Network


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

Hotspot


Dive into the research topics where Subhradip Karmakar is active.

Publication


Featured researches published by Subhradip Karmakar.


Genome Research | 2012

ChIP-seq guidelines and practices of the ENCODE and modENCODE consortia

Stephen G. Landt; Georgi K. Marinov; Anshul Kundaje; Pouya Kheradpour; Florencia Pauli; Serafim Batzoglou; Bradley E. Bernstein; Peter J. Bickel; James B. Brown; Philip Cayting; Yiwen Chen; Gilberto DeSalvo; Charles B. Epstein; Katherine I. Fisher-Aylor; Ghia Euskirchen; Mark Gerstein; Jason Gertz; Alexander J. Hartemink; Michael M. Hoffman; Vishwanath R. Iyer; Youngsook L. Jung; Subhradip Karmakar; Manolis Kellis; Peter V. Kharchenko; Qunhua Li; Tao Liu; X. Shirley Liu; Lijia Ma; Aleksandar Milosavljevic; Richard M. Myers

Chromatin immunoprecipitation (ChIP) followed by high-throughput DNA sequencing (ChIP-seq) has become a valuable and widely used approach for mapping the genomic location of transcription-factor binding and histone modifications in living cells. Despite its widespread use, there are considerable differences in how these experiments are conducted, how the results are scored and evaluated for quality, and how the data and metadata are archived for public use. These practices affect the quality and utility of any global ChIP experiment. Through our experience in performing ChIP-seq experiments, the ENCODE and modENCODE consortia have developed a set of working standards and guidelines for ChIP experiments that are updated routinely. The current guidelines address antibody validation, experimental replication, sequencing depth, data and metadata reporting, and data quality assessment. We discuss how ChIP quality, assessed in these ways, affects different uses of ChIP-seq data. All data sets used in the analysis have been deposited for public viewing and downloading at the ENCODE (http://encodeproject.org/ENCODE/) and modENCODE (http://www.modencode.org/) portals.


Blood | 2013

CUX1 is a haploinsufficient tumor suppressor gene on chromosome 7 frequently inactivated in acute myeloid leukemia.

Megan E. McNerney; Christopher D. Brown; Xiaoyue Wang; Elizabeth Bartom; Subhradip Karmakar; Chaitanya Bandlamudi; Shan Yu; Jinkyung Ko; Barry P. Sandall; Thomas Stricker; John Anastasi; Robert L. Grossman; John M. Cunningham; Michelle M. Le Beau; Kevin P. White

Loss of chromosome 7 and del(7q) [-7/del(7q)] are recurring cytogenetic abnormalities in hematologic malignancies, including acute myeloid leukemia and therapy-related myeloid neoplasms, and associated with an adverse prognosis. Despite intensive effort by many laboratories, the putative myeloid tumor suppressor(s) on chromosome 7 has not yet been identified.We performed transcriptome sequencing and SNP array analysis on de novo and therapy-related myeloid neoplasms, half with -7/del(7q). We identified a 2.17-Mb commonly deleted segment on chromosome band 7q22.1 containing CUX1, a gene encoding a homeodomain-containing transcription factor. In 1 case, CUX1 was disrupted by a translocation, resulting in a loss-of-function RNA fusion transcript. CUX1 was the most significantly differentially expressed gene within the commonly deleted segment and was expressed at haploinsufficient levels in -7/del(7q) leukemias. Haploinsufficiency of the highly conserved ortholog, cut, led to hemocyte overgrowth and tumor formation in Drosophila melanogaster. Similarly, haploinsufficiency of CUX1 gave human hematopoietic cells a significant engraftment advantage on transplantation into immunodeficient mice. Within the RNA-sequencing data, we identified a CUX1-associated cell cycle transcriptional gene signature, suggesting that CUX1 exerts tumor suppressor activity by regulating proliferative genes. These data identify CUX1 as a conserved, haploinsufficient tumor suppressor frequently deleted in myeloid neoplasms.


Cancer Cell | 2014

SPOP Promotes Tumorigenesis by Acting as a Key Regulatory Hub in Kidney Cancer

Guoqiang Li; Weimin Ci; Subhradip Karmakar; Ke Chen; Ruby Dhar; Zhixiang Fan; Zhongqiang Guo; Jing Zhang; Yuwen Ke; Lu Wang; Min Zhuang; Shengdi Hu; Xuesong Li; Liqun Zhou; Xianghong Li; Matthew F. Calabrese; Edmond R. Watson; Sandip M. Prasad; Carrie W. Rinker-Schaeffer; Thomas Stricker; Yong Tian; Brenda A. Schulman; Jiang Liu; Kevin P. White

Hypoxic stress and hypoxia-inducible factors (HIFs) play important roles in a wide range of tumors. We demonstrate that SPOP, which encodes an E3 ubiquitin ligase component, is a direct transcriptional target of HIFs in clear cell renal cell carcinoma (ccRCC). Furthermore, hypoxia results in cytoplasmic accumulation of SPOP, which is sufficient to induce tumorigenesis. This tumorigenic activity occurs through the ubiquitination and degradation of multiple regulators of cellular proliferation and apoptosis, including the tumor suppressor PTEN, ERK phosphatases, the proapoptotic molecule Daxx, and the Hedgehog pathway transcription factor Gli2. Knockdown of SPOP specifically kills ccRCC cells, indicating that it may be a promising therapeutic target. Collectively, our results indicate that SPOP serves as a regulatory hub to promote ccRCC tumorigenesis.


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

Reduced DOCK4 expression leads to erythroid dysplasia in myelodysplastic syndromes.

Sriram Sundaravel; Ryan Duggan; Tushar D. Bhagat; David Ebenezer; Hui Liu; Yiting Yu; Matthias Bartenstein; Madhu Unnikrishnan; Subhradip Karmakar; Ting Chun Liu; Ingrid Torregroza; Thomas Quenon; John Anastasi; Kathy L. McGraw; Andrea Pellagatti; Jacqueline Boultwood; Vijay Yajnik; Andrew S. Artz; Michelle M. Le Beau; Ulrich Steidl; Alan F. List; Todd Evans; Amit Verma; Amittha Wickrema

Significance Anemia is the predominant clinical manifestation of myelodysplastic syndromes (MDS). Genes that are aberrantly expressed and/or mutated that lead to the dysplastic erythroid morphology seen in −7/del(7q) MDS have not been identified. In this study, we show that reduced expression of dedicator of cytokinesis 4 (DOCK4) causes dysplasia by disrupting the actin cytoskeleton in developing red blood cells. In addition, our identification of the molecular pathway that leads to morphological defects in this type of MDS provides potential therapeutic targets downstream of DOCK4 that can be exploited to reverse the dysplasia in the erythroid lineage. Furthermore, we developed a novel single-cell multispectral flow cytometry assay for evaluation of disrupted F-actin filaments, which can be used for potential early detection of dysplastic cells in MDS. Anemia is the predominant clinical manifestation of myelodysplastic syndromes (MDS). Loss or deletion of chromosome 7 is commonly seen in MDS and leads to a poor prognosis. However, the identity of functionally relevant, dysplasia-causing, genes on 7q remains unclear. Dedicator of cytokinesis 4 (DOCK4) is a GTPase exchange factor, and its gene maps to the commonly deleted 7q region. We demonstrate that DOCK4 is underexpressed in MDS bone marrow samples and that the reduced expression is associated with decreased overall survival in patients. We show that depletion of DOCK4 levels leads to erythroid cells with dysplastic morphology both in vivo and in vitro. We established a novel single-cell assay to quantify disrupted F-actin filament network in erythroblasts and demonstrate that reduced expression of DOCK4 leads to disruption of the actin filaments, resulting in erythroid dysplasia that phenocopies the red blood cell (RBC) defects seen in samples from MDS patients. Reexpression of DOCK4 in −7q MDS patient erythroblasts resulted in significant erythropoietic improvements. Mechanisms underlying F-actin disruption revealed that DOCK4 knockdown reduces ras-related C3 botulinum toxin substrate 1 (RAC1) GTPase activation, leading to increased phosphorylation of the actin-stabilizing protein ADDUCIN in MDS samples. These data identify DOCK4 as a putative 7q gene whose reduced expression can lead to erythroid dysplasia.


PLOS ONE | 2013

Regulation of Gγ-Globin Gene by ATF2 and Its Associated Proteins through the cAMP-Response Element

Li Liu; Subhradip Karmakar; Ruby Dhar; Milind Mahajan; Alina Choudhury; Sherman M. Weissman; Betty S. Pace

The upstream Gγ-globin cAMP-response element (G-CRE) plays an important role in regulating Gγ-globin expression through binding of ATF2 and its DNA-binding partners defined in this study. ATF2 knockdown resulted in a significant reduction of γ-globin expression accompanied by decreased ATF2 binding to the G-CRE. By contrast, stable ATF2 expression in K562 cells increased γ-globin transcription which was reduced by ATF2 knockdown. Moreover, a similar effect of ATF2 on γ-globin expression was observed in primary erythroid progenitors. To understand the role of ATF2 in γ-globin expression, chromatographically purified G-CRE/ATF2-interacting proteins were subjected to mass spectrometry analysis; major binding partners included CREB1, cJun, Brg1, and histone deacetylases among others. Immunoprecipitation assays demonstrated interaction of these proteins with ATF2 and in vivo GCRE binding in CD34+ cells undergoing erythroid differentiation which was correlated with γ-globin expression during development. These results suggest synergism between developmental stage-specific recruitments of the ATF2 protein complex and expression of γ-globin during erythropoiesis. Microarray studies in K562 cells support ATF2 plays diverse roles in hematopoiesis and chromatin remodeling.


Leukemia & Lymphoma | 2018

Expression of functional folate receptors in multiple myeloma

Ying Zhou; Kenji Unno; Elizabeth Hyjek; Hui Liu; Todd M. Zimmerman; Subhradip Karmakar; Karson S. Putt; Jiayin Shen; Philip S. Low; Amittha Wickrema

Abstract Receptor-targeted delivery of imaging and therapeutic agents has emerged as an attractive strategy to diagnosis and treat many diseases including cancer. One of the most well-studied receptors for targeted therapies is the folate receptor (FR) family. FR-α and FR-β are present on many cancers with little expression in normal tissues; leading to the testing of at least six folate-targeted drugs in human clinical trials for various cancers. However, the expression of FR in blood cancers has not been fully explored with no reports of FR expression in myelomas. Herein, we report the expression of both FR-α and FR-β on CD138 + plasma cells isolated from patients with multiple myeloma. In addition, all-trans retinoic acid was shown to increase the levels of FR-α and FR-β in two myeloma cell lines. Altogether, this data suggests that folate-targeted therapies for the treatment of multiple myeloma warrants further investigation.


Single Cell Biology | 2014

Cell Based Immunotherapy: As a Promising Futuristic Solution forEffective Cancer Therapy

Subhradip Karmakar

Cell based therapy is rapidly emerging as an alternative to conventional chemotherapy based treatment for cancer. While chemotherapy, radiation therapy and surgical intervention have been the mainstay treatment regime for vast majority of cancers, they are often associated with unwanted treatment related side-effects. Quite often the side effects dramatically compromise the quality of life for the patients. Therapy related AML (t-AML) is one such consequence of mutation events associated with cytotoxic drugs. In order to make cancer therapy more tumor specific thus sparing the non-cancerous components of the body, cell based therapy harnessing the body immune system (cancer immunotherapy) is now explored in ever increasing number. This review is focused on the emergence of novel technologies that aim in engineering T lymphocytes to selectively recognize cancer specific markers on transformed cells thus facilitating their elimination without harming the normal cells of the body. Clinical data shows an enormous potential of this approach and is currently followed in several institutions worldwide against wide varieties of cancers with almost complete remission and fewer side effects.


Cancer Research | 2012

Abstract LB-411: RNA sequencing reveals CUX1 to be a conserved tumor suppressor in acute myeloid leukemia associated with loss or deletion of chromosome 7

Megan E. McNerney; Christopher D. Brown; Xiaoyue Wang; Subhradip Karmakar; Chaitanya Bandlamudi; Shan Yu; John Anastasi; Michelle M. Le Beau; Kevin P. White

Loss of chromosome 7 and del(7q) [-7/del(7q)] are recurring cytogenetic abnormalities in acute myeloid leukemia (AML), occurring in 9% of de novo AML and 50% of therapy-related myeloid neoplasms (t-MN). Abnormalities of chromosome 7 carry an adverse prognosis. Despite intensive analysis by many laboratories, and the identification of multiple minimally deleted regions on 7q, the putative tumor suppressor(s) on chromosome 7 is currently unknown. We performed transcriptome sequencing on 22 samples and SNP array analysis on 35 samples from patients with myeloid neoplasms, half of which contain -7/del(7q). Copy number analysis identified a 2.17 Mb commonly deleted segment on chromosome band 7q22.1 containing CUX1, a gene encoding a homeodomain-containing transcription factor normally expressed highly in hematopoietic stem cells. CUX1 was found to be disrupted in a translocation resulting in a chimeric RNA fusion transcript in one patient. We found the CUX1 transcript and protein to be expressed at haploinsufficent levels in -7/del(7q) leukemias. To test the tumor suppressor activity of CUX1, the drosophila homologue, CUT, was knocked-down in Drosophila melanogaster hemocytes, which led to significantly increased numbers of hemocytes and melanotic tumor formation. Restoration of CUX1 expression in a human AML cell line with -7 decreased proliferation. These data indicate that CUX1 is a conserved, haploinsufficient, tumor suppressor that promotes leukemogenesis in -7/del(7q) AML. Citation Format: {Authors}. {Abstract title} [abstract]. In: Proceedings of the 103rd Annual Meeting of the American Association for Cancer Research; 2012 Mar 31-Apr 4; Chicago, IL. Philadelphia (PA): AACR; Cancer Res 2012;72(8 Suppl):Abstract nr LB-411. doi:1538-7445.AM2012-LB-411


The Journal of Urology | 2012

145 SPECKLE-TYPE POZ PROTEIN CYTOPLASMIC MISLOCALIZATION AND OVEREXPRESSION PROMOTE TUMOR GROWTH IN AN ORTHOTOPIC MURINE RENAL CELL CANCER MODEL

Sandip M. Prasad; Subhradip Karmakar; Yi Cai; Donald VanderGriend; Kevin P. White

Collaboration


Dive into the Subhradip Karmakar'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

Hui Liu

University of Chicago

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Ruby Dhar

University of Chicago

View shared research outputs
Top Co-Authors

Avatar

Sandip M. Prasad

Medical University of South Carolina

View shared research outputs
Researchain Logo
Decentralizing Knowledge