Network


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

Hotspot


Dive into the research topics where Esha Jain is active.

Publication


Featured researches published by Esha Jain.


Cell | 2016

SIRT6 Suppresses Pancreatic Cancer through Control of Lin28b

Sita Kugel; Carlos Sebastian; Julien Fitamant; Kenneth N. Ross; Supriya K. Saha; Esha Jain; Adrianne D. Gladden; Kshitij S. Arora; Yasutaka Kato; Miguel Rivera; Sridhar Ramaswamy; Ruslan I. Sadreyev; Alon Goren; Vikram Deshpande; Nabeel Bardeesy; Raul Mostoslavsky

Chromatin remodeling proteins are frequently dysregulated in human cancer, yet little is known about how they control tumorigenesis. Here, we uncover an epigenetic program mediated by the NAD(+)-dependent histone deacetylase Sirtuin 6 (SIRT6) that is critical for suppression of pancreatic ductal adenocarcinoma (PDAC), one of the most lethal malignancies. SIRT6 inactivation accelerates PDAC progression and metastasis via upregulation of Lin28b, a negative regulator of the let-7 microRNA. SIRT6 loss results in histone hyperacetylation at the Lin28b promoter, Myc recruitment, and pronounced induction of Lin28b and downstream let-7 target genes, HMGA2, IGF2BP1, and IGF2BP3. This epigenetic program defines a distinct subset with a poor prognosis, representing 30%-40% of human PDAC, characterized by reduced SIRT6 expression and an exquisite dependence on Lin28b for tumor growth. Thus, we identify SIRT6 as an important PDAC tumor suppressor and uncover the Lin28b pathway as a potential therapeutic target in a molecularly defined PDAC subset. PAPERCLIP.


Cancer Discovery | 2017

TOX Regulates Growth, DNA Repair, and Genomic Instability in T-cell Acute Lymphoblastic Leukemia

Riadh Lobbardi; Jordan Pinder; Barbara Martinez-Pastor; Marina C. Theodorou; Jessica S. Blackburn; Brian J. Abraham; Yuka Namiki; Marc R. Mansour; Nouran S. Abdelfattah; Aleksey Molodtsov; Gabriela Alexe; Debra Toiber; Manon de Waard; Esha Jain; Myriam Boukhali; Mattia Lion; Deepak Bhere; Khalid Shah; Alejandro Gutierrez; Kimberly Stegmaier; Lewis B. Silverman; Ruslan I. Sadreyev; John M. Asara; Marjorie A. Oettinger; Wilhelm Haas; A. Thomas Look; Richard A. Young; Raul Mostoslavsky; Graham Dellaire; David M. Langenau

T-cell acute lymphoblastic leukemia (T-ALL) is an aggressive malignancy of thymocytes. Using a transgenic screen in zebrafish, thymocyte selection-associated high mobility group box protein (TOX) was uncovered as a collaborating oncogenic driver that accelerated T-ALL onset by expanding the initiating pool of transformed clones and elevating genomic instability. TOX is highly expressed in a majority of human T-ALL and is required for proliferation and continued xenograft growth in mice. Using a wide array of functional analyses, we uncovered that TOX binds directly to KU70/80 and suppresses recruitment of this complex to DNA breaks to inhibit nonhomologous end joining (NHEJ) repair. Impaired NHEJ is well known to cause genomic instability, including development of T-cell malignancies in KU70- and KU80-deficient mice. Collectively, our work has uncovered important roles for TOX in regulating NHEJ by elevating genomic instability during leukemia initiation and sustaining leukemic cell proliferation following transformation.Significance: TOX is an HMG box-containing protein that has important roles in T-ALL initiation and maintenance. TOX inhibits the recruitment of KU70/KU80 to DNA breaks, thereby inhibiting NHEJ repair. Thus, TOX is likely a dominant oncogenic driver in a large fraction of human T-ALL and enhances genomic instability. Cancer Discov; 7(11); 1336-53. ©2017 AACR.This article is highlighted in the In This Issue feature, p. 1201.


Cancer Research | 2017

Abstract 4997: Specialized microRNP and translation mechanisms in quiescent cancer cells

Syed I. A. Bukhari; Samuel S. Truesdell; Sooncheol Lee; Swapna Kollu; Anthony Classon; Myriam Boukhali; Esha Jain; Richard D. Mortensen; Akiko Yanagiya; Ruslan I. Sadreyev; Wilhelm Haas; Shobha Vasudevan

Quiescence (G0) represents an assortment of reversible, cell cycle-arrested states that are resistant to unfavorable conditions and associated with cancer persistence. G0 involves regulated gene expression with selective mRNA expression and decreased canonical translation. Low mTOR activity in G0 activates the cap complex inhibitor, eIF4EBP, and impairs canonical translation. The alternative translation mechanisms in G0 remain to be uncovered. Our data show that microRNAs, regulatory, non-coding RNAs that target distinct mRNAs to alter gene expression, can associate with alternative complexes and translation factors to regulate specific mRNA translation in G0. One subset of transcripts expressed in G0 includes specific mRNAs recruited by an FXR1a-associated microRNP (microRNA-protein complex) for translation activation in G0 mammalian cells. MicroRNPs predominantly mediate repression and downregulation; however, FXR1a-microRNP lacks conventional microRNP repressors, and instead, contains a specific RNA binding protein isoform, FXR1a. FXR1a promotes translation and is overexpressed and associated with poor prognosis in several cancers. Our data reveal that microRNA-mediated activation requires target mRNAs with unadenylated/ shortened poly(A) tails to avoid the roles of PABP in enhancing microRNA-mediated downregulation and in canonical translation that is impaired in G0. Instead of canonical translation factors that are inhibited by eIF4EBP in G0, we find alternative translation factors—a non-canonical 5’cap binding factor and an eIF4G homolog that interacts with the ribosome—are recruited by the 3’-UTR binding FXR1a-microRNP, and promote specific mRNA translation. Our data show that G0 leukemic cells are chemoresistant and their translation profile is similar to surviving leukemic cells that are isolated after clinical therapy. We find expression of critical cytokines and immune regulators in G0. Significantly, inhibiting these immune regulators in resistant G0 cancer cells reduces their survival and chemoresistance. These data reveal a specialized translation mechanism in G0 cancer cells that promotes specific mRNA translation in these conditions of reduced canonical translation, and is important for chemoresistance. Citation Format: Syed I. Bukhari, Samuel S. Truesdell, Sooncheol Lee, Swapna Kollu, Anthony Classon, Myriam Boukhali, Esha Jain, Richard D. Mortensen, Akiko Yanagiya, Ruslan Sadreyev, Wilhelm Haas, Shobha Vasudevan. Specialized microRNP and translation mechanisms in quiescent cancer cells [abstract]. In: Proceedings of the American Association for Cancer Research Annual Meeting 2017; 2017 Apr 1-5; Washington, DC. Philadelphia (PA): AACR; Cancer Res 2017;77(13 Suppl):Abstract nr 4997. doi:10.1158/1538-7445.AM2017-4997


Cancer Research | 2015

Abstract 3583: Thymocyte selection-associated HMG box protein (TOX) induces genomic instability in T-cell acute lymphoblastic leukemia

Riadh Lobbardi; Jordan Pinder; Barbara Martinez-Pastor; Jessica S. Blackburn; Brian J. Abraham; Marc R. Mansour; Nouran S. Abdelfattah; Aleksey Molodtsov; Gabriela Alexe; Debra Toiber; Manon de Waard; Esha Jain; Deepak Bhere; Khalid Shah; Alejandro Gutierrez; Kimberly Stegmaier; Lewis B. Silverman; Ruslan I. Sadreyev; John M. Asara; A. Thomas Look; Richard A. Young; Raul Mostoslavsky; Graham Dellaire; David M. Langenau

Proceedings: AACR 107th Annual Meeting 2016; April 16-20, 2016; New Orleans, LA MYC and NOTCH are major oncogenic drivers in T-cell Acute Lymphoblastic Leukemia (T-ALL), yet additional collaborating genetic lesions collaborate to induce frank malignancy. To identify these factors, a large-scale transgenic screen was completed where 28 amplified and over-expressed genes found in human T-ALL were assessed for accelerating leukemia onset in the zebrafish transgenic model. From this analysis, Thymocyte selection-associated HMG protein (TOX) synergized with both MYC and NOTCH to induce T-ALL. Here, we show that TOX is highly expressed in 95% of human primary and relapse T-ALL when compared with both normal T cells and B-ALL. TOX is highly and specifically expressed in human T-ALL due to both genomic amplification and transcriptional regulation by the master transcription factors MYB/LMO2. Characterization of zebrafish T-ALLs revealed that TOX promoted genomic instability as assessed by changes in DNA content and Whole Genome Sequencing. Effects on genomic instability were confirmed by metaphase spread following TOX expression in MEF cells, confirming roles for TOX in regulating genomic instability and elevated DNA translocation potential in a wider range of cell types. To identify TOX binding partners, Tandem Mass Spectrometry was performed in human T-ALL cells. TOX was found to interact with KU70/KU80 but not other DNA repair enzymes, a result verified by co-immunoprecipitation studies. Given that TOX elevated genomic instability in the zebrafish model, that Ku70 or Ku80 loss lead to genomic instability and T cell lymphoma in mice, and that TOX bound specifically to KU70/KU80 - the initiating factors required for Non-Homologous End Joining (NHEJ) repair - we hypothesized that TOX is a negative regulator of double-strand break repair. Fluorescent repair assays were completed in 3T3 fibroblasts and confirmed that TOX inhibits NHEJ. Dynamic real-time imaging studies showed that TOX suppresses recruitment of fluorescent-tagged KU80 to DNA breaks. Importantly, TOX loss of function increased NHEJ in human T-ALL cells and reduced time to DNA repair as assessed by fluorescent Traffic Light Reporter assays and quantitative assessment of 53BP1 and γH2A.X foci resolution following irradiation. Our results show that TOX is aberrantly re-activated in 95% of human T-ALL, thereby suppressing KU70/KU80 function to promote genomic instability and ultimately elevating rates at which acquired mutations and rearrangements are amassed in developing pre-malignant T cells. Our work shows that TOX is the major oncogenic driver of genomic instability human T-ALL and locks cells in a constant state of dampened repair. Citation Format: Riadh Lobbardi, Jordan Pinder, Barbara Martinez-Pastor, Jessica Blackburn, Brian J. Abraham, Marc Mansour, Nouran S. Abdelfattah, Aleksey Molodtsov, Gabriela Alexe, Debra Toiber, Manon de Waard, Esha Jain, Deepak Bhere, Khalid Shah, Alejandro Gutierrez, Kimberly Stegmaier, Lewis B. Silverman, Ruslan Sadreyev, John Asara, A Thomas Look, Richard A. Young, Raul Mostoslavsky, Graham Dellaire, David M. Langenau. Thymocyte selection-associated HMG box protein (TOX) induces genomic instability in T-cell acute lymphoblastic leukemia. [abstract]. In: Proceedings of the 107th Annual Meeting of the American Association for Cancer Research; 2016 Apr 16-20; New Orleans, LA. Philadelphia (PA): AACR; Cancer Res 2016;76(14 Suppl):Abstract nr 3583.


Cell | 2016

Inhibition of Dihydroorotate Dehydrogenase Overcomes Differentiation Blockade in Acute Myeloid Leukemia

David B. Sykes; Youmna Kfoury; Francois Mercier; Mathias J. Wawer; Jason M. Law; Mark K. Haynes; Tim Lewis; Amir Schajnovitz; Esha Jain; Dongjun Lee; Hanna Meyer; Kerry A. Pierce; Nicola Tolliday; Anna Waller; Steven J. Ferrara; Ashley Eheim; Detlef Stoeckigt; Katrina Maxcy; Julien M Cobert; Jacqueline Bachand; Brian A. Szekely; Siddhartha Mukherjee; Larry A. Sklar; Joanne Kotz; Clary B. Clish; Ruslan I. Sadreyev; Paul A. Clemons; Andreas Janzer; Stuart L. Schreiber; David T. Scadden


Molecular Cell | 2016

A Specialized Mechanism of Translation Mediated by FXR1a-Associated MicroRNP in Cellular Quiescence

Syed I. A. Bukhari; Samuel S. Truesdell; Sooncheol Lee; Swapna Kollu; Anthony Classon; Myriam Boukhali; Esha Jain; Richard D. Mortensen; Akiko Yanagiya; Ruslan I. Sadreyev; Wilhelm Haas; Shobha Vasudevan


Journal of Cell Biology | 2016

Single-cell transcriptional analysis of normal, aberrant, and malignant hematopoiesis in zebrafish

Finola E. Moore; Elaine G. Garcia; Riadh Lobbardi; Esha Jain; Qin Tang; John C. Moore; Mauricio Cortes; Aleksey Molodtsov; Melissa Kasheta; Christina C. Luo; Amaris J. Garcia; Ravi Mylvaganam; Jeffrey A. Yoder; Jessica S. Blackburn; Ruslan I. Sadreyev; Craig J. Ceol; Trista E. North; David M. Langenau


Cancer Research | 2018

Abstract 5371: The Metastatic Breast Cancer Project: Partnering with patients to accelerate progress in cancer research

Nikhil Wagle; Corrie Painter; Elana Anastasio; Michael Dunphy; Mary McGillicuddy; Rachel Stoddard; Esha Jain; Dewey Kim; Simona Di Lascio; Brett N. Tompson; Sara Balch; Beena Thomas; Priti Kumari; Shawn F. Johnson; Jamie Holloway; Ofir Cohen; Erik H. Knelson; Katie Larkin; Sam Pollock; Alicia Wong; Samira Bahl; Simone Maiwald; Andrew Zimmer; Esme O. Baker; Jen Hendry Lapan; Scott Sutherland; Scott Sassone; Viktor A. Adalsteinsson; Eric S. Lander; Todd R. Golub


Cancer Research | 2018

Abstract 5384: The Angiosarcoma Project: Generating the genomic landscape of an exceedingly rare cancer through a nationwide patient-driven initiative

Michael Dunphy; Esha Jain; Elana Anastasio; Mary McGillicuddy; Rachel Stoddard; Beena Thomas; Sara Balch; Kristin Anderka; Katie Larkin; Niall J. Lennon; Yen-Lin Chen; Andrew Zimmer; Esme O. Baker; Simone Maiwald; Jen Hendrey Lapan; Jason L. Hornick; Chandrajit P. Raut; George D. Demetri; Eric S. Lander; Todd R. Golub; Nikhil Wagle; Corrie Painter


Cancer Research | 2016

Abstract B16: Loss of SIRT6 reactivates the RNA-binding protein Lin28b to drive pancreatic cancer

Sita Kugel; Carlos Sebastian; Julien Fitamant; Kenneth N. Ross; Supriya K. Saha; Esha Jain; Adrianne D. Gladden; Kshitij S. Arora; Yasutaka Kato; Miguel Rivera; Sridhar Ramaswamy; Ruslan I. Sadreyev; Alon Goren; Vikram Deshpande; Nabeel Bardeesy; Raul Mostoslavsky

Collaboration


Dive into the Esha Jain'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