Network


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

Hotspot


Dive into the research topics where Sean Chen is active.

Publication


Featured researches published by Sean Chen.


The Journal of Steroid Biochemistry and Molecular Biology | 2002

Ligand and coactivator recruitment preferences of peroxisome proliferator activated receptor α

Ranjan Mukherjee; Shaoxian Sun; Linda Santomenna; Bowman Miao; Harry Walton; Boshan Liao; Kenneth T. Locke; Ji-Hu Zhang; Sonny H. Nguyen; Li Tao Zhang; Kathleen Murphy; Harold O. Ross; M.X. Xia; Christopher A. Teleha; Sean Chen; Bernard Selling; Richard Wynn; Timothy C. Burn; Peter R. Young

The mechanism by which ligands of nuclear receptors show differential effects on gene transcription is not fully understood, but is believed to result in part from the preferential recruitment and/or displacement of coactivators and corepressors. We have explored the interaction of several known ligands and the nuclear receptor (peroxisome proliferator activated receptor alpha, PPARalpha) using scintillation proximity assay (SPA) and the interaction of LXXLL containing peptides derived from three coactivators (SRC-1, CBP and PGC-1) with PPARalpha in the presence of PPARalpha agonist ligands using fluorescence resonance energy transfer (FRET). The EC(50)s of the individual ligands for recruitment showed the same rank order regardless of the coactivator peptide used, with GW2331<WY14643=ciprofibrate<L165041<gemfibrozil. Similarly, for all ligands tested, the rank order of EC(50) for peptide recruitment was CBP<PGC-1<SRC-1. These data suggest that for these LXXLL coactivator peptides, the ligands do not substantially differ in their preferences. Partial agonism was observed with ciprofibrate and PGC-1 and gemfibrozil and CBP giving a lower FRET at saturation than with the other ligands. This suggests that ciprofibrate and gemfibrozil induce a different conformation to the receptor-PGC-1 and receptor-CBP complex, respectively. In cotransfection assays, unexpected differences in potencies and efficacies were observed and the rank order of EC(50)s for activation differed from that predicted by FRET assays. In most cases, the presence of a coactivator peptide led to decrease in the EC(50)s seen in FRET assays compared to the K(i)s observed in binding to receptor only, consistent with the lower EC(50)s obtained in the transfection assays. Our data demonstrate that ligand induced coactivator preferences of PPARalpha contribute to transcription potency and efficacy.


Journal of Medicinal Chemistry | 2010

Discovery of an oxybenzylglycine based peroxisome proliferator activated receptor alpha selective agonist 2-((3-((2-(4-chlorophenyl)-5-methyloxazol-4-yl)methoxy)benzyl)(methoxycarbonyl)amino)acetic acid (BMS-687453).

Jun Li; Lawrence J. Kennedy; Yan Shi; Shiwei Tao; Xiang-Yang Ye; Stephanie Y. Chen; Ying Wang; Andres S. Hernandez; Wei Wang; Pratik Devasthale; Sean Chen; Zhi Lai; Hao Zhang; Shung Wu; Rebecca A. Smirk; Scott A. Bolton; Denis E. Ryono; Huiping Zhang; Ngiap-Kie Lim; Bang-Chi Chen; Kenneth T. Locke; Kevin O’Malley; Litao Zhang; Rai Ajit Srivastava; Bowman Miao; Daniel Meyers; Hossain Monshizadegan; Debra Search; Denise Grimm; Rongan Zhang

An 1,3-oxybenzylglycine based compound 2 (BMS-687453) was discovered to be a potent and selective peroxisome proliferator activated receptor (PPAR) alpha agonist, with an EC(50) of 10 nM for human PPARalpha and approximately 410-fold selectivity vs human PPARgamma in PPAR-GAL4 transactivation assays. Similar potencies and selectivity were also observed in the full length receptor co-transfection assays. Compound 2 has negligible cross-reactivity against a panel of human nuclear hormone receptors including PPARdelta. Compound 2 demonstrated an excellent pharmacological and safety profile in preclinical studies and thus was chosen as a development candidate for the treatment of atherosclerosis and dyslipidemia. The X-ray cocrystal structures of the early lead compound 12 and compound 2 in complex with PPARalpha ligand binding domain (LBD) were determined. The role of the crystal structure of compound 12 with PPARalpha in the development of the SAR that ultimately resulted in the discovery of compound 2 is discussed.


Journal of Pharmacology and Experimental Therapeutics | 2007

The Dual Peroxisome Proliferator-Activated Receptor α/γ Activator Muraglitazar Prevents the Natural Progression of Diabetes in db/db Mice

Effie Tozzo; Randolph Ponticiello; JoAnn Swartz; Dennis Farrelly; Rachel Zebo; Gustav Welzel; Donald M. Egan; Lori Kunselman; Andrew Peters; Liqun Gu; Michele H. French; Sean Chen; Pratik Devasthale; Evan B. Janovitz; Ada Staal; Thomas Harrity; Rene Belder; Peter T. W. Cheng; Jean M. Whaley; Simeon Taylor; Narayanan Hariharan

There are two major defects in type 2 diabetes: 1) insulin resistance and 2) insulin deficiency due to loss of β-cell function. Here we demonstrated that treatment with muraglitazar (a dual peroxisome proliferator-activated receptor α/γ activator), when initiated before or after the onset of diabetes in mice, is effective against both defects. In study 1, prediabetic db/db mice were treated for 12 weeks. The control mice developed diabetes, as evidenced by hyperglycemia, hyperinsulinemia, reduced insulin levels in the pancreas, blunted insulin response to glucose, and impaired glucose tolerance. The muraglitazar-treated mice had normal plasma glucose, and insulin levels, equivalent or higher pancreatic insulin content than normal mice, showed a robust insulin response to glucose and exhibited greater glucose tolerance. In study 2, diabetic db/db mice were treated for 4 weeks. The control mice displayed increased glucose levels, severe loss of islets, and their isolated islets secreted reduced amounts of insulin in response to glucose and exendin-4 compared with baseline. In muraglitazar-treated mice, glucose levels were reduced to normal. These mice showed reduced loss of islets, and their isolated islets secreted insulin at levels comparable to baseline. Thus, muraglitazar treatment decreased both insulin resistance and preserved β-cell function. As a result, muraglitazar treatment, when initiated before the onset of diabetes, prevented development of diabetes and, when initiated after the onset of diabetes, prevented worsening of diabetes in db/db mice.


Drug Metabolism and Disposition | 2011

Metabolism and disposition of 14C-labeled peliglitazar in humans.

Lifei Wang; Carey Munsick; Sean Chen; Samuel J. Bonacorsi; Peter T. W. Cheng; W. Griffith Humphreys; Donglu Zhang

The metabolism and disposition of dual 14C-labeled peliglitazar, a dual α/γ peroxisome proliferator-activated receptor activator, was investigated in 10 healthy male subjects with and without bile collection (groups 1 and 2) after a single 10-mg oral dose. Serial blood samples, urine, and feces (0–240 h) as well as bile samples (3–8 h after dosing from group 2 subjects) were collected. The maximum plasma concentration (Cmax) of drug was reached at approximately 1 h and the elimination half-life (t1/2) was approximately 3.5 h. The exposure to drug metabolites (Cmax and area under the plasma concentration versus time curve) was not significantly different between the two groups. The parent compound and its 1-O-β-acyl-glucuronide conjugate were the major components in plasma; other circulating metabolites, including several other glucuronide conjugates, were minor components at all time points. The major portion of the radioactive dose was recovered in feces (94% for group 1 and 32% for group 2). Approximately 24% of the radioactive dose was recovered in the bile from group 2 subjects, nearly all of which was assigned as glucuronides of peliglitazar and its oxidative metabolites (M14, M14a, M14b, M15, M15a, M15b, and M17). In contrast, fecal samples contained peliglitazar and its oxidative metabolites resulting from aliphatic/aryl hydroxylation, and O-demethylation. These results suggested that the major clearance pathway of peliglitazar was through biliary elimination of glucuronide conjugates, which were hydrolyzed to peliglitazar and its oxidative metabolites in the intestines before excretion.


ACS Medicinal Chemistry Letters | 2016

Discovery and Preclinical Evaluation of BMS-711939, an Oxybenzylglycine Based PPARα Selective Agonist

Yan Shi; Jun Li; Lawrence J. Kennedy; Shiwei Tao; Andres S. Hernandez; Zhi Lai; Sean Chen; Henry Wong; Juliang Zhu; Ashok Trehan; Ngiap-Kie Lim; Huiping Zhang; Bang-Chi Chen; Kenneth T. Locke; Kevin O’Malley; Litao Zhang; Rai Ajit Srivastava; Bowman Miao; Daniel Meyers; Hossain Monshizadegan; Debra Search; Denise Grimm; Rongan Zhang; Thomas Harrity; Lori Kunselman; Michael Cap; Jodi K. Muckelbauer; Chiehying Chang; Stanley R. Krystek; Yi-Xin Li

BMS-711939 (3) is a potent and selective peroxisome proliferator-activated receptor (PPAR) α agonist, with an EC50 of 4 nM for human PPARα and >1000-fold selectivity vs human PPARγ (EC50 = 4.5 μM) and PPARδ (EC50 > 100 μM) in PPAR-GAL4 transactivation assays. Compound 3 also demonstrated excellent in vivo efficacy and safety profiles in preclinical studies and thus was chosen for further preclinical evaluation. The synthesis, structure-activity relationship (SAR) studies, and in vivo pharmacology of 3 in preclinical animal models as well as its ADME profile are described.


Archive | 2002

Substituted acid derivatives useful as antidiabetic and antiobesity agents and method

Peter T. W. Cheng; Pratik Devasthale; Yoon T. Jeon; Sean Chen; Hao Zhang


Journal of Medicinal Chemistry | 2005

Design and synthesis of N-[(4-methoxyphenoxy)carbonyl]-N-[[4-[2-(5-methyl-2-phenyl-4-oxazolyl)ethoxy]phenyl]methyl]glycine [muraglitazar/BMS-298585], a novel peroxisome proliferator-activated receptor α/γ dual agonist with efficacious glucose and lipid-lowering activities

Pratik Devasthale; Sean Chen; Jeon Y; Fucheng Qu; Shao C; Wei Wang; Hongjian Zhang; Michael Cap; Farrelly D; Rajasree Golla; Gary J. Grover; Thomas Harrity; Zhengping Ma; Moore L; Ren J; Ramakrishna Seethala; Cheng L; Paul G. Sleph; Sun W; Tieman A; Wetterau; Arthur M. Doweyko; Chandrasena G; Chang Sy; Humphreys Wg; Sasseville Vg; Scott A. Biller; Denis E. Ryono; Selan F; Hariharan N


Archive | 2000

Oxa-and thiazole derivatives useful as antidiabetic and antiobesity agents

Peter T. W. Cheng; Pratik Devasthale; Yoon T. Jeon; Sean Chen; Hao Zhang


Diabetes | 2006

Muraglitazar, a Novel Dual (α/γ) Peroxisome Proliferator–Activated Receptor Activator, Improves Diabetes and Other Metabolic Abnormalities and Preserves β-Cell Function in db/db Mice

Thomas Harrity; Dennis Farrelly; Aaron Tieman; Cuixia Chu; Lori Kunselman; Liqun Gu; Randolph Ponticiello; Michael Cap; Fucheng Qu; Chunning Shao; Wei Wang; Hao Zhang; William Fenderson; Sean Chen; Pratik Devasthale; Yoon T. Jeon; Ramakrishna Seethala; Wen-Pin Yang; Jimmy Ren; Min Zhou; Denis E. Ryono; Scott A. Biller; Kasim A. Mookhtiar; John R. Wetterau; Richard E. Gregg; Peter T. W. Cheng; Narayanan Hariharan


Archive | 2008

1, 3 - dihydroxy substituted phenylamide glucokinase activators

Sean Chen; Peter T. W. Cheng; Rebecca A. Smirk

Collaboration


Dive into the Sean Chen's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Yan Shi

Bristol-Myers Squibb

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge