Network


Latest external collaboration on country level. Dive into details by clicking on the dots.

Hotspot


Dive into the research topics where Jin-Long Chen is active.

Publication


Featured researches published by Jin-Long Chen.


Endocrinology | 2008

Activation of G Protein-Coupled Receptor 43 in Adipocytes Leads to Inhibition of Lipolysis and Suppression of Plasma Free Fatty Acids

Hongfei Ge; Xiaofan Li; Jennifer Weiszmann; Ping Wang; Helene Baribault; Jin-Long Chen; Hui Tian; Yang Li

G protein-coupled receptor 43 (GPR43) has been identified as a receptor for short-chain fatty acids that include acetate and propionate. A potential involvement of GPR43 in immune and inflammatory response has been previously suggested because its expression is highly enriched in immune cells. GPR43 is also expressed in a number of other tissues including adipocytes; however, the functional consequences of GPR43 activation in these other tissues are not clear. In this report, we focus on the potential functions of GPR43 in adipocytes. We show that adipocytes treated with GPR43 natural ligands, acetate and propionate, exhibit a reduction in lipolytic activity. This inhibition of lipolysis is the result of GPR43 activation, because this effect is abolished in adipocytes isolated from GPR43 knockout animals. In a mouse in vivo model, we show that the activation of GPR43 by acetate results in the reduction in plasma free fatty acid levels without inducing the flushing side effect that has been observed by the activation of nicotinic acid receptor, GPR109A. These results suggest a potential role for GPR43 in regulating plasma lipid profiles and perhaps aspects of metabolic syndrome.


Proceedings of the National Academy of Sciences of the United States of America | 2001

Identification and characterization of a melanin-concentrating hormone receptor

Songzhu An; Gene Cutler; Jack Jiagang Zhao; Shu-Gui Huang; Hui Tian; Wanbo Li; Lingming Liang; Miki Rich; Amy Bakleh; Juan Du; Jin-Long Chen; Kang Dai

Melanin-concentrating hormone (MCH), a neuropeptide expressed in central and peripheral nervous systems, plays an important role in the control of feeding behaviors and energy metabolism. An orphan G protein-coupled receptor (SLC-1/GPR24) has recently been identified as a receptor for MCH (MCHR1). We report here the identification and characterization of a G protein-coupled receptor as the MCH receptor subtype 2 (MCHR2). MCHR2 has higher protein sequence homology to MCHR1 than any other G protein-coupled receptor. The expression of MCHR2 has been detected in many regions of the brain. In contrast to MCHR1, which is intronless in the coding region and is located at the chromosomal locus 22q13.3, the MCHR2 gene has multiple exons and is mapped to locus 6q21. MCHR2 is specifically activated by nanomolar concentrations of MCH, binds to MCH with high affinity, and signals through Gq protein. This discovery is important for a full understanding of MCH biology and the development of potential therapeutics for diseases involving MCH, including obesity.


Journal of Biological Chemistry | 2007

Co-receptor Requirements for Fibroblast Growth Factor-19 Signaling

Xinle Wu; Hongfei Ge; Jamila Gupte; Jennifer Weiszmann; Grant Shimamoto; Jennitte Stevens; Nessa Hawkins; Bryan Lemon; Wenyan Shen; Jing Xu; Murielle M. Véniant; Yue-Sheng Li; Richard Lindberg; Jin-Long Chen; Hui Tian; Yang Li

FGF19 is a unique member of the fibroblast growth factor (FGF) family of secreted proteins that regulates bile acid homeostasis and metabolic state in an endocrine fashion. Here we investigate the cell surface receptors required for signaling by FGF19. We show that βKlotho, a single-pass transmembrane protein highly expressed in liver and fat, induced ERK1/2 phosphorylation in response to FGF19 treatment and significantly increased the interactions between FGF19 and FGFR4. Interestingly, our results show that αKlotho, another Klotho family protein related to βKlotho, also induced ERK1/2 phosphorylation in response to FGF19 treatment and increased FGF19-FGFR4 interactions in vitro, similar to the effects of βKlotho. In addition, heparin further enhanced the effects of both αKlotho and βKlotho in FGF19 signaling and interaction experiments. These results suggest that a functional FGF19 receptor may consist of FGF receptor (FGFR) and heparan sulfate complexed with either αKlotho or βKlotho.


Journal of Biological Chemistry | 2008

C-terminal Tail of FGF19 Determines Its Specificity toward Klotho Co-receptors

Xinle Wu; Bryan Lemon; Xiaofan Li; Jamila Gupte; Jennifer Weiszmann; Jennitte Stevens; Nessa Hawkins; Wenyan Shen; Richard Lindberg; Jin-Long Chen; Hui Tian; Yang Li

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 α or βKlotho, two related single-pass transmembrane proteins, as co-receptors in relevant target tissues. We previously showed that FGF19 can bind to both α and βKlotho, whereas FGF21 and FGF23 can bind only to either βKlotho or αKlotho, 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 βKlotho and a chimera with the FGF23 C-terminal tail interacts only with αKlotho. 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 α and βKlotho 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 α or βKlotho for signaling, respectively, whereas FGF19 can signal through a Klotho chimera consisting of the N terminus of αKlotho and the C terminus of βKlotho. Our results provide the first glimpse of the regions that regulate the binding specificity between this unique family of FGFs and their co-receptors.


Journal of Molecular Biology | 2009

INT131: A Selective Modulator of PPARγ

Alykhan Motani; Zhulun Wang; Jennifer Weiszmann; Lawrence R. McGee; Gary Lee; Qingxiang Liu; Jocelyn Staunton; Zexu Fang; Helen Fuentes; Michelle Lindstrom; Jinsong Liu; Donna H.T. Biermann; Juan C. Jaen; Nigel Walker; R. Marc Learned; Jin-Long Chen; Yang Li

Summary The nuclear hormone receptor peroxisome proliferator-activated receptor γ (PPARγ; NR1C3) plays a central role in adipogenesis and is the molecular target of the thiazolidinedione class of antidiabetic drugs. To overcome the well-known shortcomings of thiazolidinediones, we have identified INT131 (formerly T131 and AMG131) as a potent selective ligand for PPARγ that is structurally and pharmacologically distinct from glitazone agonists. In vitro biochemical and cell-based functional assays showed that INT131 mediates a distinct pattern of coregulator recruitment to PPARγ. In adipocytes, INT131 showed minimal stimulation of adipocyte differentiation and partially activated PPARγ target genes involved in adipogenesis and, at the same time, showed more agonistic activity on another set of target genes that may influence insulin sensitivity directly. These unique properties of INT131 may provide a mechanistic basis for its distinct pharmacological profile. In vivo , increases in glucose tolerance were observed in Zucker ( fa/fa ) rats following a 14-day oral treatment with INT131. Although the maximal efficacies of INT131 and rosiglitazone were similar with respect to improvements in glucose tolerance, INT131 had less effect on heart and lung weights, weight gain, hemodilution, and plasma volume. Thus, INT131 appears to selectively modulate PPARγ responses in an in vivo preclinical model, showing antidiabetic efficacy while exhibiting an improved hemodynamic and cardiovascular adverse effect profile compared to the full agonist rosiglitazone. X-ray crystallography revealed that INT131 interacts with PPARγ through a distinct binding mode, forming primarily hydrophobic contacts with the ligand-binding pocket without direct hydrogen-bonding interactions to key residues in helix 12 that are characteristic of full agonists. Mutagenesis studies on Tyr473 in helix 12 demonstrated this residue as essential for rosiglitazone-induced receptor activation, but nonessential for INT131 function in vitro , providing one possible molecular determinant for INT131s distinct pharmacology. INT131 is currently being evaluated in a clinical setting as a therapeutic agent for the treatment of type 2 diabetes.


Journal of Biological Chemistry | 2008

T2384, a Novel Antidiabetic Agent with Unique Peroxisome Proliferator-activated Receptor γ Binding Properties

Yang Li; Zhulun Wang; Noboru Furukawa; Patrick Escaron; Jennifer Weiszmann; Gary Lee; Michelle Lindstrom; Jinsong Liu; Xiaohong Liu; Haoda Xu; Olga Plotnikova; Vidya Prasad; Nigel Walker; R. Marc Learned; Jin-Long Chen

The nuclear hormone receptor peroxisome proliferator-activated receptor γ (PPARγ) plays central roles in adipogenesis and glucose homeostasis and is the molecular target for the thiazolidinedione (TZD) class of antidiabetic drugs. Activation of PPARγ by TZDs improves insulin sensitivity; however, this is accompanied by the induction of several undesirable side effects. We have identified a novel synthetic PPARγ ligand, T2384, to explore the biological activities associated with occupying different regions of the receptor ligand-binding pocket. X-ray crystallography studies revealed that T2384 can adopt two distinct binding modes, which we have termed “U” and “S”, interacting with the ligand-binding pocket of PPARγ primarily via hydrophobic contacts that are distinct from full agonists. The different binding modes occupied by T2384 induced distinct patterns of coregulatory protein interaction with PPARγ in vitro and displayed unique receptor function in cell-based activity assays. We speculate that these unique biochemical and cellular activities may be responsible for the novel in vivo profile observed in animals treated systemically with T2384. When administered to diabetic KKAy mice, T2384 rapidly improved insulin sensitivity in the absence of weight gain, hemodilution, and anemia characteristics of treatment with rosiglitazone (a TZD). Moreover, upon coadministration with rosiglitazone, T2384 was able to antagonize the side effects induced by rosiglitazone treatment alone while retaining robust effects on glucose disposal. These results are consistent with the hypothesis that interactions between ligands and specific regions of the receptor ligand-binding pocket might selectively trigger a subset of receptor-mediated biological responses leading to the improvement of insulin sensitivity, without eliciting less desirable responses associated with full activation of the receptor. We suggest that T2384 may represent a prototype for a novel class of PPARγ ligand and, furthermore, that molecules sharing some of these properties would be useful for treatment of type 2 diabetes.


Bioorganic & Medicinal Chemistry | 2013

Discovery of INT131: A selective PPARγ modulator that enhances insulin sensitivity

Joshua P. Taygerly; Lawrence R. McGee; Steven M. Rubenstein; Jonathan B. Houze; Timothy D. Cushing; Yang Li; Alykhan Motani; Jin-Long Chen; Walter Frankmoelle; Guosen Ye; Marc Learned; Juan C. Jaen; Shichang Miao; Pieter B. M. W. M. Timmermans; Martin J. Thoolen; Patrick C. Kearney; John A. Flygare; Holger Beckmann; Jennifer Weiszmann; Michelle Lindstrom; Nigel Walker; Jinsong Liu; Donna H.T. Biermann; Zhulun Wang; Atsushi Hagiwara; Tetsuya Iida; Hisateru Aramaki; Yuki Kitao; Hisashi Shinkai; Noboru Furukawa

PPARγ is a member of the nuclear hormone receptor family and plays a key role in the regulation of glucose homeostasis. This Letter describes the discovery of a novel chemical class of diarylsulfonamide partial agonists that act as selective PPARγ modulators (SPPARγMs) and display a unique pharmacological profile compared to the thiazolidinedione (TZD) class of PPARγ full agonists. Herein we report the initial discovery of partial agonist 4 and the structure-activity relationship studies that led to the selection of clinical compound INT131 (3), a potent PPARγ partial agonist that displays robust glucose-lowering activity in rodent models of diabetes while exhibiting a reduced side-effects profile compared to marketed TZDs.


Bioorganic & Medicinal Chemistry Letters | 2012

Discovery of a novel melanin concentrating hormone receptor 1 (MCHR1) antagonist with reduced hERG inhibition

Jeffrey T. Mihalic; Pingchen Fan; Xiaoqi Chen; Xi Chen; Ying Fu; Alykhan Motani; Lingming Liang; Michelle Lindstrom; Liang Tang; Jin-Long Chen; Juan C. Jaen; Kang Dai; Leping Li

An initial SAR study resulted in the identification of the novel, potent MCHR1 antagonist 2. After further profiling, compound 2 was discovered to be a potent inhibitor of the hERG potassium channel, which prevented its further development. Additional optimization of this structure resulted in the discovery of the potent MCHR1 antagonist 11 with a dramatically reduced hERG liability. The decrease in hERG activity was confirmed by several in vivo preclinical cardiovascular studies examining QT prolongation. This compound demonstrated good selectivity for MCHR1 and possessed good pharmacokinetic properties across preclinical species. Compound 11 was also efficacious in reducing body weight in two in vivo mouse models. This compound was selected for clinical evaluation and was given the code AMG 076.


Bioorganic & Medicinal Chemistry Letters | 2012

Discovery and characterization of a potent and selective antagonist of melanin-concentrating hormone receptor 2

Xiaoqi Chen; Jeff Mihalic; Pingchen Fan; Lingming Liang; Michelle Lindstrom; Sylvia Wong; Qiuping Ye; Ying Fu; Juan C. Jaen; Jin-Long Chen; Kang Dai; Leping Li

A series of spiropiperidine carbazoles were synthesized and evaluated as MCHR2 antagonists using a FLIPR assay. The pharmacokinetic properties of selected compounds have also been studied. This effort led to the discovery of potent and specific MCHR2 antagonists. Compound 38 demonstrated good pharmacokinetic properties across rat, beagle dog and rhesus monkey and had a favorable selectivity profile against a number of other receptors. These MCHR2 antagonists are considered appropriate tool compounds for study of the function of MCHR2 in vivo.


Bioorganic & Medicinal Chemistry Letters | 2011

Discovery of a novel series of melanin-concentrating hormone receptor 1 antagonists for the treatment of obesity

Jeffrey T. Mihalic; Xiaoqi Chen; Pingchen Fan; Xi Chen; Ying Fu; Lingming Liang; Michael Reed; Liang Tang; Jin-Long Chen; Juan C. Jaen; Leping Li; Kang Dai

A new class of MCHR1 antagonists was discovered via a high-throughput screen. Optimization of the lead structure resulted in the identification of indole 10e. This compound possesses good pharmacokinetic properties across preclinical species and is efficacious in reducing food consumption in an MCH cannulated rat model and a cynomolgus monkey food consumption model.

Researchain Logo
Decentralizing Knowledge