Network


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

Hotspot


Dive into the research topics where Lokesh Dalasanur Nagaprashantha is active.

Publication


Featured researches published by Lokesh Dalasanur Nagaprashantha.


International Journal of Cancer | 2011

The Sensors and Regulators of Cell-Matrix Surveillance in Anoikis Resistance of Tumors

Lokesh Dalasanur Nagaprashantha; Rit Vatsyayan; Poorna Chandra Rao Lelsani; Sanjay Awasthi; Sharad S. Singhal

Normal cells continuously monitor the nature of their respective cellular microenvironment. They are equipped with an inherent molecular defense to detect changes that can precipitate and trigger an oncogenic cascade in the internal and external environment of cells. The process called anoikis unleashes many a characteristic molecular change in the cells which eventually program to cell death in response to cell detachment and inappropriate cellular attachment, both of which can otherwise potentiate the ability of cells to preferentially pursue a malignant course due to the release of molecular discipline which conforms them to a benign structural and functional spectrum. The initiation and propagation of signaling that serves as a switch to cell survival or cell death mediated by surveillance of cell microenvironment is comprised of many heterogeneous sets of molecules interacting mainly at the interface of cell–extracellular matrix. Transforming cells continuously reprogram their signaling characteristics in sensing and modulating the stimuli from cell surface molecules like integrins, cadherins and immunoglobulin family of cell adhesion molecules at adhesion complexes, which enables them to resist anoikis and metastasize to different organs. Actin cytoskeleton binds BIM and Bcl2 modifying factor (BMF), which are regulated by the adhesion status and consequent conformation of cytoskeleton in the cells. This review aims at an integrated synopsis of fundamental mechanisms of the critical interactions of cell surface molecules to facilitate a focused analysis of the differential regulation of signaling processes at cell‐ECM junctions that collectively rein the anoikis resistance, which in turn impacts metastatic aggressiveness and drug resistance of tumors originating from respective organs.


Cancer Prevention Research | 2012

Didymin Induces Apoptosis by Inhibiting N-Myc and Upregulating RKIP in Neuroblastoma

Jyotsana Singhal; Lokesh Dalasanur Nagaprashantha; Rit Vatsyayan; Ashutosh; Sanjay Awasthi; Sharad S. Singhal

Neuroblastomas arise from the neural crest cells and represent the most common solid tumors outside the nervous system in children. The amplification of N-Myc plays a primary role in the pathogenesis of neuroblastomas, whereas acquired mutations of p53 lead to refractory and relapsed cases of neuroblastomas. In this regard, dietary compounds which can target N-Myc and exert anticancer effects independent of p53 status acquire significance in the management of neuroblastomas. Hence, we investigated the anticancer properties of the flavonoid didymin in neuroblastomas. Didymin effectively inhibited proliferation and induced apoptosis irrespective of p53 status in neuroblastomas. Didymin downregulated phosphoinositide 3-kinase, pAkt, Akt, vimentin, and upregulated RKIP levels. Didymin induced G2/M arrest along with decreasing the levels of cyclin D1, CDK4, and cyclin B1. Importantly, didymin inhibited N-Myc as confirmed at protein, mRNA, and transcriptional level by promoter–reporter assays. High-performance liquid chromatography analysis of didymin-treated (2 mg/kg b.w.) mice serum revealed effective oral absorption with free didymin concentration of 2.1 μmol/L. Further in vivo mice xenograft studies revealed that didymin-treated (2 mg/kg b.w.) animals had significant reductions in tumors size compared with controls. Didymin strongly inhibited the proliferation (Ki67) and angiogenesis (CD31) markers, as well as N-Myc expression, as revealed by the histopathologic examination of paraffin-embedded section of resected tumors. Collectively, our in vitro and in vivo studies elucidated the anticancer properties and mechanisms of action of a novel, orally active, and palatable flavonoid didymin, which makes it a potential new approach for neuroblastoma therapy (NANT) to target pediatric neuroblastomas. Cancer Prev Res; 5(3); 473–83. ©2011 AACR.


Cancer Prevention Research | 2011

Targeting p53-Null Neuroblastomas through RLIP76

Jyotsana Singhal; Sushma Yadav; Lokesh Dalasanur Nagaprashantha; Rit Vatsyayan; Sharad S. Singhal; Sanjay Awasthi

The search for p53-independent mechanism of cancer cell killing is highly relevant to pediatric neuroblastomas, where successful therapy is limited by its transformation into p53-mutant and a highly drug-resistant neoplasm. Our studies on the drug-resistant p53-mutant as compared with drug-resistant p53 wild-type neuroblastoma revealed a novel mechanism for resistance to apoptosis: a direct role of p53 in regulating the cellular concentration of proapoptotic alkenals by functioning as a specific and saturable allosteric inhibitor of the alkenal–glutathione conjugate transporter, RLIP76. The RLIP76-p53 complex was showed by both immunoprecipitation analyses of purified proteins and immunofluorescence analysis. Drug transport studies revealed that p53 inhibited both basal and PKCα-stimulated transport of glutathione conjugates of 4HNE (GSHNE) and doxorubicin. Drug resistance was significantly greater for p53-mutant as compared with p53 wild-type neuroblastoma cell lines, but both were susceptible to depletion of RLIP76 by antisense alone. In addition, inhibition of RLIP76 significantly enhanced the cytotoxicity of cisplatin. Taken together, these studies provide powerful evidence for a novel mechanism for drug and apoptosis resistance in p53-mutant neuroblastoma, based on a model of regulation of p53-induced apoptosis by RLIP76, where p53 is a saturable and specific allosteric inhibitor of RLIP76, and p53 loss results in overexpression of RLIP76; thus, in the absence of p53, the drug and glutathione-conjugate transport activities of RLIP76 are enhanced. Most importantly, our findings strongly indicate RLIP76 as a novel target for therapy of drug-resistant and p53-mutant neuroblastoma. Cancer Prev Res; 4(6); 879–89. ©2011 AACR.


PLOS ONE | 2013

Proteomic Analysis of Signaling Network Regulation in Renal Cell Carcinomas with Differential Hypoxia-Inducible Factor-2α Expression

Lokesh Dalasanur Nagaprashantha; Tatjana Talamantes; Jyotsana Singhal; Jia Guo; Rit Vatsyayan; Navin Rauniyar; Sanjay Awasthi; Sharad S. Singhal; Laszlo Prokai

Background The loss of von Hippel–Lindau (VHL) protein function leads to highly vascular renal tumors characterized by an aggressive course of disease and refractoriness to chemotherapy and radiotherapy. Loss of VHL in renal tumors also differs from tumors of other organs in that the oncogenic cascade is mediated by an increase in the levels of hypoxia-inducible factor-2α (HIF2α) instead of hypoxia-inducible factor-1α (HIF1α). Methods and Principal Findings We used renal carcinoma cell lines that recapitulate the differences between mutant VHL and wild-type VHL genotypes. Utilizing a method relying on extracted peptide intensities as a label-free approach for quantitation by liquid chromatography–mass spectrometry, our proteomics study revealed regulation of key proteins important for cancer cell survival, proliferation and stress-resistance, and implicated differential regulation of signaling networks in VHL-mutant renal cell carcinoma. We also observed upregulation of cellular energy pathway enzymes and the stress-responsive mitochondrial 60-kDa heat shock protein. Finding reliance on glutaminolysis in VHL-mutant renal cell carcinoma was of particular significance, given the generally predominant dependence of tumors on glycolysis. The data have been deposited to the ProteomeXchange with identifier PXD000335. Conclusions and Significance Pathway analyses provided corroborative evidence for differential regulation of molecular and cellular functions influencing cancer energetics, metabolism and cell proliferation in renal cell carcinoma with distinct VHL genotype. Collectively, the differentially regulated proteome characterized by this study can potentially guide translational research specifically aimed at effective clinical interventions for advanced VHL-mutant, HIF2α-over-expressing tumors.


Cancer Research | 2011

Abstract 4334: Targeting p53 null neuroblastomas through RLIP76

Rit Vatsyayan; Jyotsana Singhal; Sushma Yadav; Lokesh Dalasanur Nagaprashantha; Sharad S. Singhal; Sanjay Awasthi

The search for p53-independent mechanism of cancer cell killing is highly relevant to pediatric neuroblastomas, where successful therapy is limited by its transformation into p53 mutant and a highly drug-resistant neoplasm. Our studies on the drug-resistant p53 mutant as compared with drug-resistant p53 wild-type neuroblastoma revealed a novel mechanism for resistance to apoptosis: a direct role of p53 in regulating the cellular concentration of pro-apoptotic alkenals by functioning as a specific and saturable allosteric inhibitor of the alkenal-glutathione-conjugate transporter, RLIP76. The RLIP76-p53 complex was demonstrated both by using immuno-precipitation analyses of purified proteins as well as by immuno-fluorescence analysis. Drug transport studies revealed that p53 inhibited both basal and PKCα stimulated transport of glutathione-conjugates of 4HNE (GS-HNE) and cisplatin. Drug resistance was significantly greater for p53 mutant as compared with p53 wild-type neuroblastoma cell lines, but both were susceptible to depletion of RLIP76 by antisense alone. In addition, inhibition of RLIP76 significantly enhanced the cytotoxicity of cisplatin. Taken together, these studies provide powerful evidence for a novel mechanism for drug and apoptosis resistance in p53 mutant neuroblastoma, based on a model of regulation of p53 induced apoptosis by RLIP76, where p53 is a saturable and specific allosteric inhibitor of RLIP76, and p53 loss results in over-expression of RLIP76; thus, in the absence of p53, the drug and glutathione-conjugate transport activities of RLIP76 are enhanced. Most importantly, our findings strongly indicate RLIP76 as a novel target for therapy of drug-resistant and p53 mutant neuroblastoma. (Supported in part by NIH grant CA 77495 (SA), CA 155350, Cancer Research Foundation of North Texas, and Institute for Cancer Research & the Joe & Jessie Crump Fund for Medical Education (SSS)) Citation Format: {Authors}. {Abstract title} [abstract]. In: Proceedings of the 102nd Annual Meeting of the American Association for Cancer Research; 2011 Apr 2-6; Orlando, FL. Philadelphia (PA): AACR; Cancer Res 2011;71(8 Suppl):Abstract nr 4334. doi:10.1158/1538-7445.AM2011-4334


Journal of Proteomics | 2018

2′-Hydroxyflavanone induced changes in the proteomic profile of breast cancer cells

Lokesh Dalasanur Nagaprashantha; Jyotsana Singhal; Shireen Chikara; Gabriel Gugiu; David Horne; Sanjay Awasthi; Ravi Salgia; Sharad S. Singhal

In spite of rapid advances in understanding of signaling networks associated with the incidence and therapeutic-sensitivity, breast cancer (BC) still remains the most commonly diagnosed and prevalent cancer in women. Emergence of resistance to hormonal interventions in estrogen-receptor (ER) positive BC coupled to loss of ER expression and activation of ER-independent growth factor, heat-shock, MYC and WNT pathways along with distinct mechanisms of therapeutic-resistance in HER2 over-expressing and triple-negative subtypes of BC collectively necessitates deeper profiling of the mechanistic networks regulated by potential lead anticancer compounds intended for further development to target BC. A significant part of the search for novel lead anticancer compounds for BC has focused on phytochemicals including flavonoids found in citrus fruits, which have shown promising anticancer activity. Based on the initial studies which revealed the anticancer effect of 2HF in BC, we employed an advanced TMT 10plex labeled proteomic approach to characterize the changes in non-phosphorylated and phosphorylated proteomic profile of ER+ MCF7, triple-negative MDA-MB231 and HER2+ SKBR3 BC cells, and MCF10A normal breast epithelial cells. 2HF induced significant changes in the proteins responsible for BC incidence, metastases and therapeutic sensitivity in BC cells.


Cancer Research | 2011

Abstract 3702: 2-Hydroxyflavanone inhibits renal cancer growth by inhibiting tumor cell proliferation and angiogenesis

Lokesh Dalasanur Nagaprashantha; Rit Vatsyayan; Jyotsana Singhal; Poorna Chandra Rao Lelsani; Yogesh C. Awasthi; Sanjay Awasthi; Sharad S. Singhal

Proceedings: AACR 102nd Annual Meeting 2011‐‐ Apr 2‐6, 2011; Orlando, FL Renal cell carcinoma (RCC) is one of the top ten cancers prevalent in USA. Loss-of-function mutation in the VHL is an established risk factor for RCC contributing to 75% of total reported cases of RCC. Loss of VHL leads to highly vascularized phenotype of renal tumors. Intake of oranges has been proven to reduce the risk of RCC in multi-center international clinical trials. Hence, we studied effects of 2HF, an active anti-cancer compound of oranges, in VHL- mutant RCC. Our in vitro investigations revealed that 2HF selectively inhibits VHL-mutant RCC by inhibiting EGFR signaling which is increased due to VHL mutations in RCC. Our results also revealed for the first time, that 2HF inhibits GSTπ activity. 2HF inhibited cyclin B1 and CDK4, and induced G2/M phase arrest in VHL-mutant RCC. Importantly, 2HF inhibited the angiogenesis in VHL-mutant RCC by decreasing VEGF expression. Our in vivo studies in mice xenografts confirmed our in vitro results as evident by decreased levels of proliferation marker, Ki67 and angiogenic marker CD31, in 2HF treated (0.01%, w/w) mice xenografts of VHL-mutant RCC. 2HF also increased the expression of E-cadherin in VHL-mutant RCC which would be of significance in restoring the normal epithelial phenotype. Collectively, our in vitro and in vivo results revealed the potent anti-proliferative and anti-angiogenic effects of 2HF in VHL-mutant RCC, sparing normal cells, which could have significant implications not only in the specific management of VHL-mutant RCC but also in treating other VHL syndromes with highly vascularized phenotype. (Supported in part by NIH grant CA 77495 (SA), CA 155350, Cancer Research Foundation of North Texas, and Institute for Cancer Research & the Joe & Jessie Crump Fund for Medical Education (SSS)) Citation Format: {Authors}. {Abstract title} [abstract]. In: Proceedings of the 102nd Annual Meeting of the American Association for Cancer Research; 2011 Apr 2-6; Orlando, FL. Philadelphia (PA): AACR; Cancer Res 2011;71(8 Suppl):Abstract nr 3702. doi:10.1158/1538-7445.AM2011-3702


Carcinogenesis | 2011

2′-Hydroxyflavanone inhibits proliferation, tumor vascularization and promotes normal differentiation in VHL-mutant renal cell carcinoma

Lokesh Dalasanur Nagaprashantha; Rit Vatsyayan; Jyotsana Singhal; Poorna Chandra Rao Lelsani; Laszlo Prokai; Sanjay Awasthi; Sharad S. Singhal


Cancer Letters | 2018

Oxidative stress and dietary phytochemicals: Role in cancer chemoprevention and treatment

Shireen Chikara; Lokesh Dalasanur Nagaprashantha; Jyotsana Singhal; David Horne; Sanjay Awasthi; Sharad S. Singhal


Oncotarget | 2017

2'-Hydroxyflavanone: A novel strategy for targeting breast cancer

Jyotsana Singhal; Lokesh Dalasanur Nagaprashantha; Shireen Chikara; Sanjay Awasthi; David Horne; Sharad S. Singhal

Collaboration


Dive into the Lokesh Dalasanur Nagaprashantha's collaboration.

Top Co-Authors

Avatar

Sanjay Awasthi

Texas Tech University Health Sciences Center

View shared research outputs
Top Co-Authors

Avatar

Sharad S. Singhal

University of North Texas Health Science Center

View shared research outputs
Top Co-Authors

Avatar

Jyotsana Singhal

University of North Texas Health Science Center

View shared research outputs
Top Co-Authors

Avatar

Rit Vatsyayan

University of North Texas Health Science Center

View shared research outputs
Top Co-Authors

Avatar

David Horne

City of Hope National Medical Center

View shared research outputs
Top Co-Authors

Avatar

Poorna Chandra Rao Lelsani

University of North Texas Health Science Center

View shared research outputs
Top Co-Authors

Avatar

Shireen Chikara

North Dakota State University

View shared research outputs
Top Co-Authors

Avatar

Laszlo Prokai

University of North Texas Health Science Center

View shared research outputs
Top Co-Authors

Avatar

Ravi Salgia

City of Hope National Medical Center

View shared research outputs
Top Co-Authors

Avatar

Sushma Yadav

University of Texas at Arlington

View shared research outputs
Researchain Logo
Decentralizing Knowledge