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










Publication year range
1.
Antiviral Res ; 221: 105791, 2024 01.
Article in English | MEDLINE | ID: mdl-38160942

ABSTRACT

Human respiratory syncytial virus (RSV) is a leading cause of lower respiratory tract infections (LRTI) in young children and elderly people worldwide. Recent significant progress in our understanding of the structure and function of RSV proteins has led to the discovery of several clinical candidates targeting RSV fusion and replication. These include both the development of novel small molecule interventions and the isolation of potent monoclonal antibodies. In this review, we summarize the state-of-the-art of RSV drug discovery, with a focus on the characteristics of the candidates that reached the clinical stage of development. We also discuss the lessons learned from failed and discontinued clinical developments and highlight the challenges that remain for development of RSV therapies.


Subject(s)
Respiratory Syncytial Virus Infections , Respiratory Syncytial Virus Vaccines , Respiratory Syncytial Virus, Human , Respiratory Tract Infections , Child , Humans , Aged , Child, Preschool , Antibodies, Monoclonal/therapeutic use , Respiratory Syncytial Virus Vaccines/therapeutic use , Viral Fusion Proteins , Antibodies, Viral , Antibodies, Neutralizing
2.
J Pharm Biomed Anal ; 186: 113328, 2020 Jul 15.
Article in English | MEDLINE | ID: mdl-32388224

ABSTRACT

A rabbit blood sugar bioidentity assay is required by the FDA to evaluate biological activity for all insulin and its analogs per USP<121> guideline. Not only are a large number of live animals used, but the rabbit blood sugar method is also highly variable and expensive. Our goal is to develop a functional cell-based assay to replace rabbit blood sugar method. An H4IIE G6P-Luc reporter assay was developed by utilizing insulin's role in regulating hepatic gluconeogenesis pathway. It is known that Glucose 6-phosphatase is a rate-limiting enzyme in the gluconeogenesis pathway, and the mRNA expression of its catalytic subunit, G6PC, is highly regulated by insulin. A G6P-Luc stable cell line in H4IIE hepatocytes was first generated by stably expressing luciferase reporter gene driven by human G6PC promoter via lentivirus technology. The cell-based assay was developed and optimized to demonstrate good dose-dependent responsiveness to insulin. We further qualified the assay with two analysts through multiple runs, and demonstrated excellent performance characteristics of linearity, accuracy, and precision. A robustness study was then conducted to define critical factors for assay performance. We compared this newly developed assay with a previously established cell-based pIR MSD assay, which measures insulin receptor phosphorylation (pIR) in HepG2 cell line using Meso-Scale Discovery (MSD) technology. The comparability study was conducted to compare the two assays using samples generated from forced degradation. This study showed high correlation between assays, and both are stability indicating. Compared with the pIR MSD assay, the G6P-Luc assay not only has a significantly lower variability in qualification studies, but also offers many other advantages, including ease of use in a quality control laboratory with fewer steps, lower cost, and does not depend on a single vendor. In conclusion, we have developed a physiologically relevant and robust functional cell-based assay that is suitable to replace rabbit blood sugar method.


Subject(s)
Animal Testing Alternatives , Blood Glucose/drug effects , Hypoglycemic Agents/pharmacology , Insulin Glargine/pharmacology , Animals , Blood Glucose/analysis , Cell Line , Dose-Response Relationship, Drug , Gluconeogenesis/physiology , Glucose-6-Phosphatase/genetics , Glucose-6-Phosphatase/metabolism , Hep G2 Cells , Hepatocytes/metabolism , Humans , Hypoglycemic Agents/administration & dosage , Insulin Glargine/administration & dosage , Luciferases/genetics , Promoter Regions, Genetic , Rabbits , Rats
3.
MAbs ; 12(1): 1710047, 2020.
Article in English | MEDLINE | ID: mdl-31905038

ABSTRACT

Glucose-dependent insulinotropic polypeptide (GIP) is an incretin hormone involved in regulating glucose and lipid metabolism. GIP receptor (GIPR) antagonism is believed to offer therapeutic potential for various metabolic diseases. Pharmacological intervention of GIPR, however, has limited success due to lack of effective antagonistic reagents. Previously we reported the discovery of two mouse anti-murine GIPR monoclonal antibodies (mAbs) with distinctive properties in rodent models. Here, we report the detailed structural and biochemical characterization of these two antibodies, mAb1 and mAb2. In vitro and in vivo characterizations demonstrated mAb2 is a full GIPR antagonistic antibody and mAb1 is a non-neutralizing GIPR binder. To understand the molecular basis of these two antibodies, we determined the co-crystal structures of GIPR extracellular domain in complex with mAb1 and with mAb2 at resolutions of 2.1 and 2.6 Å, respectively. While the non-neutralizing mAb1 binds to GIPR without competing with the ligand peptide, mAb2 not only partially occludes the ligand peptide binding, but also recognizes the GIPR C-terminal stalk region in a helical conformation that acts as a molecular mimic of the ligand peptide and locks GIPR in a novel auto-inhibited state. Furthermore, administration of mAb2 in diet-induced obesity mice for 7 weeks leads to both reduction in body weight gain and improvement of metabolic profiles. In contrast, mAb1 has no effect on body weight or other metabolic improvement. Together, our studies reveal the unique molecular mechanism of action underlying the superior antagonistic activity of mAb2 and signify the promising therapeutic potential of effective GIPR antagonism for the treatment of metabolic disorders.


Subject(s)
Antibodies, Monoclonal/chemistry , Antibodies, Monoclonal/pharmacology , Receptors, Gastrointestinal Hormone/antagonists & inhibitors , Weight Gain/drug effects , Animals , Diet, High-Fat/adverse effects , Male , Mice , Mice, Inbred C57BL , Obesity/etiology , Obesity/metabolism , Protein Conformation
4.
Sci Transl Med ; 10(472)2018 12 19.
Article in English | MEDLINE | ID: mdl-30567927

ABSTRACT

Glucose-dependent insulinotropic polypeptide (GIP) receptor (GIPR) has been identified in multiple genome-wide association studies (GWAS) as a contributor to obesity, and GIPR knockout mice are protected against diet-induced obesity (DIO). On the basis of this genetic evidence, we developed anti-GIPR antagonistic antibodies as a potential therapeutic strategy for the treatment of obesity and observed that a mouse anti-murine GIPR antibody (muGIPR-Ab) protected against body weight gain, improved multiple metabolic parameters, and was associated with reduced food intake and resting respiratory exchange ratio (RER) in DIO mice. We replicated these results in obese nonhuman primates (NHPs) using an anti-human GIPR antibody (hGIPR-Ab) and found that weight loss was more pronounced than in mice. In addition, we observed enhanced weight loss in DIO mice and NHPs when anti-GIPR antibodies were codosed with glucagon-like peptide-1 receptor (GLP-1R) agonists. Mechanistic and crystallographic studies demonstrated that hGIPR-Ab displaced GIP and bound to GIPR using the same conserved hydrophobic residues as GIP. Further, using a conditional knockout mouse model, we excluded the role of GIPR in pancreatic ß-cells in the regulation of body weight and response to GIPR antagonism. In conclusion, these data provide preclinical validation of a therapeutic approach to treat obesity with anti-GIPR antibodies.


Subject(s)
Glucagon-Like Peptide-1 Receptor/agonists , Obesity/drug therapy , Receptors, Gastrointestinal Hormone/antagonists & inhibitors , Adipocytes/metabolism , Animals , Antibodies/pharmacology , Antibodies/therapeutic use , Diet , Drug Therapy, Combination , Feeding Behavior , Gastric Inhibitory Polypeptide/metabolism , Glucagon-Like Peptide-1 Receptor/metabolism , Glucagon-Like Peptides/analogs & derivatives , Glucagon-Like Peptides/pharmacology , Glucagon-Like Peptides/therapeutic use , Humans , Immunoglobulin Fc Fragments/pharmacology , Immunoglobulin Fc Fragments/therapeutic use , Insulin-Secreting Cells/drug effects , Insulin-Secreting Cells/metabolism , Liraglutide/pharmacology , Liraglutide/therapeutic use , Mice, Obese , Obesity/pathology , Primates , Receptors, Gastrointestinal Hormone/metabolism , Recombinant Fusion Proteins/pharmacology , Recombinant Fusion Proteins/therapeutic use , Respiration , Weight Gain/drug effects , Weight Loss/drug effects
6.
Nature ; 550(7675): 255-259, 2017 10 12.
Article in English | MEDLINE | ID: mdl-28953886

ABSTRACT

Under homeostatic conditions, animals use well-defined hypothalamic neural circuits to help maintain stable body weight, by integrating metabolic and hormonal signals from the periphery to balance food consumption and energy expenditure. In stressed or disease conditions, however, animals use alternative neuronal pathways to adapt to the metabolic challenges of altered energy demand. Recent studies have identified brain areas outside the hypothalamus that are activated under these 'non-homeostatic' conditions, but the molecular nature of the peripheral signals and brain-localized receptors that activate these circuits remains elusive. Here we identify glial cell-derived neurotrophic factor (GDNF) receptor alpha-like (GFRAL) as a brainstem-restricted receptor for growth and differentiation factor 15 (GDF15). GDF15 regulates food intake, energy expenditure and body weight in response to metabolic and toxin-induced stresses; we show that Gfral knockout mice are hyperphagic under stressed conditions and are resistant to chemotherapy-induced anorexia and body weight loss. GDF15 activates GFRAL-expressing neurons localized exclusively in the area postrema and nucleus tractus solitarius of the mouse brainstem. It then triggers the activation of neurons localized within the parabrachial nucleus and central amygdala, which constitute part of the 'emergency circuit' that shapes feeding responses to stressful conditions. GDF15 levels increase in response to tissue stress and injury, and elevated levels are associated with body weight loss in numerous chronic human diseases. By isolating GFRAL as the receptor for GDF15-induced anorexia and weight loss, we identify a mechanistic basis for the non-homeostatic regulation of neural circuitry by a peripheral signal associated with tissue damage and stress. These findings provide opportunities to develop therapeutic agents for the treatment of disorders with altered energy demand.


Subject(s)
Body Weight/physiology , Brain Stem/metabolism , Glial Cell Line-Derived Neurotrophic Factor Receptors/metabolism , Growth Differentiation Factor 15/metabolism , Animals , Brain Stem/cytology , Brain Stem/drug effects , Central Amygdaloid Nucleus/cytology , Central Amygdaloid Nucleus/physiology , Eating/physiology , Energy Metabolism/physiology , Feeding Behavior , Female , Glial Cell Line-Derived Neurotrophic Factor Receptors/deficiency , Glial Cell Line-Derived Neurotrophic Factor Receptors/genetics , Growth Differentiation Factor 15/genetics , Growth Differentiation Factor 15/pharmacology , Homeostasis , Male , Mice , Mice, Knockout , Neurons/drug effects , Neurons/metabolism , Parabrachial Nucleus/cytology , Parabrachial Nucleus/physiology , Stress, Psychological
7.
Endocrinology ; 158(5): 1314-1327, 2017 05 01.
Article in English | MEDLINE | ID: mdl-28324011

ABSTRACT

Fibroblast growth factor (FGF) 21 is a natural hormone that modulates glucose, lipid, and energy metabolism. Previously, we engineered an Fc fusion FGF21 variant with two mutations, Fc-FGF21(RG), to extend the half-life and reduce aggregation and in vivo degradation of FGF21. We now describe a new variant developed to reduce the extreme C-terminal degradation and improve the binding affinity to ß-Klotho. We demonstrate, by introducing one additional mutation located at the C terminus of FGF21 (A180E), that the new molecule, Fc-FGF21(RGE), has gained many improved attributes. Compared with Fc-FGF21(RG), Fc-FGF21(RGE) has similar in vitro potency, preserves ß-Klotho dependency, and maintains FGF receptor selectivity and cross-species reactivity. In vivo, Fc-FGF21(RGE) showed reduced susceptibility to extreme C-terminal degradation and increased plasma levels of the bioactive intact molecule. The circulating half-life of intact Fc-FGF21(RGE) increased twofold compared with that of Fc-FGF21(RG) in mice and cynomolgus monkeys. Additionally, Fc-FGF21(RGE) exhibited threefold to fivefold enhanced binding affinity to coreceptor ß-Klotho across mouse, cynomolgus monkey, and human species. In obese and diabetic mouse and cynomolgus monkey models, Fc-FGF21(RGE) demonstrated greater efficacies to Fc-FGF21(RG), resulting in larger and more sustained improvements in multiple metabolic parameters. No increased immunogenicity was observed with Fc-FGF21(RGE). The superior biophysical, pharmacokinetic, and pharmacodynamic properties, as well as the positive metabolic effects across species, suggest that further clinical development of Fc-FGF21(RGE) as a metabolic therapy for diabetic and/or obese patients may be warranted.


