Network


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

Hotspot


Dive into the research topics where Krithika Bhuvaneshwar is active.

Publication


Featured researches published by Krithika Bhuvaneshwar.


Hepatology | 2015

βII-Spectrin (SPTBN1) suppresses progression of hepatocellular carcinoma and Wnt signaling by regulation of Wnt inhibitor kallistatin.

Xiuling Zhi; Ling Lin; Shaoxian Yang; Krithika Bhuvaneshwar; Hongkun Wang; Yuriy Gusev; Mi-Hye Lee; Bhaskar Kallakury; Narayan Shivapurkar; Katherine Cahn; Xuefei Tian; John L. Marshall; Stephen W. Byers; Aiwu R. He

βII‐Spectrin (SPTBN1) is an adapter protein for Smad3/Smad4 complex formation during transforming growth factor beta (TGF‐β) signal transduction. Forty percent of SPTBN1+/− mice spontaneously develop hepatocellular carcinoma (HCC), and most cases of human HCC have significant reductions in SPTBN1 expression. In this study, we investigated the possible mechanisms by which loss of SPTBN1 may contribute to tumorigenesis. Livers of SPTBN1+/− mice, compared to wild‐type mouse livers, display a significant increase in epithelial cell adhesion molecule‐positive (EpCAM+) cells and overall EpCAM expression. Inhibition of SPTBN1 in human HCC cell lines increased the expression of stem cell markers EpCAM, Claudin7, and Oct4, as well as decreased E‐cadherin expression and increased expression of vimentin and c‐Myc, suggesting reversion of these cells to a less differentiated state. HCC cells with decreased SPTBN1 also demonstrate increased sphere formation, xenograft tumor development, and invasion. Here we investigate possible mechanisms by which SPTBN1 may influence the stem cell traits and aggressive behavior of HCC cell lines. We found that HCC cells with decreased SPTBN1 express much less of the Wnt inhibitor kallistatin and exhibit decreased β‐catenin phosphorylation and increased β‐catenin nuclear localization, indicating Wnt signaling activation. Restoration of kallistatin expression in these cells reversed the observed Wnt activation. Conclusion: SPTBN1 expression in human HCC tissues is positively correlated with E‐cadherin and kallistatin levels, and decreased SPTBN1 and kallistatin gene expression is associated with decreased relapse‐free survival. Our data suggest that loss of SPTBN1 activates Wnt signaling, which promotes acquisition of stem cell‐like features, and ultimately contributes to malignant tumor progression. (Hepatology 2015;61:598‐612)


Frontiers in Genetics | 2013

Genome-wide multi-omics profiling of colorectal cancer identifies immune determinants strongly associated with relapse

Subha Madhavan; Yuriy Gusev; Thanemozhi G. Natarajan; Lei Song; Krithika Bhuvaneshwar; Robinder Gauba; Abhishek Pandey; Bassem R. Haddad; David Goerlitz; Amrita K. Cheema; Hartmut Juhl; Bhaskar Kallakury; John L. Marshall; Stephen W. Byers; Louis M. Weiner

The use and benefit of adjuvant chemotherapy to treat stage II colorectal cancer (CRC) patients is not well understood since the majority of these patients are cured by surgery alone. Identification of biological markers of relapse is a critical challenge to effectively target treatments to the ~20% of patients destined to relapse. We have integrated molecular profiling results of several “omics” data types to determine the most reliable prognostic biomarkers for relapse in CRC using data from 40 stage I and II CRC patients. We identified 31 multi-omics features that highly correlate with relapse. The data types were integrated using multi-step analytical approach with consecutive elimination of redundant molecular features. For each data type a systems biology analysis was performed to identify pathways biological processes and disease categories most affected in relapse. The biomarkers detected in tumors urine and blood of patients indicated a strong association with immune processes including aberrant regulation of T-cell and B-cell activation that could lead to overall differences in lymphocyte recruitment for tumor infiltration and markers indicating likelihood of future relapse. The immune response was the biologically most coherent signature that emerged from our analyses among several other biological processes and corroborates other studies showing a strong immune response in patients less likely to relapse.


Computational and structural biotechnology journal | 2015

A case study for cloud based high throughput analysis of NGS data using the globus genomics system

Krithika Bhuvaneshwar; Dinanath Sulakhe; Robinder Gauba; Alex Rodriguez; Ravi K. Madduri; Utpal J. Dave; Lukasz Lacinski; Ian T. Foster; Yuriy Gusev; Subha Madhavan

Next generation sequencing (NGS) technologies produce massive amounts of data requiring a powerful computational infrastructure, high quality bioinformatics software, and skilled personnel to operate the tools. We present a case study of a practical solution to this data management and analysis challenge that simplifies terabyte scale data handling and provides advanced tools for NGS data analysis. These capabilities are implemented using the “Globus Genomics” system, which is an enhanced Galaxy workflow system made available as a service that offers users the capability to process and transfer data easily, reliably and quickly to address end-to-endNGS analysis requirements. The Globus Genomics system is built on Amazon s cloud computing infrastructure. The system takes advantage of elastic scaling of compute resources to run multiple workflows in parallel and it also helps meet the scale-out analysis needs of modern translational genomics research.


BMC Bioinformatics | 2016

G-DOC Plus – an integrative bioinformatics platform for precision medicine

Krithika Bhuvaneshwar; Anas Belouali; Varun Singh; Robert M. Johnson; Lei Song; Adil Alaoui; Michael Harris; Robert Clarke; Louis M. Weiner; Yuriy Gusev; Subha Madhavan

BackgroundG-DOC Plus is a data integration and bioinformatics platform that uses cloud computing and other advanced computational tools to handle a variety of biomedical BIG DATA including gene expression arrays, NGS and medical images so that they can be analyzed in the full context of other omics and clinical information.ResultsG-DOC Plus currently holds data from over 10,000 patients selected from private and public resources including Gene Expression Omnibus (GEO), The Cancer Genome Atlas (TCGA) and the recently added datasets from REpository for Molecular BRAin Neoplasia DaTa (REMBRANDT), caArray studies of lung and colon cancer, ImmPort and the 1000 genomes data sets. The system allows researchers to explore clinical-omic data one sample at a time, as a cohort of samples; or at the level of population, providing the user with a comprehensive view of the data.G-DOC Plus tools have been leveraged in cancer and non-cancer studies for hypothesis generation and validation; biomarker discovery and multi-omics analysis, to explore somatic mutations and cancer MRI images; as well as for training and graduate education in bioinformatics, data and computational sciences. Several of these use cases are described in this paper to demonstrate its multifaceted usability.ConclusionG-DOC Plus can be used to support a variety of user groups in multiple domains to enable hypothesis generation for precision medicine research. The long-term vision of G-DOC Plus is to extend this translational bioinformatics platform to stay current with emerging omics technologies and analysis methods to continue supporting novel hypothesis generation, analysis and validation for integrative biomedical research. By integrating several aspects of the disease and exposing various data elements, such as outpatient lab workup, pathology, radiology, current treatments, molecular signatures and expected outcomes over a web interface, G-DOC Plus will continue to strengthen precision medicine research. G-DOC Plus is available at: https://gdoc.georgetown.edu.


Carcinogenesis | 2014

Transcriptional regulation of STAT3 by SPTBN1 and SMAD3 in HCC through cAMP-response element-binding proteins ATF3 and CREB2

Ling Lin; Zhixing Yao; Krithika Bhuvaneshwar; Yuriy Gusev; Bhaskar Kallakury; Shaoxian Yang; Kirti Shetty; Aiwu R uth He

The cytoskeletal protein Spectrin, beta, non-erythrocytic 1 (SPTBN1), an adapter protein to SMAD3 in TGF-β signaling, may prevent hepatocellular carcinoma (HCC) development by downregulating the expression of signal transducer and activator of transcription 3 (STAT3). To elucidate the as yet undefined mechanisms that regulate this process, we demonstrate that higher levels of STAT3 transcription are found in livers of heterozygous SPTBN1(+/-) mice as compared to that of wild type mice. We also found increased levels of STAT3 mRNA, STAT3 protein, and p-STAT3 in human HCC cell-lines after knockdown of SPTBN1 or SMAD3, which promoted cell colony formation. Inhibition of STAT3 overrode the increase in cell colony formation due to knockdown of SPTBN1 or SMAD3. We also found that inhibition of SPTBN1 or SMAD3 upregulated STAT3 promoter activity in HCC cell-lines, which is dependent upon the cAMP-response element (CRE) and STAT-binding element (SBE) sites of the STAT3 promoter. Mechanistically, suppression of SPTBN1 and SMAD3 augmented the transcription of STAT3 by upregulating the CRE-binding proteins ATF3 and CREB2 and augmented the binding of those proteins to the regions within or upstream of the CRE site of the STAT3 promoter. Finally, in human HCC tissues, SPTBN1 expression correlated negatively with expression levels of STAT3, ATF3, and CREB2; SMAD3 expression correlated negatively with STAT3 expression; and the level of phosphorylated SMAD3 (p-SMAD3) correlated negatively with ATF3 and CREB2 protein levels. SPTBN1 and SMAD3 collaborate with CRE-binding transcription factors to inhibit STAT3, thereby preventing HCC development.


Cancer Informatics | 2013

In Silico Discovery of Mitosis Regulation Networks Associated with Early Distant Metastases in Estrogen Receptor Positive Breast Cancers

Yuriy Gusev; Rebecca B. Riggins; Krithika Bhuvaneshwar; Robinder Gauba; Laura Sheahan; Robert Clarke; Subha Madhavan

The aim of this study was to perform comparative analysis of multiple public datasets of gene expression in order to identify common genes as potential prognostic biomarkers. Additionally, the study sought to identify biological processes and pathways that are most significantly associated with early distant metastases (<5 years) in women with estrogen receptor-positive (ER+) breast tumors. Datasets from three published studies were selected for in silico analysis of gene expression profiles of ER+ breast cancer, using time to distant metastasis as the clinical endpoint. A subset of 44 differently expressed genes (DEGs) was found common to all three studies and characterized by mitotic checkpoint genes and pathways that regulate mitotic spindle and chromosome dynamics. DEG promoter regions were enriched with NFY binding sites. Analysis of miRNA target sites identified significant enrichment of miR-192, miR-193B, and miR-16–1 targets. Aberrant mitotic regulation could drive increased genomic instability leading to a progression towards an early onset metastatic phenotype. The relative importance of mitotic instability may reflect the clinical utility of mitotic poisons in metastatic breast cancer, including poisons such as the taxanes, epothilones, and vinca alkaloids.


Pharmacogenetics and Genomics | 2014

Pharmacogenomic characterization of gemcitabine response--a framework for data integration to enable personalized medicine.

Michael Harris; Krithika Bhuvaneshwar; Thanemozhi G. Natarajan; Laura Sheahan; Difei Wang; Mahlet G. Tadesse; Ira Shoulson; Ross Filice; Kenneth Steadman; Michael J. Pishvaian; Subha Madhavan; John F. Deeken

Objectives Response to the oncology drug gemcitabine may be variable in part due to genetic differences in the enzymes and transporters responsible for its metabolism and disposition. The aim of our in-silico study was to identify gene variants significantly associated with gemcitabine response that may help to personalize treatment in the clinic. Methods We analyzed two independent data sets: (a) genotype data from NCI-60 cell lines using the Affymetrix DMET 1.0 platform combined with gemcitabine cytotoxicity data in those cell lines, and (b) genome-wide association studies (GWAS) data from 351 pancreatic cancer patients treated on an NCI-sponsored phase III clinical trial. We also performed a subset analysis on the GWAS data set for 135 patients who were given gemcitabine+placebo. Statistical and systems biology analyses were performed on each individual data set to identify biomarkers significantly associated with gemcitabine response. Results Genetic variants in the ABC transporters (ABCC1, ABCC4) and the CYP4 family members CYP4F8 and CYP4F12, CHST3, and PPARD were found to be significant in both the NCI-60 and GWAS data sets. We report significant association between drug response and variants within members of the chondroitin sulfotransferase family (CHST) whose role in gemcitabine response is yet to be delineated. Conclusion Biomarkers identified in this integrative analysis may contribute insights into gemcitabine response variability. As genotype data become more readily available, similar studies can be conducted to gain insights into drug response mechanisms and to facilitate clinical trial design and regulatory reviews.


Genome Biology | 2011

G-CODE: enabling systems medicine through innovative informatics

Subha Madhavan; Yuriy Gusev; Michael Harris; David M. Tanenbaum; Robinder Gauba; Krithika Bhuvaneshwar; Andrew Shinohara; Kevin Rosso; Lavinia A. Carabet; Lei Song; Rebecca B. Riggins; Sivanesan Dakshanamurthy; Yue Wang; Stephen W. Byers; Robert Clarke; Louis M. Weiner

The new and emerging field of systems medicine, an application of systems biology approaches to biomedical problems in the clinical setting, leverages complex computational tools and high dimensional data to derive personalized assessments of disease risk. Systems medicine offers the potential for more effective individualized diagnosis, prognosis and treatment options. The Georgetown Clinical & Omics Development Engine (G-CODE) is a generic and flexible web-based platform that serves to allow basic, translational and clinical research activities by integrating patient characteristics and clinical outcome data with a variety of high-throughput research data in a unified environment to enable systems medicine. Through this modular, extensible and flexible infrastructure, we can quickly and easily assemble new translational web applications with both analytic and generic administrative features. New analytic functionalities specific to the needs of a particular disease community can easily be added within this modular architecture. With G-CODE, we hope to help enable the creation of new disease-centric portals, as well as the widespread use of biomedical informatics tools by basic, clinical and translational researchers, through providing powerful analytic tools and capabilities within easy-to-use interfaces that can be customized to the needs of each research community. This infrastructure was first deployed in the form of the Georgetown Database of Cancer (G-DOC) [1], which includes a broad collection of bioinformatics and systems biology tools for analysis and visualization of four major omics types: DNA, mRNA, microRNA and metabolites. Although several rich data repositories for high dimensional research data exist in the public domain, most focus on a single data type and do not support integration across multiple technologies. G-DOC contains data for more than 2,500 patients with breast cancer and almost 800 patients with gastrointestinal cancer, all of which are handled in a manner that allows maximum integration. We believe that G-DOC will help facilitate systems medicine by allowing easy identification of trends and patterns in integrated datasets and will hence facilitate the use of better targeted therapies for cancer. One obvious area for expansion of the G-CODE/G-DOC platform infrastructure is to support next-generation sequencing (NGS), which is a highly enabling and transformative emerging technology for the biomedical sciences. Nonetheless, effective utilization of these data is impeded by the substantial handling, manipulation and analysis requirements that are entailed. We have concluded that cloud computing is well positioned to fill these gaps, as this type of infrastructure permits rapid scaling with low input costs. As such, the Georgetown University team is exploring the use of the Amazon EC2 cloud and the Galaxy platform to process whole exome, whole genome, RNA-Seq and chromatin immunoprecipitation (ChIP)-Seq NGS data. The processed NGS data will be integrated into G-DOC to ensure that they can be analyzed in the full context of other omics data. Likewise, all G-CODE projects will simultaneously benefit from these advances in NGS data handling. Through technology re-use, the G-CODE infrastructure will accelerate progress in a variety of ongoing programs that are in need of integrative multi-omics analysis and will advance our opportunities to practice effective systems medicine in the near future.


Oncotarget | 2017

EGR1 regulates cellular metabolism and survival in endocrine resistant breast cancer

Ayesha N. Shajahan-Haq; Simina M. Boca; Lu Jin; Krithika Bhuvaneshwar; Yuriy Gusev; Amrita K. Cheema; Diane Demas; Kristopher S. Raghavan; Ryan D. Michalek; Subha Madhavan; Robert B. Clarke

About 70% of all breast cancers are estrogen receptor alpha positive (ER+; ESR1). Many are treated with antiestrogens. Unfortunately, de novo and acquired resistance to antiestrogens is common but the underlying mechanisms remain unclear. Since growth of cancer cells is dependent on adequate energy and metabolites, the metabolomic profile of endocrine resistant breast cancers likely contains features that are deterministic of cell fate. Thus, we integrated data from metabolomic and transcriptomic analyses of ER+ MCF7-derived breast cancer cells that are antiestrogen sensitive (LCC1) or resistant (LCC9) that resulted in a gene-metabolite network associated with EGR1 (early growth response 1). In human ER+ breast tumors treated with endocrine therapy, higher EGR1 expression was associated with a more favorable prognosis. Mechanistic studies showed that knockdown of EGR1 inhibited cell growth in both cells and EGR1 overexpression did not affect antiestrogen sensitivity. Comparing metabolite profiles in LCC9 cells following perturbation of EGR1 showed interruption of lipid metabolism. Tolfenamic acid, an anti-inflammatory drug, decreased EGR1 protein levels and synergized with antiestrogens in inhibiting cell proliferation in LCC9 cells. Collectively, these findings indicate that EGR1 is an important regulator of breast cancer cell metabolism and is a promising target to prevent or reverse endocrine resistance.About 70% of all breast cancers are estrogen receptor alpha positive (ER+; ESR1). Many are treated with antiestrogens. Unfortunately, de novo and acquired resistance to antiestrogens is common but the underlying mechanisms remain unclear. Since growth of cancer cells is dependent on adequate energy and metabolites, the metabolomic profile of endocrine resistant breast cancers likely contains features that are deterministic of cell fate. Thus, we integrated data from metabolomic and transcriptomic analyses of ER+ MCF7-derived breast cancer cells that are antiestrogen sensitive (LCC1) or resistant (LCC9) that resulted in a gene-metabolite network associated with EGR1 (early growth response 1). In human ER+ breast tumors treated with endocrine therapy, higher EGR1 expression was associated with a more favorable prognosis. Mechanistic studies showed that knockdown of EGR1 inhibited cell growth in both cells and EGR1 overexpression did not affect antiestrogen sensitivity. Comparing metabolite profiles in LCC9 cells following perturbation of EGR1 showed interruption of lipid metabolism. Tolfenamic acid, an anti-inflammatory drug, decreased EGR1 protein levels and synergized with antiestrogens in inhibiting cell proliferation in LCC9 cells. Collectively, these findings indicate that EGR1 is an important regulator of breast cancer cell metabolism and is a promising target to prevent or reverse endocrine resistance.


Cancer Informatics | 2017

CINdex: A Bioconductor Package for Analysis of Chromosome Instability in DNA Copy Number Data

Lei Song; Krithika Bhuvaneshwar; Yue Wang; Yuanjian Feng; Ie Ming Shih; Subha Madhavan; Yuriy Gusev

The CINdex Bioconductor package addresses an important area of high-throughput genomic analysis. It calculates the chromosome instability (CIN) index, a novel measurement that quantitatively characterizes genome-wide copy number alterations (CNAs) as a measure of CIN. The advantage of this package is an ability to compare CIN index values between several groups for patients (case and control groups), which is a typical use case in translational research. The differentially changed cytobands or chromosomes can then be linked to genes located in the affected genomic regions, as well as pathways. This enables in-depth systems biology–based network analysis and assessment of the impact of CNA on various biological processes or clinical outcomes. This package was successfully applied to analysis of DNA copy number data in colorectal cancer as a part of multi-omics integrative study as well as for analysis of several other cancer types. The source code, along with an end-to-end tutorial, and example data are freely available in Bioconductor at http://bioconductor.org/packages/CINdex/.

Collaboration


Dive into the Krithika Bhuvaneshwar's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Lei Song

Georgetown University Medical Center

View shared research outputs
Top Co-Authors

Avatar

Robert Clarke

University of Washington

View shared research outputs
Top Co-Authors

Avatar

Louis M. Weiner

Georgetown University Medical Center

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Robinder Gauba

Georgetown University Medical Center

View shared research outputs
Top Co-Authors

Avatar

Amrita K. Cheema

Georgetown University Medical Center

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge