Network


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

Hotspot


Dive into the research topics where Kshipra M. Gharpure is active.

Publication


Featured researches published by Kshipra M. Gharpure.


Nature Communications | 2013

Src activation by adrenoreceptors is a key switch for tumour metastasis

Guillermo N. Armaiz-Pena; Julie K. Allen; Anthony Cruz; Rebecca L. Stone; Alpa M. Nick; Yvonne G. Lin; Liz Y. Han; Lingegowda S. Mangala; Gabriel J. Villares; Pablo Vivas-Mejia; Cristian Rodriguez-Aguayo; Archana S. Nagaraja; Kshipra M. Gharpure; Zheng Wu; Robert D. English; Kizhake V. Soman; Mian M.K. Shahzad; Maya Zigler; Michael T. Deavers; Alexander Zien; Theodoros Soldatos; David B. Jackson; John E. Wiktorowicz; Madeline Torres-Lugo; Tom Young; Koen De Geest; Gary E. Gallick; Menashe Bar-Eli; Gabriel Lopez-Berestein; Steve W. Cole

Norepinephrine (NE) can modulate multiple cellular functions important for cancer progression; however, how this single extracellular signal regulates such a broad array of cellular processes is unknown. Here, we identify Src as a key regulator of phosphoproteomic signaling networks activated in response to beta-adrenergic signaling in cancer cells. These results also identify a new mechanism of Src phosphorylation that mediates beta-adrenergic/PKA regulation of downstream networks, thereby enhancing tumor cell migration, invasion and growth. In human ovarian cancer samples, high tumoral NE levels were correlated with high pSrcY419 levels. Moreover, among cancer patients, the use of beta blockers was significantly associated with reduced cancer-related mortality. Collectively, these data provide a pivotal molecular target for disrupting neural signaling in the tumor microenvironment.


Nature Communications | 2014

Hypoxia-mediated downregulation of miRNA biogenesis promotes tumour progression

Rajesha Rupaimoole; Sherry Y. Wu; Sunila Pradeep; Cristina Ivan; Chad V. Pecot; Kshipra M. Gharpure; Archana S. Nagaraja; Guillermo N. Armaiz-Pena; Michael McGuire; Behrouz Zand; Heather J. Dalton; Justyna Filant; Justin Bottsford Miller; Chunhua Lu; Nouara C. Sadaoui; Lingegowda S. Mangala; Morgan Taylor; Twan van den Beucken; Elizabeth Koch; Cristian Rodriguez-Aguayo; Li Huang; Menashe Bar-Eli; Bradly G. Wouters; Milan Radovich; Mircea Ivan; George A. Calin; Wei Zhang; Gabriel Lopez-Berestein; Anil K. Sood

Cancer-related deregulation of miRNA biogenesis has been suggested, but the underlying mechanisms remain elusive. Here we report a previously unrecognized effect of hypoxia in the downregulation of Drosha and Dicer in cancer cells that leads to dysregulation of miRNA biogenesis and increased tumour progression. We show that hypoxia-mediated downregulation of Drosha is dependent on ETS1/ELK1 transcription factors. Moreover, mature miRNA array and deep sequencing studies reveal altered miRNA maturation in cells under hypoxic conditions. At a functional level, this phenomenon results in increased cancer progression in vitro and in vivo, and data from patient samples are suggestive of miRNA biogenesis downregulation in hypoxic tumours. Rescue of Drosha by siRNAs targeting ETS1/ELK1 in vivo results in significant tumour regression. These findings provide a new link in the mechanistic understanding of global miRNA downregulation in the tumour microenvironment. MicroRNAs play important roles in the maintenance of cellular homeostasis through the post-transcriptional regulation of gene expression. Here, the authors implicate loss of the miRNA biogenesis factor Drosha and altered miRNA maturation in tumour progression under hypoxic conditions.


Cell Reports | 2015

Long Noncoding RNA Ceruloplasmin Promotes Cancer Growth by Altering Glycolysis

Rajesha Rupaimoole; Jaehyuk Lee; Monika Haemmerle; Hui Ling; Rebecca A. Previs; Sunila Pradeep; Sherry Y. Wu; Cristina Ivan; Manuela Ferracin; Jennifer B. Dennison; Niki Zacharias Millward; Archana S. Nagaraja; Kshipra M. Gharpure; Michael McGuire; Nidhin Sam; Guillermo N. Armaiz-Pena; Nouara C. Sadaoui; Cristian Rodriguez-Aguayo; George A. Calin; Ronny Drapkin; Jeffery Kovacs; Gordon B. Mills; Wei Zhang; Gabriel Lopez-Berestein; Pratip Bhattacharya; Anil K. Sood

Long noncoding RNAs (lncRNAs) significantly influence the development and regulation of genome expression in cells. Here, we demonstrate the role of lncRNA ceruloplasmin (NRCP) in cancer metabolism and elucidate functional effects leading to increased tumor progression. NRCP was highly upregulated in ovarian tumors, and knockdown of NRCP resulted in significantly increased apoptosis, decreased cell proliferation, and decreased glycolysis compared with control cancer cells. In an orthotopic mouse model of ovarian cancer, siNRCP delivered via a liposomal carrier significantly reduced tumor growth compared with control treatment. We identified NRCP as an intermediate binding partner between STAT1 and RNA polymerase II, leading to increased expression of downstream target genes such as glucose-6-phosphate isomerase. Collectively, we report a previously unrecognized role of the lncRNA NRCP in modulating cancer metabolism. As demonstrated, DOPC nanoparticle-incorporated siRNA-mediated silencing of this lncRNA in vivo provides therapeutic avenue toward modulating lncRNAs in cancer.


Journal of Clinical Investigation | 2013

ATP11B mediates platinum resistance in ovarian cancer

Myrthala Moreno-Smith; Jyotsnabaran Halder; Paul S. Meltzer; Tamas A. Gonda; Lingegowda S. Mangala; Rajesha Rupaimoole; Chunhua Lu; Archana S. Nagaraja; Kshipra M. Gharpure; Yu Kang; Cristian Rodriguez-Aguayo; Pablo Vivas-Mejia; Behrouz Zand; Rosemarie Schmandt; Hua Wang; Robert R. Langley; Nicholas B. Jennings; Cristina Ivan; Jeremy E. Coffin; Guillermo N. Armaiz; Justin Bottsford-Miller; Sang Bae Kim; Margaret S. Halleck; Mary J.C. Hendrix; William Bornman; Menashe Bar-Eli; Ju Seog Lee; Zahid H. Siddik; Gabriel Lopez-Berestein; Anil K. Sood

Platinum compounds display clinical activity against a wide variety of solid tumors; however, resistance to these agents is a major limitation in cancer therapy. Reduced platinum uptake and increased platinum export are examples of resistance mechanisms that limit the extent of DNA damage. Here, we report the discovery and characterization of the role of ATP11B, a P-type ATPase membrane protein, in cisplatin resistance. We found that ATP11B expression was correlated with higher tumor grade in human ovarian cancer samples and with cisplatin resistance in human ovarian cancer cell lines. ATP11B gene silencing restored the sensitivity of ovarian cancer cell lines to cisplatin in vitro. Combined therapy of cisplatin and ATP11B-targeted siRNA significantly decreased cancer growth in mice bearing ovarian tumors derived from cisplatin-sensitive and -resistant cells. In vitro mechanistic studies on cellular platinum content and cisplatin efflux kinetics indicated that ATP11B enhances the export of cisplatin from cells. The colocalization of ATP11B with fluorescent cisplatin and with vesicular trafficking proteins, such as syntaxin-6 (STX6) and vesicular-associated membrane protein 4 (VAMP4), strongly suggests that ATP11B contributes to secretory vesicular transport of cisplatin from Golgi to plasma membrane. In conclusion, inhibition of ATP11B expression could serve as a therapeutic strategy to overcome cisplatin resistance.


Nature Communications | 2016

A miR-192-EGR1-HOXB9 regulatory network controls the angiogenic switch in cancer

Sherry Y. Wu; Rajesha Rupaimoole; Fangrong Shen; Sunila Pradeep; Chad V. Pecot; Cristina Ivan; Archana S. Nagaraja; Kshipra M. Gharpure; Elizabeth Pham; Hiroto Hatakeyama; Michael McGuire; Monika Haemmerle; Viviana Vidal-Anaya; Courtney Olsen; Cristian Rodriguez-Aguayo; Justyna Filant; Ehsan A. Ehsanipour; Shelley M. Herbrich; Sourindra Maiti; Li Huang; Ji Hoon Kim; Xinna Zhang; Hee Dong Han; Guillermo N. Armaiz-Pena; Elena G. Seviour; Susan L. Tucker; Min Zhang; Da Yang; Laurence J.N. Cooper; Rouba Ali-Fehmi

A deeper mechanistic understanding of tumour angiogenesis regulation is needed to improve current anti-angiogenic therapies. Here we present evidence from systems-based miRNA analyses of large-scale patient data sets along with in vitro and in vivo experiments that miR-192 is a key regulator of angiogenesis. The potent anti-angiogenic effect of miR-192 stems from its ability to globally downregulate angiogenic pathways in cancer cells through regulation of EGR1 and HOXB9. Low miR-192 expression in human tumours is predictive of poor clinical outcome in several cancer types. Using 1,2-dioleoyl-sn-glycero-3-phosphatidylcholine (DOPC) nanoliposomes, we show that miR-192 delivery leads to inhibition of tumour angiogenesis in multiple ovarian and renal tumour models, resulting in tumour regression and growth inhibition. This anti-angiogenic and anti-tumour effect is more robust than that observed with an anti-VEGF antibody. Collectively, these data identify miR-192 as a central node in tumour angiogenesis and support the use of miR-192 in an anti-angiogenesis therapy.


Oncogene | 2016

Hypoxia-upregulated microRNA-630 targets Dicer, leading to increased tumor progression.

Rajesha Rupaimoole; Cristina Ivan; Da Yang; Kshipra M. Gharpure; Sherry Y. Wu; Chad V. Pecot; Rebecca A. Previs; Archana S. Nagaraja; Guillermo N. Armaiz-Pena; Michael McGuire; Sunila Pradeep; Lingegowda S. Mangala; Cristian Rodriguez-Aguayo; Li Huang; Menashe Bar-Eli; Wei Zhang; Gabriel Lopez-Berestein; George A. Calin; Anil K. Sood

MicroRNAs (miRNAs) are small RNA molecules that affect cellular processes by controlling gene expression. Recent studies have shown that hypoxia downregulates Drosha and Dicer, key enzymes in miRNA biogenesis, causing a decreased pool of miRNAs in cancer and resulting in increased tumor growth and metastasis. Here we demonstrate a previously unrecognized mechanism by which hypoxia downregulates Dicer. We found that miR-630, which is upregulated under hypoxic conditions, targets and downregulates Dicer expression. In an orthotopic mouse model of ovarian cancer, delivery of miR-630 using 1,2-dioleoyl-sn-glycero-3-phosphocholine (DOPC) nanoliposomes resulted in increased tumor growth and metastasis, and decreased Dicer expression. Treatment with the combination of anti-miR-630 and anti-vascular endothelial growth factor antibody in mice resulted in rescue of Dicer expression and significantly decreased tumor growth and metastasis. These results indicate that targeting miR-630 is a promising approach to overcome Dicer deregulation in cancer. As demonstrated in the study, use of DOPC nanoliposomes for anti-miR delivery serves as a better alternative approach to cell line-based overexpression of sense or antisense miRNAs, while avoiding potential in vitro selection effects. Findings from this study provide a new understanding of miRNA biogenesis downregulation observed under hypoxia and suggest therapeutic avenues to target this dysregulation in cancer.


Clinical Cancer Research | 2014

Molecular Biomarkers of Residual Disease after Surgical Debulking of High-Grade Serous Ovarian Cancer

Susan L. Tucker; Kshipra M. Gharpure; Shelley M. Herbrich; Anna K. Unruh; Alpa M. Nick; Erin K. Crane; Robert L. Coleman; Jamie Guenthoer; Heather J. Dalton; Sherry Y. Wu; R. Rupaimoole; Gabriel Lopez-Berestein; Bulent Ozpolat; Cristina Ivan; Wei Hu; Keith A. Baggerly; Anil K. Sood

Purpose: Residual disease following primary cytoreduction is associated with adverse overall survival in patients with epithelial ovarian cancer. Accurate identification of patients at high risk of residual disease has been elusive, lacking external validity and prompting many to undergo unnecessary surgical exploration. Our goal was to identify and validate molecular markers associated with high rates of residual disease. Methods: We interrogated two publicly available datasets from chemonaïve primary high-grade serous ovarian tumors for genes overexpressed in patients with residual disease and significant at a 10% false discovery rate (FDR) in both datasets. We selected genes with wide dynamic range for validation in an independent cohort using quantitative RT-PCR to assay gene expression, followed by blinded prediction of a patient subset at high risk for residual disease. Predictive success was evaluated using a one-sided Fisher exact test. Results: Forty-seven probe sets met the 10% FDR criterion in both datasets. These included FABP4 and ADH1B, which tracked tightly, showed dynamic ranges >16-fold and had high expression levels associated with increased incidence of residual disease. In the validation cohort (n = 139), FABP4 and ADH1B were again highly correlated. Using the top quartile of FABP4 PCR values as a prespecified threshold, we found 30 of 35 cases of residual disease in the predicted high-risk group (positive predictive value = 86%) and 54 of 104 among the remaining patients (P = 0.0002; OR, 5.5). Conclusion: High FABP4 and ADH1B expression is associated with significantly higher risk of residual disease in high-grade serous ovarian cancer. Patients with high tumoral levels of these genes may be candidates for neoadjuvant chemotherapy. Clin Cancer Res; 20(12); 3280–8. ©2014 AACR.


Molecular Cancer Therapeutics | 2014

Therapeutic Silencing of KRAS Using Systemically Delivered siRNAs

Chad V. Pecot; Sherry Y. Wu; Seth Bellister; Justyna Filant; Rajesha Rupaimoole; Takeshi Hisamatsu; Rajat Bhattacharya; Anshumaan Maharaj; Salma H. Azam; Cristian Rodriguez-Aguayo; Archana S. Nagaraja; Maria Pia Morelli; Kshipra M. Gharpure; Trent A. Waugh; Vianey Gonzalez-Villasana; Behrouz Zand; Heather J. Dalton; Scott Kopetz; Gabriel Lopez-Berestein; Lee M. Ellis; Anil K. Sood

Despite being among the most common oncogenes in human cancer, to date, there are no effective clinical options for inhibiting KRAS activity. We investigated whether systemically delivered KRAS siRNAs have therapeutic potential in KRAS-mutated cancer models. We identified KRAS siRNA sequences with notable potency in knocking down KRAS expression. Using lung and colon adenocarcinoma cell lines, we assessed antiproliferative effects of KRAS silencing in vitro. For in vivo experiments, we used a nanoliposomal delivery platform, DOPC, for systemic delivery of siRNAs. Various lung and colon cancer models were used to determine efficacy of systemic KRAS siRNA based on tumor growth, development of metastasis, and downstream signaling. KRAS siRNA sequences induced >90% knockdown of KRAS expression, significantly reducing viability in mutant cell lines. In the lung cancer model, KRAS siRNA treatment demonstrated significant reductions in primary tumor growth and distant metastatic disease, while the addition of CDDP was not additive. Significant reductions in Ki-67 indices were seen in all treatment groups, whereas significant increases in caspase-3 activity were only seen in the CDDP treatment groups. In the colon cancer model, KRAS siRNA reduced tumor KRAS and pERK expression. KRAS siRNAs significantly reduced HCP1 subcutaneous tumor growth, as well as outgrowth of liver metastases. Our studies demonstrate a proof-of-concept approach to therapeutic KRAS targeting using nanoparticle delivery of siRNA. This study highlights the potential translational impact of therapeutic RNA interference, which may have broad applications in oncology, especially for traditional “undruggable” targets. Mol Cancer Ther; 13(12); 2876–85. ©2014 AACR.


Clinical Cancer Research | 2015

Nanotechnology: Future of Oncotherapy

Kshipra M. Gharpure; Sherry Y. Wu; Chun Li; Gabriel Lopez-Berestein; Anil K. Sood

Recent advances in nanotechnology have established its importance in several areas including medicine. The myriad of applications in oncology range from detection and diagnosis to drug delivery and treatment. Although nanotechnology has attracted a lot of attention, the practical application of nanotechnology to clinical cancer care is still in its infancy. This review summarizes the role that nanotechnology has played in improving cancer therapy, its potential for affecting all aspects of cancer care, and the challenges that must be overcome to realize its full promise. Clin Cancer Res; 21(14); 3121–30. ©2015 AACR.


Oncogene | 2016

Sustained adrenergic signaling leads to increased metastasis in ovarian cancer via increased PGE2 synthesis

Archana S. Nagaraja; Piotr L. Dorniak; Nouara C. Sadaoui; Yu Kang; Tan Lin; Guillermo N. Armaiz-Pena; Sherry Y. Wu; Rajesha Rupaimoole; Julie K. Allen; Kshipra M. Gharpure; Sunila Pradeep; Behrouz Zand; Rebecca A. Previs; Jean M. Hansen; Cristina Ivan; Cristian Rodriguez-Aguayo; Peiying Yang; Gabriel Lopez-Berestein; Susan K. Lutgendorf; Steve W. Cole; Anil K. Sood

Adrenergic stimulation adversely affects tumor growth and metastasis, but the underlying mechanisms are not well understood. Here, we uncovered a novel mechanism by which catecholamines induce inflammation by increasing prostaglandin E2 (PGE2) levels in ovarian cancer cells. Metabolic changes in tumors isolated from patients with depression and mice subjected to restraint stress showed elevated PGE2 levels. Increased metabolites, PTGS2 and PTGES protein levels were found in Skov3-ip1 and HeyA8 cells treated with norepinephrine (NE), and these changes were shown to be mediated by ADRB2 receptor signaling. Silencing PTGS2 resulted in significantly decreased migration and invasion in ovarian cancer cells in the presence of NE and decreased tumor burden and metastasis in restraint stress orthotopic models. In human ovarian cancer samples, concurrent increased ADRB2, PTGS2 and PTGES expression was associated with reduced overall and progression-free patient survival. In conclusion, increased adrenergic stimulation results in increased PGE2 synthesis via ADRB2–Nf-kB–PTGS2 axis, which drives tumor growth and metastasis.

Collaboration


Dive into the Kshipra M. Gharpure's collaboration.

Top Co-Authors

Avatar

Archana S. Nagaraja

University of Texas MD Anderson Cancer Center

View shared research outputs
Top Co-Authors

Avatar

Anil K. Sood

University of Texas MD Anderson Cancer Center

View shared research outputs
Top Co-Authors

Avatar

Gabriel Lopez-Berestein

University of Texas MD Anderson Cancer Center

View shared research outputs
Top Co-Authors

Avatar

Rajesha Rupaimoole

University of Texas MD Anderson Cancer Center

View shared research outputs
Top Co-Authors

Avatar

Sherry Y. Wu

University of Texas MD Anderson Cancer Center

View shared research outputs
Top Co-Authors

Avatar

Sunila Pradeep

University of Texas MD Anderson Cancer Center

View shared research outputs
Top Co-Authors

Avatar

Cristian Rodriguez-Aguayo

University of Texas MD Anderson Cancer Center

View shared research outputs
Top Co-Authors

Avatar

Cristina Ivan

University of Texas MD Anderson Cancer Center

View shared research outputs
Top Co-Authors

Avatar

Lingegowda S. Mangala

University of Texas MD Anderson Cancer Center

View shared research outputs
Top Co-Authors

Avatar

Guillermo N. Armaiz-Pena

University of Texas MD Anderson Cancer Center

View shared research outputs
Researchain Logo
Decentralizing Knowledge