Network


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

Hotspot


Dive into the research topics where Jenny Xie is active.

Publication


Featured researches published by Jenny Xie.


Journal of Immunology | 2003

Sphingosine-1-Phosphate Receptor Agonism Impairs the Efficiency of the Local Immune Response by Altering Trafficking of Naive and Antigen-Activated CD4+ T Cells

Jenny Xie; Naomi Nomura; Sam L. Koprak; Elizabeth J. Quackenbush; Michael J. Forrest; Hugh Rosen

FTY720 (2-amino-[2-(4-octylphenyl) ethyl]-1,3-propanediol hydrochloride) is an immunosuppressive agent that inhibits allograft rejection. We recently demonstrated that FTY-phosphate, the active metabolite of FTY720, acts as a full agonist for sphingosine-1-phosphate (S1P) receptors. Furthermore, activation of S1P receptors with their natural ligand, S1P, as well as pharmacological ligands leads to lymphopenia, probably due to sequestration of lymphocytes in secondary lymphoid organs. In the present study we used a local Ag-challenged mouse model to examine the effects of FTY720 on T cell activation in the draining lymph node (DLN) and on the release of activated T cells to the peripheral blood compartment. We showed that the number of Ag-activated CD4+ T cells in the DLN after injection of Ag and CFA into a footpad was dramatically reduced after FTY720 treatment. However, T cell proliferation, both in vitro and in vivo, was not impaired by FTY720. Our results suggest that the reduced efficiency of T cell responses in the DLN in response to a local Ag is probably due to a defective recirculation of naive T cells caused by FTY720 treatment. Furthermore, we found that the numbers of naive and Ag-activated CD4+ T cells in the peripheral blood of Ag-challenged mice were equally reduced with FTY720 treatment, suggesting that both T cell subsets are sequestered in the DLNs. Thus, FTY720 induces immunosuppression through inhibition of both the recirculation of naive T cells and the release of Ag-activated T cells from the DLN to lymph and to the blood compartment.


Journal of Immunology | 2014

Engineering of a Novel Anti-CD40L Domain Antibody for Treatment of Autoimmune Diseases

Jenny Xie; Aaron P. Yamniuk; Virna Borowski; Robert Kuhn; Vojkan Susulic; Sandra Rex-Rabe; Xiaoxia Yang; Xiadi Zhou; Yifan Zhang; Kathleen M. Gillooly; Ruth Brosius; Rathna Ravishankar; Kimberly Waggie; Kathy Mink; Laura A. Price; Robert Rehfuss; James Tamura; Yongmi An; Lin Cheng; Bozena Abramczyk; Olga Ignatovich; Philip Drew; Steven Grant; James W. Bryson; Suzanne J. Suchard; Luisa Salter-Cid; Steven G. Nadler; Anish Suri

CD40–CD40L interactions play a critical role in regulating immune responses. Blockade of CD40L by Abs, such as the anti-CD40L Ab 5c8, demonstrated positive clinical effects in patients with autoimmune diseases; however, incidents of thromboembolism (TE) precluded further development of these molecules. In this study, we examined the role of the Fc domain interaction with FcγRs in modulating platelet activation and potential for TE. Our results show that the interaction of the 5c8 wild-type IgG1 Fc domain with FcγRs is responsible for platelet activation, as measured by induction of PAC-1 and CD62P. A version of 5c8 with a mutated IgG1 tail was identified that showed minimal FcγR binding and platelet activation while maintaining full binding to CD40L. To address whether Fc effector function is required for immunosuppression, a potent Ab fragment, termed a “domain Ab” (dAb), against murine CD40L was identified and fused to a murine IgG1 Fc domain containing a D265A mutation that lacks Fc effector function. In vitro, this dAb–Fc demonstrated comparable potency to the benchmark mAb MR-1 in inhibiting B cell and dendritic cell activation. Furthermore, the anti-CD40L dAb–Fc exhibited a notable efficacy comparable to MR-1 in various preclinical models, such as keyhole limpet hemocyanin–induced Ab responses, alloantigen-induced T cell proliferation, “heart-to-ear” transplantation, and NZB × NZW F1 spontaneous lupus. Thus, our data show that immunosuppression and TE can be uncoupled and that a CD40L dAb with an inert Fc tail is expected to be efficacious for treating autoimmune diseases, with reduced risk for TE.


Journal of Medicinal Chemistry | 2016

Potent and Selective Agonists of Sphingosine 1-Phosphate 1 (S1P1): Discovery and SAR of a Novel Isoxazole Based Series

Scott H. Watterson; Junqing Guo; Steve Spergel; Charles M. Langevine; Robert V. Moquin; Ding Ren Shen; Melissa Yarde; Mary Ellen Cvijic; Dana Banas; Richard Liu; Suzanne J. Suchard; Kathleen M. Gillooly; Tracy L. Taylor; Sandra Rex-Rabe; David J. Shuster; Kim W. McIntyre; Georgia Cornelius; Celia D’Arienzo; Anthony Marino; Praveen Balimane; Bethanne M. Warrack; Luisa Salter-Cid; Murray McKinnon; Joel C. Barrish; Percy H. Carter; William J. Pitts; Jenny Xie; Alaric J. Dyckman

Sphingosine 1-phosphate (S1P) is the endogenous ligand for the sphingosine 1-phosphate receptors (S1P1-5) and evokes a variety of cellular responses through their stimulation. The interaction of S1P with the S1P receptors plays a fundamental physiological role in a number of processes including vascular development and stabilization, lymphocyte migration, and proliferation. Agonism of S1P1, in particular, has been shown to play a significant role in lymphocyte trafficking from the thymus and secondary lymphoid organs, resulting in immunosuppression. This article will detail the discovery and SAR of a potent and selective series of isoxazole based full agonists of S1P1. Isoxazole 6d demonstrated impressive efficacy when administered orally in a rat model of arthritis and in a mouse experimental autoimmune encephalomyelitis (EAE) model of multiple sclerosis.


Journal of Medicinal Chemistry | 2016

Discovery and Structure–Activity Relationship (SAR) of a Series of Ethanolamine-Based Direct-Acting Agonists of Sphingosine-1-phosphate (S1P1)

John L. Gilmore; James E. Sheppeck; Scott H. Watterson; Lauren Haque; Parag Mukhopadhyay; Andrew J. Tebben; Michael A. Galella; Ding Ren Shen; Melissa Yarde; Mary Ellen Cvijic; Virna Borowski; Kathleen M. Gillooly; Tracy L. Taylor; Kim W. McIntyre; Bethanne M. Warrack; Paul Levesque; Julia P. Li; Georgia Cornelius; Celia D’Arienzo; Anthony Marino; Praveen Balimane; Luisa Salter-Cid; Joel C. Barrish; William J. Pitts; Percy H. Carter; Jenny Xie; Alaric J. Dyckman

Sphingosine-1-phosphate (S1P) is a bioactive sphingolipid metabolite that regulates a multitude of physiological processes such as lymphocyte trafficking, cardiac function, vascular development, and inflammation. Because of the ability of S1P1 receptor agonists to suppress lymphocyte egress, they have great potential as therapeutic agents in a variety of autoimmune diseases. In this article, the discovery of selective, direct acting S1P1 agonists utilizing an ethanolamine scaffold containing a terminal carboxylic acid is described. Potent S1P1 agonists such as compounds 18a and 19a which have greater than 1000-fold selectivity over S1P3 are described. These compounds efficiently reduce blood lymphocyte counts in rats through 24 h after single doses of 1 and 0.3 mpk, respectively. Pharmacodynamic properties of both compounds are discussed. Compound 19a was further studied in two preclinical models of disease, exhibiting good efficacy in both the rat adjuvant arthritis model (AA) and the mouse experimental autoimmune encephalomyelitis model (EAE).


ACS Medicinal Chemistry Letters | 2016

Identification and Preclinical Pharmacology of BMS-986104: A Differentiated S1P1 Receptor Modulator in Clinical Trials

T. G. Murali Dhar; Hai-Yun Xiao; Jenny Xie; Lois D. Lehman-McKeeman; Dauh-Rurng Wu; Marta Dabros; Xiaoxia Yang; Tracy L. Taylor; Xia D. Zhou; Elizabeth M. Heimrich; Rochelle Thomas; Kim W. McIntyre; Bethanne M. Warrack; Hong Shi; Paul Levesque; Jia L. Zhu; James K. Hennan; Praveen Balimane; Zheng Yang; Anthony Marino; Georgia Cornelius; Celia D’Arienzo; Arvind Mathur; Ding Ren Shen; Mary Ellen Cvijic; Luisa Salter-Cid; Joel C. Barrish; Percy H. Carter; Alaric J. Dyckman

Clinical validation of S1P receptor modulation therapy was achieved with the approval of fingolimod (Gilenya, 1) as the first oral therapy for relapsing remitting multiple sclerosis. However, 1 causes a dose-dependent reduction in the heart rate (bradycardia), which occurs within hours after first dose. We disclose the identification of clinical compound BMS-986104 (3d), a novel S1P1 receptor modulator, which demonstrates ligand-biased signaling and differentiates from 1 in terms of cardiovascular and pulmonary safety based on preclinical pharmacology while showing equivalent efficacy in a T-cell transfer colitis model.


Journal of Pharmacology and Experimental Therapeutics | 2015

Dual Inhibition of Interleukin-23 and Interleukin-17 Offers Superior Efficacy in Mouse Models of Autoimmunity

Paul R. Mangan; Linhui Julie Su; Victoria Jenny; Andrea L. Tatum; Caryn Picarillo; Stacey Skala; Noah Ditto; Zheng Lin; Xiaoxia Yang; Pete Z. Cotter; David J. Shuster; Yunling Song; Virna Borowski; Rochelle Thomas; Elizabeth M. Heimrich; Brigitte Devaux; Ruchira Das Gupta; Irvith M. Carvajal; Kim W. McIntyre; Jenny Xie; Qihong Zhao; Mary Struthers; Luisa Salter-Cid

Therapies targeting either interleukin (IL)-23 or IL-17 have shown promise in treating T helper 17 (Th17)–driven autoimmune diseases. Although IL-23 is a critical driver of IL-17, recognition of nonredundant and independent functions of IL-23 and IL-17 has prompted the notion that dual inhibition of both IL-23 and IL-17 could offer even greater efficacy for treating autoimmune diseases relative to targeting either cytokine alone. To test this hypothesis, we generated selective inhibitors of IL-23 and IL-17 and tested the effect of either treatment alone compared with their combination in vitro and in vivo. In vitro, using a novel culture system of murine Th17 cells and NIH/3T3 fibroblasts, we showed that inhibition of both IL-23 and IL-17 completely suppressed IL-23–dependent IL-22 production from Th17 cells and cooperatively blocked IL-17–dependent IL-6 secretion from the NIH/3T3 cells to levels below either inhibitor alone. In vivo, in the imiquimod induced skin inflammation model, and in the myelin oligodendrocyte glycoprotein peptide–induced experimental autoimmune encephalomyelitis model, we demonstrated that dual inhibition of IL-17 and IL-23 was more efficacious in reducing disease than targeting either cytokine alone. Together, these data support the hypothesis that neutralization of both IL-23 and IL-17 may provide enhanced benefit against Th17 mediated autoimmunity and provide a basis for a therapeutic strategy aimed at dual targeting IL-23 and IL-17.


Bioorganic & Medicinal Chemistry Letters | 2016

Discovery and SAR of pyrrolo[2,1-f][1,2,4]triazin-4-amines as potent and selective PI3Kδ inhibitors.

Rajeev S. Bhide; James Neels; Lan-Ying Qin; Zheming Ruan; Sylwia Stachura; Carolyn A. Weigelt; John S. Sack; Kevin Stefanski; Xiaomei Gu; Jenny Xie; Christine Goldstine; Stacey Skala; Donna L. Pedicord; Stefan Ruepp; T. G. Murali Dhar; Percy H. Carter; Luisa Salter-Cid; Michael A. Poss; Paul Davies

Aberrant Class I PI3K signaling is a key factor contributing to many immunological disorders and cancers. We have identified 4-amino pyrrolotriazine as a novel chemotype that selectively inhibits PI3Kδ signaling despite not binding to the specificity pocket of PI3Kδ isoform. Structure activity relationship (SAR) led to the identification of compound 30 that demonstrated efficacy in mouse Keyhole Limpet Hemocyanin (KLH) and collagen induced arthritis (CIA) models.


Journal of Pharmacology and Experimental Therapeutics | 2015

Integrated Pharmacokinetic/Pharmacodynamic Analysis for Determining the Minimal Anticipated Biological Effect Level of a Novel Anti-CD28 Receptor Antagonist BMS-931699

Zheng Yang; Hugh Wang; T. W. Salcedo; Suzanne J. Suchard; Jenny Xie; L. A. Schneeweis; C. A. Fleener; J. D. Calore; Rong Shi; S. X. Y. Zhang; Rodrigues Ad; Bruce D. Car; Punit Marathe; Steven G. Nadler

BMS-931699 (lulizumab pegol), a domain antibody (dAb) conjugated with 40-kDa branched polyethylene glycol, is a human anti-CD28 receptor antagonist under development for the treatment of inflammatory and autoimmune diseases. In the present work, the minimal anticipated biologic effect level (MABEL) was determined for BMS-931699 by integrating all the available preclinical data. The relevance of the in vitro mixed lymphocyte reaction (MLR) assay to a whole blood CD28 receptor occupancy (RO) assessment, as well as the relationship between the CD28 RO and the inhibition of T-cell-dependent antibody response to keyhole limpet hemocyanin in vivo, was demonstrated through an integrated pharmacokinetic/pharmacodynamic analysis using anti-hCD28 dAb-001 (differing from BMS-931699 by two additional amino acids at the N-terminus) and a mouse surrogate. Based on this analysis, the EC10 value (0.32 nM) from the human MLR assay and the human plasma volume (0.04 l/kg) were employed to calculate the MABEL (0.01 mg) of BMS-931699 in humans, with a CD28 RO predicted to be ≤10%. The estimated MABEL dose was threefold higher than the value derived from the binding constant and twofold less than the MABEL converted from animal efficacy studies based on the body surface area. Furthermore, it was 2900-fold lower than the human equivalent dose derived from the no observed adverse effect level in monkeys (15 mg/kg/week for 5 doses, intravenous dosing) with a 10-fold safety factor applied. Therefore, the MABEL dose represented a sound approach to mitigate any potential risk in targeting CD28 and was successfully used as the first-in-human starting dose for BMS-931699.


Journal of Leukocyte Biology | 2017

A novel function of CXCL10 in mediating monocyte production of proinflammatory cytokines

Qihong Zhao; Taeg Kim; Jian Pang; Wendy Sun; Xiaoxia Yang; Jinhong Wang; Yunling Song; Hongwei Zhang; Huadong Sun; Vangipuram S. Rangan; Shrikant Deshpande; Huaping Tang; Mary Ellen Cvijic; Richard A. Westhouse; Timothy Olah; Jenny Xie; Mary Struthers; Luisa Salter-Cid

IFN‐γ–inducible protein 10 (CXCL10), a chemokine that is abundantly secreted in response to inflammatory stimuli, has been implicated in the pathogenesis of multiple inflammatory diseases, such as inflammatory bowel disease. Whereas CXCL10 is traditionally recognized for recruiting pathogenic T cells to inflamed sites, its nonchemotactic role during inflammation remains poorly defined. In this report, we identified a novel function of CXCL10 in the regulation of the inflammatory potential of human monocytes to produce cytokines. We found that CXCL10 was necessary and sufficient for IFN‐γ–primed human monocytes to induce a robust production of proinflammatory cytokines, such as IL‐12 and IL‐23. CXCL10‐induced monocyte production of these cytokines depended on CXCR3 receptor engagement as well as on the Iκ B kinase and p38 MAPK signaling pathways. By using an innate‐mediated murine colitis model, we demonstrated that anti‐CXCL10 Ab treatment robustly suppressed the local production of myeloid‐derived inflammatory cytokines and intestinal tissue damage. Together, our data unravel a previously unappreciated role of CXCL10 in the amplification of myeloid cell–mediated inflammatory responses. Targeting CXCL10 is therefore an attractive approach to treating inflammatory diseases that are driven by innate and adaptive immunity.


Journal of Biomolecular Screening | 2014

Integrating High-Content Analysis into a Multiplexed Screening Approach to Identify and Characterize GPCR Agonists

Yingjie Zhu; John Watson; Mengjie Chen; Ding Ren Shen; Melissa Yarde; Michele Agler; Neil T. Burford; Andrew Alt; Sukhanya Jayachandra; Mary Ellen Cvijic; Litao Zhang; Alaric J. Dyckman; Jenny Xie; Jonathan O’Connell; Martyn Banks; Andrea Weston

G protein–coupled receptors (GPCRs) are one of the most popular and proven target classes for therapeutic intervention. The increased appreciation for allosteric modulation, receptor oligomerization, and biased agonism has led to the development of new assay platforms that seek to capitalize on these aspects of GPCR biology. High-content screening is particularly well suited for GPCR drug discovery given the ability to image and quantify changes in multiple cellular parameters, to resolve subcellular structures, and to monitor events within a physiologically relevant environment. Focusing on the sphingosine-1-phosphate (S1P1) receptor, we evaluated the utility of high-content approaches in hit identification efforts by developing and applying assays to monitor β-arrestin translocation, GPCR internalization, and GPCR recycling kinetics. Using these approaches in combination with more traditional GPCR screening assays, we identified compounds whose unique pharmacological profiles would have gone unnoticed if using a single platform. In addition, we identified a compound that induces an atypical pattern of β-arrestin translocation and GPCR recycling kinetics. Our results highlight the value of high-content imaging in GPCR drug discovery efforts and emphasize the value of a multiassay approach to study pharmacological properties of compounds of interest.

Collaboration


Dive into the Jenny Xie'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
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge