is increased after FGF21 treatment [62]. PGC-1α knockout mice fail to induce glucose-6-phosphatase catalytic subunit and phosphoenopyruvate carboxykinase mRNA expression after FGF21 injection [60]. Taking together, PGC-1α may play an important role in FGF21 promoting hepatic gluconeogenesis. In contract to those findings, a study using mice with liver-specific deletion of PGC-1α reveals that liver PGC-1α is unnecessary in the effects of FGF21 on the gluconeogenesis [61]. The difference results from those two studies can be explained by the mouse models. One is a whole-body PGC-1α knockout and the other is a liver-specific knockout. It suggests that PGC-1α expressed in tissues other than liver affects FGF21 regulatory effects on the gluconeogenesis. For example, a study has shown that FGF21 induces PGC-1α via an indirect mechanism of central nervous system [63]. So the importance of PGC-1α induction for FGF21 action remains in question [60, 61].
Ketone bodies are produced in the liver and delivered to the brain as the major source of energy during fasting period. FGF21 is a molecule regulating lipid metabolism in response to fasting [18, 19]. Increased FGF21 promotes adipose lipolysis in white adipose tissue in an endocrine manner, and increases the fatty acids transport to the liver where they are directly oxidized for energy production or utilized as a source for ketone body formation [18]. FGF21 transgenic mice have higher serum concentrations of β-hydroxybutyrate even in the fed state [18]. PPARα is responsible for coordinating lipid oxidation and ketogenesis in the liver during starvation. Although FGF21 is one of PPARα target genes, FGF21 induces ketone body production through a mechanism distinct from that previously described for PPARα. As we mentioned above, FGF21 induces ketogenesis by stimulating lipolysis, thereby increasing the supply of free fatty acids to the liver [18, 24]. Both carnitine palmitoyl transferase-1a (CPT-1a) and hydroxymethyl glutaryl-CoA synthase-2 (HMGCS2) are rate-limiting enzymes in ketogenesis [64]. Their genes are directly changed by PPARα [65]. However, FGF21 cannot regulate their gene expressions, but increases their protein levels through a posttranscriptional mechanism [18].
Mice fed a high-fat, low-carbohydrate ketogenic diet exhibit marked increases in FGF21 expression in the liver [20, 58, 66] and in white adipose tissue (WAT) by fasting–refeeding regimens [67, 68]. These responses in the liver and WAT are likely mediated by carbohydrate response element-binding protein and PPARγ, respectively [2023]. Notably, unlike the fasting response that elicits FGF21 release from the liver into circulation, feeding induction of FGF21 in WAT do not cause a corresponding increase in circulating levels of FGF21, so FGF21 secreted by adipose tissue promotes fatty acid synthesis in adipose tissue in a paracrine or autocrine manner [68], and liver generated FGF21 promotes adipose lipolysis in white adipose tissue in an endocrine manner [18]. So FGF21 regulates lipogenesis and lipolysis by distinct modes. Recent studies found that FGF21 acts as a negative feedback signal to terminate GH-stimulated lipolysis in adipocytes and hepatocytes. In the liver, GH stimulates transcription of the FGF21 through the signal transducer and activator of transcription 5 (STAT 5) signaling pathway [69, 70].

Conclusions and implications

FGF15/19 and FGF21 acting on the heels of metabolic regulation factors to regulate metabolism in response to nutritional status. FGF15/19 is secreted in response to feeding. FGF21 is induced in response to diverse nutrition stressors, especially fasting. Therefore, they can be considered “late-acting’’ fed- and fasted-state hormones respectively. So we conclude that FGF15/19 and FGF21 play significant roles in coordinating nutrition homeostasis under a variety of physiological conditions.
Both FGF15/19 and FGF21 have distinct physiological effects on nutrient metabolism, though they belong to the same subfamily. FGF15/19 mainly regulates gallbladder filling, bile acid synthesis, and inhibits hepatic gluconeogenesis and lipogenesis in fed state, whereas FGF21 regulates glucose uptake, glycogen synthesis, and ketogenesis in the fed/fast state. Interestingly, it has been shown that FGF15/19 and FGF21 have potential roles in metabolic diseases, such as nonalcoholic fatty liver disease, bile acid diarrhea, cardiovascular disease and diabetes. Given the high variability in inter-individuals and interspecies, further studies are urgently needed to evaluate the legitimate therapeutic roles of FGF15/19 and FGF21 in malnutrition associated diseases.

Authors’ contributions

DG and LZ contributed to the literature search, synthesis of information and preparation of the manuscript. DC and BY were involved in refining the study design and literature search. JY critically reviewed, edited and revised the manuscript. All authors read and approved the final manuscript.

Acknowledgements

This work was supported by Key Project from Sichuan Department of Education (13ZA0261) and National Key Basic Research Program of China (2012CB124701).

Competing interests

The authors declare that they have no competing interests.
Open AccessThis article is distributed under the terms of the Creative Commons Attribution 4.0 International License (http://​creativecommons.​org/​licenses/​by/​4.​0/​), which permits unrestricted use, distribution, and reproduction in any medium, provided you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons license, and indicate if changes were made. The Creative Commons Public Domain Dedication waiver (http://​creativecommons.​org/​publicdomain/​zero/​1.​0/​) applies to the data made available in this article, unless otherwise stated.
Literatur
1.
Zurück zum Zitat Itoh N. Hormone-like (endocrine) Fgfs: their evolutionary history and roles in development, metabolism, and disease. Cell Tissue Res. 2010;342:1–11.CrossRefPubMedPubMedCentral Itoh N. Hormone-like (endocrine) Fgfs: their evolutionary history and roles in development, metabolism, and disease. Cell Tissue Res. 2010;342:1–11.CrossRefPubMedPubMedCentral
3.
Zurück zum Zitat Itoh N, Ohta H, Konishi M. Endocrine FGFs: evolution, physiology, pathophysiology, and pharmacotherapy. Front Endocrinol (Lausanne). 2015;6:154. Itoh N, Ohta H, Konishi M. Endocrine FGFs: evolution, physiology, pathophysiology, and pharmacotherapy. Front Endocrinol (Lausanne). 2015;6:154.
4.
Zurück zum Zitat Presta M. Dell’Era P, Mitola S, Moroni E, Ronca R, Rusnati M: fibroblast growth factor/fibroblast growth factor receptor system in angiogenesis. Cytokine Growth Factor Rev. 2005;16:159–78.CrossRefPubMed Presta M. Dell’Era P, Mitola S, Moroni E, Ronca R, Rusnati M: fibroblast growth factor/fibroblast growth factor receptor system in angiogenesis. Cytokine Growth Factor Rev. 2005;16:159–78.CrossRefPubMed
5.
Zurück zum Zitat Itoh N, Ornitz DM. Evolution of the Fgf and Fgfr gene families. Trends Genet. 2004;20:563–9.CrossRefPubMed Itoh N, Ornitz DM. Evolution of the Fgf and Fgfr gene families. Trends Genet. 2004;20:563–9.CrossRefPubMed
6.
Zurück zum Zitat Itoh N, Ornitz DM. Functional evolutionary history of the mouse Fgf gene family. Dev Dyn. 2008;237:18–27.CrossRefPubMed Itoh N, Ornitz DM. Functional evolutionary history of the mouse Fgf gene family. Dev Dyn. 2008;237:18–27.CrossRefPubMed
7.
Zurück zum Zitat Goldfarb M, Schoorlemmer J, Williams A, Diwakar S, Wang Q, Huang X, Giza J, Tchetchik D, Kelley K, Vega A, et al. Fibroblast growth factor homologous factors control neuronal excitability through modulation of voltage-gated sodium channels. Neuron. 2007;55:449–63.CrossRefPubMedPubMedCentral Goldfarb M, Schoorlemmer J, Williams A, Diwakar S, Wang Q, Huang X, Giza J, Tchetchik D, Kelley K, Vega A, et al. Fibroblast growth factor homologous factors control neuronal excitability through modulation of voltage-gated sodium channels. Neuron. 2007;55:449–63.CrossRefPubMedPubMedCentral
8.
9.
Zurück zum Zitat Cicione C, Degirolamo C, Moschetta A. Emerging role of fibroblast growth factors 15/19 and 21 as metabolic integrators in the liver. Hepatology. 2012;56:2404–11.CrossRefPubMed Cicione C, Degirolamo C, Moschetta A. Emerging role of fibroblast growth factors 15/19 and 21 as metabolic integrators in the liver. Hepatology. 2012;56:2404–11.CrossRefPubMed
10.
Zurück zum Zitat Itoh N, Ornitz DM. Fibroblast growth factors: from molecular evolution to roles in development, metabolism and disease. J Biochem. 2011;149:121–30.CrossRefPubMedPubMedCentral Itoh N, Ornitz DM. Fibroblast growth factors: from molecular evolution to roles in development, metabolism and disease. J Biochem. 2011;149:121–30.CrossRefPubMedPubMedCentral
11.
Zurück zum Zitat Owen BM, Mangelsdorf DJ, Kliewer SA. Tissue-specific actions of the metabolic hormones FGF15/19 and FGF21. Trends Endocrinol Metab. 2015;26:22–9.CrossRefPubMedPubMedCentral Owen BM, Mangelsdorf DJ, Kliewer SA. Tissue-specific actions of the metabolic hormones FGF15/19 and FGF21. Trends Endocrinol Metab. 2015;26:22–9.CrossRefPubMedPubMedCentral
12.
Zurück zum Zitat Zhang F, Yu L, Lin X, Cheng P, He L, Li X, Lu X, Tan Y, Yang H, Cai L. Minireview: roles of fibroblast growth factors 19 and 21 in metabolic regulation and chronic diseases. Mol Endocrinol. 2015;29:1400–13.CrossRefPubMed Zhang F, Yu L, Lin X, Cheng P, He L, Li X, Lu X, Tan Y, Yang H, Cai L. Minireview: roles of fibroblast growth factors 19 and 21 in metabolic regulation and chronic diseases. Mol Endocrinol. 2015;29:1400–13.CrossRefPubMed
13.
Zurück zum Zitat Potthoff MJ, Boney-Montoya J, Choi M, He T, Sunny NE, Satapati S, Suino-Powell K, Xu HE, Gerard RD, Finck BN. FGF15/19 regulates hepatic glucose metabolism by inhibiting the CREB-PGC-1α pathway. Cell Metab. 2011;13:729–38.CrossRefPubMedPubMedCentral Potthoff MJ, Boney-Montoya J, Choi M, He T, Sunny NE, Satapati S, Suino-Powell K, Xu HE, Gerard RD, Finck BN. FGF15/19 regulates hepatic glucose metabolism by inhibiting the CREB-PGC-1α pathway. Cell Metab. 2011;13:729–38.CrossRefPubMedPubMedCentral
14.
15.
Zurück zum Zitat Choi M, Moschetta A, Bookout AL, Peng L, Umetani M, Holmstrom SR, Suino-Powell K, Xu HE, Richardson JA, Gerard RD. Identification of a hormonal basis for gallbladder filling. Nat Med. 2006;12:1253–5.CrossRefPubMed Choi M, Moschetta A, Bookout AL, Peng L, Umetani M, Holmstrom SR, Suino-Powell K, Xu HE, Richardson JA, Gerard RD. Identification of a hormonal basis for gallbladder filling. Nat Med. 2006;12:1253–5.CrossRefPubMed
16.
Zurück zum Zitat Goetz R, Beenken A, Ibrahimi OA, Kalinina J, Olsen SK, Eliseenkova AV, Xu C, Neubert TA, Zhang F, Linhardt RJ, et al. Molecular insights into the klotho-dependent, endocrine mode of action of fibroblast growth factor 19 subfamily members. Mol Cell Biol. 2007;27:3417–28.CrossRefPubMedPubMedCentral Goetz R, Beenken A, Ibrahimi OA, Kalinina J, Olsen SK, Eliseenkova AV, Xu C, Neubert TA, Zhang F, Linhardt RJ, et al. Molecular insights into the klotho-dependent, endocrine mode of action of fibroblast growth factor 19 subfamily members. Mol Cell Biol. 2007;27:3417–28.CrossRefPubMedPubMedCentral
17.
Zurück zum Zitat Kharitonenkov A, Shiyanova TL, Koester A, Ford AM, Micanovic R, Galbreath EJ, Sandusky GE, Hammond LJ, Moyers JS, Owens RA, et al. FGF-21 as a novel metabolic regulator. J Clin Invest. 2005;115:1627–35.CrossRefPubMedPubMedCentral Kharitonenkov A, Shiyanova TL, Koester A, Ford AM, Micanovic R, Galbreath EJ, Sandusky GE, Hammond LJ, Moyers JS, Owens RA, et al. FGF-21 as a novel metabolic regulator. J Clin Invest. 2005;115:1627–35.CrossRefPubMedPubMedCentral
18.
Zurück zum Zitat Inagaki T, Dutchak P, Zhao G, Ding X, Gautron L, Parameswara V, Li Y, Goetz R, Mohammadi M, Esser V, et al. Endocrine regulation of the fasting response by PPARalpha-mediated induction of fibroblast growth factor 21. Cell Metab. 2007;5:415–25.CrossRefPubMed Inagaki T, Dutchak P, Zhao G, Ding X, Gautron L, Parameswara V, Li Y, Goetz R, Mohammadi M, Esser V, et al. Endocrine regulation of the fasting response by PPARalpha-mediated induction of fibroblast growth factor 21. Cell Metab. 2007;5:415–25.CrossRefPubMed
19.
Zurück zum Zitat 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.CrossRefPubMed 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.CrossRefPubMed
20.
Zurück zum Zitat 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.CrossRefPubMed 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.CrossRefPubMed
21.
Zurück zum Zitat 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.CrossRefPubMed 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.CrossRefPubMed
22.
Zurück zum Zitat 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.CrossRefPubMedPubMedCentral 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.CrossRefPubMedPubMedCentral
23.
Zurück zum Zitat Iizuka K, Takeda J, Horikawa Y. Glucose induces FGF21 mRNA expression through ChREBP activation in rat hepatocytes. FEBS Lett. 2009;583:2882–6.CrossRefPubMed Iizuka K, Takeda J, Horikawa Y. Glucose induces FGF21 mRNA expression through ChREBP activation in rat hepatocytes. FEBS Lett. 2009;583:2882–6.CrossRefPubMed
24.
Zurück zum Zitat Hotta Y, Nakamura H, Konishi M, Murata Y, Takagi H, Matsumura S, Inoue K, Fushiki T, Itoh N. Fibroblast growth factor 21 regulates lipolysis in white adipose tissue but is not required for ketogenesis and triglyceride clearance in liver. Endocrinology. 2009;150:4625–33.CrossRefPubMed Hotta Y, Nakamura H, Konishi M, Murata Y, Takagi H, Matsumura S, Inoue K, Fushiki T, Itoh N. Fibroblast growth factor 21 regulates lipolysis in white adipose tissue but is not required for ketogenesis and triglyceride clearance in liver. Endocrinology. 2009;150:4625–33.CrossRefPubMed
25.
Zurück zum Zitat Lee PL, Johnson DE, Cousens LS, Fried VA, Williams LT. Purification and complementary DNA cloning of a receptor for basic fibroblast growth factor. Science. 1989;245:57–60.CrossRefPubMed Lee PL, Johnson DE, Cousens LS, Fried VA, Williams LT. Purification and complementary DNA cloning of a receptor for basic fibroblast growth factor. Science. 1989;245:57–60.CrossRefPubMed
26.
Zurück zum Zitat Johnson DE, Williams LT. Structural and functional diversity in the FGF receptor multigene family. Adv Cancer Res. 1993;60:1–41.CrossRefPubMed Johnson DE, Williams LT. Structural and functional diversity in the FGF receptor multigene family. Adv Cancer Res. 1993;60:1–41.CrossRefPubMed
27.
Zurück zum Zitat Powers CJ, McLeskey SW, Wellstein A. Fibroblast growth factors, their receptors and signaling. Endocr Relat Cancer. 2000;7:165–97.CrossRefPubMed Powers CJ, McLeskey SW, Wellstein A. Fibroblast growth factors, their receptors and signaling. Endocr Relat Cancer. 2000;7:165–97.CrossRefPubMed
28.
Zurück zum Zitat Ornitz DM. FGFs, heparan sulfate and FGFRs: complex interactions essential for development. Bioessays. 2000;22(2):108–12.CrossRefPubMed Ornitz DM. FGFs, heparan sulfate and FGFRs: complex interactions essential for development. Bioessays. 2000;22(2):108–12.CrossRefPubMed
29.
Zurück zum Zitat Yang C, Jin C, Li X, Wang F, McKeehan WL, Luo Y. Differential specificity of endocrine FGF19 and FGF21 to FGFR1 and FGFR4 in complex with KLB. PLoS ONE. 2012;7:e33870.CrossRefPubMedPubMedCentral Yang C, Jin C, Li X, Wang F, McKeehan WL, Luo Y. Differential specificity of endocrine FGF19 and FGF21 to FGFR1 and FGFR4 in complex with KLB. PLoS ONE. 2012;7:e33870.CrossRefPubMedPubMedCentral
30.
Zurück zum Zitat Kurosu H, Choi M, Ogawa Y, Dickson AS, Goetz R, Eliseenkova AV, Mohammadi M, Rosenblatt KP, Kliewer SA, Kuro-o M. Tissue-specific expression of betaKlotho and fibroblast growth factor (FGF) receptor isoforms determines metabolic activity of FGF19 and FGF21. J Biol Chem. 2007;282:26687–95.CrossRefPubMedPubMedCentral Kurosu H, Choi M, Ogawa Y, Dickson AS, Goetz R, Eliseenkova AV, Mohammadi M, Rosenblatt KP, Kliewer SA, Kuro-o M. Tissue-specific expression of betaKlotho and fibroblast growth factor (FGF) receptor isoforms determines metabolic activity of FGF19 and FGF21. J Biol Chem. 2007;282:26687–95.CrossRefPubMedPubMedCentral
31.
Zurück zum Zitat Kuro-o M, Matsumura Y, Aizawa H, Kawaguchi H, Suga T, Utsugi T, Ohyama Y, Kurabayashi M, Kaname T, Kume E, et al. Mutation of the mouse klotho gene leads to a syndrome resembling ageing. Nature. 1997;390:45–51.CrossRefPubMed Kuro-o M, Matsumura Y, Aizawa H, Kawaguchi H, Suga T, Utsugi T, Ohyama Y, Kurabayashi M, Kaname T, Kume E, et al. Mutation of the mouse klotho gene leads to a syndrome resembling ageing. Nature. 1997;390:45–51.CrossRefPubMed
32.
Zurück zum Zitat Lin BC, Wang M, Blackmore C, Desnoyers LR. Liver-specific activities of FGF19 require Klotho beta. J Biol Chem. 2007;282:27277–84.CrossRefPubMed Lin BC, Wang M, Blackmore C, Desnoyers LR. Liver-specific activities of FGF19 require Klotho beta. J Biol Chem. 2007;282:27277–84.CrossRefPubMed
33.
Zurück zum Zitat Lin BC, Desnoyers LR. FGF19 and cancer endocrine FGFs and Klothos. Springer US. 2012;73:183–94. Lin BC, Desnoyers LR. FGF19 and cancer endocrine FGFs and Klothos. Springer US. 2012;73:183–94.
34.
Zurück zum Zitat Wu X, Ge H, Lemon B, Weiszmann J, Gupte J, Hawkins N, Li X, Tang J, Lindberg R, Li Y. Selective activation of FGFR4 by an FGF19 variant does not improve glucose metabolism in ob/ob mice. Proc Natl Acad Sci USA. 2009;106:14379–84.CrossRefPubMedPubMedCentral Wu X, Ge H, Lemon B, Weiszmann J, Gupte J, Hawkins N, Li X, Tang J, Lindberg R, Li Y. Selective activation of FGFR4 by an FGF19 variant does not improve glucose metabolism in ob/ob mice. Proc Natl Acad Sci USA. 2009;106:14379–84.CrossRefPubMedPubMedCentral
35.
Zurück zum Zitat Suzuki M, Uehara Y, Motomura-Matsuzaka K, Oki J, Koyama Y, Kimura M, Asada M, Komi-Kuramochi A, Oka S, Imamura T. betaKlotho is required for fibroblast growth factor (FGF) 21 signaling through FGF receptor (FGFR) 1c and FGFR3c. Mol Endocrinol. 2008;22:1006–14.CrossRefPubMed Suzuki M, Uehara Y, Motomura-Matsuzaka K, Oki J, Koyama Y, Kimura M, Asada M, Komi-Kuramochi A, Oka S, Imamura T. betaKlotho is required for fibroblast growth factor (FGF) 21 signaling through FGF receptor (FGFR) 1c and FGFR3c. Mol Endocrinol. 2008;22:1006–14.CrossRefPubMed
36.
Zurück zum Zitat Ogawa Y, Kurosu H, Yamamoto M, Nandi A, Rosenblatt KP, Goetz R, Eliseenkova AV, Mohammadi M, Kuro-o M. BetaKlotho is required for metabolic activity of fibroblast growth factor 21. Proc Natl Acad Sci USA. 2007;104:7432–7.CrossRefPubMedPubMedCentral Ogawa Y, Kurosu H, Yamamoto M, Nandi A, Rosenblatt KP, Goetz R, Eliseenkova AV, Mohammadi M, Kuro-o M. BetaKlotho is required for metabolic activity of fibroblast growth factor 21. Proc Natl Acad Sci USA. 2007;104:7432–7.CrossRefPubMedPubMedCentral
37.
Zurück zum Zitat Yie J, Hecht R, Patel J, Stevens J, Wang W, Hawkins N, Steavenson S, Smith S, Winters D, Fisher S, et al. FGF21 N- and C-termini play different roles in receptor interaction and activation. FEBS Lett. 2009;583:19–24.CrossRefPubMed Yie J, Hecht R, Patel J, Stevens J, Wang W, Hawkins N, Steavenson S, Smith S, Winters D, Fisher S, et al. FGF21 N- and C-termini play different roles in receptor interaction and activation. FEBS Lett. 2009;583:19–24.CrossRefPubMed
38.
Zurück zum Zitat Tomiyama K, Maeda R, Urakawa I, Yamazaki Y, Tanaka T, Ito S, Nabeshima Y, Tomita T, Odori S, Hosoda K, et al. Relevant use of Klotho in FGF19 subfamily signaling system in vivo. Proc Natl Acad Sci USA. 2010;107:1666–71.CrossRefPubMedPubMedCentral Tomiyama K, Maeda R, Urakawa I, Yamazaki Y, Tanaka T, Ito S, Nabeshima Y, Tomita T, Odori S, Hosoda K, et al. Relevant use of Klotho in FGF19 subfamily signaling system in vivo. Proc Natl Acad Sci USA. 2010;107:1666–71.CrossRefPubMedPubMedCentral
39.
Zurück zum Zitat Asada M, Shinomiya M, Suzuki M, Honda E, Sugimoto R, Ikekita M, Imamura T. Glycosaminoglycan affinity of the complete fibroblast growth factor family. Biochim Biophys Acta. 2009;1790:40–8.CrossRefPubMed Asada M, Shinomiya M, Suzuki M, Honda E, Sugimoto R, Ikekita M, Imamura T. Glycosaminoglycan affinity of the complete fibroblast growth factor family. Biochim Biophys Acta. 2009;1790:40–8.CrossRefPubMed
40.
Zurück zum Zitat Holt JA, Luo G, Billin AN, Bisi J, McNeill YY, Kozarsky KF, Donahee M, Wang DY, Mansfield TA, Kliewer SA, et al. Definition of a novel growth factor-dependent signal cascade for the suppression of bile acid biosynthesis. Genes Dev. 2003;17:1581–91.CrossRefPubMedPubMedCentral Holt JA, Luo G, Billin AN, Bisi J, McNeill YY, Kozarsky KF, Donahee M, Wang DY, Mansfield TA, Kliewer SA, et al. Definition of a novel growth factor-dependent signal cascade for the suppression of bile acid biosynthesis. Genes Dev. 2003;17:1581–91.CrossRefPubMedPubMedCentral
41.
Zurück zum Zitat Inagaki T, Choi M, Moschetta A, Peng L, Cummins CL, McDonald JG, Luo G, Jones SA, Goodwin B, Richardson JA, et al. Fibroblast growth factor 15 functions as an enterohepatic signal to regulate bile acid homeostasis. Cell Metab. 2005;2:217–25.CrossRefPubMed Inagaki T, Choi M, Moschetta A, Peng L, Cummins CL, McDonald JG, Luo G, Jones SA, Goodwin B, Richardson JA, et al. Fibroblast growth factor 15 functions as an enterohepatic signal to regulate bile acid homeostasis. Cell Metab. 2005;2:217–25.CrossRefPubMed
42.
Zurück zum Zitat Kir S, Kliewer SA, Mangelsdorf DJ. Roles of FGF19 in liver metabolism. Cold Spring Harb Symp Quant Biol. 2011;76:139–44.CrossRefPubMed Kir S, Kliewer SA, Mangelsdorf DJ. Roles of FGF19 in liver metabolism. Cold Spring Harb Symp Quant Biol. 2011;76:139–44.CrossRefPubMed
43.
Zurück zum Zitat Lundasen T, Galman C, Angelin B, Rudling M. Circulating intestinal fibroblast growth factor 19 has a pronounced diurnal variation and modulates hepatic bile acid synthesis in man. J Intern Med. 2006;260:530–6.CrossRefPubMed Lundasen T, Galman C, Angelin B, Rudling M. Circulating intestinal fibroblast growth factor 19 has a pronounced diurnal variation and modulates hepatic bile acid synthesis in man. J Intern Med. 2006;260:530–6.CrossRefPubMed
44.
Zurück zum Zitat Goodwin B, Jones SA, Price RR, Watson MA, Mckee DD, Moore LB, et al. A regulatory cascade of the nuclear receptors FXR, SHP-1, and LRH-1 represses bile acid biosynthesis. Mol Cell. 2000;6(3):517–26.CrossRefPubMed Goodwin B, Jones SA, Price RR, Watson MA, Mckee DD, Moore LB, et al. A regulatory cascade of the nuclear receptors FXR, SHP-1, and LRH-1 represses bile acid biosynthesis. Mol Cell. 2000;6(3):517–26.CrossRefPubMed
45.
Zurück zum Zitat Lu TT, Makishima M, Repa JJ, Schoonjans K, Keer TA, Auwerx J, et al. Molecular basis for feedback regulation of bile acid synthesis by nuclear receptors. Mol Cell. 2000;6(3):507–15.CrossRefPubMed Lu TT, Makishima M, Repa JJ, Schoonjans K, Keer TA, Auwerx J, et al. Molecular basis for feedback regulation of bile acid synthesis by nuclear receptors. Mol Cell. 2000;6(3):507–15.CrossRefPubMed
46.
Zurück zum Zitat De Fabiani E, Mitro N, Anzulovich AC, Pinelli A, Galli G, Crestani M. The negative effects of bile acids and tumor necrosis factor-alpha on the transcription of cholesterol 7alpha-hydroxylase gene (CYP7A1) converge to hepatic nuclear factor-4: a novel mechanism of feedback regulation of bile acid synthesis mediated by nuclear receptors. J Biol Chem. 2001;276:30708–16.CrossRefPubMed De Fabiani E, Mitro N, Anzulovich AC, Pinelli A, Galli G, Crestani M. The negative effects of bile acids and tumor necrosis factor-alpha on the transcription of cholesterol 7alpha-hydroxylase gene (CYP7A1) converge to hepatic nuclear factor-4: a novel mechanism of feedback regulation of bile acid synthesis mediated by nuclear receptors. J Biol Chem. 2001;276:30708–16.CrossRefPubMed
47.
Zurück zum Zitat Kerr TA, Saeki S, Schneider M, Schaefer K, Berdy S, Redder T, et al. Loss of nuclear receptor SHP impairs but does not eliminate negative feedback regulation of bile acid synthesis. Dev Cell. 2002;2(6):713–20.CrossRefPubMedPubMedCentral Kerr TA, Saeki S, Schneider M, Schaefer K, Berdy S, Redder T, et al. Loss of nuclear receptor SHP impairs but does not eliminate negative feedback regulation of bile acid synthesis. Dev Cell. 2002;2(6):713–20.CrossRefPubMedPubMedCentral
48.
Zurück zum Zitat Wang L, Lee YK, Bundman D, Han Y, Thevananther S, Kim C, et al. Redundant pathways for negative feedback regulation of bile acid production. Dev Cell. 2002;2(6):721–31.CrossRefPubMed Wang L, Lee YK, Bundman D, Han Y, Thevananther S, Kim C, et al. Redundant pathways for negative feedback regulation of bile acid production. Dev Cell. 2002;2(6):721–31.CrossRefPubMed
49.
Zurück zum Zitat Kok T, Hulzebos CV, Wolters H, Havinga R, Agellon LB, Stellaard F, Shan B, Schwarz M, Kuipers F. Enterohepatic circulation of bile salts in farnesoid X receptor-deficient mice: efficient intestinal bile salt absorption in the absence of ileal bile acid-binding protein. J Biol Chem. 2003;278:41930–7.CrossRefPubMed Kok T, Hulzebos CV, Wolters H, Havinga R, Agellon LB, Stellaard F, Shan B, Schwarz M, Kuipers F. Enterohepatic circulation of bile salts in farnesoid X receptor-deficient mice: efficient intestinal bile salt absorption in the absence of ileal bile acid-binding protein. J Biol Chem. 2003;278:41930–7.CrossRefPubMed
50.
Zurück zum Zitat Song KH, Li T, Owsley E, Strom S, Chiang JY. Bile acids activate fibroblast growth factor 19 signaling in human hepatocytes to inhibit cholesterol 7alpha-hydroxylase gene expression. Hepatology. 2009;49:297–305.CrossRefPubMedPubMedCentral Song KH, Li T, Owsley E, Strom S, Chiang JY. Bile acids activate fibroblast growth factor 19 signaling in human hepatocytes to inhibit cholesterol 7alpha-hydroxylase gene expression. Hepatology. 2009;49:297–305.CrossRefPubMedPubMedCentral
51.
Zurück zum Zitat Vergnes L, Lee JM, Chin RG, Auwerx J, Reue K. Diet1 functions in the FGF15/19 enterohepatic signaling axis to modulate bile acid and lipid levels. Cell Metab. 2013;17:916–28.CrossRefPubMedPubMedCentral Vergnes L, Lee JM, Chin RG, Auwerx J, Reue K. Diet1 functions in the FGF15/19 enterohepatic signaling axis to modulate bile acid and lipid levels. Cell Metab. 2013;17:916–28.CrossRefPubMedPubMedCentral
52.
Zurück zum Zitat Kim I, Ahn SH, Inagaki T, Choi M, Ito S, Guo GL, Kliewer SA, Gonzalez FJ. Differential regulation of bile acid homeostasis by the farnesoid X receptor in liver and intestine. J Lipid Res. 2007;48:2664–72.CrossRefPubMed Kim I, Ahn SH, Inagaki T, Choi M, Ito S, Guo GL, Kliewer SA, Gonzalez FJ. Differential regulation of bile acid homeostasis by the farnesoid X receptor in liver and intestine. J Lipid Res. 2007;48:2664–72.CrossRefPubMed
53.
Zurück zum Zitat Kir S, Beddow SA, Samuel VT, Miller P, Previs SF, Suino-Powell K, Xu HE, Shulman GI, Kliewer SA, Mangelsdorf DJ. FGF19 as a postprandial, insulin-independent activator of hepatic protein and glycogen synthesis. Science. 2011;331:1621–4.CrossRefPubMedPubMedCentral Kir S, Beddow SA, Samuel VT, Miller P, Previs SF, Suino-Powell K, Xu HE, Shulman GI, Kliewer SA, Mangelsdorf DJ. FGF19 as a postprandial, insulin-independent activator of hepatic protein and glycogen synthesis. Science. 2011;331:1621–4.CrossRefPubMedPubMedCentral
54.
Zurück zum Zitat Shin DJ, Osborne TF. FGF15/FGFR4 integrates growth factor signaling with hepatic bile acid metabolism and insulin action. J Biol Chem. 2009;284:11110–20.CrossRefPubMedPubMedCentral Shin DJ, Osborne TF. FGF15/FGFR4 integrates growth factor signaling with hepatic bile acid metabolism and insulin action. J Biol Chem. 2009;284:11110–20.CrossRefPubMedPubMedCentral
55.
Zurück zum Zitat Stejskal D, Karpisek M, Hanulova Z, Stejskal P. Fibroblast growth factor-19: development, analytical characterization and clinical evaluation of a new ELISA test. Scand J Clin Lab Invest. 2008;68:501–7.CrossRefPubMed Stejskal D, Karpisek M, Hanulova Z, Stejskal P. Fibroblast growth factor-19: development, analytical characterization and clinical evaluation of a new ELISA test. Scand J Clin Lab Invest. 2008;68:501–7.CrossRefPubMed
56.
Zurück zum Zitat Reiche M, Bachmann A, Lossner U, Bluher M, Stumvoll M, Fasshauer M. Fibroblast growth factor 19 serum levels: relation to renal function and metabolic parameters. Horm Metab Res. 2010;42:178–81.CrossRefPubMed Reiche M, Bachmann A, Lossner U, Bluher M, Stumvoll M, Fasshauer M. Fibroblast growth factor 19 serum levels: relation to renal function and metabolic parameters. Horm Metab Res. 2010;42:178–81.CrossRefPubMed
57.
Zurück zum Zitat Mráz M, Lacinová Z, Kaválková P, Haluziková D, Trachta P, Drápalová J, et al. Serum concentrations of fibroblast growth factor 19 in patients with obesity and type 2 diabetes mellitus: the influence of acute hyperinsulinemia, very-low calorie diet and PPAR-alpha agonist treatment. Physiol Res. 2011;60(4):627–36.PubMed Mráz M, Lacinová Z, Kaválková P, Haluziková D, Trachta P, Drápalová J, et al. Serum concentrations of fibroblast growth factor 19 in patients with obesity and type 2 diabetes mellitus: the influence of acute hyperinsulinemia, very-low calorie diet and PPAR-alpha agonist treatment. Physiol Res. 2011;60(4):627–36.PubMed
58.
Zurück zum Zitat 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;5:426–37.CrossRefPubMed 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;5:426–37.CrossRefPubMed
59.
Zurück zum Zitat Lundasen T, Hunt MC, Nilsson LM, Sanyal S, Angelin B, Alexson SE, Rudling M. PPARalpha is a key regulator of hepatic FGF21. Biochem Biophys Res Commun. 2007;360:437–40.CrossRefPubMed Lundasen T, Hunt MC, Nilsson LM, Sanyal S, Angelin B, Alexson SE, Rudling M. PPARalpha is a key regulator of hepatic FGF21. Biochem Biophys Res Commun. 2007;360:437–40.CrossRefPubMed
60.
Zurück zum Zitat Potthoff MJ, Inagaki T, Satapati S, Ding X, He T, Goetz R, Mohammadi M, Finck BN, Mangelsdorf DJ, Kliewer SA, Burgess SC. FGF21 induces PGC-1alpha and regulates carbohydrate and fatty acid metabolism during the adaptive starvation response. Proc Natl Acad Sci USA. 2009;106:10853–8.CrossRefPubMedPubMedCentral Potthoff MJ, Inagaki T, Satapati S, Ding X, He T, Goetz R, Mohammadi M, Finck BN, Mangelsdorf DJ, Kliewer SA, Burgess SC. FGF21 induces PGC-1alpha and regulates carbohydrate and fatty acid metabolism during the adaptive starvation response. Proc Natl Acad Sci USA. 2009;106:10853–8.CrossRefPubMedPubMedCentral
61.
Zurück zum Zitat 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.CrossRefPubMedPubMedCentral 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.CrossRefPubMedPubMedCentral
62.
Zurück zum Zitat Yoon JC, Puigserver P, Chen GX, Donovan J, Wu ZD, Rhee J, Adelmant G, Stafford J, Kahn CR, Granner DK, et al. Control of hepatic gluconeogenesis through the transcriptional coactivator PGC-1. Nature. 2001;413:131–8.CrossRefPubMed Yoon JC, Puigserver P, Chen GX, Donovan J, Wu ZD, Rhee J, Adelmant G, Stafford J, Kahn CR, Granner DK, et al. Control of hepatic gluconeogenesis through the transcriptional coactivator PGC-1. Nature. 2001;413:131–8.CrossRefPubMed
63.
Zurück zum Zitat Sarruf DA, Thaler JP, Morton GJ, German J, Fischer JD, Ogimoto K, Schwartz MW. Fibroblast growth factor 21 action in the brain increases energy expenditure and insulin sensitivity in obese rats. Diabetes. 2010;59:1817–24.CrossRefPubMedPubMedCentral Sarruf DA, Thaler JP, Morton GJ, German J, Fischer JD, Ogimoto K, Schwartz MW. Fibroblast growth factor 21 action in the brain increases energy expenditure and insulin sensitivity in obese rats. Diabetes. 2010;59:1817–24.CrossRefPubMedPubMedCentral
64.
65.
Zurück zum Zitat Erol E, Kumar LS, Cline GW, Shulman GI, Kelly DP, Binas B. Liver fatty acid-binding protein is required for high rates of hepatic fatty acid oxidation but not for the action of PPAR-alpha in fasting mice. Faseb J. 2003;17:347. Erol E, Kumar LS, Cline GW, Shulman GI, Kelly DP, Binas B. Liver fatty acid-binding protein is required for high rates of hepatic fatty acid oxidation but not for the action of PPAR-alpha in fasting mice. Faseb J. 2003;17:347.
66.
Zurück zum Zitat Uebanso T, Taketani Y, Yamamoto H, Amo K, Ominami H, Arai H, Takei Y, Masuda M, Tanimura A, Harada N, et al. Paradoxical regulation of human FGF21 by both fasting and feeding signals: is FGF21 a nutritional adaptation factor? PLoS ONE. 2011;6:e22976.CrossRefPubMedPubMedCentral Uebanso T, Taketani Y, Yamamoto H, Amo K, Ominami H, Arai H, Takei Y, Masuda M, Tanimura A, Harada N, et al. Paradoxical regulation of human FGF21 by both fasting and feeding signals: is FGF21 a nutritional adaptation factor? PLoS ONE. 2011;6:e22976.CrossRefPubMedPubMedCentral
67.
Zurück zum Zitat Oishi K, Konishi M, Murata Y, Itoh N. Time-imposed daily restricted feeding induces rhythmic expression of Fgf21 in white adipose tissue of mice. Biochem Biophys Res Commun. 2011;412:396–400.CrossRefPubMed Oishi K, Konishi M, Murata Y, Itoh N. Time-imposed daily restricted feeding induces rhythmic expression of Fgf21 in white adipose tissue of mice. Biochem Biophys Res Commun. 2011;412:396–400.CrossRefPubMed
68.
Zurück zum Zitat 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.CrossRefPubMedPubMedCentral 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.CrossRefPubMedPubMedCentral
69.
Zurück zum Zitat Chen W, Hoo RL, Konishi M, Itoh N, Lee PC, Ye HY, Lam KS, Xu A. Growth hormone induces hepatic production of fibroblast growth factor 21 through a mechanism dependent on lipolysis in adipocytes. J Biol Chem. 2011;286:34559–66.CrossRefPubMedPubMedCentral Chen W, Hoo RL, Konishi M, Itoh N, Lee PC, Ye HY, Lam KS, Xu A. Growth hormone induces hepatic production of fibroblast growth factor 21 through a mechanism dependent on lipolysis in adipocytes. J Biol Chem. 2011;286:34559–66.CrossRefPubMedPubMedCentral
70.
Zurück zum Zitat Yu J, Zhao L, Wang A, Eleswarapu S, Ge X, Chen D, Jiang H. Growth hormone stimulates transcription of the fibroblast growth factor 21 gene in the liver through the signal transducer and activator of transcription 5. Endocrinology. 2012;153:750–8.CrossRefPubMed Yu J, Zhao L, Wang A, Eleswarapu S, Ge X, Chen D, Jiang H. Growth hormone stimulates transcription of the fibroblast growth factor 21 gene in the liver through the signal transducer and activator of transcription 5. Endocrinology. 2012;153:750–8.CrossRefPubMed
Metadaten
Titel
Regulation of fibroblast growth factor 15/19 and 21 on metabolism: in the fed or fasted state
Publikationsdatum
01.12.2016
Erschienen in
Journal of Translational Medicine / Ausgabe 1/2016
Elektronische ISSN: 1479-5876
DOI
https://doi.org/10.1186/s12967-016-0821-0

Weitere Artikel der Ausgabe 1/2016

Journal of Translational Medicine 1/2016 Zur Ausgabe

Leitlinien kompakt für die Innere Medizin

Mit medbee Pocketcards sicher entscheiden.

Seit 2022 gehört die medbee GmbH zum Springer Medizin Verlag

Update Innere Medizin

Bestellen Sie unseren Fach-Newsletter und bleiben Sie gut informiert.