Network


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

Hotspot


Dive into the research topics where Shomir Ghosh is active.

Publication


Featured researches published by Shomir Ghosh.


Immunity | 2014

Small molecule RORγt antagonists inhibit T helper 17 cell transcriptional network by divergent mechanisms

Sheng Xiao; Nir Yosef; Jianfei Yang; Yonghui Wang; Ling Zhou; Chen Zhu; Chuan Wu; Erkan Baloglu; Darby Schmidt; Radha Ramesh; Mercedes Lobera; Mark S. Sundrud; Pei-Yun Tsai; Zhijun Xiang; Jinsong Wang; Yan Xu; Xichen Lin; Karsten Kretschmer; Peter B. Rahl; Richard A. Young; Zhong Zhong; David A. Hafler; Aviv Regev; Shomir Ghosh; Alexander Marson; Vijay K. Kuchroo

We identified three retinoid-related orphan receptor gamma t (RORγt)-specific inhibitors that suppress T helper 17 (Th17) cell responses, including Th17-cell-mediated autoimmune disease. We systemically characterized RORγt binding in the presence and absence of drugs with corresponding whole-genome transcriptome sequencing. RORγt acts as a direct activator of Th17 cell signature genes and a direct repressor of signature genes from other T cell lineages; its strongest transcriptional effects are on cis-regulatory sites containing the RORα binding motif. RORγt is central in a densely interconnected regulatory network that shapes the balance of T cell differentiation. Here, the three inhibitors modulated the RORγt-dependent transcriptional network to varying extents and through distinct mechanisms. Whereas one inhibitor displaced RORγt from its target loci, the other two inhibitors affected transcription predominantly without removing DNA binding. Our work illustrates the power of a system-scale analysis of transcriptional regulation to characterize potential therapeutic compounds that inhibit pathogenic Th17 cells and suppress autoimmunity.


Nature Chemical Biology | 2013

Selective class IIa histone deacetylase inhibition via a nonchelating zinc-binding group

Mercedes Lobera; Kevin P. Madauss; Denise Teotico Pohlhaus; Quentin G Wright; Mark Trocha; Darby Schmidt; Erkan Baloglu; Ryan P. Trump; Martha S. Head; Glenn A. Hofmann; Monique Murray-Thompson; Benjamin Schwartz; Subhas Chakravorty; Zining Wu; Palwinder K. Mander; Laurens Kruidenier; Robert A. Reid; William Burkhart; Brandon J Turunen; James X. Rong; Craig D. Wagner; Mary Moyer; Carrow Wells; Xuan Hong; John T. Moore; Jon D. Williams; Dulce Soler; Shomir Ghosh; Michael A. Nolan

In contrast to studies on class I histone deacetylase (HDAC) inhibitors, the elucidation of the molecular mechanisms and therapeutic potential of class IIa HDACs (HDAC4, HDAC5, HDAC7 and HDAC9) is impaired by the lack of potent and selective chemical probes. Here we report the discovery of inhibitors that fill this void with an unprecedented metal-binding group, trifluoromethyloxadiazole (TFMO), which circumvents the selectivity and pharmacologic liabilities of hydroxamates. We confirm direct metal binding of the TFMO through crystallographic approaches and use chemoproteomics to demonstrate the superior selectivity of the TFMO series relative to a hydroxamate-substituted analog. We further apply these tool compounds to reveal gene regulation dependent on the catalytic active site of class IIa HDACs. The discovery of these inhibitors challenges the design process for targeting metalloenzymes through a chelating metal-binding group and suggests therapeutic potential for class IIa HDAC enzyme blockers distinct in mechanism and application compared to current HDAC inhibitors.


Journal of Immunology | 2014

Pharmacologic Inhibition of RORγt Regulates Th17 Signature Gene Expression and Suppresses Cutaneous Inflammation In Vivo

Jill Skepner; Radha Ramesh; Mark Trocha; Darby Schmidt; Erkan Baloglu; Mercedes Lobera; Thaddeus Carlson; Jonathan Hill; Lisa A. Orband-Miller; Ashley Barnes; Mohamed Boudjelal; Mark S. Sundrud; Shomir Ghosh; Jianfei Yang

IL-17–producing CD4+Th17 cells, CD8+Tc17 cells, and γδ T cells play critical roles in the pathogenesis of autoimmune psoriasis. RORγt is required for the differentiation of Th17 cells and expression of IL-17. In this article, we describe a novel, potent, and selective RORγt inverse agonist (TMP778), and its inactive diastereomer (TMP776). This chemistry, for the first time to our knowledge, provides a unique and powerful set of tools to probe RORγt-dependent functions. TMP778, but not TMP776, blocked human Th17 and Tc17 cell differentiation and also acutely modulated IL-17A production and inflammatory Th17-signature gene expression (Il17a, Il17f, Il22, Il26, Ccr6, and Il23) in mature human Th17 effector/memory T cells. In addition, TMP778, but not TMP776, inhibited IL-17A production in both human and mouse γδ T cells. IL-23–induced IL-17A production was also blocked by TMP778 treatment. In vivo targeting of RORγt in mice via TMP778 administration reduced imiquimod-induced psoriasis-like cutaneous inflammation. Further, TMP778 selectively regulated Th17-signature gene expression in mononuclear cells isolated from both the blood and affected skin of psoriasis patients. In summary, to our knowledge, we are the first to demonstrate that RORγt inverse agonists: 1) inhibit Tc17 cell differentiation, as well as IL-17 production by γδ T cells and CD8+ Tc17 cells; 2) block imiquimod-induced cutaneous inflammation; 3) inhibit Th17 signature gene expression by cells isolated from psoriatic patient samples; and 4) block IL-23–induced IL-17A expression. Thus, RORγt is a tractable drug target for the treatment of cutaneous inflammatory disorders, which may afford additional therapeutic benefit over existing modalities that target only IL-17A.


Immunology | 2015

In vivo regulation of gene expression and T helper type 17 differentiation by RORγt inverse agonists.

Jill Skepner; Mark Trocha; Radha Ramesh; Xiaoyan A. Qu; Darby Schmidt; Erkan Baloglu; Mercedes Lobera; Scott P. Davis; Michael A. Nolan; Thaddeus Carlson; Jonathan Hill; Shomir Ghosh; Mark S. Sundrud; Jianfei Yang

The orphan nuclear receptor, retinoic acid receptor‐related orphan nuclear receptor γt (RORγt), is required for the development and pathogenic function of interleukin‐17A‐secreting CD4+ T helper type 17 (Th17) cells. Whereas small molecule RORγt antagonists impair Th17 cell development and attenuate autoimmune inflammation in vivo, the broader effects of these inhibitors on RORγt‐dependent gene expression in vivo has yet to be characterized. We show that the RORγt inverse agonist TMP778 acts potently and selectively to block mouse Th17 cell differentiation in vitro and to impair Th17 cell development in vivo upon immunization with the myelin antigen MOG35–55 plus complete Freunds adjuvant. Importantly, we show that TMP778 acts in vivo to repress the expression of more than 150 genes, most of which fall outside the canonical Th17 transcriptional signature and are linked to a variety of inflammatory pathologies in humans. Interestingly, more than 30 genes are related with SMAD3, a transcription factor involved in the Th17 cell differentiation. These results reveal novel disease‐associated genes regulated by RORγt during inflammation in vivo, and provide an early read on potential disease indications and safety concerns associated with pharmacological targeting of RORγt.


Annual Reports in Medicinal Chemistry | 2011

Targeting Th17 and Treg Signaling Pathways in Autoimmunity

Shomir Ghosh; Mercedes Lobera; Mark S. Sundrud

Publisher Summary This chapter focuses on the Th17 and T regulatory (Treg) signaling pathways in autoimmunity. Treg cells are distinguished from conventional naive T cells by their constitutive expression of the transcription factor Forkhead box, winged-helix protein 3 (Foxp3). Current paradigms suggest that the balance between conventional naive T cell activation and Treg-mediated immune suppression controls whether immune responses are ultimately protective, ineffective, or pathogenic. Early clinical results utilizing Treg cellular therapy support the notion that increasing Treg numbers in autoimmune patients can support tolerance. Evidence from both autoimmune mouse models and human patients reveals that Treg cells are necessary to prevent spontaneous autoimmunity throughout life, thereby indicating that Treg cells can also regulate immune responses to pathogens and developing tumors. T cell–driven autoimmune disorders continue to present a significant unmet clinical need. Advances in the understanding of T cell activation, differentiation, and regulation have yielded novel approaches to specifically dampen pathogenic immune reactions without creating potentially dangerous states of general immune suppression. Specific modulation of Th17 and Treg cells affords broad therapeutic promise for the treatment of autoimmune and chronic inflammatory conditions.


Archive | 2013

Methods of treatment

Shomir Ghosh; Mercedes Lobera


Journal of clinical & cellular immunology | 2013

Small Molecule Inhibitors Targeting the Th17 Cell Transcription Factor RORγt for the Treatment of Autoimmune Diseases

Jianfei Yang; Jill Skepner; Mark Trocha; Shomir Ghosh


Archive | 2013

COMPOUNDS AND METHODS TO INHIBIT HISTONE DEACETYLASE (HDAC) ENZYMES

Shomir Ghosh; Mercedes Lobera; Trump, Ryan, Paul; Cunyu Zhang


Archive | 2009

Bicyclic compounds and methods of making and using same

Shomir Ghosh; Gali Golan; Boaz Inbal; Vincent Jacques; Mercedes Lobera


Archive | 2011

Oxadiazole HDAC inhibitors

Erkan Baloglu; Shomir Ghosh; Mercedes Lobera; Darby Schmidt

Collaboration


Dive into the Shomir Ghosh's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Mark S. Sundrud

Boston Children's Hospital

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge