Network


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

Hotspot


Dive into the research topics where Shailendra Kumar Dhar Dwivedi is active.

Publication


Featured researches published by Shailendra Kumar Dhar Dwivedi.


Genes and Diseases | 2015

Bmi-1: At the crossroads of physiological and pathological biology

Resham Bhattacharya; Soumyajit Banerjee Mustafi; Mark Street; Anindya Dey; Shailendra Kumar Dhar Dwivedi

Bmi-1 is a member of the Polycomb repressor complex 1 that mediates gene silencing by regulating chromatin structure and is indispensable for self-renewal of both normal and cancer stem cells. Despite three decades of research that have elucidated the transcriptional regulation, post-translational modifications and functions of Bmi-1 in regulating the DNA damage response, cellular bioenergetics, and pathologies, the entire potential of a protein with such varied functions remains to be realized. This review attempts to synthesize the current knowledge on Bmi-1 with an emphasis on its role in both normal physiology and cancer. Additionally, since cancer stem cells are emerging as a new paradigm for therapy resistance, the role of Bmi-1 in this perspective is also highlighted. The wide spectrum of malignancies that implicate Bmi-1 as a signature for stemness and oncogenesis also make it a suitable candidate for therapy. Nonetheless, new approaches are vitally needed to further characterize physiological roles of Bmi-1 with the long-term goal of using Bmi-1 as a prognostic marker and a therapeutic target.


The FASEB Journal | 2016

Cystathionine β-synthase regulates endothelial function via protein S-sulfhydration

Sounik Saha; Prabir K. Chakraborty; Xunhao Xiong; Shailendra Kumar Dhar Dwivedi; Soumyajit Banerjee Mustafi; Noah R. Leigh; Ramani Ramchandran; Priyabrata Mukherjee; Resham Bhattacharya

Deficiencies of the human cystathionine β‐synthase (CBS) enzyme are characterized by a plethora of vascular disorders and hyperhomocysteinemia. However, several clinical trials demonstrated that despite reduction in homocysteine levels, disease outcome remained unaffected, thus the mechanism of endothelial dysfunction is poorly defined. Here, we show that the loss of CBS function in endothelial cells (ECs) leads to a significant down‐regulation of cellular hydrogen sulfide (H2S) by 50% and of glutathione (GSH) by 40%. Silencing CBS in ECs compromised phenotypic and signaling responses to the VEGF that were potentiated by decreased transcription of VEGF receptor (VEGFR)‐2 and neuropilin (NRP)‐1, the primary receptors regulating endothelial function. Transcriptional down‐regulation of VEGFR‐2 and NRP‐1 was mediated by a lack in stability of the transcription factor specificity protein 1 (Spl), which is a sulfhydration target of H2S at residues Cys68 and Cys755. Reinstating H2S but not GSH in CBS‐silenced ECs restored Sp1 levels and its binding to the VEGFR‐2 promoter and VEGFR‐2, NRP‐1 expression, VEGF‐dependent proliferation, and migration phenotypes. Thus, our study emphasizes the importance of CBS‐mediated protein S‐sulfhydration in maintaining vascular health and function.—Saha, S., Chakraborty, P. K., Xiong, X., Dwivedi, S. K. D., Mustafi, S. B., Leigh, N.R., Ramchandran, R., Mukherjee, P., Bhattacharya, R. Cystathionine β‐synthase regulates endothelial function via protein S‐sulfhydration. FASEB J. 30, 441‐456 (2016). www.fasebj.org


Oncotarget | 2016

Therapeutic evaluation of microRNA-15a and microRNA-16 in ovarian cancer

Shailendra Kumar Dhar Dwivedi; Soumyajit Banerjee Mustafi; Lingegowda S. Mangala; Dahai Jiang; Sunila Pradeep; Cristian Rodriguez-Aguayo; Hui Ling; Cristina Ivan; Priyabrata Mukherjee; George A. Calin; Gabriel Lopez-Berestein; Anil K. Sood; Resham Bhattacharya

Treatment of chemo-resistant ovarian cancer (OvCa) remains clinically challenging and there is a pressing need to identify novel therapeutic strategies. Here we report that multiple mechanisms that promote OvCa progression and chemo-resistance could be inhibited by ectopic expression of miR-15a and miR-16. Significant correlations between low expression of miR-16, high expression of BMI1 and shortened overall survival (OS) were noted in high grade serous (HGS) OvCa patients upon analysis of The Cancer Genome Atlas (TCGA). Targeting BMI1, in vitro with either microRNA reduced clonal growth of OvCa cells. Additionally, epithelial to mesenchymal transition (EMT) as well as expression of the cisplatin transporter ATP7B were inhibited by miR-15a and miR-16 resulting in decreased degradation of the extra-cellular matrix and enhanced sensitization of OvCa cells to cisplatin. Nanoliposomal delivery of the miR-15a and miR-16 combination, in a pre-clinical chemo-resistant orthotopic mouse model of OvCa, demonstrated striking reduction in tumor burden compared to cisplatin alone. Thus, with the advent of miR replacement therapy some of which are in Phase 2 clinical trials, miR-15a and miR-16 represent novel ammunition in the anti-OvCa arsenal.


ACS Nano | 2016

Gold Nanoparticle Reprograms Pancreatic Tumor Microenvironment and Inhibits Tumor Growth

Sounik Saha; Xunhao Xiong; Prabir K. Chakraborty; Khader Shameer; Rochelle R. Arvizo; Rachel A. Kudgus; Shailendra Kumar Dhar Dwivedi; Nazir Hossen; Elizabeth Gillies; J. David Robertson; Joel T. Dudley; Raul Urrutia; Russell G. Postier; Resham Bhattacharya; Priyabrata Mukherjee

Altered tumor microenvironment (TME) arising from a bidirectional crosstalk between the pancreatic cancer cells (PCCs) and the pancreatic stellate cells (PSCs) is implicated in the dismal prognosis in pancreatic ductal adenocarcinoma (PDAC), yet effective strategies to disrupt the crosstalk is lacking. Here, we demonstrate that gold nanoparticles (AuNPs) inhibit proliferation and migration of both PCCs and PSCs by disrupting the bidirectional communication via alteration of the cell secretome. Analyzing the key proteins identified from a functional network of AuNP-altered secretome in PCCs and PSCs, we demonstrate that AuNPs impair secretions of major hub node proteins in both cell types and transform activated PSCs toward a lipid-rich quiescent phenotype. By reducing activation of PSCs, AuNPs inhibit matrix deposition, enhance angiogenesis, and inhibit tumor growth in an orthotopic co-implantation model in vivo. Auto- and heteroregulations of secretory growth factors/cytokines are disrupted by AuNPs resulting in reprogramming of the TME. By utilizing a kinase dead mutant of IRE1-α, we demonstrate that AuNPs alter the cellular secretome through the ER-stress-regulated IRE1-dependent decay pathway (RIDD) and identify endostatin and matrix metalloproteinase 9 as putative RIDD targets. Thus, AuNPs could potentially be utilized as a tool to effectively interrogate bidirectional communications in the tumor microenvironment, reprogram it, and inhibit tumor growth by its therapeutic function.


Biochimica et Biophysica Acta | 2016

MDR1 mediated chemoresistance: BMI1 and TIP60 in action.

Soumyajit Banerjee Mustafi; Prabir K. Chakraborty; Sarwat Naz; Shailendra Kumar Dhar Dwivedi; Mark Street; Rumki Basak; Da Yang; Kai Ding; Priyabrata Mukherjee; Resham Bhattacharya

Chemotherapy-induced emergence of drug resistant cells is frequently observed and is exemplified by the expression of family of drug resistance proteins including, multidrug resistance protein 1 (MDR1). However, a concise mechanism for chemotherapy-induced MDR1 expression is unclear. Mechanistically, mutational selection, epigenetic alteration, activation of the Wnt pathway or impaired p53 function have been implicated. The present study describes that the surviving fraction of cisplatin resistant cells co- upregulate MDR1, BMI1 and acetyl transferase activity of TIP60. Using complementary gain and loss of function approaches, we demonstrate that the expression of MDR1 is positively regulated by BMI1, a stem-cell factor classically known as a transcriptional repressor. Our study establishes a functional interaction between TIP60 and BMI-1 resulting in upregulation of MDR1 expression. Chromatin immunoprecipitation (ChIP) assays further establish that the proximal MDR1 promoter responds to cisplatin in a BMI1 dependent manner. BMI1 interacts with a cluster of E-box elements on the MDR1 promoter and recruits TIP60 resulting in acetylation of histone H2A and H3. Collectively, our data establish a hitherto unknown liaison among MDR1, BMI1 and TIP60 and provide mechanistic insights into cisplatin-induced MDR1 expression resulting in acquired cross-resistance against paclitaxel, doxorubicin and likely other drugs. In conclusion, our results advocate utilizing anti-BMI1 strategies to alleviate acquired resistance to chemotherapy.


Autophagy | 2016

Inhibition of BMI1 induces autophagy-mediated necroptosis

Anindya Dey; Soumyajit Banerjee Mustafi; Sounik Saha; Shailendra Kumar Dhar Dwivedi; Priyabrata Mukherjee; Resham Bhattacharya

ABSTRACT The clonal self-renewal property conferred by BMI1 is instrumental in maintenance of not only normal stem cells but also cancer-initiating cells from several different malignancies that represent a major challenge to chemotherapy. Realizing the immense pathological significance, PTC-209, a small molecule inhibitor of BMI1 transcription has recently been described. While targeting BMI1 in various systems significantly decreases clonal growth, the mechanisms differ, are context-dependent, and somewhat unclear. We report here that genetic or pharmacological inhibition of BMI1 significantly impacts clonal growth without altering CDKN2A/INK4/ARF or CCNG2 and induces autophagy in ovarian cancer (OvCa) cells through ATP depletion. While autophagy can promote survival or induce cell death, targeting BMI1 engages the PINK1-PARK2-dependent mitochondrial pathway and induces a novel mode of nonapoptotic, necroptosis-mediated cell death. In OvCa, necroptosis is potentiated by activation of the RIPK1-RIPK3 complex that phosphorylates its downstream substrate, MLKL. Importantly, genetic or pharmacological inhibitors of autophagy or RIPK3 rescue clonal growth in BMI1 depleted cells. Thus, we have established a novel molecular link between BMI1, clonal growth, autophagy and necroptosis. In chemoresistant OvCa where apoptotic pathways are frequently impaired, necroptotic cell death modalities provide an important alternate strategy that leverage overexpression of BMI1.


Nature Communications | 2017

MICU1 drives glycolysis and chemoresistance in ovarian cancer.

Prabir K. Chakraborty; Soumyajit Banerjee Mustafi; Xunhao Xiong; Shailendra Kumar Dhar Dwivedi; Vasyl Nesin; Sounik Saha; Min Zhang; Danny N. Dhanasekaran; Muralidharan Jayaraman; Robert S. Mannel; Kathleen N. Moore; Scott McMeekin; Da Yang; Rosemary E. Zuna; Kai Ding; Leonidas Tsiokas; Resham Bhattacharya; Priyabrata Mukherjee

Cancer cells actively promote aerobic glycolysis to sustain their metabolic requirements through mechanisms not always clear. Here, we demonstrate that the gatekeeper of mitochondrial Ca2+ uptake, Mitochondrial Calcium Uptake 1 (MICU1/CBARA1) drives aerobic glycolysis in ovarian cancer. We show that MICU1 is overexpressed in a panel of ovarian cancer cell lines and that MICU1 overexpression correlates with poor overall survival (OS). Silencing MICU1 in vitro increases oxygen consumption, decreases lactate production, inhibits clonal growth, migration and invasion of ovarian cancer cells, whereas silencing in vivo inhibits tumour growth, increases cisplatin efficacy and OS. Mechanistically, silencing MICU1 activates pyruvate dehydrogenase (PDH) by stimulating the PDPhosphatase-phosphoPDH-PDH axis. Forced-expression of MICU1 in normal cells phenocopies the metabolic aberrations of malignant cells. Consistent with the in vitro and in vivo findings we observe a significant correlation between MICU1 and pPDH (inactive form of PDH) expression with poor prognosis. Thus, MICU1 could serve as an important therapeutic target to normalize metabolic aberrations responsible for poor prognosis in ovarian cancer.


Molecular Cancer | 2017

BMI1, a new target of CK2α

Soumyajit Banerjee Mustafi; Prabir K. Chakraborty; Shailendra Kumar Dhar Dwivedi; Kai Ding; Katherine Moxley; Priyabrata Mukherjee; Resham Bhattacharya

BackgroundThe polycomb group protein, BMI1 plays important roles in chromatin modification, stem cell function, DNA damage repair and mitochondrial bioenergetics. Such diverse cellular functions of BMI1 could be, in part, due to post-translational modifications, especially phosphorylation. To date, AKT has been reported as a kinase that by site specific phosphorylation of BMI1 modulates its oncogenic functions.MethodsImmunoprecipitation in conjunction with kinase assay and mass spectrometry was used to determine association with and site specific phosphorylation of BMI1 by CK2α. Functional implications of the BMI1/CK2α axis was examined in cancer cells utilizing siRNA and exogenous gene expression followed by biochemical and phenotypic studies. Correlations between expression of CK2α and BMI1 were determined from cell lines and formalin fixed paraffin embedded tissues representing the normal fallopian tube epithelium and high grade serous ovarian cancer samples.ResultsHere we report that CK2α, a nuclear serine threonine kinase, phosphorylates BMI1 at Serine 110 as determined by in-vitro/ex-vivo kinase assay and mass spectrometry. In ovarian cancer cell lines, expression of CK2α correlated with the phospho-species, as well as basal BMI1 levels. Preventing phosphorylation of BMI1 at Serine 110 significantly decreased half-life and stability of the protein. Additionally, re-expression of the phosphorylatable but not non-phosphorylatable BMI1 rescued clonal growth in endogenous BMI1 silenced cancer cells leading us to speculate that CK2α-mediated phosphorylation stabilizes BMI1 and promotes its oncogenic function. Clinically, compared to normal fallopian tube epithelial tissues, the expression of both BMI1 and CK2α were significantly higher in tumor tissues obtained from high-grade serous ovarian cancer patients. Among tumor samples, the expression of BMI1 and CK2α positively correlated (Spearman coefficient = 0.62, P = 0.0021) with each other.ConclusionTaken together, our findings establish an important regulatory role of CK2α on BMI1 phosphorylation and stability and implicate the CK2α/BMI1 axis in ovarian cancer.


The FASEB Journal | 2016

Mitochondrial BMI1 maintains bioenergetic homeostasis in cells

Soumyajit Banerjee Mustafi; Nicolas Aznar; Shailendra Kumar Dhar Dwivedi; Prabir K. Chakraborty; Rumki Basak; Priyabrata Mukherjee; Pradipta Ghosh; Resham Bhattacharya

The polycomb complex proto‐oncogene BMI1 [B lymphoma Mo‐MLV insertion region 1 homolog (mouse)] is essential for self‐renewal ofnormal andcancer stemcells. BMI1‐nullmice showseveredefects ingrowth, development, and survival. Although BMI1 is known to exert its effect in the nucleus via repression of 2 potent cellcycle regulators that are encoded by the Ink4a/Arf locus, deletion of this locus only partially rescues BMI1‐null phenotypes, which is indicative of alternatemechanisms of action of BMI1. Here, we showthat an extranuclearpool of BMI1 localizes to inner mitochondrial membrane and directly regulates mitochondrial RNA (mtRNA) homeostasis andbioenergetics. Thesemitochondrial functions of BMI1 are independent of itspreviouslydescribednuclear functions because a nuclear localization‐defective mutant BMI1 rescued several bioenergetic defects that we observed in BMI1‐depleted cells, for example, mitochondrial respiration, cytochrome c oxidase activity, and ATP production. Mechanistically, BMI1 coprecipitated with polynucleotide phosphorylase, a ribonuclease that is responsible for decay of mtRNA transcripts. Loss of BMI1 enhanced ribonuclease activity of polynucleotide phosphorylase and reduced mtRNA stability. These findings not only establish a novel extranuclear role of BMI1 in the regulation of mitochondrial bioenergetics, but also provide new mechanistic insights into the role of this protooncogene instemcell differentiation, neuronal aging, and cancer.—Banerjee Mustafi, S., Aznar, N., Dwivedi, S. K. D., Chakraborty, P. K., Basak, R., Mukherjee, P., Ghosh, P., Bhattacharya, R. Mitochondrial BMI1 maintains bioenergetic homeostasis in cells. FASEB J. 30, 4042–4055 (2016). www.fasebj.org


Molecular Cancer Therapeutics | 2018

Inhibition of BMI1, a Therapeutic Approach in Endometrial Cancer

Megan Buechel; Anindya Dey; Shailendra Kumar Dhar Dwivedi; A.K. Crim; Kai Ding; Roy Zhang; Priyabrata Mukherjee; Kathleen N. Moore; Liangxian Cao; Arthur Branstrom; Marla Weetall; John Baird; Resham Bhattacharya

With rising incidence rates, endometrial cancer is one of the most common gynecologic malignancies in the United States. Although surgery provides significant survival benefit to early-stage patients, those with advanced or recurrent metastatic disease have a dismal prognosis. Limited treatment options include chemotherapy and radiotherapy. Hence, there is a compelling need for developing molecularly targeted therapy. Here, we show that the polycomb ring finger protein BMI1, also known as a stem cell factor, is significantly overexpressed in endometrial cancer cell lines, endometrial cancer patient tissues as well as in nonendometrioid histologies and associated with poor overall survival. PTC-028, a second-generation inhibitor of BMI1 function, decreases invasion of endometrial cancer cells and potentiates caspase-dependent apoptosis, while normal cells with minimal expression of BMI1 remain unaffected. In an aggressive uterine carcinosarcoma xenograft model, single-agent PTC-028 significantly delayed tumor growth and increased tumor doubling time compared with the standard carboplatin/paclitaxel therapy. Therefore, anti-BMI1 strategies may represent a promising targeted approach in patients with advanced or recurrent endometrial cancer, a population where treatment options are limited. Mol Cancer Ther; 17(10); 2136–43. ©2018 AACR.

Collaboration


Dive into the Shailendra Kumar Dhar Dwivedi's collaboration.

Top Co-Authors

Avatar

Resham Bhattacharya

University of Oklahoma Health Sciences Center

View shared research outputs
Top Co-Authors

Avatar

Priyabrata Mukherjee

University of Oklahoma Health Sciences Center

View shared research outputs
Top Co-Authors

Avatar

Soumyajit Banerjee Mustafi

University of Oklahoma Health Sciences Center

View shared research outputs
Top Co-Authors

Avatar

Anindya Dey

University of Oklahoma Health Sciences Center

View shared research outputs
Top Co-Authors

Avatar

Prabir K. Chakraborty

Saha Institute of Nuclear Physics

View shared research outputs
Top Co-Authors

Avatar

Kai Ding

University of Oklahoma

View shared research outputs
Top Co-Authors

Avatar

A.K. Crim

University of Oklahoma Health Sciences Center

View shared research outputs
Top Co-Authors

Avatar

Sounik Saha

University of Oklahoma

View shared research outputs
Top Co-Authors

Avatar

Xunhao Xiong

University of Oklahoma Health Sciences Center

View shared research outputs
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge