Network


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

Hotspot


Dive into the research topics where Vinee Purohit is active.

Publication


Featured researches published by Vinee Purohit.


Proceedings of the National Academy of Sciences of the United States of America | 2012

MUC1 mucin stabilizes and activates hypoxia-inducible factor 1 alpha to regulate metabolism in pancreatic cancer

Nina V. Chaika; Teklab Gebregiworgis; Michelle E. Lewallen; Vinee Purohit; Prakash Radhakrishnan; Xiang Liu; Bo Zhang; Kamiya Mehla; Roger B. Brown; Thomas C. Caffrey; Fang Yu; Keith R. Johnson; Robert Powers; Michael A. Hollingsworth; Pankaj K. Singh

Aberrant glucose metabolism is one of the hallmarks of cancer that facilitates cancer cell survival and proliferation. Here, we demonstrate that MUC1, a large, type I transmembrane protein that is overexpressed in several carcinomas including pancreatic adenocarcinoma, modulates cancer cell metabolism to facilitate growth properties of cancer cells. MUC1 occupies the promoter elements of multiple genes directly involved in glucose metabolism and regulates their expression. Furthermore, MUC1 expression enhances glycolytic activity in pancreatic cancer cells. We also demonstrate that MUC1 expression enhances in vivo glucose uptake and expression of genes involved in glucose uptake and metabolism in orthotopic implantation models of pancreatic cancer. The MUC1 cytoplasmic tail is known to activate multiple signaling pathways through its interactions with several transcription factors/coregulators at the promoter elements of various genes. Our results indicate that MUC1 acts as a modulator of the hypoxic response in pancreatic cancer cells by regulating the expression/stability and activity of hypoxia-inducible factor-1α (HIF-1α). MUC1 physically interacts with HIF-1α and p300 and stabilizes the former at the protein level. By using a ChIP assay, we demonstrate that MUC1 facilitates recruitment of HIF-1α and p300 on glycolytic gene promoters in a hypoxia-dependent manner. Also, by metabolomic studies, we demonstrate that MUC1 regulates multiple metabolite intermediates in the glucose and amino acid metabolic pathways. Thus, our studies indicate that MUC1 acts as a master regulator of the metabolic program and facilitates metabolic alterations in the hypoxic environments that help tumor cells survive and proliferate under such conditions.


Cancer Letters | 2012

Graviola: A novel promising natural-derived drug that inhibits tumorigenicity and metastasis of pancreatic cancer cells in vitro and in vivo through altering cell metabolism

María P. Torres; Satyanarayana Rachagani; Vinee Purohit; Poomy Pandey; Suhasini Joshi; Erik D. Moore; Sonny L. Johansson; Pankaj K. Singh; Apar Kishor Ganti; Surinder K. Batra

Pancreatic tumors are resistant to conventional chemotherapies. The present study was aimed at evaluating the potential of a novel plant-derived product as a therapeutic agent for pancreatic cancer (PC). The effects of an extract from the tropical tree Annona Muricata, commonly known as Graviola, was evaluated for cytotoxicity, cell metabolism, cancer-associated protein/gene expression, tumorigenicity, and metastatic properties of PC cells. Our experiments revealed that Graviola induced necrosis of PC cells by inhibiting cellular metabolism. The expression of molecules related to hypoxia and glycolysis in PC cells (i.e. HIF-1α, NF-κB, GLUT1, GLUT4, HKII, and LDHA) were downregulated in the presence of the extract. In vitro functional assays further confirmed the inhibition of tumorigenic properties of PC cells. Overall, the compounds that are naturally present in a Graviola extract inhibited multiple signaling pathways that regulate metabolism, cell cycle, survival, and metastatic properties in PC cells. Collectively, alterations in these parameters led to a decrease in tumorigenicity and metastasis of orthotopically implanted pancreatic tumors, indicating promising characteristics of the natural product against this lethal disease.


Cancer and Metabolism | 2014

Metabolic reprogramming induced by ketone bodies diminishes pancreatic cancer cachexia

Surendra K. Shukla; Teklab Gebregiworgis; Vinee Purohit; Nina V. Chaika; Venugopal Gunda; Prakash Radhakrishnan; Kamiya Mehla; Iraklis I. Pipinos; Robert Powers; Fang Yu; Pankaj K. Singh

BackgroundAberrant energy metabolism is a hallmark of cancer. To fulfill the increased energy requirements, tumor cells secrete cytokines/factors inducing muscle and fat degradation in cancer patients, a condition known as cancer cachexia. It accounts for nearly 20% of all cancer-related deaths. However, the mechanistic basis of cancer cachexia and therapies targeting cancer cachexia thus far remain elusive. A ketogenic diet, a high-fat and low-carbohydrate diet that elevates circulating levels of ketone bodies (i.e., acetoacetate, β-hydroxybutyrate, and acetone), serves as an alternative energy source. It has also been proposed that a ketogenic diet leads to systemic metabolic changes. Keeping in view the significant role of metabolic alterations in cancer, we hypothesized that a ketogenic diet may diminish glycolytic flux in tumor cells to alleviate cachexia syndrome and, hence, may provide an efficient therapeutic strategy.ResultsWe observed reduced glycolytic flux in tumor cells upon treatment with ketone bodies. Ketone bodies also diminished glutamine uptake, overall ATP content, and survival in multiple pancreatic cancer cell lines, while inducing apoptosis. A decrease in levels of c-Myc, a metabolic master regulator, and its recruitment on glycolytic gene promoters, was in part responsible for the metabolic phenotype in tumor cells. Ketone body-induced intracellular metabolomic reprogramming in pancreatic cancer cells also leads to a significantly diminished cachexia in cell line models. Our mouse orthotopic xenograft models further confirmed the effect of a ketogenic diet in diminishing tumor growth and cachexia.ConclusionsThus, our studies demonstrate that the cachectic phenotype is in part due to metabolic alterations in tumor cells, which can be reverted by a ketogenic diet, causing reduced tumor growth and inhibition of muscle and body weight loss.


PLOS ONE | 2012

Differential Expression of Metabolic Genes in Tumor and Stromal Components of Primary and Metastatic Loci in Pancreatic Adenocarcinoma

Nina V. Chaika; Fang Yu; Vinee Purohit; Kamiya Mehla; Audrey J. Lazenby; Dominick J. DiMaio; Judy M. Anderson; Jen Jen Yeh; Keith R. Johnson; Michael A. Hollingsworth; Pankaj K. Singh

Background Pancreatic cancer is the fourth leading cause of cancer related deaths in the United States with a five-year survival rate of 6%. It is characterized by extremely aggressive tumor growth rate and high incidence of metastasis. One of the most common and profound biochemical phenotypes of animal and human cancer cells is their ability to metabolize glucose at high rates, even under aerobic conditions. However, the contribution of metabolic interrelationships between tumor cells and cells of the surrounding microenvironment to the progression of cancer is not well understood. We evaluated differential expression of metabolic genes and, hence, metabolic pathways in primary tumor and metastases of patients with pancreatic adenocarcinoma. Methods and Findings We analyzed the metabolic gene (those involved in glycolysis, tri-carboxylic acid pathway, pentose-phosphate pathway and fatty acid metabolism) expression profiles of primary and metastatic lesions from pancreatic cancer patients by gene expression arrays. We observed two principal results: genes that were upregulated in primary and most of the metastatic lesions; and genes that were upregulated only in specific metastatic lesions in a site-specific manner. Immunohistochemical (IHC) analyses of several metabolic gene products confirmed the gene expression patterns at the protein level. The IHC analyses also revealed differential tumor and stromal expression patterns of metabolic enzymes that were correlated with the metastasis sites. Conclusions Here, we present the first comprehensive studies that establish differential metabolic status of tumor and stromal components and elevation of aerobic glycolysis gene expression in pancreatic cancer.


Oncotarget | 2015

Active YAP promotes pancreatic cancer cell motility, invasion and tumorigenesis in a mitotic phosphorylation-dependent manner through LPAR3

Shuping Yang; Lin Zhang; Vinee Purohit; Surendra K. Shukla; Xingcheng Chen; Fang Yu; Kai Fu; Yuanhong Chen; Joyce C. Solheim; Pankaj K. Singh; Wei Song; Jixin Dong

The transcriptional co-activator Yes-associated protein, YAP, is a main effector in the Hippo tumor suppressor pathway. We recently defined a mechanism for positive regulation of YAP through CDK1-mediated mitotic phosphorylation. Here, we show that active YAP promotes pancreatic cancer cell migration, invasion and anchorage-independent growth in a mitotic phosphorylation-dependent manner. Mitotic phosphorylation is essential for YAP-driven tumorigenesis in animals. YAP reduction significantly impairs cell migration and invasion. Immunohistochemistry shows significant upregulation and nuclear localization of YAP in metastases when compared with primary tumors and normal tissue in human. Mitotic phosphorylation of YAP controls a unique transcriptional program in pancreatic cells. Expression profiles reveal LPAR3 (lysophosphatidic acid receptor 3) as a mediator for mitotic phosphorylation-driven pancreatic cell motility and invasion. Together, this work identifies YAP as a novel regulator of pancreatic cancer cell motility, invasion and metastasis, and as a potential therapeutic target for invasive pancreatic cancer.


Cancer Cell | 2017

MUC1 and HIF-1alpha Signaling Crosstalk Induces Anabolic Glucose Metabolism to Impart Gemcitabine Resistance to Pancreatic Cancer.

Surendra K. Shukla; Vinee Purohit; Kamiya Mehla; Venugopal Gunda; Nina V. Chaika; Enza Vernucci; Ryan J. King; Jaime Abrego; Gennifer Goode; Aneesha Dasgupta; Alysha L. Illies; Teklab Gebregiworgis; Bingbing Dai; Jithesh J. Augustine; Divya Murthy; Kuldeep S. Attri; Oksana Mashadova; Paul M. Grandgenett; Robert Powers; Quan P. Ly; Audrey J. Lazenby; Jean L. Grem; Fang Yu; José M. Matés; John M. Asara; Jung Whan Kim; Jordan Hankins; Colin D. Weekes; Michael A. Hollingsworth; Natalie J. Serkova

Poor response to cancer therapy due to resistance remains a clinical challenge. The present study establishes a widely prevalent mechanism of resistance to gemcitabine in pancreatic cancer, whereby increased glycolytic flux leads to glucose addiction in cancer cells and a corresponding increase in pyrimidine biosynthesis to enhance the intrinsic levels of deoxycytidine triphosphate (dCTP). Increased levels of dCTP diminish the effective levels of gemcitabine through molecular competition. We also demonstrate that MUC1-regulated stabilization of hypoxia inducible factor-1α (HIF-1α) mediates such metabolic reprogramming. Targeting HIF-1α or de novo pyrimidine biosynthesis, in combination with gemcitabine, strongly diminishes tumor burden. Finally, reduced expression of TKT and CTPS, which regulate flux into pyrimidine biosynthesis, correlates with better prognosis in pancreatic cancer patients on fluoropyrimidine analogs.


PLOS ONE | 2017

MUC1 facilitates metabolomic reprogramming in triple-negative breast cancer

Gennifer Goode; Venugopal Gunda; Nina V. Chaika; Vinee Purohit; Fang Yu; Pankaj K. Singh

Background Mucin1 (MUC1), a glycoprotein associated with chemoresistance and an aggressive cancer phenotype, is aberrantly overexpressed in triple-negative breast cancer (TNBC). Recent studies suggest that MUC1 plays a role in modulating cancer cell metabolism and thereby supports tumor growth. Herein, we examined the role of MUC1 in metabolic reprogramming in TNBC. Methods MUC1 was stably overexpressed in MDA-MB-231 TNBC cells and stably knocked down in MDA-MB-468 cells. We performed liquid chromatography-coupled tandem mass spectrometry-assisted metabolomic analyses and physiological assays, which indicated significant alterations in the metabolism of TNBC cells due to MUC1 expression. Results Differential analyses identified significant differences in metabolic pathways implicated in cancer cell growth. In particular, MUC1 expression altered glutamine dependency of the cells, which can be attributed in part to the changes in the expression of genes that regulate glutamine metabolism, as observed by real-time PCR analysis. Furthermore, MUC1 expression altered the sensitivity of cells to transaminase inhibitor aminooxyacetate (AOA), potentially by altering glutamine metabolism. Conclusions Collectively, these results suggest that MUC1 serves as a metabolic regulator in TNBC, facilitating the metabolic reprogramming of glutamine utilization that influences TNBC tumor growth.


Journal of Proteome Research | 2017

Glucose Limitation Alters Glutamine Metabolism in MUC1-Overexpressing Pancreatic Cancer Cells

Teklab Gebregiworgis; Vinee Purohit; Surendra K. Shukla; Saber Tadros; Nina V. Chaika; Jaime Abrego; Scott E. Mulder; Venugopal Gunda; Pankaj K. Singh; Robert Powers

Pancreatic cancer cells overexpressing Mucin 1 (MUC1) rely on aerobic glycolysis and, correspondingly, are dependent on glucose for survival. Our NMR metabolomics comparative analysis of control (S2–013.Neo) and MUC1-overexpressing (S2–013.MUC1) cells demonstrates that MUC1 reprograms glutamine metabolism upon glucose limitation. The observed alteration in glutamine metabolism under glucose limitation was accompanied by a relative decrease in the proliferation of MUC1-overexpressing cells compared with steady-state conditions. Moreover, glucose limitation induces G1 phase arrest where S2–013.MUC1 cells fail to enter S phase and synthesize DNA because of a significant disruption in pyrimidine nucleotide biosynthesis. Our metabolomics analysis indicates that glutamine is the major source of oxaloacetate in S2–013.Neo and S2–013.MUC1 cells, where oxaloacetate is converted to aspartate, an important metabolite for pyrimidine nucleotide biosynthesis. However, glucose limitation impedes the flow of glutamine carbons into the pyrimidine nucleotide rings and instead leads to a significant accumulation of glutamine-derived aspartate in S2–013.MUC1 cells.


Cancer Cell | 2017

Erratum: MUC1 and HIF-1alpha Signaling Crosstalk Induces Anabolic Glucose Metabolism to Impart Gemcitabine Resistance to Pancreatic Cancer (Cancer Cell (2017) 32(1) (71–87.e7) (S1535610817302544) (10.1016/j.ccell.2017.06.004))

Surendra K. Shukla; Vinee Purohit; Kamiya Mehla; Venugopal Gunda; Nina V. Chaika; Enza Vernucci; Ryan J. King; Jaime Abrego; Gennifer Goode; Aneesha Dasgupta; Alysha L. Illies; Teklab Gebregiworgis; Bingbing Dai; Jithesh J. Augustine; Divya Murthy; Kuldeep S. Attri; Oksana Mashadova; Paul M. Grandgenett; Robert Powers; Quan P. Ly; Audrey J. Lazenby; Jean L. Grem; Fang Yu; José M. Matés; John M. Asara; Jung Whan Kim; Jordan Hankins; Colin D. Weekes; Michael A. Hollingsworth; Natalie J. Serkova

Surendra K. Shukla, Vinee Purohit, Kamiya Mehla, Venugopal Gunda, Nina V. Chaika, Enza Vernucci, Ryan J. King, Jaime Abrego, Gennifer D. Goode, Aneesha Dasgupta, Alysha L. Illies, Teklab Gebregiworgis, Bingbing Dai, Jithesh J. Augustine, Divya Murthy, Kuldeep S. Attri, Oksana Mashadova, Paul M. Grandgenett, Robert Powers, Quan P. Ly, Audrey J. Lazenby, Jean L. Grem, Fang Yu, José M. Matés, John M. Asara, Jung-whan Kim, Jordan H. Hankins, Colin Weekes, Michael A. Hollingsworth, Natalie J. Serkova, Aaron R. Sasson, Jason B. Fleming, Jennifer M. Oliveto, Costas A. Lyssiotis, Lewis C. Cantley, Lyudmyla Berim, and Pankaj K. Singh* *Correspondence: [email protected] http://dx.doi.org/10.1016/j.ccell.2017.08.008


PLOS ONE | 2017

Correction: MUC1 facilitates metabolomic reprogramming in triple-negative breast cancer

Gennifer Goode; Venugopal Gunda; Nina V. Chaika; Vinee Purohit; Fang Yu; Pankaj K. Singh

[This corrects the article DOI: 10.1371/journal.pone.0176820.].

Collaboration


Dive into the Vinee Purohit's collaboration.

Top Co-Authors

Avatar

Pankaj K. Singh

University of Nebraska Medical Center

View shared research outputs
Top Co-Authors

Avatar

Nina V. Chaika

University of Nebraska Medical Center

View shared research outputs
Top Co-Authors

Avatar

Fang Yu

University of Nebraska Medical Center

View shared research outputs
Top Co-Authors

Avatar

Robert Powers

University of Nebraska–Lincoln

View shared research outputs
Top Co-Authors

Avatar

Teklab Gebregiworgis

University of Nebraska–Lincoln

View shared research outputs
Top Co-Authors

Avatar

Venugopal Gunda

University of Nebraska Medical Center

View shared research outputs
Top Co-Authors

Avatar

Kamiya Mehla

University of Nebraska Medical Center

View shared research outputs
Top Co-Authors

Avatar

Michael A. Hollingsworth

University of Nebraska Medical Center

View shared research outputs
Top Co-Authors

Avatar

Surendra K. Shukla

University of Nebraska Medical Center

View shared research outputs
Top Co-Authors

Avatar

Keith R. Johnson

Eppley Institute for Research in Cancer and Allied Diseases

View shared research outputs
Researchain Logo
Decentralizing Knowledge