Network


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

Hotspot


Dive into the research topics where Mercy Gohil is active.

Publication


Featured researches published by Mercy Gohil.


Nature Immunology | 2013

Natural and inducible TH17 cells are regulated differently by Akt and mTOR pathways

Jiyeon S. Kim; Tammarah Sklarz; Lauren B. Banks; Mercy Gohil; Adam T. Waickman; Nicolas Skuli; Bryan L. Krock; Chong T Luo; Weihong Hu; Kristin N Pollizzi; Ming O. Li; Jeffrey C. Rathmell; Morris J. Birnbaum; Jonathan D. Powell; Martha S. Jordan; Gary A. Koretzky

Natural T helper 17 (nTH17) cells are a population of interleukin 17 (IL-17)-producing cells that acquire effector function in the thymus during development. Here we demonstrate that the serine/threonine kinase Akt plays a critical role in regulating nTH17 cell development. While Akt and the downstream mTORC1–ARNT–HIFα axis were required for inducible TH17 (iTH17) cell generation in the periphery, nTH17 cells developed independently of mTORC1. In contrast, mTORC2 and inhibition of Foxo proteins were critical for nTH17 cell development. Moreover, Akt controlled TH17 subsets through distinct isoforms, as deletion of Akt2, but not Akt1, led to defective iTH17 cell generation. These findings reveal novel mechanisms regulating nTH17 cell development and previously unknown roles of Akt and mTOR in shaping T cell subsets.Natural T helper 17 (nTH17) cells are a population of interleukin 17 (IL-17)-producing cells that acquire effector function in the thymus during development. Here we demonstrate that the serine/threonine kinase Akt has a critical role in regulating nTH17 cell development. Although Akt and the downstream mTORC1–ARNT–HIFα axis were required for generation of inducible TH17 (iTH17) cells, nTH17 cells developed independently of mTORC1. In contrast, mTORC2 and inhibition of Foxo proteins were critical for development of nTH17 cells. Moreover, distinct isoforms of Akt controlled the generation of TH17 cell subsets, as deletion of Akt2, but not of Akt1, led to defective generation of iTH17 cells. These findings define mechanisms regulating nTH17 cell development and reveal previously unknown roles of Akt and mTOR in shaping subsets of T cells.


Blood | 2010

T-cell receptor signals direct the composition and function of the memory CD8+ T-cell pool.

Jennifer E. Smith-Garvin; Jeremy C. Burns; Mercy Gohil; Tao Zou; Jiyeon S. Kim; Jonathan S. Maltzman; E. John Wherry; Gary A. Koretzky; Martha S. Jordan

SH2 domain-containing leukocyte phosphoprotein of 76 kDa (SLP-76) nucleates a signaling complex critical for T-cell receptor (TCR) signal propagation. Mutations in the tyrosines of SLP-76 result in graded defects in TCR-induced signals depending on the tyrosine(s) affected. Here we use 2 strains of genomic knock-in mice expressing tyrosine to phenylalanine mutations to examine the role of TCR signals in the differentiation of effector and memory CD8(+) T cells in response to infection in vivo. Our data support a model in which altered TCR signals can determine the rate of memory versus effector cell differentiation independent of initial T-cell expansion. Furthermore, we show that TCR signals sufficient to promote CD8(+) T-cell differentiation are different from those required to elicit inflammatory cytokine production.


Proceedings of the National Academy of Sciences of the United States of America | 2015

Deregulation of the Hippo pathway in soft-tissue sarcoma promotes FOXM1 expression and tumorigenesis.

T.S. Karin Eisinger-Mathason; Vera Mucaj; Kevin Biju; Michael S. Nakazawa; Mercy Gohil; Timothy P. Cash; Sam S. Yoon; Nicolas Skuli; Kyung Park; Sharon Gerecht; M. Celeste Simon

Significance Soft-tissue sarcomas are aggressive, often lethal tumors, which are understudied. Few therapies beyond standard resection and traditional chemotherapy/radiation are available. Sarcomas are diverse malignancies, including ∼65 distinct histological subtypes. The existence of common mechanisms underlying multiple subtypes has not previously been shown. We demonstrate that the Hippo pathway, an important regulator of cell proliferation, is deregulated in ≥25% of sarcomas, encompassing multiple commonly diagnosed subtypes. When control of the Hippo pathway is lost, expression of the effector protein Yes-Associated Protein (YAP) is stabilized, resulting in higher levels of proliferation. For the first time, to our knowledge, we show that YAP interacts with the forkhead box transcription factor FOXM1 to coregulate critical components of sarcomagenesis, specifically in fibrosarcoma, undifferentiated pleomorphic sarcomas, and liposarcomas. Genetic aberrations responsible for soft-tissue sarcoma formation in adults are largely unknown, with targeted therapies sorely needed for this complex and heterogeneous family of diseases. Here we report that that the Hippo pathway is deregulated in many soft-tissue sarcomas, resulting in elevated expression of the effector molecule Yes-Associated Protein (YAP). Based on data gathered from human sarcoma patients, a novel autochthonous mouse model, and mechanistic analyses, we determined that YAP-dependent expression of the transcription factor forkhead box M1 (FOXM1) is necessary for cell proliferation/tumorigenesis in a subset of soft-tissue sarcomas. Notably, FOXM1 directly interacts with the YAP transcriptional complex via TEAD1, resulting in coregulation of numerous critical pro-proliferation targets that enhance sarcoma progression. Finally, pharmacologic inhibition of FOXM1 decreases tumor size in vivo, making FOXM1 an attractive therapeutic target for the treatment of some sarcoma subtypes.


Nature Medicine | 2018

Determinants of response and resistance to CD19 chimeric antigen receptor (CAR) T cell therapy of chronic lymphocytic leukemia

Joseph A. Fraietta; Simon F. Lacey; Elena Orlando; Iulian Pruteanu-Malinici; Mercy Gohil; Stefan Lundh; Alina C. Boesteanu; Yan Wang; Roddy S. O’Connor; Wei-Ting Hwang; Edward Pequignot; David E Ambrose; Changfeng Zhang; Nicholas Wilcox; Felipe Bedoya; Corin Dorfmeier; Fang Chen; Lifeng Tian; Harit Parakandi; Minnal Gupta; Regina M. Young; F. Brad Johnson; Irina Kulikovskaya; Li Liu; Jun Xu; Sadik Kassim; Megan M. Davis; Bruce L. Levine; Noelle V. Frey; Don L. Siegel

Tolerance to self-antigens prevents the elimination of cancer by the immune system1,2. We used synthetic chimeric antigen receptors (CARs) to overcome immunological tolerance and mediate tumor rejection in patients with chronic lymphocytic leukemia (CLL). Remission was induced in a subset of subjects, but most did not respond. Comprehensive assessment of patient-derived CAR T cells to identify mechanisms of therapeutic success and failure has not been explored. We performed genomic, phenotypic and functional evaluations to identify determinants of response. Transcriptomic profiling revealed that CAR T cells from complete-responding patients with CLL were enriched in memory-related genes, including IL-6/STAT3 signatures, whereas T cells from nonresponders upregulated programs involved in effector differentiation, glycolysis, exhaustion and apoptosis. Sustained remission was associated with an elevated frequency of CD27+CD45RO–CD8+ T cells before CAR T cell generation, and these lymphocytes possessed memory-like characteristics. Highly functional CAR T cells from patients produced STAT3-related cytokines, and serum IL-6 correlated with CAR T cell expansion. IL-6/STAT3 blockade diminished CAR T cell proliferation. Furthermore, a mechanistically relevant population of CD27+PD-1–CD8+ CAR T cells expressing high levels of the IL-6 receptor predicts therapeutic response and is responsible for tumor control. These findings uncover new features of CAR T cell biology and underscore the potential of using pretreatment biomarkers of response to advance immunotherapies.An IL-6/STAT3 signature and memory CD8 T cell subset in preinfusion chimeric antigen receptor–expressing T cells associate with response in patients with high-risk chronic lymphocytic leukemia.


Blood | 2015

The aryl hydrocarbon receptor nuclear translocator is an essential regulator of murine hematopoietic stem cell viability.

Bryan L. Krock; Tzipora S. Eisinger-Mathason; Dionysios N. Giannoukos; Jessica E.S. Shay; Mercy Gohil; David S.M. Lee; Michael S. Nakazawa; Julie Sesen; Nicolas Skuli; M. Celeste Simon

Hypoxia-inducible factors (HIFs) are master regulators of the transcriptional response to low oxygen and play essential roles in embryonic development, tissue homeostasis, and disease. Recent studies have demonstrated that hematopoietic stem cells (HSCs) within the bone marrow localize to a hypoxic niche and that HIF-1α promotes HSC adaptation to stress. Because the related factor HIF-2α is also expressed in HSCs, the combined role of HIF-1α and HIF-2α in HSC maintenance is unclear. To this end, we have conditionally deleted the HIF-α dimerization partner, the aryl hydrocarbon receptor nuclear translocator (ARNT) in the hematopoietic system to ablate activity of both HIF-1α and HIF-2α and assessed the functional consequence of ARNT deficiency on fetal liver and adult hematopoiesis. We determined that ARNT is essential for adult and fetal HSC viability and homeostasis. Importantly, conditional knockout of both Hif-1α and Hif-2α phenocopied key aspects of these HSC phenotypes, demonstrating that the impact of Arnt deletion is primarily HIF dependent. ARNT-deficient long-term HSCs underwent apoptosis, potentially because of reduced B-cell lymphoma 2 (BCL-2) and vascular endothelial growth factor A (VEGF-A) expression. Our results suggest that HIF activity may regulate HSC homeostasis through these prosurvival factors.


Nature | 2018

Disruption of TET2 promotes the therapeutic efficacy of CD19-targeted T cells

Joseph A. Fraietta; Christopher L. Nobles; Morgan A. Sammons; Stefan Lundh; Shannon A. Carty; Tyler J. Reich; Alexandria P. Cogdill; Jennifer J.D. Morrissette; Jamie E. DeNizio; Shantan Reddy; Young Hwang; Mercy Gohil; Irina Kulikovskaya; Farzana Nazimuddin; Minnal Gupta; Fang Chen; John K. Everett; Katherine A. Alexander; Enrique Lin-Shiao; Marvin H. Gee; Xiaojun Liu; Regina M. Young; David E Ambrose; Yan Wang; Jun Xu; Martha S. Jordan; Katherine T. Marcucci; Bruce L. Levine; K. Christopher Garcia; Yangbing Zhao

Cancer immunotherapy based on genetically redirecting T cells has been used successfully to treat B cell malignancies1–3. In this strategy, the T cell genome is modified by integration of viral vectors or transposons encoding chimaeric antigen receptors (CARs) that direct tumour cell killing. However, this approach is often limited by the extent of expansion and persistence of CAR T cells4,5. Here we report mechanistic insights from studies of a patient with chronic lymphocytic leukaemia treated with CAR T cells targeting the CD19 protein. Following infusion of CAR T cells, anti-tumour activity was evident in the peripheral blood, lymph nodes and bone marrow; this activity was accompanied by complete remission. Unexpectedly, at the peak of the response, 94% of CAR T cells originated from a single clone in which lentiviral vector-mediated insertion of the CAR transgene disrupted the methylcytosine dioxygenase TET2 gene. Further analysis revealed a hypomorphic mutation in this patient’s second TET2 allele. TET2-disrupted CAR T cells exhibited an epigenetic profile consistent with altered T cell differentiation and, at the peak of expansion, displayed a central memory phenotype. Experimental knockdown of TET2 recapitulated the potency-enhancing effect of TET2 dysfunction in this patient’s CAR T cells. These findings suggest that the progeny of a single CAR T cell induced leukaemia remission and that TET2 modification may be useful for improving immunotherapies.Genetically engineered T cells that induced remission in a patient with chronic lymphocytic leukaemia were found to have disruption of the TET2 gene, which caused T cell changes that potentiated their anti-tumour effects.


Journal of Immunology | 2018

The Loss of TET2 Promotes CD8+ T Cell Memory Differentiation

Shannon A. Carty; Mercy Gohil; Lauren B. Banks; Renee M. Cotton; Matthew E. Johnson; Erietta Stelekati; Andrew D. Wells; E. John Wherry; Gary A. Koretzky; Martha S. Jordan

T cell differentiation requires appropriate regulation of DNA methylation. In this article, we demonstrate that the methylcytosine dioxygenase ten-eleven translocation (TET)2 regulates CD8+ T cell differentiation. In a murine model of acute viral infection, TET2 loss promotes early acquisition of a memory CD8+ T cell fate in a cell-intrinsic manner without disrupting Ag-driven cell expansion or effector function. Upon secondary recall, TET2-deficient memory CD8+ T cells demonstrate superior pathogen control. Genome-wide methylation analysis identified a number of differentially methylated regions in TET2-deficient versus wild-type CD8+ T cells. These differentially methylated regions did not occur at the loci of differentially expressed memory markers; rather, several hypermethylated regions were identified in known transcriptional regulators of CD8+ T cell memory fate. Together, these data demonstrate that TET2 is an important regulator of CD8+ T cell fate decisions.


Journal of Immunology | 2011

The SLP-76 Src Homology 2 Domain Is Required for T Cell Development and Activation

Jeremy C. Burns; Evann Corbo; Janine Degen; Mercy Gohil; Christine Anterasian; Burkart Schraven; Gary A. Koretzky; Stefanie Kliche; Martha S. Jordan

The adapter protein Src homology 2 (SH2) domain-containing leukocyte protein of 76 kDa (SLP-76) is critical for multiple aspects of T cell development and function. Through its protein-binding domains, SLP-76 serves as a platform for the assembly of multiple enzymes and adapter proteins that function together to activate second messengers required for TCR signal propagation. The N terminus of SLP-76, which contains three tyrosines that serve as docking sites for SH2 domain-containing proteins, and the central proline-rich region of SLP-76 have been well studied and are known to be important for both thymocyte selection and activation of peripheral T cells. Less is known about the function of the C-terminal SH2 domain of SLP-76. This region inducibly associates with ADAP and HPK1. Combining regulated deletion of endogenous SLP-76 with transgenic expression of a SLP-76 SH2 domain mutant, we demonstrate that the SLP-76 SH2 domain is required for peripheral T cell activation and positive selection of thymocytes, a function not previously attributed to this region. This domain is also important for T cell proliferation, IL-2 production, and phosphorylation of protein kinase D and IκB. ADAP-deficient T cells display similar, but in some cases less severe, defects despite phosphorylation of a negative regulatory site on SLP-76 by HPK1, a function that is lost in SLP-76 SH2 domain mutant T cells.


European Journal of Immunology | 2017

mTORC2 regulates multiple aspects of NKT-cell development and function

Tammarah Sklarz; Peng Guan; Mercy Gohil; Renee M. Cotton; Moyar Q. Ge; Angela Haczku; Rupali Das; Martha S. Jordan

Invariant NKT (iNKT) cells bridge innate and adaptive immunity by rapidly secreting cytokines and lysing targets following TCR recognition of lipid antigens. Based on their ability to secrete IFN‐γ, IL‐4 and IL‐17A, iNKT‐cells are classified as NKT‐1, NKT‐2, and NKT‐17 subsets, respectively. The molecular pathways regulating iNKT‐cell fate are not fully defined. Recent studies implicate Rictor, a required component of mTORC2, in the development of select iNKT‐cell subsets, however these reports are conflicting. To resolve these questions, we used Rictorfl/fl CD4cre+ mice and found that Rictor is required for NKT‐17 cell development and normal iNKT‐cell cytolytic function. Conversely, Rictor is not absolutely required for IL‐4 and IFN‐γ production as peripheral iNKT‐cells make copious amounts of these cytokines. Overall iNKT‐cell numbers are dramatically reduced in the absence of Rictor. We provide data indicating Rictor regulates cell survival as well as proliferation of developing and mature iNKT‐cells. Thus, mTORC2 regulates multiple aspects of iNKT‐cell development and function.


Cancer Research | 2015

Abstract 960: Deregulation of the Hippo pathway in soft tissue sarcoma promotes FOXM1 expression and tumorigenesis

T.S. Karin Eisinger; Vera Mucaj; Kevin Biju; Michael S. Nakazawa; Mercy Gohil; Timothy P. Cash; Sam S. Yoon; Nicolas Skuli; Kyung Park; Sharon Gerecht; Celeste Simon

The genetic aberrations responsible for soft tissue sarcoma formation in adults are largely unknown, and targeted therapies are sorely needed for this complex and heterogeneous family of diseases. Here we report that that the Hippo pathway is deregulated in many soft tissue sarcomas, resulting in elevated expression of the effector molecule YAP. Based on data gathered from human sarcoma patients, we concluded that more than 40% of human sarcomas have lost one or more copies of upstream HIPPO pathway regulators including SAV1, LATS2, and NF2. Loss of these molecules deactivates the HIPPO pathway, permitting uncontrolled proliferation and growth. Expression of active YAP is the direct result of this copy number loss and we have found YAP to be elevated in primary human fibrosarscomas, liposarcomas, and pleomorphic tumors. Pharmacologic inhibition of YAP significantly impaired primary sarcomagenesis in a murine allograft model. Using a novel autochthonous mouse model and mechanistic analyses in human and murine sarcoma cells, we determined that YAP-dependent expression of the transcription factor, FOXM1, is necessary for sarcoma cell proliferation and tumorigenesis. FOXM1 expression is elevated in multiple human soft tissue sarcoma subtypes. Notably, FOXM1 directly interacts with the YAP transcriptional complex via TEAD1, resulting in co-regulation of numerous critical pro-proliferation targets that enhance sarcoma progression. Finally, pharmacologic inhibition of FOXM1 decreases tumor size in vivo, making FOXM1 an attractive therapeutic target for the treatment of sarcomas. Citation Format: T.S. Karin Eisinger, Vera Mucaj, Kevin Biju, Michael Nakazawa, Mercy Gohil, Timothy Cash, Sam Yoon, Nicolas Skuli, Kyung Min park, Sharon Gerecht, Celeste Simon. Deregulation of the Hippo pathway in soft tissue sarcoma promotes FOXM1 expression and tumorigenesis. [abstract]. In: Proceedings of the 106th Annual Meeting of the American Association for Cancer Research; 2015 Apr 18-22; Philadelphia, PA. Philadelphia (PA): AACR; Cancer Res 2015;75(15 Suppl):Abstract nr 960. doi:10.1158/1538-7445.AM2015-960

Collaboration


Dive into the Mercy Gohil's collaboration.

Top Co-Authors

Avatar

Martha S. Jordan

University of Pennsylvania

View shared research outputs
Top Co-Authors

Avatar

Gary A. Koretzky

University of Pennsylvania

View shared research outputs
Top Co-Authors

Avatar

E. John Wherry

University of Pennsylvania

View shared research outputs
Top Co-Authors

Avatar

Lauren B. Banks

University of Pennsylvania

View shared research outputs
Top Co-Authors

Avatar

Shannon A. Carty

University of Pennsylvania

View shared research outputs
Top Co-Authors

Avatar

Bruce L. Levine

University of Pennsylvania

View shared research outputs
Top Co-Authors

Avatar

Bryan L. Krock

University of Pennsylvania

View shared research outputs
Top Co-Authors

Avatar

David E Ambrose

University of Pennsylvania

View shared research outputs
Top Co-Authors

Avatar

Erietta Stelekati

University of Pennsylvania

View shared research outputs
Top Co-Authors

Avatar

Fang Chen

University of Pennsylvania

View shared research outputs
Researchain Logo
Decentralizing Knowledge