Network


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

Hotspot


Dive into the research topics where Abhishek A. Chakraborty is active.

Publication


Featured researches published by Abhishek A. Chakraborty.


Nature | 2016

On-target efficacy of a HIF-2α antagonist in preclinical kidney cancer models

Hyejin Cho; Xinlin Du; James P. Rizzi; Ella Liberzon; Abhishek A. Chakraborty; Wenhua Gao; Ingrid Carvo; Sabina Signoretti; Richard K. Bruick; John A. Josey; Eli M. Wallace; William G. Kaelin

Clear cell renal cell carcinoma, the most common form of kidney cancer, is usually linked to inactivation of the pVHL tumour suppressor protein and consequent accumulation of the HIF-2α transcription factor (also known as EPAS1). Here we show that a small molecule (PT2399) that directly inhibits HIF-2α causes tumour regression in preclinical mouse models of primary and metastatic pVHL-defective clear cell renal cell carcinoma in an on-target fashion. pVHL-defective clear cell renal cell carcinoma cell lines display unexpectedly variable sensitivity to PT2399, however, suggesting the need for predictive biomarkers to be developed to use this approach optimally in the clinic.


Genes & Development | 2014

Prolyl hydroxylation by EglN2 destabilizes FOXO3a by blocking its interaction with the USP9x deubiquitinase

Xingnan Zheng; Bo Zhai; Peppi Koivunen; Sandra J. Shin; Gang Lu; Jiayun Liu; Christoph Geisen; Abhishek A. Chakraborty; Javid Moslehi; David M. Smalley; Xin Wei; Xian Chen; Zhengming Chen; Justine M. Beres; Jing Zhang; Jen Lan Tsao; Mitchell C. Brenner; Yuqing Zhang; Cheng Fan; Ronald A. DePinho; Ji Hye Paik; Steven P. Gygi; William G. Kaelin; Qing Zhang

The three EglN prolyl hydroxylases (EglN1, EglN2, and EglN3) regulate the stability of the HIF transcription factor. We recently showed that loss of EglN2, however, also leads to down-regulation of Cyclin D1 and decreased cell proliferation in a HIF-independent manner. Here we report that EglN2 can hydroxylate FOXO3a on two specific prolyl residues in vitro and in vivo. Hydroxylation of these sites prevents the binding of USP9x deubiquitinase, thereby promoting the proteasomal degradation of FOXO3a. FOXO transcription factors can repress Cyclin D1 transcription. Failure to hydroxylate FOXO3a promotes its accumulation in cells, which in turn suppresses Cyclin D1 expression. These findings provide new insights into post-transcriptional control of FOXO3a and provide a new avenue for pharmacologically altering Cyclin D1 activity.


Cell | 2016

EGLN1 Inhibition and Rerouting of α-Ketoglutarate Suffice for Remote Ischemic Protection

Benjamin A. Olenchock; Javid Moslehi; Alan H. Baik; Shawn M. Davidson; Jeremy Williams; William J. Gibson; Abhishek A. Chakraborty; Kerry A. Pierce; Christine M. Miller; Eric A. Hanse; Ameeta Kelekar; Lucas B. Sullivan; Amy J. Wagers; Clary B. Clish; Matthew G. Vander Heiden; William G. Kaelin

Ischemic preconditioning is the phenomenon whereby brief periods of sublethal ischemia protect against a subsequent, more prolonged, ischemic insult. In remote ischemic preconditioning (RIPC), ischemia to one organ protects others organs at a distance. We created mouse models to ask if inhibition of the alpha-ketoglutarate (αKG)-dependent dioxygenase Egln1, which senses oxygen and regulates the hypoxia-inducible factor (HIF) transcription factor, could suffice to mediate local and remote ischemic preconditioning. Using somatic gene deletion and a pharmacological inhibitor, we found that inhibiting Egln1 systemically or in skeletal muscles protects mice against myocardial ischemia-reperfusion (I/R) injury. Parabiosis experiments confirmed that RIPC in this latter model was mediated by a secreted factor. Egln1 loss causes accumulation of circulating αKG, which drives hepatic production and secretion of kynurenic acid (KYNA) that is necessary and sufficient to mediate cardiac ischemic protection in this setting.


Science | 2016

pVHL suppresses kinase activity of Akt in a proline-hydroxylation–dependent manner

Jianping Guo; Abhishek A. Chakraborty; Pengda Liu; Wenjian Gan; Xingnan Zheng; Hiroyuki Inuzuka; Bin Wang; Jinfang Zhang; Linli Zhang; Min Yuan; Jesse Novak; Jin Q. Cheng; Alex Toker; Sabina Signoretti; Qing Zhang; John M. Asara; William G. Kaelin; Wenyi Wei

Activation of the serine-threonine kinase Akt promotes the survival and proliferation of various cancers. Hypoxia promotes the resistance of tumor cells to specific therapies. We therefore explored a possible link between hypoxia and Akt activity. We found that Akt was prolyl-hydroxylated by the oxygen-dependent hydroxylase EglN1. The von Hippel–Lindau protein (pVHL) bound directly to hydroxylated Akt and inhibited Akt activity. In cells lacking oxygen or functional pVHL, Akt was activated to promote cell survival and tumorigenesis. We also identified cancer-associated Akt mutations that impair Akt hydroxylation and subsequent recognition by pVHL, thus leading to Akt hyperactivation. Our results show that microenvironmental changes, such as hypoxia, can affect tumor behaviors by altering Akt activation, which has a critical role in tumor growth and therapeutic resistance.


Proceedings of the National Academy of Sciences of the United States of America | 2008

Adenovirus E1A targets p400 to induce the cellular oncoprotein Myc

Kathryn A Tworkowski; Abhishek A. Chakraborty; Andrew V. Samuelson; Yvette R. Seger; Masako Narita; Gregory J. Hannon; Scott W. Lowe; William P. Tansey

Adenovirus E1A drives oncogenesis by targeting key regulatory pathways that are critical for cellular growth control. The interaction of E1A with p400 is essential for many E1A activities, but the downstream target of this interaction is unknown. Here, we present evidence that the oncoprotein transcription factor Myc is the target of this interaction. We show that E1A stabilizes Myc protein via p400 and promotes the coassociation of Myc and p400 at Myc target genes, leading to their transcriptional induction. We also show that E1A requires Myc for its ability to activate Myc-dependent gene expression and induce apoptosis, and that forced expression of Myc is sufficient to rescue the activity of an E1A-mutant defective in p400 binding. Together, these findings establish that Myc, via p400, is an essential downstream target of E1A.


Cancer Research | 2009

Adenoviral E1A Function through Myc

Abhishek A. Chakraborty; William P. Tansey

The study of DNA tumor viruses has been invaluable in uncovering the cellular nodes and pathways that contribute to oncogenesis. Perhaps one of the best-studied oncoproteins encoded by a DNA tumor virus is adenovirus E1A, which modifies the function of key regulatory proteins such as retinoblastoma (Rb) and the chromatin remodeling protein p400. Although the interaction of E1A with Rb has long been known to target regulation of the E2F transcription factors, the downstream target of the E1A-p400 interaction has remained elusive. We have recently reported that a critical downstream link of the E1A-p400 nexus is the oncoprotein transcription factor c-Myc. Through its interaction with p400, E1A stabilizes Myc and promotes formation of Myc-p400 complexes on chromatin, leading to activation of Myc target genes. These findings point to an important role for p400 in Myc function and reveal that E1A drives oncogenesis by tapping into two important transcriptional networks: those of E2F and Myc.


Experimental Cell Research | 2009

Inference of cell cycle-dependent proteolysis by laser scanning cytometry.

Abhishek A. Chakraborty; William P. Tansey

Mechanisms that couple protein turnover to cell cycle progression are critical for coordinating the events of cell duplication and division. Despite the importance of cell cycle-regulated proteolysis, however, technologies to measure this phenomenon are limited, and typically involve monitoring cells that are released back into the cell cycle after synchronization. We describe here the use of laser scanning cytometry (LSC), a technical merger between fluorescence microscopy and flow cytometry, to determine cell cycle-dependent changes in protein stability in unperturbed, asynchronous, cultures of mammalian cells. In this method, the ability of the LSC to accurately measure whole cell fluorescence is employed, together with RNA fluorescence in situ hybridization and immunofluorescence, to relate abundance of a particular RNA and protein in a cell to its point at the cell cycle. Parallel monitoring of RNA and protein levels is used, together with protein synthesis inhibitors, to reveal cell cycle-specific changes in protein turnover. We demonstrate the viability of this method by analyzing the proteolysis of two prominent human oncoproteins, Myc and Cyclin E, and argue that this LSC-based approach offers several practical advantages over traditional cell synchronization methods.


Oncogene | 2015

A common functional consequence of tumor-derived mutations within c-MYC

Abhishek A. Chakraborty; Claudio Scuoppo; Soumyadeep Dey; Lance R. Thomas; Shelly L. Lorey; Scott W. Lowe; William P. Tansey

The relevance of changes to the coding sequence of the c-MYC oncogene to malignancy is controversial. Overexpression of a pristine form of MYC is observed in many cancers and is sufficient to drive tumorigenesis in most contexts. Yet missense changes to MYC are found in ~50% of Burkitt’s lymphomas, aggregate within an amino-terminal degron important for proteasomal destruction of MYC, and where examined profoundly enhance the tumorigenic properties of MYC in vitro and in vivo. Much of the controversy surrounding these mutants stems from the limited number of mutations that have been evaluated and their clustering within a single region of the MYC protein; the highly-conserved Myc box I (MbI) element. Here, by analysis of extant genomic data sets, we identify a previously unrecognized hotspot for tumor-associated MYC mutations, located in a conserved central portion of the protein. We show that, despite their distal location in MYC, mutations in this region precisely phenocopy those in MbI in terms of stability, in vitro transformation, growth-promoting properties, in vivo tumorigenesis and ability to escape p53-dependent tumor surveillance mechanisms. The striking parallels between the behavior of tumor-derived mutations in disparate regions of the MYC protein reveals that a common molecular process is disrupted by these mutations, implying an active role for these mutations in tumorigenesis and suggesting that different therapeutic strategies may be needed for treatment of lymphomas expressing wild type versus mutant forms of MYC protein.


Science Translational Medicine | 2017

HIF activation causes synthetic lethality between the VHL tumor suppressor and the EZH1 histone methyltransferase.

Abhishek A. Chakraborty; Eijiro Nakamura; Jun Qi; Amanda Creech; Jacob D. Jaffe; Joshiawa Paulk; Jesse Novak; Kshithija Nagulapalli; Samuel K. McBrayer; Glenn S. Cowley; Javier Pineda; Jiaxi Song; Yaoyu E. Wang; Steven A. Carr; David E. Root; Sabina Signoretti; James Bradner; William G. Kaelin

Dysregulated H3K27 demethylase activity in pVHL-deficient cells increases dependence on the EZH1 H3K27 methyltransferase. Renal cancer’s loss is patients’ gain Clear cell renal cell carcinoma is the most common form of kidney cancer, and it is typically linked to the loss of von Hippel–Lindau protein (pVHL), a tumor suppressor. This loss results in activation of genes that are normally induced by hypoxia, including some histone demethylases. Chakraborty et al. discovered that increased H3K27 demethylase activity renders pVHL-deficient cells hyperdependent on an opposing H3K27 methyltransferase, EZH1, for survival. Inhibition of EZH1 is lethal for these cells. Inactivation of the von Hippel–Lindau tumor suppressor protein (pVHL) is the signature lesion in the most common form of kidney cancer, clear cell renal cell carcinoma (ccRCC). pVHL loss causes the transcriptional activation of hypoxia-inducible factor (HIF) target genes, including many genes that encode histone lysine demethylases. Moreover, chromatin regulators are frequently mutated in this disease. We found that ccRCC displays increased H3K27 acetylation and a shift toward mono- or unmethylated H3K27 caused by an HIF-dependent increase in H3K27 demethylase activity. Using a focused short hairpin RNA library, as well as CRISPR (clustered regularly interspaced short palindromic repeats)/Cas9 (CRISPR-associated protein 9) and a pharmacological inhibitor, we discovered that pVHL-defective ccRCC cells are hyperdependent on the H3K27 methyltransferase EZH1 for survival. Therefore, targeting EZH1 could be therapeutically useful in ccRCC.


Cell | 2016

Erratum: EGLN1 Inhibition and Rerouting of α-Ketoglutarate Suffice for Remote Ischemic Protection (Cell (2016) 164 (884-895))

Benjamin A. Olenchock; Javid Moslehi; Alan H. Baik; Shawn M. Davidson; Jeremy Williams; William J. Gibson; Abhishek A. Chakraborty; Kerry A. Pierce; Christine M. Miller; Eric Hanse; Ameeta Kelekar; Lucas B. Sullivan; Amy J. Wagers; Clary B. Clish; Matthew G. Vander Heiden; William G. Kaelin

Due to an oversight in the preparation of this article, the authors inadvertently neglected to include Abhishek A. Chakraborty in the author list. He has been added to the author list, and his contributions are now noted in the Author Contributions section of the article online.

Collaboration


Dive into the Abhishek A. Chakraborty's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Javid Moslehi

Vanderbilt University Medical Center

View shared research outputs
Top Co-Authors

Avatar

Matthew G. Vander Heiden

Massachusetts Institute of Technology

View shared research outputs
Top Co-Authors

Avatar

Sabina Signoretti

Brigham and Women's Hospital

View shared research outputs
Top Co-Authors

Avatar

Alan H. Baik

University of California

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge