Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 23
Filter
Add more filters











Publication year range
1.
Hepatol Commun ; 8(5)2024 05 01.
Article in English | MEDLINE | ID: mdl-38696369

ABSTRACT

BACKGROUND: Human genetic studies have identified several mitochondrial amidoxime-reducing component 1 (MTARC1) variants as protective against metabolic dysfunction-associated steatotic liver disease. The MTARC1 variants are associated with decreased plasma lipids and liver enzymes and reduced liver-related mortality. However, the role of mARC1 in fatty liver disease is still unclear. METHODS: Given that mARC1 is mainly expressed in hepatocytes, we developed an N-acetylgalactosamine-conjugated mouse Mtarc1 siRNA, applying it in multiple in vivo models to investigate the role of mARC1 using multiomic techniques. RESULTS: In ob/ob mice, knockdown of Mtarc1 in mouse hepatocytes resulted in decreased serum liver enzymes, LDL-cholesterol, and liver triglycerides. Reduction of mARC1 also reduced liver weight, improved lipid profiles, and attenuated liver pathological changes in 2 diet-induced metabolic dysfunction-associated steatohepatitis mouse models. A comprehensive analysis of mARC1-deficient liver from a metabolic dysfunction-associated steatohepatitis mouse model by metabolomics, proteomics, and lipidomics showed that Mtarc1 knockdown partially restored metabolites and lipids altered by diet. CONCLUSIONS: Taken together, reducing mARC1 expression in hepatocytes protects against metabolic dysfunction-associated steatohepatitis in multiple murine models, suggesting a potential therapeutic approach for this chronic liver disease.


Subject(s)
Disease Models, Animal , Gene Knockdown Techniques , Hepatocytes , Animals , Mice , Hepatocytes/metabolism , Liver/metabolism , Male , RNA, Small Interfering/genetics , Mitochondrial Proteins/genetics , Mitochondrial Proteins/metabolism , Non-alcoholic Fatty Liver Disease/genetics , Non-alcoholic Fatty Liver Disease/metabolism , Non-alcoholic Fatty Liver Disease/prevention & control , Mice, Inbred C57BL
2.
EBioMedicine ; 15: 173-183, 2017 Feb.
Article in English | MEDLINE | ID: mdl-28041926

ABSTRACT

Pharmacological doses of fibroblast growth factor (FGF) 21 effectively normalize glucose, lipid and energy homeostasis in multiple animal models with many benefits translating to obese humans with type 2 diabetes. However, a role for FGF21 in the regulation of bile acid metabolism has not been reported. Herein, we demonstrate AAV-mediated FGF21 overexpression in mice increases liver expression of the key bile acid producing enzyme, Cyp7a1, resulting in an increased bile acid pool. Furthermore, in cholecystectomized mice, FGF21-mediated bile acid pool increase led to increased transit of bile acids into colon. We elucidate that the mechanism of FGF21 induced bile acid changes is mainly through antagonizing FGF15/19 function on liver ßKlotho/FGFR4 receptor complex; thus inhibiting FGF15/19-mediated suppression of Cyp7a1 expression. In conclusion, these data reveal a previously unidentified role for FGF21 on bile acid metabolism and may be relevant to understand the effects of FGF21 analogs in clinical studies.


Subject(s)
Bile Acids and Salts/biosynthesis , Fibroblast Growth Factors/genetics , Fibroblast Growth Factors/metabolism , Gene Expression , Animals , Biomarkers , Body Weight , Cell Line , Cholesterol 7-alpha-Hydroxylase/genetics , Cholesterol 7-alpha-Hydroxylase/metabolism , Fibroblast Growth Factors/antagonists & inhibitors , Gene Expression Regulation , Glucose/metabolism , Homeostasis , Klotho Proteins , Lipid Metabolism , Membrane Proteins/metabolism , Mice , Mice, Knockout , Models, Animal , Protein Binding , Receptor, Fibroblast Growth Factor, Type 4/genetics , Receptor, Fibroblast Growth Factor, Type 4/metabolism , Signal Transduction
3.
J Biol Chem ; 292(5): 1951-1969, 2017 02 03.
Article in English | MEDLINE | ID: mdl-28011645

ABSTRACT

The discovery of brown adipose tissue (BAT) as a key regulator of energy expenditure has sparked interest in identifying novel soluble factors capable of activating inducible BAT (iBAT) to combat obesity. Using a high content cell-based screen, we identified fibroblast growth factor 16 (FGF16) as a potent inducer of several physical and transcriptional characteristics analogous to those of both "classical" BAT and iBAT. Overexpression of Fgf16 in vivo recapitulated several of our in vitro findings, specifically the significant induction of the Ucp1 gene and UCP1 protein expression in inguinal white adipose tissue (iWAT), a common site for emergent active iBAT. Despite significant UCP1 up-regulation in iWAT and dramatic weight loss, the metabolic improvements observed due to Fgf16 overexpression in vivo were not the result of increased energy expenditure, as measured by indirect calorimetric assessment. Instead, a pattern of reduced food and water intake, combined with feces replete with lipid and bile acid, indicated a phenotype more akin to that of starvation and intestinal malabsorption. Gene expression analysis of the liver and ileum indicated alterations in several steps of bile acid metabolism, including hepatic synthesis and reabsorption. Histological analysis of intestinal tissue revealed profound abnormalities in support of this conclusion. The in vivo data, together with FGF receptor binding analysis, indicate that the in vivo outcome observed is the likely result of both direct and indirect mechanisms and probably involves multiple receptors. These results highlight the complexity of FGF signaling in the regulation of various metabolic processes.


Subject(s)
Adipose Tissue, White/metabolism , Fibroblast Growth Factors/metabolism , Signal Transduction , Thermogenesis , Ubiquitin-Specific Proteases/biosynthesis , Adipose Tissue, White/pathology , Animals , Cell Line , Dietary Fats/adverse effects , Dietary Fats/pharmacology , Fibroblast Growth Factors/genetics , Humans , Mice , Obesity/chemically induced , Obesity/genetics , Obesity/metabolism , Ubiquitin-Specific Proteases/genetics
4.
EBioMedicine ; 2(3): 214-24, 2015 Mar.
Article in English | MEDLINE | ID: mdl-26151067

ABSTRACT

The polarization of tissue resident macrophages toward the alternatively activated, anti-inflammatory M2 phenotype is believed to positively impact obesity and insulin resistance. Here we show that the soluble form of the extracellular domain (ECD) of C-type lectin-like receptor 2, CLEC2, regulates Kupffer cell polarization in the liver and improves glucose and lipid parameters in diabetic animal models. Over-expression of Fc-CLEC2(ECD) in mice via in vivo gene delivery, or injection of recombinant Fc-CLEC2(ECD) protein, results in a reduction of blood glucose and liver triglyceride levels and improves glucose tolerance. Furthermore, Fc-CLEC2(ECD) treatment improves cytokine profiles and increases both the M2 macrophage population and the genes involved in the oxidation of lipid metabolism in the liver. These data reveal a previously unidentified role for CLEC2 as a regulator of macrophage polarity, and establish CLEC2 as a promising therapeutic target for treatment of diabetes and liver disease.


Subject(s)
Glucose/metabolism , Kupffer Cells/metabolism , Lectins, C-Type/metabolism , Lipid Metabolism/physiology , Animals , Cell Polarity , Fatty Liver/genetics , Fatty Liver/metabolism , Homeostasis/drug effects , Humans , Kupffer Cells/cytology , Kupffer Cells/drug effects , Lectins, C-Type/genetics , Lipid Metabolism/drug effects , Macrophage Activation/drug effects , Macrophages/drug effects , Macrophages/metabolism , Male , Mice, Inbred C57BL , Mice, Inbred Strains , Protein Structure, Tertiary , Receptors, Fc/genetics , Recombinant Proteins/genetics , Recombinant Proteins/pharmacology , Solubility
5.
PLoS One ; 10(6): e0126924, 2015.
Article in English | MEDLINE | ID: mdl-26083576

ABSTRACT

Trefoil factor 3 (TFF3), also called intestinal trefoil factor or Itf, is a 59 amino acid peptide found as a homodimer predominantly along the gastrointestinal tract and in serum. TFF3 expression is elevated during gastrointestinal adenoma progression and has been shown to promote mucosal wound healing. Here we show that in contrast to other trefoil factor family members, TFF1 and TFF2, TFF3 is highly expressed in mouse duodenum, jejunum and ileum and that its expression is regulated by food intake. Overexpression of TFF3 using a recombinant adeno-associated virus (AAV) vector, or daily administration of recombinant TFF3 protein in vivo improved glucose tolerance in a diet-induced obesity mouse model. Body weight, fasting insulin, triglyceride, cholesterol and leptin levels were not affected by TFF3 treatment. Induction of mucinous metaplasia was observed in mice with AAV-mediated TFF3 overexpression, however, no such adverse histological effect was seen after the administration of recombinant TFF3 protein. Altogether these results suggest that the therapeutic potential of targeting TFF3 to treat T2D may be limited.


Subject(s)
Blood Glucose/metabolism , Eating/genetics , Genetic Vectors/adverse effects , Metaplasia/genetics , Mucins/genetics , Obesity/genetics , Animals , Cholesterol/blood , Dependovirus/genetics , Diet, High-Fat , Duodenum/metabolism , Duodenum/pathology , Gene Expression , Gene Expression Regulation , Genetic Vectors/chemistry , Genetic Vectors/metabolism , Glucose Tolerance Test , Humans , Ileum/metabolism , Ileum/pathology , Insulin/blood , Jejunum/metabolism , Jejunum/pathology , Leptin/blood , Male , Metaplasia/etiology , Metaplasia/metabolism , Metaplasia/pathology , Mice , Mucins/administration & dosage , Mucins/metabolism , Obesity/etiology , Obesity/metabolism , Obesity/pathology , Recombinant Proteins/administration & dosage , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Signal Transduction , Trefoil Factor-2 , Trefoil Factor-3 , Triglycerides/blood
6.
Expert Opin Ther Targets ; 18(11): 1253-64, 2014 Nov.
Article in English | MEDLINE | ID: mdl-25287216

ABSTRACT

OBJECTIVE: Although the human genome encodes ∼ 20,000 protein-coding genes, only a very small fraction of these have been explored as potential targets for therapeutic development. The challenge of identifying and validating new protein targets has contributed to the significant reduction in the productivity of the pharmaceutical industry in the recent decade, highlighting the continued need to find new therapeutic targets. RESEARCH DESIGN AND METHODS: The traditional methods to discover new targets are expensive, low throughput and time consuming, usually taking years to validate or invalidate a target. To address these limitations, as a proof of concept, we explored the hydrodynamic tail vein (HTV) injection as a gene delivery method for direct in vivo phenotypic screening of novel secreted factor targets for Type II diabetes therapeutics. RESULTS: High levels and sustained expression of target proteins were observed in diabetic mouse models tested, allowing us to identify multiple novel hormones that may regulate glucose metabolism. CONCLUSIONS: These results suggest that HTV is a low-cost, high-throughput method for direct in vivo phenotypic drug screening in metabolic disorders and could be applicable to many other disease areas as well. This method if combined with other approaches such as human genetic studies could provide a significant value to future drug discovery.


Subject(s)
Diabetes Mellitus, Experimental/drug therapy , Diabetes Mellitus, Type 2/drug therapy , Hypoglycemic Agents/pharmacology , Proteome , Animals , Diabetes Mellitus, Experimental/physiopathology , Diabetes Mellitus, Type 2/physiopathology , Drug Discovery/methods , Gene Transfer Techniques , Glucose/metabolism , High-Throughput Screening Assays/methods , Humans , Hydrodynamics , Injections, Intravenous , Male , Mice , Mice, Inbred C57BL , Phenotype , Tail/blood supply
7.
J Biol Chem ; 289(44): 30470-30480, 2014 Oct 31.
Article in English | MEDLINE | ID: mdl-25204652

ABSTRACT

The role of fibroblast growth factor receptor 4 (FGFR4) in regulating bile acid synthesis has been well defined; however, its reported role on glucose and energy metabolism remains unresolved. Here, we show that FGFR4 deficiency in mice leads to improvement in glucose metabolism, insulin sensitivity, and reduction in body weight under high fat conditions. Mechanism of action studies in FGFR4-deficient mice suggest that the effects are mediated in part by increased plasma levels of adiponectin and the endocrine FGF factors FGF21 and FGF15, the latter of which increase in response to an elevated bile acid pool. Direct actions of increased bile acids on bile acid receptors, and other potential indirect mechanisms, may also contribute to the observed metabolic changes. The results described herein suggest that FGFR4 antagonists alone, or in combination with other agents, could serve as a novel treatment for diabetes.


Subject(s)
Diet, High-Fat/adverse effects , Insulin Resistance , Obesity/metabolism , Receptor, Fibroblast Growth Factor, Type 4/genetics , Adiponectin/blood , Adipose Tissue/metabolism , Animals , Female , Fibroblast Growth Factors/administration & dosage , Fibroblast Growth Factors/blood , Glucose/metabolism , HEK293 Cells , Humans , Ileum/metabolism , Liver/metabolism , Male , Mice, Inbred C57BL , Mice, Knockout , Obesity/etiology , Receptor, Fibroblast Growth Factor, Type 4/deficiency , Transcriptome
8.
PLoS One ; 8(4): e61432, 2013.
Article in English | MEDLINE | ID: mdl-23630589

ABSTRACT

The endocrine hormone FGF21 has attracted considerable interest as a potential therapeutic for treating diabetes and obesity. As an alternative to the native cytokine, we generated bispecific Avimer polypeptides that bind with high affinity and specificity to one of the receptor and coreceptor pairs used by FGF21, FGFR1c and ß-Klotho. These Avimers exhibit FGF21-like activity in in vitro assays with potency greater than FGF21. In a study conducted in obese male cynomolgus monkeys, animals treated with an FGFR1c/ß-Klotho bispecific Avimer showed improved metabolic parameters and reduced body weight comparable to the effects seen with FGF21. These results not only demonstrate the essential roles of FGFR1c and ß-Klotho in mediating the metabolic effects of FGF21, they also describe a first bispecific activator of this unique receptor complex and provide validation for a novel therapeutic approach to target this potentially important pathway for treating diabetes and obesity.


Subject(s)
Anti-Obesity Agents/pharmacology , Fibroblast Growth Factors/physiology , Membrane Proteins/antagonists & inhibitors , Obesity/drug therapy , Peptides/pharmacology , Receptor, Fibroblast Growth Factor, Type 1/antagonists & inhibitors , Amino Acid Sequence , Animals , Anti-Obesity Agents/pharmacokinetics , Binding Sites , Binding, Competitive , Body Weight/drug effects , Cell Line , Drug Evaluation, Preclinical , Fibroblast Growth Factors/chemistry , Insulin/blood , Klotho Proteins , Macaca fascicularis , Male , Membrane Proteins/biosynthesis , Mice , Molecular Mimicry , Molecular Sequence Data , Obesity/blood , Peptides/pharmacokinetics , Protein Binding , Rats , Receptor, Fibroblast Growth Factor, Type 4/chemistry , Recombinant Fusion Proteins/antagonists & inhibitors , Recombinant Fusion Proteins/biosynthesis , Serum Albumin/pharmacokinetics , Serum Albumin/pharmacology , Signal Transduction , Triglycerides/blood
9.
BioDrugs ; 27(2): 159-66, 2013 Apr.
Article in English | MEDLINE | ID: mdl-23456652

ABSTRACT

BACKGROUND AND OBJECTIVE: Fibroblast growth factor 21 (FGF21) has potent effects on normalizing glucose, lipid, and energy homeostasis, and represents an attractive novel therapy for type 2 diabetes mellitus and obesity. Approaches to improve the pharmacokinetic properties of FGF21, such as conjugation with polyethylene glycol, have been explored for therapeutic development. However, not only is there room for further pharmacokinetic improvements, additional re-engineering approaches to improve the potency and stability of FGF21 have not been reported. Here, we describe a novel approach to modify and improve the function of FGF21 by altering its C-terminal ßKlotho interaction domain. METHODS: We first identified Avimer proteins that are capable of binding ßKlotho. Then we explored replacing the C-terminal ßKlotho interaction domain of FGF21 with a ßKlotho-binding Avimer protein. RESULTS: Such a ßKlotho-binding Avimer protein was able to fully complement the C-terminal domain function of FGF21. The resulting FGF21-Avimer fusion is functionally indistinguishable from wild type FGF21, and more tolerant of C-terminal modification. CONCLUSION: These results demonstrate a viable strategy to modulate the affinity, potency, and engineering of FGF21, paving the way for further improvements of FGF21 as a therapeutic.


Subject(s)
Anti-Obesity Agents/pharmacology , Fibroblast Growth Factors/pharmacology , Hypoglycemic Agents/pharmacology , Protein Engineering/methods , Recombinant Fusion Proteins/pharmacology , Amino Acid Sequence , Animals , Anti-Obesity Agents/chemistry , Anti-Obesity Agents/therapeutic use , Blood Glucose/analysis , Fibroblast Growth Factors/genetics , Fibroblast Growth Factors/standards , Fibroblast Growth Factors/therapeutic use , Humans , Hypoglycemic Agents/chemistry , Hypoglycemic Agents/therapeutic use , Male , Mice , Mice, Inbred Strains , Molecular Sequence Data , Obesity/blood , Obesity/drug therapy , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/standards , Recombinant Fusion Proteins/therapeutic use
10.
J Lipid Res ; 54(2): 325-32, 2013 Feb.
Article in English | MEDLINE | ID: mdl-23204296

ABSTRACT

Elevated triglyceride (TG) and cholesterol levels are risk factors for cardiovascular disease and are often associated with diabetes and metabolic syndrome. Recent reports suggest that fibroblast growth factor (FGF)19 and FGF21 can dramatically improve metabolic dysfunction, including hyperglycemia, hypertriglyceridemia, and hypercholesterolemia. Due to their similar receptor specificities and co-receptor requirements, FGF19 and FGF21 share many common properties and have been thought to be interchangeable in metabolic regulation. Here we directly compared how pharmacological administration of recombinant FGF19 or FGF21 proteins affect metabolism in B6.V-Lep(ob)/J leptin-deficient mice. FGF19 and FGF21 equally improved glucose parameters; however, we observed increased serum TG and cholesterol levels after treatment with FGF19 but not with FGF21. Increases in serum TGs were also observed after a 4-day treatment with FGF19 in C57BL6/J mice on a high-fat diet. This is in contrast to many literature reports that showed significant improvements in hyperlipidemia after chronic treatment with FGF19 or FGF21 in high-fat diet models. We propose that FGF19 has lipid-raising and lipid-lowering actions mediated through different FGF receptors and target tissues, and the results described here provide a potential mechanism that may explain the inconsistency in the reported effects of FGF19 on lipid metabolism.


Subject(s)
Fibroblast Growth Factors/pharmacology , Lipid Metabolism/drug effects , Animals , Cell Line , Cholesterol/blood , Diet/adverse effects , Fibroblast Growth Factors/chemistry , Liver/drug effects , Liver/metabolism , Male , Mice , Obesity/blood , Obesity/etiology , Receptor, Fibroblast Growth Factor, Type 4/metabolism , Triglycerides/blood
11.
Sci Transl Med ; 4(162): 162ra153, 2012 Nov 28.
Article in English | MEDLINE | ID: mdl-23197570

ABSTRACT

Fibroblast growth factor 21 (FGF21) is a distinctive member of the FGF family with potent beneficial effects on lipid, body weight, and glucose metabolism and has attracted considerable interest as a potential therapeutic for treating diabetes and obesity. As an alternative to native FGF21, we have developed a monoclonal antibody, mimAb1, that binds to ßKlotho with high affinity and specifically activates signaling from the ßKlotho/FGFR1c (FGF receptor 1c) receptor complex. In obese cynomolgus monkeys, injection of mimAb1 led to FGF21-like metabolic effects, including decreases in body weight, plasma insulin, triglycerides, and glucose during tolerance testing. Mice with adipose-selective FGFR1 knockout were refractory to FGF21-induced improvements in glucose metabolism and body weight. These results in obese monkeys (with mimAb1) and in FGFR1 knockout mice (with FGF21) demonstrated the essential role of FGFR1c in FGF21 function and suggest fat as a critical target tissue for the cytokine and antibody. Because mimAb1 depends on ßKlotho to activate FGFR1c, it is not expected to induce side effects caused by activating FGFR1c alone. The unexpected finding of an antibody that can activate FGF21-like signaling through cell surface receptors provided preclinical validation for an innovative therapeutic approach to diabetes and obesity.


Subject(s)
Antibodies, Monoclonal/therapeutic use , Diabetes Mellitus/drug therapy , Fibroblast Growth Factors/immunology , Glucuronidase/metabolism , Obesity/drug therapy , Receptor, Fibroblast Growth Factor, Type 1/metabolism , Adipose Tissue/drug effects , Adipose Tissue/metabolism , Adipose Tissue/pathology , Animals , Antibodies, Monoclonal/pharmacology , Body Weight/genetics , Diabetes Mellitus/blood , Epitopes/chemistry , Glucose/metabolism , Glucuronidase/immunology , Humans , Klotho Proteins , Macaca fascicularis , Mice , Obesity/blood , Obesity/complications , Phosphates/blood , Receptor, Fibroblast Growth Factor, Type 1/agonists , Signal Transduction/drug effects , Triglycerides/blood
12.
FEBS Lett ; 586(8): 1214-9, 2012 Apr 24.
Article in English | MEDLINE | ID: mdl-22575658

ABSTRACT

Adhesion G-protein-coupled receptors (GPCR) are special members of GPCRs with long N-termini containing multiple domains. We overexpressed our collection of receptors together with G-proteins in mammalian cell lines and measured the concentrations of intracellular signaling molecules, such as inositol phosphate and cAMP. Our results show that a subset of tested adhesion GPCRs has constitutive activities and is capable of coupling to a variety of G-proteins. In addition, we have identified a small molecule compound that specifically activates one of the subfamily members, GPR97, and the activation was confirmed by an independent GTPγS assay. These findings suggest classical GPCR screening assays could be applied to de-orphanize these receptors, and provide pharmacological tools to improve understanding of the physiological functions of these receptors.


Subject(s)
Receptors, G-Protein-Coupled/metabolism , Signal Transduction , Animals , CHO Cells , Cell Line , Cricetinae , Cyclic AMP/metabolism , Guanosine 5'-O-(3-Thiotriphosphate)/metabolism , Humans , Inositol Phosphates/metabolism , Transfection
13.
PLoS One ; 7(3): e33603, 2012.
Article in English | MEDLINE | ID: mdl-22457778

ABSTRACT

Diabetes and associated metabolic conditions have reached pandemic proportions worldwide, and there is a clear unmet medical need for new therapies that are both effective and safe. FGF19 and FGF21 are distinctive members of the FGF family that function as endocrine hormones. Both have potent effects on normalizing glucose, lipid, and energy homeostasis, and therefore, represent attractive potential next generation therapies for combating the growing epidemics of type 2 diabetes and obesity. The mechanism responsible for these impressive metabolic effects remains unknown. While both FGF19 and FGF21 can activate FGFRs 1c, 2c, and 3c in the presence of co-receptor ßKlotho in vitro, which receptor is responsible for the metabolic activities observed in vivo remains unknown. Here we have generated a variant of FGF19, FGF19-7, that has altered receptor specificity with a strong bias toward FGFR1c. We show that FGF19-7 is equally efficacious as wild type FGF19 in regulating glucose, lipid, and energy metabolism in both diet-induced obesity and leptin-deficient mouse models. These results are the first direct demonstration of the central role of the ßKlotho/FGFR1c receptor complex in glucose and lipid regulation, and also strongly suggest that activation of this receptor complex alone might be sufficient to achieve all the metabolic functions of endocrine FGF molecules.


Subject(s)
Fibroblast Growth Factors/metabolism , Glucose/metabolism , Receptor, Fibroblast Growth Factor, Type 1/physiology , Animals , Blotting, Western , Cell Line , Dependovirus/genetics , Fibroblast Growth Factors/genetics , Genetic Vectors , Glucose Tolerance Test , Liver/metabolism , Male , Mice , Protein Binding , Receptor, Fibroblast Growth Factor, Type 1/metabolism , Signal Transduction
14.
J Mol Biol ; 418(1-2): 82-9, 2012 Apr 20.
Article in English | MEDLINE | ID: mdl-22370560

ABSTRACT

Three fibroblast growth factor (FGF) molecules, FGF19, FGF21, and FGF23, form a unique subfamily that functions as endocrine hormones. FGF19 and FGF21 can regulate glucose, lipid, and energy metabolism, while FGF23 regulates phosphate homeostasis. The FGF receptors and co-receptors for these three FGF molecules have been identified, and domains important for receptor interaction and specificity determination are beginning to be elucidated. However, a number of questions remain unanswered, such as the identification of fibroblast growth factor receptor responsible for glucose regulation. Here, we have generated a variant of FGF23: FGF23-21c, where the C-terminal domain of FGF23 was replaced with the corresponding regions from FGF21. FGF23-21c showed a number of interesting and unexpected properties in vitro. In contrast to wild-type FGF23, FGF23-21c gained the ability to activate FGFR1c and FGFR2c in the presence of ßKlotho and was able to stimulate glucose uptake into adipocytes in vitro and lower glucose levels in ob/ob diabetic mice model to similar extent as FGF21 in vivo. These results suggest that ßKlotho/FGFR1c or FGFR2c receptor complexes are sufficient for glucose regulation. Interestingly, without the FGF23 C-terminal domain, FGF23-21c was still able to activate fibroblast growth factor receptors in the presence of αKlotho. This suggests not only that sequences outside of the C-terminal region may also contribute to the interaction with co-receptors but also that FGF23-21c may be able to regulate both glucose and phosphate metabolisms. This raises an interesting concept of designing an FGF molecule that may be able to address multiple diseases simultaneously. Further understanding of FGF/receptor interactions may allow the development of exciting opportunities for novel therapeutic discovery.


Subject(s)
Fibroblast Growth Factors/metabolism , Glucuronidase/metabolism , Membrane Proteins/metabolism , Receptor, Fibroblast Growth Factor, Type 1/metabolism , Receptor, Fibroblast Growth Factor, Type 2/metabolism , Receptors, Cell Surface/metabolism , Signal Transduction , Adipocytes/metabolism , Animals , Blood Glucose/drug effects , Cell Line , Female , Fibroblast Growth Factor-23 , Fibroblast Growth Factors/pharmacology , Glucose/metabolism , Klotho Proteins , Male , Mice , Mice, Obese
15.
J Mol Biol ; 408(3): 491-502, 2011 May 06.
Article in English | MEDLINE | ID: mdl-21392510

ABSTRACT

FGF21 is a member of a unique subfamily of fibroblast growth factors that function as endocrine hormones and regulate a variety of metabolic activities. Unlike paracrine FGFs, FGF21 does not bind heparin and requires ßKlotho as a co-receptor to activate FGFR signaling. In the presence of ßKlotho, FGF21 is able to activate FGFRs 1c, 2c and 3c but not FGFR4. Chimeric FGFR1c/FGFR4 receptors were constructed to identify domains that confer this specificity and to understand regions important for FGF21-induced receptor activation. With these chimeras, we showed that domain 3 of the FGFR1c extracellular domain plays a critical role in specificity determination and receptor activation by FGF21. Furthermore, we were able to narrow down the sequences important for this function to a six amino acid region within domain 3 of FGFR1c. It is interesting to note that this region falls into the ßC'-ßE loop, which has been shown to be important for receptor specificity determination in paracrine FGFs, suggesting a common principle in both endocrine and paracrine FGF receptor interaction and activation.


Subject(s)
Fibroblast Growth Factors/metabolism , Receptor, Fibroblast Growth Factor, Type 1/metabolism , Receptor, Fibroblast Growth Factor, Type 4/metabolism , Animals , Cell Line , Models, Molecular , Protein Binding , Protein Interaction Domains and Motifs , Protein Structure, Quaternary , Protein Structure, Tertiary , Rats , Receptor, Fibroblast Growth Factor, Type 1/genetics , Receptor, Fibroblast Growth Factor, Type 4/genetics , Recombinant Proteins/genetics , Recombinant Proteins/metabolism
16.
Proc Natl Acad Sci U S A ; 107(32): 14158-63, 2010 Aug 10.
Article in English | MEDLINE | ID: mdl-20660733

ABSTRACT

FGF19 and FGF21 are distinctive members of the FGF family that function as endocrine hormones. Their potent effects on normalizing glucose, lipid, and energy homeostasis in disease models have made them an interesting focus of research for combating the growing epidemics of diabetes and obesity. Despite overlapping functions, FGF19 and FGF21 have many discrete effects, the most important being that FGF19 has both metabolic and proliferative effects, whereas FGF21 has only metabolic effects. Here we identify the structural determinants dictating differential receptor interactions that explain and distinguish these two physiological functions. We also have generated FGF19 variants that have lost the ability to induce hepatocyte proliferation but that still are effective in lowering plasma glucose levels and improving insulin sensitivity in mice. Our results add valuable insight into the structure-function relationship of FGF19/FGF21 and identify the structural basis underpinning the distinct proliferative feature of FGF19 compared with FGF21. In addition, these studies provide a road map for engineering FGF19 as a potential therapeutic candidate for treating diabetes and obesity.


Subject(s)
Blood Glucose , Cell Proliferation , Fibroblast Growth Factors/physiology , Hepatocytes/cytology , Amino Acid Sequence , Animals , Diabetes Mellitus/drug therapy , Genetic Variation , Insulin Resistance , Mice , Obesity/drug therapy , Peptide Fragments/pharmacology , Recombinant Fusion Proteins/pharmacology , Recombinant Proteins/pharmacology
17.
J Biol Chem ; 285(8): 5165-70, 2010 Feb 19.
Article in English | MEDLINE | ID: mdl-20018895

ABSTRACT

FGF19 and FGF21, unique members of the fibroblast growth factor (FGF) family, are hormones that regulate glucose, lipid, and energy homeostasis. Increased hepatocyte proliferation and liver tumor formation have also been observed in FGF19 transgenic mice. Here, we report that, in contrast to FGF19, FGF21 does not induce hepatocyte proliferation in vivo. To identify the mechanism for FGF19-induced hepatocyte proliferation, we explored similarities and differences in receptor specificity between FGF19 and FGF21. We find that although both are able to activate FGF receptors (FGFRs) 1c, 2c, and 3c, only FGF19 activates FGFR4, the predominant receptor in the liver. Using a C-terminal truncation mutant of FGF19 and a series of FGF19/FGF21 chimeric molecules, we determined that amino acids residues 38-42 of FGF19 are sufficient to confer both FGFR4 activation and increased hepatocyte proliferation in vivo to FGF21. These data suggest that activation of FGFR4 is the mechanism whereby FGF19 can increase hepatocyte proliferation and induce hepatocellular carcinoma formation.


Subject(s)
Carcinoma, Hepatocellular/mortality , Cell Proliferation , Cell Transformation, Neoplastic/metabolism , Fibroblast Growth Factors/metabolism , Hepatocytes/metabolism , Receptor, Fibroblast Growth Factor, Type 4/metabolism , 3T3-L1 Cells , Amino Acid Sequence , Animals , Carcinoma, Hepatocellular/genetics , Cell Transformation, Neoplastic/genetics , Female , Fibroblast Growth Factors/genetics , Fibroblast Growth Factors/pharmacology , Humans , Mice , Mice, Transgenic , Receptor, Fibroblast Growth Factor, Type 4/genetics , Sequence Deletion
18.
Proc Natl Acad Sci U S A ; 106(34): 14379-84, 2009 Aug 25.
Article in English | MEDLINE | ID: mdl-19706524

ABSTRACT

FGF19 is a hormone that regulates bile acid and glucose homeostasis. Progress has been made in identifying cofactors for receptor activation. However, several functions of FGF19 have not yet been fully defined, including the actions of FGF19 on target tissues, its FGF receptor specificity, and the contributions of other cofactors, such as heparin. Here, we explore the requirements for FGF19-FGFR/co-receptor interactions and signaling in detail. We show that betaKlotho was essential for FGF19 interaction with FGFRs 1c, 2c, and 3c, but FGF19 was able to interact directly with FGFR4 in the absence of betaKlotho in a heparin-dependent manner. Further, FGF19 activated FGFR4 signaling in the presence or absence of betaKlotho, but activation of FGFRs 1c, 2c, or 3c was completely betaKlotho dependent. We then generated an FGF19 molecule, FGF19dCTD, which has a deletion of the C-terminal region responsible for betaKlotho interaction. We determined that betaKlotho-dependent FGFR1c, 2c, and 3c interactions and activation were abolished, and betaKlotho-independent FGFR4 activation was preserved; therefore, FGF19dCTD is an FGFR4-specific activator. This unique FGF19 molecule specifically activated FGFR4-dependent signaling in liver and suppressed CYP7A1 expression in vivo, but was unable to activate signaling in adipose where FGFR4 expression is very low. Interestingly, unlike FGF19, treatment of ob/ob mice with FGF19dCTD failed to improve glucose levels and insulin sensitivity. These results suggest that FGF19-regulated liver bile acid metabolism could be independent of its glucose-lowering effect, and direct FGFR activation in adipose tissue may play an important role in the regulation of glucose homeostasis.


Subject(s)
Fibroblast Growth Factors/metabolism , Glucose/metabolism , Obesity/metabolism , Receptor, Fibroblast Growth Factor, Type 4/metabolism , Adipose Tissue/metabolism , Animals , Blotting, Western , Cell Line , Fibroblast Growth Factors/genetics , Fibroblast Growth Factors/pharmacokinetics , Glucose/pharmacokinetics , Glucose Tolerance Test , Klotho Proteins , Liver/metabolism , Membrane Proteins/genetics , Membrane Proteins/metabolism , Mice , Mice, Inbred C57BL , Mice, Obese , Obesity/genetics , Protein Binding , Receptor, Fibroblast Growth Factor, Type 1/genetics , Receptor, Fibroblast Growth Factor, Type 1/metabolism , Receptor, Fibroblast Growth Factor, Type 2/genetics , Receptor, Fibroblast Growth Factor, Type 2/metabolism , Receptor, Fibroblast Growth Factor, Type 3/genetics , Receptor, Fibroblast Growth Factor, Type 3/metabolism , Receptor, Fibroblast Growth Factor, Type 4/genetics , Reverse Transcriptase Polymerase Chain Reaction , Tissue Distribution , Transcriptional Activation
19.
J Biol Chem ; 283(48): 33304-9, 2008 Nov 28.
Article in English | MEDLINE | ID: mdl-18829467

ABSTRACT

FGF19 subfamily proteins (FGF19, FGF21, and FGF23) are unique members of fibroblast growth factors (FGFs) that regulate energy, bile acid, glucose, lipid, phosphate, and vitamin D homeostasis in an endocrine fashion. Their activities require the presence of alpha or betaKlotho, two related single-pass transmembrane proteins, as co-receptors in relevant target tissues. We previously showed that FGF19 can bind to both alpha and betaKlotho, whereas FGF21 and FGF23 can bind only to either betaKlotho or alphaKlotho, respectively in vitro. To determine the mechanism regulating the binding and specificity among FGF19 subfamily members to Klotho family proteins, chimeric proteins between FGF19 subfamily members or chimeric proteins between Klotho family members were constructed to probe the interaction between those two families. Our results showed that a chimera of FGF19 with the FGF21 C-terminal tail interacts only with betaKlotho and a chimera with the FGF23 C-terminal tail interacts only with alphaKlotho. FGF signaling assays also reflected the change of specificity we observed for the chimeras. These results identified the C-terminal tail of FGF19 as a region necessary for its recognition of Klotho family proteins. In addition, chimeras between alpha and betaKlotho were also generated to probe the regions in Klotho proteins that are important for signaling by this FGF subfamily. Both FGF23 and FGF21 require intact alpha or betaKlotho for signaling, respectively, whereas FGF19 can signal through a Klotho chimera consisting of the N terminus of alphaKlotho and the C terminus of betaKlotho. Our results provide the first glimpse of the regions that regulate the binding specificity between this unique family of FGFs and their co-receptors.


Subject(s)
Fibroblast Growth Factors/metabolism , Glucuronidase/metabolism , Signal Transduction/physiology , Cell Line , Fibroblast Growth Factor-23 , Fibroblast Growth Factors/genetics , Glucuronidase/genetics , Humans , Klotho Proteins , Protein Binding/genetics , Protein Structure, Tertiary/physiology , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism
20.
J Lipid Res ; 49(4): 797-803, 2008 Apr.
Article in English | MEDLINE | ID: mdl-18174606

ABSTRACT

GPR81 is an orphan G protein-coupled receptor (GPCR) that has a high degree of homology to the nicotinic acid receptor GPR109A. GPR81 expression is highly enriched and specific in adipocytes. However, the function and signaling properties of GPR81 are unknown because of the lack of natural or synthetic ligands. Using chimeric G proteins that convert Gi-coupled receptors to Gq-mediated inositol phosphate (IP) accumulation, we show that GPR81 can constitutively increase IP accumulation in HEK293 cells and suggest that GPR81 couples to the Gi signaling pathway. We also constructed a chimeric receptor that expresses the extracellular domains of cysteinyl leukotriene 2 receptor (CysLT2R) and the intracellular domains of GPR81. We show that the CysLT2R ligand, leukotriene D(4) (LTD4), is able to activate this chimeric receptor through activation of the Gi pathway. In addition, LTD4 is able to inhibit lipolysis in adipocytes expressing this chimeric receptor. These results suggest that GPR81 couples to the Gi signaling pathway and that activation of the receptor may regulate adipocyte function and metabolism. Hence, targeting GPR81 may lead to the development of a novel and effective therapy for dyslipidemia and a better side effect profile than nicotinic acid.


Subject(s)
Receptors, G-Protein-Coupled/metabolism , Signal Transduction , Adipocytes/cytology , Adipocytes/drug effects , Adipocytes/metabolism , Amino Acid Sequence , Animals , Cell Differentiation , Cell Line , Cricetinae , GTP-Binding Protein alpha Subunits, Gi-Go/metabolism , Humans , Leukotriene D4/pharmacology , Mice , Molecular Sequence Data , Organ Specificity , Receptors, G-Protein-Coupled/chemistry , Receptors, G-Protein-Coupled/genetics , Sequence Alignment , Sequence Homology, Amino Acid , Transcription, Genetic/genetics , Up-Regulation
SELECTION OF CITATIONS
SEARCH DETAIL