Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 14 de 14
Filter
Add more filters










Publication year range
1.
Cell Metab ; 35(2): 274-286.e10, 2023 02 07.
Article in English | MEDLINE | ID: mdl-36630958

ABSTRACT

GDF15 and its receptor GFRAL/RET form a non-homeostatic system that regulates food intake and body weight in preclinical species. Here, we describe a GDF15 analog, LY3463251, a potent agonist at the GFRAL/RET receptor with prolonged pharmacokinetics. In rodents and obese non-human primates, LY3463251 decreased food intake and body weight with no signs of malaise or emesis. In a first-in-human study in healthy participants, single subcutaneous LY3463251 injections showed a safety and pharmacokinetic profile supporting further clinical development with dose-dependent nausea and emesis in a subset of individuals. A subsequent 12-week multiple ascending dose study in overweight and obese participants showed that LY3463251 induced significant decreases in food intake and appetite scores associated with modest body weight reduction independent of nausea and emesis (clinicaltrials.gov: NCT03764774). These observations demonstrate that agonism of the GFRAL/RET system can modulate energy balance in humans, though the decrease in body weight is surprisingly modest, suggesting challenges in leveraging the GDF15 system for clinical weight-loss applications.


Subject(s)
Obesity , Weight Loss , Animals , Humans , Body Weight , Obesity/drug therapy , Vomiting , Growth Differentiation Factor 15
2.
Proc Natl Acad Sci U S A ; 119(13): e2116506119, 2022 03 29.
Article in English | MEDLINE | ID: mdl-35333651

ABSTRACT

SignificanceTirzepatide is a dual agonist of the glucose-dependent insulinotropic polypeptide receptor (GIPR) and the glucagon-like peptide-1 receptor (GLP-1R), which are incretin receptors that regulate carbohydrate metabolism. This investigational agent has proven superior to selective GLP-1R agonists in clinical trials in subjects with type 2 diabetes mellitus. Intriguingly, although tirzepatide closely resembles native GIP in how it activates the GIPR, it differs markedly from GLP-1 in its activation of the GLP-1R, resulting in less agonist-induced receptor desensitization. We report how cryogenic electron microscopy and molecular dynamics simulations inform the structural basis for the unique pharmacology of tirzepatide. These studies reveal the extent to which fatty acid modification, combined with amino acid sequence, determines the mode of action of a multireceptor agonist.


Subject(s)
Diabetes Mellitus, Type 2 , Receptors, Gastrointestinal Hormone , Diabetes Mellitus, Type 2/drug therapy , Diabetes Mellitus, Type 2/metabolism , Gastric Inhibitory Polypeptide/metabolism , Gastric Inhibitory Polypeptide/pharmacology , Gastric Inhibitory Polypeptide/therapeutic use , Glucagon-Like Peptide-1 Receptor/metabolism , Humans , Incretins/pharmacology , Receptors, Gastrointestinal Hormone/agonists , Receptors, Gastrointestinal Hormone/metabolism , Receptors, Gastrointestinal Hormone/therapeutic use
3.
Elife ; 92020 07 29.
Article in English | MEDLINE | ID: mdl-32723474

ABSTRACT

The cytokine, GDF15, is produced in pathological states which cause cellular stress, including cancer. When over expressed, it causes dramatic weight reduction, suggesting a role in disease-related anorexia. Here, we demonstrate that the GDF15 receptor, GFRAL, is located in a subset of cholecystokinin neurons which span the area postrema and the nucleus of the tractus solitarius of the mouse. GDF15 activates GFRALAP/NTS neurons and supports conditioned taste and place aversions, while the anorexia it causes can be blocked by a monoclonal antibody directed at GFRAL or by disrupting CCK neuronal signalling. The cancer-therapeutic drug, cisplatin, induces the release of GDF15 and activates GFRALAP/NTS neurons, as well as causing significant reductions in food intake and body weight in mice. These metabolic effects of cisplatin are abolished by pre-treatment with the GFRAL monoclonal antibody. Our results suggest that GFRAL neutralising antibodies or antagonists may provide a co-treatment opportunity for patients undergoing chemotherapy.


Subject(s)
Anorexia/genetics , Brain Stem/physiology , Growth Differentiation Factor 15/genetics , Neurons/physiology , Pica/genetics , Signal Transduction , Animals , Cholecystokinin/metabolism , Growth Differentiation Factor 15/administration & dosage , Growth Differentiation Factor 15/metabolism , Male , Mice , Mice, Inbred C57BL , Random Allocation , Rats , Rats, Sprague-Dawley , Recombinant Proteins/administration & dosage
4.
Endocrinology ; 158(10): 3090-3096, 2017 10 01.
Article in English | MEDLINE | ID: mdl-28938451

ABSTRACT

The increased prevalence of obesity and its cardiometabolic implications demonstrates the imperative to identify novel therapeutic targets able to effect meaningful metabolic changes in this population. Antibody-mediated targeting of fibroblast growth factor receptor 1c isoform (FGFR1c) has been shown to ameliorate hyperglycemia and protect from diet- and genetically-induced obesity in rodents and nonhuman primates. However, it is currently unknown which tissue(s) contribute to this glucose-lowering effect. Thus, to elucidate this effect, we treated euglycemic mice with H7, a monoclonal antibody that selectively targets FGFR1c, and used whole-body positron emission computed tomography with a glucose tracer (18F-fluorodeoxyglucose). Treatment with H7 increased basal glucose uptake in white adipose tissue (WAT), brown adipose tissue (BAT), the brain, and liver but reduced it in the quadriceps muscles. Consequentially, blood glucose was significantly reduced in response to treatment. Under insulin-stimulated conditions, the effects of H7 were maintained in WAT, BAT, liver, and muscle. Treatment with H7 decreased triglyceride (TG) content and increased adipose TG lipase content in white adipose tissue, while increasing activation of acetyl coenzyme A carboxylase, suggesting futile cycling of TGs, albeit favoring net hydrolysis. We demonstrated, in vitro, this is a direct effect of treatment in adipose tissue, as basal cellular respiration and glucose uptake were increased in response to treatment. Taken together, these data suggest that antibody-mediated targeting of FGFR1c exerts its powerful glucose-lowering efficacy primarily due to increased glucose uptake in adipose tissue.


Subject(s)
Adipose Tissue, Brown/metabolism , Adipose Tissue, White/metabolism , Antibodies, Monoclonal/administration & dosage , Receptor, Fibroblast Growth Factor, Type 1/antagonists & inhibitors , Acetyl-CoA Carboxylase/metabolism , Animals , Antibodies, Monoclonal/therapeutic use , Blood Glucose/analysis , Brain/drug effects , Brain/metabolism , Enzyme Activation/drug effects , Glucose/metabolism , Insulin/pharmacology , Lipase/analysis , Liver/drug effects , Liver/metabolism , Male , Mice , Mice, Inbred C57BL , Muscle, Skeletal/drug effects , Muscle, Skeletal/metabolism , Obesity/metabolism , Protein Isoforms , Receptor, Fibroblast Growth Factor, Type 1/immunology , Triglycerides/metabolism
5.
PLoS One ; 8(3): e58575, 2013.
Article in English | MEDLINE | ID: mdl-23536797

ABSTRACT

Fibroblast growth factor 21 is a novel hormonal regulator with the potential to treat a broad variety of metabolic abnormalities, such as type 2 diabetes, obesity, hepatic steatosis, and cardiovascular disease. Human recombinant wild type FGF21 (FGF21) has been shown to ameliorate metabolic disorders in rodents and non-human primates. However, development of FGF21 as a drug is challenging and requires re-engineering of its amino acid sequence to improve protein expression and formulation stability. Here we report the design and characterization of a novel FGF21 variant, LY2405319. To enable the development of a potential drug product with a once-daily dosing profile, in a preserved, multi-use formulation, an additional disulfide bond was introduced in FGF21 through Leu118Cys and Ala134Cys mutations. FGF21 was further optimized by deleting the four N-terminal amino acids, His-Pro-Ile-Pro (HPIP), which was subject to proteolytic cleavage. In addition, to eliminate an O-linked glycosylation site in yeast a Ser167Ala mutation was introduced, thus allowing large-scale, homogenous protein production in Pichia pastoris. Altogether re-engineering of FGF21 led to significant improvements in its biopharmaceutical properties. The impact of these changes was assessed in a panel of in vitro and in vivo assays, which confirmed that biological properties of LY2405319 were essentially identical to FGF21. Specifically, subcutaneous administration of LY2405319 in ob/ob and diet-induced obese (DIO) mice over 7-14 days resulted in a 25-50% lowering of plasma glucose coupled with a 10-30% reduction in body weight. Thus, LY2405319 exhibited all the biopharmaceutical and biological properties required for initiation of a clinical program designed to test the hypothesis that administration of exogenous FGF21 would result in effects on disease-related metabolic parameters in humans.


Subject(s)
Fibroblast Growth Factors/pharmacology , Recombinant Proteins , 3T3 Cells , Amino Acid Substitution , Animals , Cell Line , Drug Design , Fibroblast Growth Factors/chemistry , Fibroblast Growth Factors/genetics , Gene Expression , Genetic Variation , Hep G2 Cells , Humans , Klotho Proteins , Male , Membrane Proteins/metabolism , Mice , Models, Molecular , Pichia/genetics , Pichia/metabolism , Protein Conformation , Protein Stability , Temperature
6.
PLoS One ; 7(5): e38438, 2012.
Article in English | MEDLINE | ID: mdl-22675463

ABSTRACT

Fibroblast growth factors 19 (FGF19) and 21 (FGF21) have emerged as key regulators of energy metabolism. Several studies have been conducted to understand the mechanism of FGF19 and FGF21 action, however, the data presented has often been inconsistent and at times contradictory. Here in a single study we compare the mechanisms mediating FGF19/FGF21 actions, and how similarities/differences in actions at the cellular level between these two factors translate to common/divergent physiological outputs. Firstly, we show that in cell culture FGF19/FGF21 are very similar, however, key differences are still observed differentiating the two. In vitro we found that both FGF's activate FGFRs in the context of ßKlotho (KLB) expression. Furthermore, both factors alter ERK phosphorylation and glucose uptake with comparable potency. Combination treatment of cells with both factors did not have additive effects and treatment with a competitive inhibitor, the FGF21 delta N17 mutant, also blocked FGF19's effects, suggestive of a shared receptor activation mechanism. The key differences between FGF21/FGF19 were noted at the receptor interaction level, specifically the unique ability of FGF19 to bind/signal directly via FGFR4. To determine if differential effects on energy homeostasis and hepatic mitogenicity exist we treated DIO and ob/ob mice with FGF19/FGF21. We find comparable efficacy of the two proteins to correct body weight and serum glucose in both DIO and ob/ob mice. Nevertheless, FGF21 and FGF19 had distinctly different effects on proliferation in the liver. Interestingly, in vivo blockade of FGF21 signaling in mice using ΔN17 caused profound changes in glycemia indicative of the critical role KLB and FGF21 play in the regulation of glucose homeostasis. Overall, our data demonstrate that while subtle differences exist in vitro the metabolic effects in vivo of FGF19/FGF21 are indistinguishable, supporting a shared mechanism of action for these two hormones in the regulation of energy balance.