Subject(s)
Anti-Obesity Agents/therapeutic use , Diabetes Mellitus, Experimental/drug therapy , Fibroblast Growth Factors/therapeutic use , Immunoglobulin Fc Fragments/therapeutic use , Membrane Proteins/metabolism , Obesity/drug therapy , 3T3-L1 Cells , Animals , Anti-Obesity Agents/chemical synthesis , Anti-Obesity Agents/metabolism , Diabetes Mellitus, Experimental/metabolism , Disease Models, Animal , Drug Stability , Fibroblast Growth Factors/genetics , Fibroblast Growth Factors/metabolism , HEK293 Cells , Half-Life , Humans , Immunoglobulin Fc Fragments/genetics , Immunoglobulin Fc Fragments/metabolism , Klotho Proteins , Macaca fascicularis , Male , Mice , Mice, Inbred C57BL , Mutation , Obesity/metabolism , Protein Binding , Protein Engineering/methods , Proteolysis , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , Recombinant Fusion Proteins/therapeutic use , Treatment Outcome
8.
Bioconjug Chem ; 24(6): 915-25, 2013 Jun 19.
Article in English | MEDLINE | ID: mdl-23594041

ABSTRACT

Fibroblast growth factor 21 (FGF21) is involved in regulating energy metabolism, and it has shown significant promise as a treatment for type II diabetes; however, the native protein has a very short circulating half-life necessitating frequent injections to maintain a physiological effect. Polyethylene glycol (PEG) conjugation to proteins has been used as a method for extending the circulating half-life of many pharmaceutical proteins; however, PEG does carry the risk of vacuole formation, particularly in the renal tubular epithelium. Since renal vacuole formation may be particularly problematic for diabetic patients, we engineered site-directed PEGylated variants of FGF21 with sustained potency and minimized vacuole formation. This was accomplished both by probing the site of PEGylation on FGF21 as well as by examining various PEG configurations. While the site of PEGylation has a significant impact on the bioactivity of FGF21, it has only a marginal impact on vacuole formation; however, the configuration and number of PEGs conjugated to the protein has a much more profound effect on vacuologenesis.


Subject(s)
Fibroblast Growth Factors/chemistry , Polyethylene Glycols/chemistry , Protein Engineering , Vacuoles/metabolism , Animals , Fibroblast Growth Factors/metabolism , HEK293 Cells , Humans , Male , Mice , Mice, Obese , Models, Molecular , Polyethylene Glycols/metabolism , Vacuoles/genetics
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.
PLoS One ; 7(11): e49345, 2012.
Article in English | MEDLINE | ID: mdl-23209571

ABSTRACT

Fibroblast growth factor 21 (FGF21) is a promising drug candidate for the treatment of type 2 diabetes. However, the use of wild type native FGF21 is challenging due to several limitations. Among these are its short half-life, its susceptibility to in vivo proteolytic degradation and its propensity to in vitro aggregation. We here describe a rationale-based protein engineering approach to generate a potent long-acting FGF21 analog with improved resistance to proteolysis and aggregation. A recombinant Fc-FGF21 fusion protein was constructed by fusing the Fc domain of human IgG1 to the N-terminus of human mature FGF21 via a linker peptide. The Fc positioned at the N-terminus was determined to be superior to the C-terminus as the N-terminal Fc fusion retained the ßKlotho binding affinity and the in vitro and in vivo potency similar to native FGF21. Two specific point mutations were introduced into FGF21. The leucine to arginine substitution at position 98 (L98R) suppressed FGF21 aggregation at high concentrations and elevated temperatures. The proline to glycine replacement at position 171 (P171G) eliminated a site-specific proteolytic cleavage of FGF21 identified in mice and cynomolgus monkeys. The derived Fc-FGF21(RG) molecule demonstrated a significantly improved circulating half-life while maintaining the in vitro activity similar to that of wild type protein. The half-life of Fc-FGF21(RG) was 11 h in mice and 30 h in monkeys as compared to 1-2 h for native FGF21 or Fc-FGF21 wild type. A single administration of Fc-FGF21(RG) in diabetic mice resulted in a sustained reduction in blood glucose levels and body weight gains up to 5-7 days, whereas the efficacy of FGF21 or Fc-FGF21 lasted only for 1 day. In summary, we engineered a potent and efficacious long-acting FGF21 analog with a favorable pharmaceutical property for potential clinical development.


Subject(s)
Diabetes Mellitus, Experimental/metabolism , Diabetes Mellitus, Type 2/metabolism , Fibroblast Growth Factors/pharmacology , Hypoglycemic Agents/pharmacology , Protein Engineering , Recombinant Fusion Proteins/pharmacology , Animals , Diabetes Mellitus, Experimental/drug therapy , Diabetes Mellitus, Type 2/drug therapy , Disease Models, Animal , Fibroblast Growth Factors/genetics , Fibroblast Growth Factors/metabolism , Humans , Macaca fascicularis , Male , Mice , Mutation , Proteolysis , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism
11.
Chem Biol Drug Des ; 79(4): 398-410, 2012 Apr.
Article in English | MEDLINE | ID: mdl-22248288

ABSTRACT

The endocrine fibroblast growth factor 21 (FGF21) requires both fibroblast growth factor receptor (FGFR) and ß-Klotho for signaling. In this study, we sought to understand the inter-molecular physical interactions in the FGF21/FGFR/ß-Klotho complex by deleting key regions in FGFR1c or FGF21. Deletion of the D1 and the D1-D2 linker (the D1/linker region) from FGFR1c led to ß-Klotho-independent receptor activation by FGF21, suggesting that there may be a direct interaction between FGF21 and the D1/linker region-deficient FGFR1c. Consistent with this, the extracellular portion of FGFR1c lacking the D1/linker region blocked FGF21 action in a reporter assay, presumably by binding to and sequestering FGF21 from acting on cell surface receptor complex. In addition, the D1/linker region-deficient FGFR1c had enhanced interaction with ß-Klotho. Further, we demonstrated that deletion of the D1/linker region enhanced the formation of the FGF21/ß-Klotho/FGFR1c ternary complex in both Biacore and asymmetrical flow field flow fractionation studies. Finally, we found that the N-terminus of FGF21 is involved in the interaction with FGFR1c and FGF21/ß-Klotho/FGFR1c ternary complex formation. Taken together, our data suggest that the D1/linker region regulates both the FGF21/FGFR1c and FGFR1c/ß-Klotho interaction, and a direct interaction of FGF21 with FGFR1c may be an important step in receptor-mediated FGF21 signaling.


Subject(s)
Fibroblast Growth Factors/metabolism , Membrane Proteins/metabolism , Receptor, Fibroblast Growth Factor, Type 1/metabolism , Cell Line , Fibroblast Growth Factors/chemistry , Humans , Klotho Proteins , Membrane Proteins/chemistry , Protein Interaction Domains and Motifs , Receptor, Fibroblast Growth Factor, Type 1/chemistry , Signal Transduction
12.
FEBS Lett ; 583(1): 19-24, 2009 Jan 05.
Article in English | MEDLINE | ID: mdl-19059246

ABSTRACT

Fibroblast growth factor-21 (FGF21) signaling requires the presence of beta-Klotho, a co-receptor with a very short cytoplasmic domain. Here we show that FGF21 binds directly to beta-Klotho through its C-terminus. Serial C-terminal truncations of FGF21 weakened or even abrogated its interaction with beta-Klotho in a Biacore assay, and led to gradual loss of potency in a luciferase reporter assay but with little effect on maximal response. In contrast, serial N-terminal truncations of FGF21 had no impact on beta-Klotho binding. Interestingly, several of them exhibited characteristics of partial agonists with minimal effects on potency. These data demonstrate that the C-terminus of FGF21 is critical for binding to beta-Klotho and the N-terminus is critical for fibroblast growth factor receptor (FGFR) activation.


Subject(s)
Fibroblast Growth Factors/metabolism , Membrane Proteins/metabolism , Receptor, Fibroblast Growth Factor, Type 1/metabolism , Amino Acid Sequence , Cell Line , Fibroblast Growth Factors/chemistry , Fibroblast Growth Factors/genetics , Genes, Reporter , Humans , Klotho Proteins , Luciferases/genetics , Membrane Proteins/genetics , Molecular Sequence Data , Protein Structure, Tertiary
13.
Am J Physiol Endocrinol Metab ; 295(5): E1142-51, 2008 Nov.
Article in English | MEDLINE | ID: mdl-18728225

ABSTRACT

c-Jun NH(2)-terminal kinase (JNK) plays an important role in insulin resistance; however, identification of pharmacologically potent and selective small molecule JNK inhibitors has been limited. Compound A has a cell IC(50) of 102 nM and is at least 100-fold selective against related kinases and 27-fold selective against glycogen synthase kinase-3beta and cyclin-dependent kinase-2. In C57BL/6 mice, compound A reduced LPS-mediated increases in both plasma cytokine levels and phosphorylated c-Jun in adipose tissue. Treatment of mice fed a high-fat diet with compound A for 3 wk resulted in a 13.1 +/- 1% decrease in body weight and a 9.3 +/- 1.5% decrease in body fat, compared with a 6.6 +/- 2.1% increase in body weight and a 6.7 +/- 2.1% increase in body fat in vehicle-treated mice. Mice pair fed to those that received compound A exhibited a body weight decrease of 7 +/- 1% and a decrease in body fat of 1.6 +/- 1.3%, suggesting that reductions in food intake could not account solely for the reductions in adiposity observed. Compound A dosed at 30 mg/kg for 13 days in high-fat fed mice resulted in a significant decrease in phosphorylated c-Jun in adipose tissue accompanied by a decrease in weight and reductions in glucose and triglycerides and increases in insulin sensitivity to levels comparable with those in lean control mice. The ability of compound A to reduce the insulin-stimulated phosphorylation of insulin receptor substrate-1 (IRS-1) von Ser307 and partially reverse the free fatty acid inhibition of glucose uptake in 3T3L1 adipocytes, suggests that enhancement of insulin signaling in addition to weight loss may contribute to the effects of compound A on insulin sensitization in vivo. Pharmacological inhibition of JNK using compound A may therefore offer an effective therapy for type 2 diabetes mediated at least in part via weight reduction.


Subject(s)
Aminopyridines/pharmacology , JNK Mitogen-Activated Protein Kinases/antagonists & inhibitors , Protein Kinase Inhibitors/pharmacology , 3T3-L1 Cells , Adipose Tissue, White/drug effects , Adipose Tissue, White/metabolism , Aminopyridines/pharmacokinetics , Animals , Blood Glucose/metabolism , Body Weight/drug effects , Cytokines/blood , Dietary Fats/administration & dosage , Dietary Fats/pharmacology , Eating/drug effects , Humans , Insulin/blood , Insulin/pharmacology , Insulin Receptor Substrate Proteins/metabolism , Insulin Resistance , JNK Mitogen-Activated Protein Kinases/metabolism , Lipopolysaccharides/pharmacology , Male , Mice , Mice, Inbred C57BL , Mitogen-Activated Protein Kinase 8/antagonists & inhibitors , Mitogen-Activated Protein Kinase 8/metabolism , Obesity/drug therapy , Obesity/etiology , Obesity/pathology , Phosphorylation/drug effects , Protein Kinase Inhibitors/pharmacokinetics , Proto-Oncogene Proteins c-jun/metabolism , Tumor Necrosis Factor-alpha/pharmacology , U937 Cells
14.
J Biol Chem ; 277(44): 41925-30, 2002 Nov 01.
Article in English | MEDLINE | ID: mdl-12200443

ABSTRACT

Peroxisome proliferator-activated receptor (PPAR) gamma is a nuclear receptor that is a key regulator of adipogenesis and is present in two isoforms generated by alternative splicing, PPARgamma1 and PPARgamma2. Studies of the ability of each isoform to stimulate fat differentiation have yielded ambiguous results, in part because PPARgamma stimulates its own expression. We have thus undertaken a formal genetic analysis using PPARgamma-null fibroblast cell lines to assess the specific role of each individual isoform in adipogenesis. We show here that both PPARgamma1 and PPARgamma2 have the intrinsic ability to stimulate robust adipogenesis. Adipose cells stimulated by either PPARgamma1 or PPARgamma2 express a similar gene profile and show similar responses to insulin. However, in response to low ligand concentrations, PPARgamma2 shows a quantitatively greater ability to induce adipogenesis. Analyses involving coactivator binding and transcriptional assays indicate that PPARgamma2 has an enhanced ability to bind components of the DRIP/TRAP complex, coactivators required for fat differentiation.


Subject(s)
Adipocytes/physiology , Receptors, Cytoplasmic and Nuclear/physiology , Transcription Factors/physiology , Animals , Carrier Proteins/metabolism , Cell Differentiation , Cells, Cultured , Mediator Complex Subunit 1 , Mice , Protein Isoforms , Receptors, Cytoplasmic and Nuclear/genetics , Receptors, Thyroid Hormone/metabolism , Transcription Factors/genetics , Transcription Factors/metabolism , Transcription, Genetic
SELECTION OF CITATIONS
SEARCH DETAIL
...