Network


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

Hotspot


Dive into the research topics where Jay P. Powers is active.

Publication


Featured researches published by Jay P. Powers.


Journal of Immunology | 2009

Elucidation of CXCR7-Mediated Signaling Events and Inhibition of CXCR4-Mediated Tumor Cell Transendothelial Migration by CXCR7 Ligands

Brian A. Zabel; Yu Wang; Susanna Lewén; Robert D. Berahovich; Mark E.T. Penfold; Penglie Zhang; Jay P. Powers; Bretton Summers; Zhenhua Miao; Bin Zhao; Ali Jalili; Anna Janowska-Wieczorek; Juan C. Jaen; Thomas J. Schall

CXCR7 binds chemokines CXCL11 (I-TAC) and CXCL12 (SDF-1) but does not act as a classical chemoattractant receptor. Using CCX771, a novel small molecule with high affinity and selectivity for CXCR7, we found that, although CXCR7 is dispensable for “bare filter” in vitro chemotaxis, CXCR7 plays an essential role in the CXCL12/CXCR4-mediated transendothelial migration (TEM) of CXCR4+CXCR7+ human tumor cells. Importantly, although CXCL11 is unable to stimulate directly the migration of these cells, it acts as a potent antagonist of their CXCL12-induced TEM. Furthermore, even though this TEM is driven by CXCR4, the CXCR7 ligand CCX771 is substantially more potent at inhibiting it than the CXCR4 antagonist AMD3100, which is more than 100 times weaker at inhibiting TEM when compared with its ability to block bare filter chemotaxis. Far from being a “silent” receptor, we show that CXCR7 displays early hallmark events associated with intracellular signaling. Upon cognate chemokine binding, CXCR7 associates with β-arrestin2, an interaction that can be blocked by CXCR7-specific mAbs. Remarkably, the synthetic CXCR7 ligand CCX771 also potently stimulates β-arrestin2 recruitment to CXCR7, with greater potency and efficacy than the endogenous chemokine ligands. These results indicate that CXCR7 can regulate CXCL12-mediated migratory cues, and thus may play a critical role in driving CXCR4+CXCR7+ tumor cell metastasis and tissue invasion. CXCR7 ligands, such as the chemokine CXCL11 and the newly described synthetic molecule CCX771, may represent novel therapeutic opportunities for the control of such cells.


Journal of The American Society of Nephrology | 2014

C5a Receptor (CD88) Blockade Protects against MPO-ANCA GN

Hong Xiao; Daniel J. Dairaghi; Jay P. Powers; Linda Ertl; Trageen Baumgart; Yu Wang; Lisa C. Seitz; Mark E.T. Penfold; Lin Gan; Peiqi Hu; B. Lu; Norma P. Gerard; Craig Gerard; Thomas J. Schall; Juan C. Jaen; Ronald J. Falk; J. Charles Jennette

Necrotizing and crescentic GN (NCGN) with a paucity of glomerular immunoglobulin deposits is associated with ANCA. The most common ANCA target antigens are myeloperoxidase (MPO) and proteinase 3. In a manner that requires activation of the alternative complement pathway, passive transfer of antibodies to mouse MPO (anti-MPO) induces a mouse model of ANCA NCGN that closely mimics human disease. Here, we confirm the importance of C5aR/CD88 in the mediation of anti-MPO-induced NCGN and report that C6 is not required. We further demonstrate that deficiency of C5a-like receptor (C5L2) has the reverse effect of C5aR/CD88 deficiency and results in more severe disease, indicating that C5aR/CD88 engagement enhances inflammation and C5L2 engagement suppresses inflammation. Oral administration of CCX168, a small molecule antagonist of human C5aR/CD88, ameliorated anti-MPO-induced NCGN in mice expressing human C5aR/CD88. These observations suggest that blockade of C5aR/CD88 might have therapeutic benefit in patients with ANCA-associated vasculitis and GN.


American Journal of Physiology-renal Physiology | 2013

CCR2 antagonist CCX140-B provides renal and glycemic benefits in diabetic transgenic human CCR2 knockin mice

Tim Sullivan; Zhenhua Miao; Daniel J. Dairaghi; Antoni Krasinski; Yu Wang; Bin N. Zhao; Trageen Baumgart; Linda Ertl; Andrew M. K. Pennell; Lisa Seitz; Jay P. Powers; Ruiping Zhao; Solomon Ungashe; Zheng Wei; Landin Boring; Chia-Lin Tsou; Israel F. Charo; Robert D. Berahovich; Thomas J. Schall; Juan C. Jaen

Chemokine (C-C motif) receptor 2 (CCR2) is central for the migration of monocytes into inflamed tissues. The novel CCR2 antagonist CCX140-B, which is currently in two separate phase 2 clinical trials in diabetic nephropathy, has recently been shown to reduce hemoglobin A1c and fasting blood glucose levels in type 2 diabetics. In this report, we describe the effects of this compound on glycemic and renal function parameters in diabetic mice. Since CCX140-B has a low affinity for mouse CCR2, transgenic human CCR2 knockin mice were generated and rendered diabetic with either a high-fat diet (diet-induced obesity) or by deletion of the leptin receptor gene (db/db). CCX140-B treatment in both models resulted in decreased albuminuria, which was associated with decreased glomerular hypertrophy and increased podocyte density. Moreover, treatment of diet-induced obese mice with CCX140-B resulted in decreased levels of fasting blood glucose and insulin, normalization of homeostatic model assessment of insulin resistance values, and decreased numbers of adipose tissue inflammatory macrophages. Unlike other CCR2 antagonists, CCX140-B had no effect on plasma levels of the CCR2 ligand CCL2 or on the numbers of blood monocytes. These results support the ongoing evaluation of this molecule in diabetic subjects with impaired renal function.


Journal of Medicinal Chemistry | 2013

Rational Design and Binding Mode Duality of MDM2–p53 Inhibitors

Felix Gonzalez-Lopez de Turiso; Daqing Sun; Yosup Rew; Michael D. Bartberger; Hilary P. Beck; Jude Canon; Ada Chen; David Chow; Tiffany L. Correll; Xin Huang; Lisa Julian; Frank Kayser; Mei-Chu Lo; Alexander M. Long; Dustin L. McMinn; Jonathan D. Oliner; Tao Osgood; Jay P. Powers; Anne Y. Saiki; Steve Schneider; Paul Shaffer; Shou-Hua Xiao; Peter Yakowec; Xuelei Yan; Qiuping Ye; Dongyin Yu; Xiaoning Zhao; Jing Zhou; Julio C. Medina; Steven H. Olson

Structural analysis of both the MDM2-p53 protein-protein interaction and several small molecules bound to MDM2 led to the design and synthesis of tetrasubstituted morpholinone 10, an MDM2 inhibitor with a biochemical IC50 of 1.0 μM. The cocrystal structure of 10 with MDM2 inspired two independent optimization strategies and resulted in the discovery of morpholinones 16 and 27 possessing distinct binding modes. Both analogues were potent MDM2 inhibitors in biochemical and cellular assays, and morpholinone 27 (IC50 = 0.10 μM) also displayed suitable PK profile for in vivo animal experiments. A pharmacodynamic (PD) experiment in mice implanted with human SJSA-1 tumors showed p21(WAF1) mRNA induction (2.7-fold over vehicle) upon oral dosing of 27 at 300 mg/kg.


Blood | 2012

CCR1 blockade reduces tumor burden and osteolysis in vivo in a mouse model of myeloma bone disease

Daniel J. Dairaghi; Babatunde O. Oyajobi; Anjana Gupta; Brandon McCluskey; Shichang Miao; Jay P. Powers; Lisa C. Seitz; Yu Wang; Yibin Zeng; Penglie Zhang; Thomas J. Schall; Juan C. Jaen

The chemokine CCL3/MIP-1α is a risk factor in the outcome of multiple myeloma (MM), particularly in the development of osteolytic bone disease. This chemokine, highly overexpressed by MM cells, can signal mainly through 2 receptors, CCR1 and CCR5, only 1 of which (CCR1) is responsive to CCL3 in human and mouse osteoclast precursors. CCR1 activation leads to the formation of osteolytic lesions and facilitates tumor growth. Here we show that formation of mature osteoclasts is blocked by the highly potent and selective CCR1 antagonist CCX721, an analog of the clinical compound CCX354. We also show that doses of CCX721 selected to completely inhibit CCR1 produce a profound decrease in tumor burden and osteolytic damage in the murine 5TGM1 model of MM bone disease. Similar effects were observed when the antagonist was used prophylactically or therapeutically, with comparable efficacy to that of zoledronic acid. 5TGM1 cells were shown to express minimal levels of CCR1 while secreting high levels of CCL3, suggesting that the therapeutic effects of CCX721 result from CCR1 inhibition on non-MM cells, most likely osteoclasts and osteoclast precursors. These results provide a strong rationale for further development of CCR1 antagonists for the treatment of MM and associated osteolytic bone disease.


Journal of Medicinal Chemistry | 2008

Discovery of Novel, Potent Benzamide Inhibitors of 11β-Hydroxysteroid Dehydrogenase Type 1 (11β-HSD1) Exhibiting Oral Activity in an Enzyme Inhibition ex Vivo Model

Lisa Julian; Zhulun Wang; Tracy Bostick; Seb Caille; Rebekah Choi; Michael DeGraffenreid; Yongmei Di; Xiao He; Randall W. Hungate; Juan C. Jaen; Jinsong Liu; Mario Monshouwer; Dustin Mcminn; Yosup Rew; Athena Sudom; Daqing Sun; Hua Tu; Stefania Ursu; Nigel Walker; Xuelei Yan; Qiuping Ye; Jay P. Powers

We report the discovery of potent benzamide inhibitors of 11beta-hydroxysteroid dehydrogenase (11beta-HSD1). The optimization and correlation of in vitro and in vivo metabolic stability will be described. Through modifications to our initial lead 2, we discovered pyridyl compound 13. This compound has a favorable pharmacokinetic profile across three species and showed a dose-dependent decrease in adipose 11beta-HSD1 activity in a monkey ex vivo pharmacodynamic model.


Bioorganic & Medicinal Chemistry Letters | 2008

Discovery and Initial SAR of Arylsulfonylpiperazine Inhibitors of 11β-Hydroxysteroid Dehydrogenase Type 1 (11β-HSD1)

Daqing Sun; Zhulun Wang; Yongmei Di; Juan C. Jaen; Marc Labelle; Ji Ma; Shichang Miao; Athena Sudom; Liang Tang; Craig Tomooka; Hua Tu; Stefania Ursu; Nigel Walker; Xuelei Yan; Qiuping Ye; Jay P. Powers

High-throughput screening of a small-molecule compound library resulted in the identification of a series of arylsulfonylpiperazines that are potent and selective inhibitors of human 11beta-Hydroxysteroid Dehydrogenase Type 1 (11beta-HSD1). Optimization of the initial lead resulted in the discovery of compound (R)-45 (11beta-HSD1 IC(50)=3nM).


Metabolism-clinical and Experimental | 2013

Experimental evidence for the use of CCR2 antagonists in the treatment of type 2 diabetes

Timothy J. Sullivan; Zhenhua Miao; Bin N. Zhao; Linda Ertl; Yu Wang; Antoni Krasinski; Matthew J. Walters; Jay P. Powers; Daniel J. Dairaghi; Trageen Baumgart; Lisa Seitz; Robert D. Berahovich; Thomas J. Schall; Juan C. Jaen

OBJECTIVE CCR2 inhibition has produced promising experimental and clinical anti-hyperglycemic effects. These results support the thesis that insulin resistance and Type 2 diabetes (T2D) are associated with chronic unresolved inflammation. The aim of this study was to provide a broad analysis of the various physiological changes occurring in mouse models of T2D in connection with pharmacological CCR2 inhibition. MATERIALS/METHODS A mouse-active chemical analogue of the clinical candidate CCX140-B was tested in diet-induced obese (DIO) mice and db/db mice. Measurements included: adipose tissue inflammatory macrophage counts; peripheral blood glucose levels at steady-state and after glucose and insulin challenges; peripheral blood insulin and adiponectin levels; 24-h urine output and urinary glucose levels; pancreatic islet number and size; hepatic triglyceride and glycogen content; and hepatic glucose-6-phosphatase levels. RESULTS In DIO mice, the CCR2 antagonist completely blocked the recruitment of inflammatory macrophages to visceral adipose tissue. The mice exhibited reduced hyperglycemia and insulinemia, improved insulin sensitivity, increased circulating adiponectin levels, decreased pancreatic islet size and increased islet number. It also reduced urine output, glucose excretion, hepatic glycogen and triglyceride content and glucose 6-phosphatase levels. Similar effects were observed in the db/db diabetic mice. CONCLUSIONS These data indicate that pharmacological inhibition of CCR2 in models of T2D can reduce inflammation in adipose tissue, alter hepatic metabolism and ameliorate multiple diabetic parameters. These mechanisms may contribute to the promising anti-diabetic effects seen in humans with at least one CCR2 antagonist.


Bioorganic & Medicinal Chemistry Letters | 2009

Discovery and optimization of piperidyl benzamide derivatives as a novel class of 11β-HSD1 inhibitors

Yosup Rew; Dustin L. McMinn; Zhulun Wang; Xiao He; Randall W. Hungate; Juan C. Jaen; Athena Sudom; Daqing Sun; Hua Tu; Stefania Ursu; Elisia Villemure; Nigel Walker; Xuelei Yan; Qiuping Ye; Jay P. Powers

Discovery and optimization of a piperidyl benzamide series of 11beta-HSD1 inhibitors is described. This series was derived from a cyclohexyl benzamide lead structures to address PXR selectivity, high non-specific protein binding, poor solubility, limited in vivo exposure, and in vitro cytotoxicity issues observed with the cyclohexyl benzamide structures. These efforts led to the discovery of piperidyl benzamide 15 which features improved properties over the cyclohexyl benzamide derivatives.


Bioorganic & Medicinal Chemistry | 2008

Distinctive molecular inhibition mechanisms for selective inhibitors of human 11β-hydroxysteroid dehydrogenase type 1

Hua Tu; Jay P. Powers; Jinsong Liu; Stefania Ursu; Athena Sudom; Xuelei Yan; Haoda Xu; David Park Meininger; Michael DeGraffenreid; Xiao He; Juan C. Jaen; Daqing Sun; Marc Labelle; Hiroshi Yamamoto; Bei Shan; Nigel Walker; Zhulun Wang

11beta-hydroxysteroid dehydrogenase type 1 (11beta-HSD1) catalyzes the NADPH dependent interconversion of inactive cortisone to active cortisol. Excess 11beta-HSD1 or cortisol leads to insulin resistance and metabolic syndrome in animal models and in humans. Inhibiting 11beta-HSD1 activity signifies a promising therapeutic strategy in the treatment of Type 2 diabetes and related diseases. Herein, we report two highly potent and selective small molecule inhibitors of human 11beta-HSD1. While compound 1, a sulfonamide, functions as a simple substrate competitive inhibitor, compound 2, a triazole, shows the kinetic profile of a mixed inhibitor. Co-crystal structures reveal that both compounds occupy the 11beta-HSD1 catalytic site, but present distinct molecular interactions with the protein. Strikingly, compound 2 interacts much closer to the cofactor NADP+ and likely modifies its binding. Together, the structural and kinetic analyses demonstrate two distinctive molecular inhibition mechanisms, providing valuable information for future inhibitor design.

Collaboration


Dive into the Jay P. Powers's collaboration.

Researchain Logo
Decentralizing Knowledge