Subject(s)
Fibroblast Growth Factors/pharmacology , Animals , Body Weight/drug effects , Cell Line , Cell Proliferation/drug effects , Eating/drug effects , Fibroblast Growth Factors/administration & dosage , Glucuronidase/metabolism , Hepatocytes/drug effects , Hepatocytes/metabolism , Klotho Proteins , Male , Mice , Mice, Inbred C57BL , Receptors, Fibroblast Growth Factor/metabolism , Signal Transduction/drug effects
7.
Adv Exp Med Biol ; 728: 214-28, 2012.
Article in English | MEDLINE | ID: mdl-22396172

ABSTRACT

The prevalence of obesity and diabetes has been dramatically increasing during the last decade suggesting a greater patient need for more efficacious and safer drugs. Large molecule therapy has played an important role in diabetes since the discovery of insulin. This legacy was continued upon the introduction of Humulin (first recombinant insulin), Humalog (first engineered insulin) and Byetta (first incretin mimetic). Several other protein therapeutics, such as leptin, adiponectin, bone morphogenic protein-9 and others, are currently in or considered for therapeutic development. Among them, FGF21 is one of the most promising candidates given its outstanding pharmacologic benefits for nearly each and every abnormality of a metabolic disease and lack of apparent side effects in a variety of animal models. Thus, FGF21 represents a novel and appealing therapeutic reagent for Type 2 diabetes mellitus, obesity, dyslipidemia, cardiovascular and fatty liver diseases. The in vitro biology, genetic animal models and in vivo pharmacology of FGF21 will be discussed in this chapter.


Subject(s)
Fibroblast Growth Factors/therapeutic use , Animals , Cell Line , Drug Administration Schedule , Fibroblast Growth Factors/administration & dosage , Fibroblast Growth Factors/genetics , Fibroblast Growth Factors/pharmacology , Humans , Indicators and Reagents/administration & dosage , Indicators and Reagents/pharmacology , Indicators and Reagents/therapeutic use
8.
J Cell Physiol ; 219(2): 227-34, 2009 May.
Article in English | MEDLINE | ID: mdl-19117008

ABSTRACT

Fibroblast growth factor 21 is a member of endocrine FGFs subfamily, along with FGF19 and FGF23. It is emerging as a novel regulator with beneficial effects on a variety of metabolic parameters, including glucose and lipid control. FGF21 activity depends on membrane protein betaKlotho that physically complexes with various FGF receptors, thus conferring them the ability to bind FGF21 and activate downstream signaling pathways. FGF21, like other FGFs, folds to a beta-trefoil-like core region, with disordered N- and C-termini. In order to investigate their role in the activity of FGF21, we have constructed a series of deletion mutants and tested them for their ability to (1) bind betaKlotho, analyzed by surface plasmon resonance spectroscopy (2) signal through MAPK phosphorylation and inhibit apoptosis in 3T3-L1/betaKlotho fibroblasts (3) stimulate GLUT1 mRNA upregulation and glucose uptake in 3T3-L1 adipocytes. Binding studies with betaKlotho revealed that the interaction with the co-receptor involves the C-terminus, as progressive removal of amino acids from the carboxy end decreased affinity for betaKlotho. By contrast, removal of up to 17 amino acids from the N-terminus had no effect on the interaction with betaKlotho. Terminal deletions had greater effect on function, as deletions of six amino acids from the amino-terminus and only four from the carboxy-terminus each significantly impacted activity (10-fold). Of the extreme terminal truncations, with no detectable activity, DeltaN17 acted as competitive antagonist while DeltaC20 did not. Our structure/function studies show that the C-terminus is important for betaKlotho interaction whereas the N-terminus likely interacts directly with FGF receptors.


Subject(s)
Fibroblast Growth Factors/chemistry , Fibroblast Growth Factors/metabolism , Protein Structure, Secondary , Protein Structure, Tertiary , Receptors, Fibroblast Growth Factor/metabolism , 3T3 Cells , Adipocytes/cytology , Adipocytes/physiology , Animals , Apoptosis/physiology , Fibroblast Growth Factor-23 , Fibroblast Growth Factors/genetics , Fibroblasts/cytology , Fibroblasts/physiology , Glucose Transporter Type 1/genetics , Glucose Transporter Type 1/metabolism , Klotho Proteins , MAP Kinase Signaling System/physiology , Membrane Proteins/genetics , Membrane Proteins/metabolism , Mice , Mitogen-Activated Protein Kinases/genetics , Mitogen-Activated Protein Kinases/metabolism , Mutation , Surface Plasmon Resonance
9.
EMBO J ; 28(3): 261-73, 2009 Feb 04.
Article in English | MEDLINE | ID: mdl-19131967

ABSTRACT

By screening a fetal brain two-hybrid library with the death domain of the p75 neurotrophin receptor (NTR), we identified the Sall2 transcription factor as a novel interacting protein. Sall2 is a unique member of the Sall gene family, which is believed to be a tumour suppressor. Here, we show that Sall2 contains a p75NTR interaction domain not found in other Sall proteins and that p75NTR/Sall2 complexes co-immunoprecipitate from brain lysates. NGF dissociates p75NTR/Sall2 complexes and activates TrkA, which has an obligate function in the nuclear translocation of Sall2. NGF also increases Sall2 expression and this is mediated by p75NTR, but may not require TrkA. Depletion of Sall2 from cells decreases the expression and activity of p21(WAF1/CIP1), as well as the ability of NGF to induce growth arrest and the development of neurites. Overexpression of Sall2 activates p21(WAF1/CIP1), induces growth arrest, and promotes neurite outgrowth independently of NGF. These data establish Sall2 as a link between NTRs and transcriptional events that regulate the growth and development of neuronal cells.


Subject(s)
Cell Cycle/drug effects , Nerve Growth Factor/pharmacology , Neurites/drug effects , Neurites/metabolism , Receptor, Nerve Growth Factor/metabolism , Signal Transduction/drug effects , Transcription Factors/metabolism , Animals , Cell Differentiation/drug effects , Cell Proliferation/drug effects , Cyclin-Dependent Kinase Inhibitor p21/metabolism , DNA-Binding Proteins , Gene Silencing/drug effects , Hippocampus/cytology , Humans , Intracellular Signaling Peptides and Proteins/metabolism , Mice , Neurons/cytology , Neurons/drug effects , PC12 Cells , Protein Binding/drug effects , Protein Structure, Tertiary , Protein Transport/drug effects , Rats , Receptor, Nerve Growth Factor/chemistry , Receptor, trkA/metabolism
10.
Endocrinology ; 149(12): 6018-27, 2008 Dec.
Article in English | MEDLINE | ID: mdl-18687777

ABSTRACT

Fibroblast growth factor 21 (FGF21) is a metabolic regulator that provides efficient and durable glycemic and lipid control in various animal models. However, its potential to treat obesity, a major health concern affecting over 30% of the population, has not been fully explored. Here we report that systemic administration of FGF21 for 2 wk in diet-induced obese and ob/ob mice lowered their mean body weight by 20% predominantly via a reduction in adiposity. Although no decrease in total caloric intake or effect on physical activity was observed, FGF21-treated animals exhibited increased energy expenditure, fat utilization, and lipid excretion, reduced hepatosteatosis, and ameliorated glycemia. Transcriptional and blood cytokine profiling studies revealed effects consistent with the ability of FGF21 to ameliorate insulin and leptin resistance, enhance fat oxidation and suppress de novo lipogenesis in liver as well as to activate futile cycling in adipose. Overall, these data suggest that FGF21 exhibits the therapeutic characteristics necessary for an effective treatment of obesity and fatty liver disease and provides novel insights into the metabolic determinants of these activities.


Subject(s)
Fibroblast Growth Factors/pharmacology , Obesity/drug therapy , Adiposity/drug effects , Animals , Body Weight/drug effects , Dietary Fats/administration & dosage , Energy Intake/drug effects , Energy Metabolism/drug effects , Insulin/blood , Insulin Resistance , Leptin/blood , Male , Mice , Mice, Inbred C57BL , Motor Activity/drug effects , Obesity/blood , Obesity/etiology
11.
FEBS Lett ; 582(12): 1725-30, 2008 May 28.
Article in English | MEDLINE | ID: mdl-18460341

ABSTRACT

Fibroblast growth factor 21 (FGF21) is active in murine adipocytes and has beneficial metabolic effects in animal models of type 2 diabetes mellitus. We assessed whether FGF21 influences lipolysis in human adipocytes and 3T3-L1 cells. FGF21 had no short-time effect (h) while a 3-day incubation with FGF21 attenuated hormone-stimulated lipolysis. FGF21 did not influence the mRNA expression of genes involved in regulating lipolysis, but significantly reduced the expression of the lipid droplet-associated phosphoprotein perilipin without affecting differentiation. Via reduced release of fatty acids into the circulation, the anti-lipolytic effect could be a mechanism through which FGF21 promotes insulin sensitivity in man.


Subject(s)
Adipocytes/metabolism , Fibroblast Growth Factors/physiology , Glucuronidase/metabolism , Insulin Resistance , Lipolysis , 3T3-L1 Cells , Adipocytes/drug effects , Animals , Carrier Proteins , Cell Differentiation/drug effects , Fatty Acids/blood , Fatty Acids/metabolism , Fibroblast Growth Factors/pharmacology , Gene Expression/drug effects , Humans , Klotho Proteins , Lipolysis/drug effects , Lipolysis/genetics , Mice , Perilipin-1 , Phosphoproteins/genetics , Phosphoproteins/metabolism
12.
J Cell Physiol ; 215(1): 1-7, 2008 Apr.
Article in English | MEDLINE | ID: mdl-18064602

ABSTRACT

Fibroblast growth factor-21 (FGF-21) is a metabolic regulator that can influence glucose and lipid control in diabetic rodents and primates. We demonstrate that betaKlotho is an integral part of an activated FGF-21-betaKlotho-FGF receptor (FGFR) complex thus a critical subunit of the FGF-21 receptor. Cells lacking betaKlotho did not respond to FGF-21; the introduction of betaKlotho to these cells conferred FGF-21-responsiveness and recapitulated the entire scope of FGF-21 signaling observed in naturally responsive cells. Interestingly, FGF-21-mediated effects are heparin independent suggesting that betaKlotho plays a role in FGF-21 activity similar to the one played by heparin in the signaling of conventional FGFs. Moreover, in addition to conferring specificity for FGF-21, betaKlotho appears to support FGF-19 activity and mediates the receptor selectivity profile of FGF-19. All together, these results indicate that betaKlotho and FGFRs form the cognate FGF-21 receptor complex, mediating FGF-21 cellular specificity and physiological effects.


Subject(s)
Fibroblast Growth Factors/metabolism , Membrane Proteins/metabolism , Receptors, Fibroblast Growth Factor/metabolism , 3T3-L1 Cells , Animals , Fibroblast Growth Factors/pharmacology , Humans , Klotho Proteins , Mice , Protein Binding
13.
Cancer Res ; 67(2): 450-4, 2007 Jan 15.
Article in English | MEDLINE | ID: mdl-17234751

ABSTRACT

Hdm2 is elevated in numerous types of malignancies and is thought to impede the function of wild-type p53. Reactivation of p53 by disrupting the association with Hdm2 was the impetus for the development of Nutlin3. Although regulation of p53 has been the central focus of Hdm2 activity, it also binds other proteins through its p53-binding domain. Here, we show that hypoxia-inducible factor 1alpha (HIF1alpha) binds to Hdm2 in the domain designated to bind p53. HIF1alpha and p53 share a conserved motif that is required to bind Hdm2. Distinct complexes form between Hdm2-HIF1alpha and Hdm2-p53 as determined by immunoprecipitation of nuclear extracts and in vitro. The Hdm2 antagonist Nutlin3 prevents the association between Hdm2 and HIF1alpha. The vascular endothelial growth factor (VEGF) gene is a transcriptional target of HIF1alpha, and under normoxic or hypoxic conditions, Hdm2 increases HIF1alpha activity to induce VEGF production. Blocking the association of Hdm2 and HIF1alpha by Nutlin3, or ablating Hdm2 expression, diminished the level of VEGF under conditions of normoxia or hypoxia. Our findings establish a unique role for Nutlin3 in attenuating VEGF induction by preventing the association of Hdm2 with HIF1alpha.


Subject(s)
Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Imidazoles/pharmacology , Piperazines/pharmacology , Proto-Oncogene Proteins c-mdm2/metabolism , Vascular Endothelial Growth Factor A/biosynthesis , Amino Acid Sequence , Animals , Cell Line, Tumor , Conserved Sequence , Humans , Hypoxia-Inducible Factor 1, alpha Subunit/antagonists & inhibitors , Hypoxia-Inducible Factor 1, alpha Subunit/genetics , Protein Binding , Protein Structure, Tertiary , Proto-Oncogene Proteins c-mdm2/antagonists & inhibitors , Proto-Oncogene Proteins c-mdm2/genetics , RNA, Small Interfering/genetics , Rabbits , Recombinant Proteins/biosynthesis , Recombinant Proteins/genetics , Recombinant Proteins/pharmacology , Sequence Alignment , Transfection , Tumor Suppressor Protein p53/metabolism , Vascular Endothelial Growth Factor A/antagonists & inhibitors , Vascular Endothelial Growth Factor A/genetics
14.
J Cell Physiol ; 210(1): 1-6, 2007 Jan.
Article in English | MEDLINE | ID: mdl-17063460

ABSTRACT

Fibroblast growth factor (FGF)-21 is a novel regulator of insulin-independent glucose transport in 3T3-L1 adipocytes and has glucose and triglyceride lowering effects in rodent models of diabetes. The precise mechanisms whereby FGF-21 regulates metabolism remain to be determined. Here we describe the early signaling events triggered by FGF-21 treatment of 3T3-L1 adipocytes and reveal a functional interplay between FGF-21 and peroxisome proliferator-activated receptor gamma (PPARgamma) pathways that leads to a marked stimulation of glucose transport. While the early actions of FGF-21 on 3T3-L1 adipocytes involve rapid accumulation of intracellular calcium and phosphorylation of Akt, GSK-3, p70(S6K), SHP-2, MEK1/2, and Stat3, continuous treatment for 72 h induces an increase in PPARgamma protein expression. Moreover, chronic activation of the PPARgamma pathway in 3T3-L1 adipocytes with the PPARgamma agonist and anti-diabetic agent, rosiglitazone (BRL 49653), enhances FGF-21 action to induce tyrosine phosphorylation of FGF receptor-2. Strikingly, treatment of cells with FGF-21 and rosiglitazone in combination leads to a pronounced increase in expression of the GLUT1 glucose transporter and a marked synergy in stimulation of glucose transport. Together these results reveal a novel synergy between two regulators of glucose homeostasis, FGF-21 and PPARgamma, and further define FGF-21 mechanism of action.


Subject(s)
Adipocytes/drug effects , Fibroblast Growth Factors/pharmacology , Glucose/metabolism , Hypoglycemic Agents/pharmacology , PPAR gamma/drug effects , Receptor Cross-Talk , Signal Transduction/drug effects , 3T3-L1 Cells , Adipocytes/cytology , Adipocytes/metabolism , Animals , Calcium Signaling/drug effects , Cell Differentiation/drug effects , Dose-Response Relationship, Drug , Drug Synergism , Fibroblast Growth Factors/genetics , Glucose Transporter Type 1/metabolism , Intracellular Signaling Peptides and Proteins/metabolism , Mice , PPAR gamma/metabolism , Phosphorylation , Receptor, Fibroblast Growth Factor, Type 2/metabolism , Recombinant Proteins/pharmacology , Rosiglitazone , Thiazolidinediones/pharmacology , Time Factors , Up-Regulation/drug effects
SELECTION OF CITATIONS
SEARCH DETAIL
...