Network


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

Hotspot


Dive into the research topics where Kolaparthi Venkatasubbarao is active.

Publication


Featured researches published by Kolaparthi Venkatasubbarao.


Journal of Biological Chemistry | 2005

Trichostatin A induces transforming growth factor β type II receptor promoter activity and acetylation of Sp1 by recruitment of PCAF/p300 to a Sp1.NF-Y complex

Weiqi Huang; Shujie Zhao; Sudhakar Ammanamanchi; Michael G. Brattain; Kolaparthi Venkatasubbarao; James W. Freeman

Transforming growth factor β type II receptor (TβRII) is a tumor suppressor gene that can be transcriptionally silenced by histone deacetylases (HDACs) in cancer cells. In this report, we demonstrated the mechanism by which trichostatin A (TSA), an inhibitor of HDAC, induces the expression of TβRII in human pancreatic cancer cell lines by modulating the transcriptional components that bind a specific DNA region of the TβRII promoter. This region of the TβRII promoter possesses Sp1 and NF-Y binding sites in close proximity (located at –102 and –83, respectively). Treatment of cells with TSA activates the TβRII promoter in a time-dependent manner through the recruitment of p300 and PCAF into a Sp1·NF-Y·HDAC complex that binds this DNA element. The recruitment of p300 and PCAF into the complex is associated with a concomitant acetylation of Sp1 and an overall decrease in the amount of HDAC associated with the complex. Transient overexpression of p300 or PCAF potentiated TSA-induced TβRII promoter activity. The effect of PCAF was dependent on its histone acetyltransferase activity, whereas that of p300 was independent. Stable transfection of PCAF caused an increase in TβRII mRNA expression, the association of PCAF with TβRII promoter, and the acetylation of Sp1. Taken together, these results showed that TSA treatment of pancreatic cancer cells leads to transcriptional activation of the TβRII promoter through modulation of the components of a Sp1·NF-Y·p300·PCAF·HDAC-1 multiprotein complex. Moreover, the interaction of NF-Y with the Sp1-associated complex may further explain why this specific Sp1 site mediates transcriptional responsiveness to TSA.


Cancer Research | 2008

Inhibition of STAT3Tyr705 Phosphorylation by Smad4 Suppresses Transforming Growth Factor β–Mediated Invasion and Metastasis in Pancreatic Cancer Cells

Shujie Zhao; Kolaparthi Venkatasubbarao; Jillian W. Lazor; Jane Sperry; Changqing Jin; Lin Cao; James W. Freeman

The role of Smad4 in transforming growth factor beta (TGFbeta)-mediated epithelial-mesenchymal transition (EMT), invasion, and metastasis was investigated using isogenically matched pancreatic cancer cell lines that differed only in expression of Smad4. Cells expressing Smad4 showed an enhanced TGFbeta-mediated EMT as determined by increased expression of vimentin and decreased expression of beta-catenin and E-cadherin. TGFbeta-mediated invasion was suppressed in Smad4-intact cells as determined by in vitro assays, and these cells showed a reduced metastasis in an orthotopic model of pancreatic cancer. Interestingly, TGFbeta inhibited STAT3(Tyr705) phosphorylation in Smad4-intact cells. The decrease in STAT3(Tyr705) phosphorylation was linked to a TGFbeta/Smad4-dependent and enhanced activation of extracellular signal-regulated kinases, which caused an increase in serine phosphorylation of STAT3(Ser727). Down-regulating signal transducer and activator of transcription 3 (STAT3) expression by short hairpin RNA in Smad4-deficient cells prevented TGFbeta-induced invasion. Conversely, expressing a constitutively activated form of STAT3 (STAT3-C) in Smad4-intact cells enhanced invasion. This study indicates the requirement of STAT3 activity for TGFbeta-induced invasion in pancreatic cancer cells and implicates Smad4-dependent signaling in regulating STAT3 activity. These findings further suggest that loss of Smad4, leading to aberrant activation of STAT3, contributes to the switch of TGFbeta from a tumor-suppressive to a tumor-promoting pathway in pancreatic cancer.


Journal of Biological Chemistry | 1997

Ionizing radiation-inducible apoptosis in the absence of p53 linked to transcription factor EGR-1.

Mansoor M. Ahmed; Stephen F. Sells; Kolaparthi Venkatasubbarao; Sana M. Fruitwala; Sumathi Muthukkumar; Cindy Harp; Mohammed Mohiuddin; Vivek M. Rangnekar

The tumor suppressor protein p53 is a pivotal regulator of apoptosis, and prostate cancer cells that lack p53 protein are moderately resistant to apoptotic death by ionizing radiation. Genes encoding the transcription factor early growth response-1 (EGR-1) and cytokine tumor necrosis factor-α (TNF-α) were induced upon irradiation of prostate cancer cells, and inhibition of EGR-1 function resulted in abrogation of both TNF-α induction and apoptosis. Induction of the TNF-α gene by ionizing radiation and EGR-1 was mediated via a GC-rich EGR-1-binding motif in the TNF-α promoter. Because TNF-α induces apoptosis in prostate cancer cells, these findings suggest that, in the absence of p53, ionizing radiation-inducible apoptosis is mediated by EGR-1 via TNF-α transactivation.


Journal of Biological Chemistry | 1996

EGR-1 INDUCTION IS REQUIRED FOR MAXIMAL RADIOSENSITIVITY IN A375-C6 MELANOMA CELLS

Mansoor M. Ahmed; Kolaparthi Venkatasubbarao; Sana M. Fruitwala; Sumathi Muthukkumar; David P. Wood; Stephen F. Sells; Mohammed Mohiuddin; Vivek M. Rangnekar

Exposure to ionizing radiation leads to induction of the immediate-early gene, early growth response-1 (Egr-1). Previous studies have suggested distinct cell type- and inducer-specific roles for EGR-1 protein in cellular growth inhibition. The present study was undertaken to determine the functional role of EGR-1 in growth inhibition caused by exposure of tumor cells to ionizing radiation. Exposure to ionizing radiation caused induction of EGR-1 protein in human melanoma cells A375-C6. Inhibition of either the function of EGR-1 protein by stable transfection with a dominant-negative mutant or the expression of EGR-1 by transient transfection with an antisense oligomer resulted in a diminished growth-inhibitory response to ionizing radiation. Because previous studies have suggested that mutations in the tumor-suppressor gene p53 confer radio-resistance, we examined the p53 status of A375-C6 cells. Interestingly, both the parental and the transfected A375-C6 cells showed trisomy for wild-type p53 alleles. Exposure to ionizing radiation resulted in induction of p53 protein that localized to the nucleus in A375-C6 cells. These data suggest that inhibition of EGR-1 function confers radio resistance despite the induction of wild-type nuclear p53. Thus, EGR-1 is required for the growth-inhibitory response to ionizing radiation in A375-C6 cells.


Cancer Research | 2005

Farnesyl transferase inhibitor (R115777)-induced inhibition of STAT3(Tyr705) phosphorylation in human pancreatic cancer cell lines require extracellular signal-regulated kinases.

Kolaparthi Venkatasubbarao; Ahsan Choudary; James W. Freeman

In this study, we report that R115777, a nonpeptidomimetic farnesyl transferase inhibitor, suppresses the growth of human pancreatic adenocarcinoma cell lines and that this growth inhibition is associated with modulation in the phosphorylation levels of signal transducers and activators of transcription 3 (STAT3) and extracellular signal-regulated kinases (ERK). Treatment of cells with R115777 inhibited the tyrosine phosphorylation of STAT3((Tyr705)), while increasing the serine phosphorylation of STAT3((Ser727)). We found the differential phosphorylation of STAT3 was due to an increased and prolonged activation of ERKs. The biological significance of ERK-mediated inhibition of STAT3((Tyr705)) phosphorylation was further assessed by treating the cells with an inhibitor (PD98059) of mitogen-activated protein kinase kinase (MEK) or by transfecting the cells with a vector that expresses constitutively active MEK-1. Expression of constitutively active MEK-1 caused an increase of ERK activity and inhibited STAT3((Tyr705)) phosphorylation. Conversely, inhibition of ERK activity by PD98059 reversed the R115777-induced inhibition of STAT3((Tyr705)) phosphorylation. R115777 also caused the inhibition of the binding of STAT3 to its consensus binding element. An increase in the activation of ERKs either by overexpressing MEK-1 or treatment of cells with R115777 caused an up-regulation in the levels of a cyclin-dependent kinase (cdk) inhibitor, p21(cip1/waf1). These observations suggest that R115777-induced growth inhibition is partly due to the prolonged activation of ERKs that mediates an inhibition of STAT3((Tyr705)) phosphorylation and an increase in the levels of p21(cip1/waf1) in human pancreatic adenocarcinoma cell lines.


Genes, Chromosomes and Cancer | 1998

Novel mutations in the polyadenine tract of the transforming growth factor β type II receptor gene are found in a subpopulation of human pancreatic adenocarcinomas

Kolaparthi Venkatasubbarao; Mansoor M. Ahmed; Carol Swiderski; Cindy Harp; Eun Y. Lee; Patrick C. McGrath; Mohammed Mohiuddin; William E. Strodel; James W. Freeman

In this study, we determined the incidence of microsatellite instability (MIN) in pancreatic adenocarcinoma and determined whether MIN might target, for mutations, the simple nucleotide repeats of the transforming growth factor β type II receptor (TGFBR2) gene. Forty‐eight surgically resected pancreatic tumor tissue samples and two normal pancreas tissue samples were analyzed in this study. Microsatellite analysis was performed for six loci in 14 of the 48 tumor specimens for which we had matching normal genomic DNA. Only four of the 14 tumors (29%) were MIN‐positive as determined by the presence of microsatellite variations in more than one locus. Interestingly, eight of the 14 specimens (57%) showed microsatellite variations or loss of heterozygosity at D18S34, suggesting that this locus may be a critical region of genetic instability in pancreatic tumorigenesis. Of the 48 tumors, only two (4%) showed mutations in the polyA region, one of the MIN‐targeted sites of the TGFBR2 gene. DNA sequence analysis of these two specimens showed the presence of a two‐base deletion in one tumor specimen and the other tumor specimen showed a base substitution in the polyA tract at codon 128 of the TGFBR2 gene. The fact that these mutations occurred in the polyA tract of some pancreatic tumors suggests that a subpopulation of these tumors may be susceptible to MIN‐targeted mutations. The incidence of these mutations are low and similar to that reported for nonhereditary, sporadic colon cancers. Genes Chromosomes Cancer 22:138–144, 1998.


PLOS ONE | 2014

A miRNA signature of chemoresistant mesenchymal phenotype identifies novel molecular targets associated with advanced pancreatic cancer.

Alakesh Bera; Kolaparthi Venkatasubbarao; Muthu Saravanan Manoharan; Ping Hill; James W. Freeman

In this study a microRNA (miRNA) signature was identified in a gemcitabine resistant pancreatic ductal adenocarcinoma (PDAC) cell line model (BxPC3-GZR) and this signature was further examined in advanced PDAC tumor specimens from The Cancer Genome Atlas (TCGA) database. BxPC3-GZR showed a mesenchymal phenotype, expressed high levels of CD44 and showed a highly significant deregulation of 17 miRNAs. Based on relevance to cancer, a seven-miRNA signature (miR-100, miR-125b, miR-155, miR-21, miR-205, miR-27b and miR-455-3p) was selected for further studies. A strong correlation was observed for six of the seven miRNAs in 43 advanced tumor specimens compared to normal pancreas tissue. To assess the functional relevance we initially focused on miRNA-125b, which is over-expressed in both the BxPC3-GZR model and advanced PDAC tumor specimens. Knockdown of miRNA-125b in BxPC3-GZR and Panc-1 cells caused a partial reversal of the mesenchymal phenotype and enhanced response to gemcitabine. Moreover, RNA-seq data from each of 40 advanced PDAC tumor specimens from the TCGA data base indicate a negative correlation between expression of miRNA-125b and five of six potential target genes (BAP1, BBC3, NEU1, BCL2, STARD13). Thus far, two of these target genes, BBC3 and NEU1, that are tumor suppressor genes but not yet studied in PDAC, appear to be functional targets of miR-125b since knockdown of miR125b caused their up regulation. These miRNAs and their molecular targets may serve as targets to enhance sensitivity to chemotherapy and reduce metastatic spread.


International Journal of Cancer | 2008

Downregulation of PAR-4, a pro-apoptotic gene, in pancreatic tumors harboring K-ras mutation

Mansoor M. Ahmed; David G. Sheldon; Mushtaq A. Fruitwala; Kolaparthi Venkatasubbarao; Eun Y. Lee; Seema Gupta; Craig Wood; Mohammed Mohiuddin; William E. Strodel

Oncogenic ras is known to inhibit cell death and growth inhibitory genes and activate prosurvival genes. Proapoptotic gene PAR‐4, has been found to be downregulated by oncogenic ras. Since pancreatic tumors harbor a high incidence of K‐ras point mutations, we hypothesized that oncogenic K‐ras might influence the function and expression of PAR‐4. PAR‐4 expression levels were analyzed in 4 established pancreatic tumor cell lines, 10 normal pancreatic tissues, 44 frozen tumor tissues and 25 paraffin‐embedded pancreatic adenocarcinoma samples by Real Time RT‐PCR, Western blot analysis and immunohistochemistry. K‐ras mutational status was analyzed by allele‐specific oligonucleotide‐hybridization. Expression levels of PAR‐4 were correlated with the K‐ras mutational status and clinical characteristics. Further, modulation of endogenous PAR‐4 was tested by transiently expressing oncogenic ras in a wild‐type K‐ras pancreatic cancer cell line, BxPC‐3. Three cell lines with K‐ras mutations showed low levels of PAR‐4 when compared to a normal pancreatic tissue. Of 44 frozen tumors, 16 showed appreciable upregulation of Par mRNA and 27 showed significant downregulation of PAR‐4 mRNA when compared to normal pancreatic tissue and 1 had levels equivalent to normal pancreatic tissue. Of 25 paraffin‐embedded tumors, 9 showed downregulation of PAR‐4 protein and this downregulation of PAR‐4 correlated significantly with K‐ras mutational status (p < 0.00002). In addition, the presence of PAR‐4 mRNA or protein expression in pancreatic tumors correlated with prolonged survival. Transient overexpression of oncogenic ras in wild‐type K‐ras BxPC‐3 cells significantly downregulated the endogenous PAR‐4 protein levels and conferred accelerated growth. Thus, downregulation or loss of PAR‐4 expression by oncogenic ras may provide a selective survival advantage for pancreatic tumors, through inhibition of proapoptotic pathway mediated by PAR‐4.


Molecular Cancer | 2013

Inhibiting signal transducer and activator of transcription-3 increases response to gemcitabine and delays progression of pancreatic cancer

Kolaparthi Venkatasubbarao; Lindsay L. Peterson; Shujie Zhao; Ping Hill; Lin Cao; Qing Zhou; Steffan T. Nawrocki; James W. Freeman

BackgroundAmong the solid tumors, human pancreatic ductal adenocarcinoma (PDAC) has the worst prognosis. Gemcitabine is the standard first line of therapy for pancreatic cancer but has limited efficacy due to inherent or rapid development of resistance and combining EGFR inhibitors with this regimen results in only a modest clinical benefit. The goal of this study was to identify molecular targets that are activated during gemcitabine therapy alone or in combination with an EGFR inhibitor.MethodsPDAC cell lines were used to determine molecular changes and rates of growth after treatment with gemcitabine or an EGFR inhibitor, AG1478, by Western blot analysis and MTT assays respectively. Flow cytometric analysis was performed to study the cell cycle progression and rate of apoptosis after gemcitabine treatment. ShRNA was used to knockdown STAT3. An in vivo orthotopic animal model was used to evaluate STAT3 as a target. Immunohistochemical analysis was performed to analyze Ki67 and STAT3 expression in tumors.ResultsTreatment with gemcitabine increased the levels of EGFRTyr1068 and ERK phosphorylation in the PDAC cell lines tested. The constitutive STAT3Tyr705 phosphorylation observed in PDAC cell lines was not altered by treatment with gemcitabine. Treatment of cells with gemcitabine or AG1478 resulted in differential rate of growth inhibition. AG1478 efficiently blocked the phosphorylation of EGFRTyr1068 and inhibited the phosphorylation of down-stream effectors AKT and ERKs, while STAT3Tyr705 phosphorylation remained unchanged. Combining these two agents neither induced synergistic growth suppression nor inhibited STAT3Tyr705 phosphorylation, thus prompting further studies to assess whether targeting STAT3 improves the response to gemcitabine or AG1478. Indeed, knockdown of STAT3 increased sensitivity to gemcitabine by inducing pro-apoptotic signals and by increasing G1 cell cycle arrest. However, knockdown of STAT3 did not enhance the growth inhibitory potential of AG1478. In vivo orthotopic animal model results show that knockdown of STAT3 caused a significant reduction in tumor burden and delayed tumor progression with increased response to gemcitabine associated with a decrease in the Ki-67 positive cells.ConclusionsThis study suggests that STAT3 should be considered an important molecular target for therapy of PDAC for enhancing the response to gemcitabine.


Frontiers in Bioscience | 2004

Alterations of cell signaling pathways in pancreatic cancer.

James W. Freeman; Daniel T. DeArmond; Michael Lake; Weiqi Huang; Kolaparthi Venkatasubbarao; Shujie Zhao

Pancreatic ductal adenocarcinomas continue to have the worst prognosis of any adult malignancy with a five-year survival rate of less than 4%. One approach to improve patient survival from pancreatic cancer is to identify new biological targets that contribute to the aggressive pathogenecity of this disease and to develop reagents that will interfere with the function of these targets. Apart from the identification of the genetic profile of pancreatic cancer, a number of studies have focused on aberrant cell signaling pathways and their role in pancreatic cancer biology and response to therapy. This review, although not comprehensive, will discuss the salient features of several of these pathways. These include the roles of TGF beta signaling in both tumor suppression and tumor promotion and the effects of deregulation of phosphotyrosine kinase receptor signaling pathways in pancreatic cancer.

Collaboration


Dive into the Kolaparthi Venkatasubbarao's collaboration.

Top Co-Authors

Avatar

James W. Freeman

University of Texas Health Science Center at San Antonio

View shared research outputs
Top Co-Authors

Avatar

Mansoor M. Ahmed

National Institutes of Health

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Shujie Zhao

University of Texas Health Science Center at San Antonio

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Eun Y. Lee

University of Kentucky

View shared research outputs
Top Co-Authors

Avatar

Cindy Harp

University of Kentucky

View shared research outputs
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge