Network


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

Hotspot


Dive into the research topics where Eugene A. Volpe is active.

Publication


Featured researches published by Eugene A. Volpe.


Oncogene | 2007

Disruption of transforming growth factor-β signaling through β-spectrin ELF leads to hepatocellular cancer through cyclin D1 activation

Krit Kitisin; Natarajan Ganesan; Yi Tang; Wilma Jogunoori; Eugene A. Volpe; Sang-Soo Kim; Varalakshmi Katuri; Bhaskar Kallakury; Michael J. Pishvaian; Chris Albanese; Jonathan Mendelson; Michael Zasloff; Asif Rashid; T. Fishbein; Stephen R.T. Evans; A. Sidawy; E P Reddy; Bibhuti Mishra; Lynt B. Johnson; Kirti Shetty; Lopa Mishra

Transforming growth factor-β (TGF-β) signaling members, TGF-β receptor type II (TBRII), Smad2, Smad4 and Smad adaptor, embryonic liver fodrin (ELF), are prominent tumor suppressors in gastrointestinal cancers. Here, we show that 40% of elf+/− mice spontaneously develop hepatocellular cancer (HCC) with markedly increased cyclin D1, cyclin-dependent kinase 4 (Cdk4), c-Myc and MDM2 expression. Reduced ELF but not TBRII, or Smad4 was observed in 8 of 9 human HCCs (P<0.017). ELF and TBRII are also markedly decreased in human HCC cell lines SNU-398 and SNU-475. Restoration of ELF and TBRII in SNU-398 cells markedly decreases cyclin D1 as well as hyperphosphorylated-retinoblastoma (hyperphosphorylated-pRb). Thus, we show that TGF-β signaling and Smad adaptor ELF suppress human hepatocarcinogenesis, potentially through cyclin D1 deregulation. Loss of ELF could serve as a primary event in progression toward a fully transformed phenotype and could hold promise for new therapeutic approaches in human HCCs.


Oncogene | 2005

Inactivation of ELF/TGF- β signaling in human gastrointestinal cancer

Varalakshmi Katuri; Yi Tang; Blair Marshall; Asif Rashid; Wilma Jogunoori; Eugene A. Volpe; Anton N. Sidawy; Stephen R.T. Evans; Jonathan Blay; G. Ian Gallicano; E. Premkumar Reddy; Lopa Mishra; Bibhuti Mishra

TGF-β/Smads regulate a wide variety of biological responses through transcriptional regulation of target genes. ELF, a β-spectrin, plays a key role in the transmission of TGF-β-mediated transcriptional response through Smads. ELF was originally identified as a key protein involved in endodermal stem/progenitor cells committed to foregut lineage. Also, as a major dynamic adaptor and scaffolding protein, ELF is important for the generation of functionally distinct membranes, protein sorting and the development of polarized differentiated epithelial cells. Disruption of elf results in the loss of Smad3/Smad4 activation and, therefore, a disruption of the TGF-β pathway. These observations led us to pursue the function of ELF in gastrointestinal (GI) epithelial cell–cell adhesion and tumor suppression. Here, we show a significant loss of ELF and reduced Smad4 expression in human gastric cancer tissue samples. Also, of the six human gastric cancer cell lines examined, three show deficient ELF expression. Furthermore, we demonstrate the rescue of E-cadherin-dependent homophilic cell–cell adhesion by ectopic expression of full-length elf. Our results suggest that ELF has an essential role in tumor suppression in GI cancers.


Oncogene | 2006

RING finger-dependent ubiquitination by PRAJA is dependent on TGF- β and potentially defines the functional status of the tumor suppressor ELF

T. Saha; D. Vardhini; Yi Tang; Varalakshmi Katuri; Wilma Jogunoori; Eugene A. Volpe; Dale S. Haines; A. Sidawy; X. Zhou; I. Gallicano; Richard Schlegel; Bibhuti Mishra; Lopa Mishra

In gastrointestinal cells, biological signals for transforming growth factor-beta (TGF-β) are transduced through transmembrane serine/threonine kinase receptors that signal to Smad proteins. Smad4, a tumor suppressor, is often mutated in human gastrointestinal cancers. The mechanism of Smad4 inactivation, however, remains uncertain and could be through E3-mediated ubiquitination of Smad4/adaptor protein complexes. Disruption of ELF (embryonic liver fodrin), a Smad4 adaptor protein, modulates TGF-β signaling. We have found that PRAJA, a RING-H2 protein, interacts with ELF in a TGF-β-dependent manner, with a fivefold increase of PRAJA expression and a subsequent decrease in ELF and Smad4 expression, in gastrointestinal cancer cell lines (P<0.05). Strikingly, PRAJA manifests substantial E3-dependent ubiquitination of ELF and Smad3, but not Smad4. Δ-PRAJA, which has a deleted RING finger domain at the C terminus, abolishes ubiquitination of ELF. A stable cell line that overexpresses PRAJA exhibits low levels of ELF in comparison to a Δ-PRAJA stable cell line, where ELF expression is high compared to normal controls. The alteration of ELF and/or Smad4 expression and/or function in the TGF-β signaling pathway may be induced by enhancement of ELF degradation, which is mediated by a high-level expression of PRAJA in gastrointestinal cancers. In hepatocytes, half-life (t1/2) and rate constant for degradation (kD) of ELF is 1.91 h and 21.72 min−1 when coupled with ectopic expression of PRAJA in cells stimulated by TGF-β, compared to PRAJA-transfected unstimulated cells (t1/2=4.33 h and kD=9.6 min−1). These studies reveal a mechanism for tumorigenesis whereby defects in adaptor proteins for Smads, such as ELF, can undergo degradation by PRAJA, through the ubiquitin-mediated pathway.


Gastroenterology | 2008

W1964 Telomerase Reverse Transcriptase Regulation By TGF-β Signaling Through Adaptor ELF and SMAD3 That Is Independent of C-MYC

Zhixing Yao; Young-Woo Kim; Rupen Amin; Eugene A. Volpe; Wilma Jogunoori; Lopa Mishra; Bibhutibushan Mishra

3442 Transforming Growth Factor-β (TGF-β) induced antiproliferative responses is a hallmark of many cancer cells. Smad proteins as intracellular mediators for TGF-β signaling and regulate target gene expression by activating or repressing gene transcription. Smads 2, 3 and 4 inhibit G1/S cell cycle progression mostly by suppression of c-Myc and cyclin-dependent kinases (cdks). Myc amplification, telomere maintenance, and telomerase reverse transcriptase (TERT) reactivation are common features of human foregut cancers, such as hepatocellular carcinoma (HCC). Emerging evidence indicates that ELF, a Smad3/Smad4 adaptor protein required for TGF-β signaling, is a powerful tumor suppressor [Science, 2005, 310(5745):68-71, Oncogene, 2007, 26(50):7103-10]. Elf+/-, elf+/-/Smad3+/- and elf+/-/Smad4+/-, (but not Smad3-/-) mice dramatically develop foregut cancers, including hepatocellular, pancreatic and gastric cancers. However, the specific role(s) of h-TERT, c-MYC and Elf, and their relation to the TGF-β pathway, in foregut cancer formation are poorly understood. Deletion of ELF results in a dramatic and spontaneous formation of liver and gastrointestinal cancers, with exon 15 mutations in 11% of human HCC and gastric cancer cell lines tested. Elf+/-andelf+/-/Smad3+/- mice develop visceromegaly and multiple GI cancers (70% of mice), spontaneously. This phenotype provides compelling evidence that elf+/-and elf+/-/Smad3+/- mice are a model of the hereditary human cancer syndrome Beckwith-Wiedemann (BWS). Aims: In this study, we investigated the mechanism of role of Elf, Smad3 and c-Myc in regulating human TERT gene expression by TGF-β in HepG2, PLC/PRF/5, SNU 298 cell lines and elf+/-/Smad3+/- mice tumor tissues in vitro and in vivo. Results show that: 1) TheElf+/-/Smad3+/-mice develop visceromegaly and multiple cancers, including metastatic pancreatic, hepatocellular, small bowel lymphomas, adrenocortical carcinomas, renal carcinomas and tumor tissues from elf+/-/Smad3+/- mice show a dramatic decrease of ELF mRNA; 2) TERT and c-Myc are markedly elevated in elf+/-andelf+/-/Smad3+/- mice; Interestingly, TERT levels are far higher than can be accounted for by c-Myc levels in elf+/-andelf+/-/Smad3+/- tumors; 3)Ectopic ELF and Smad3 suppress TERT greater than c-Myc in the absence of TGF-β. 4) Both ELF and Smad3 associate with c-Myc in TGF-β stimulated hepatocytes and suppress TERT. 5)Overexpression of ELF and/or Smad3 decreases hTERT RNA levels in PLC/PRF/5 and SNU 298 human cell lines. Conclusions: Taken together our preliminary data suggest that divergent pathways converge on ELF and Smad3 that then regulate TERT. Inactivation of the TGF-β signaling pathway with TERT activation provides a strong strategy for generating targeted therapeutics at TERT in these lethal human cancers.


Journal of the American Chemical Society | 2004

Isoxazolyl-Serine-Based Agonists of Peroxisome Proliferator-Activated Receptor: Design, Synthesis, and Effects on Cardiomyocyte Differentiation

Zhi Liang Wei; Pavel A. Petukhov; Fero Bizik; Joaquim Teixeira; Mark Mercola; Eugene A. Volpe; Robert I. Glazer; Timothy M. Willson; Alan P. Kozikowski


Biochemical and Biophysical Research Communications | 2006

Inactivation of TGF-β signaling in lung cancer results in increased CDK4 activity that can be rescued by ELF

Hye Jung Baek; Sang-Soo Kim; Fabio May da Silva; Eugene A. Volpe; Stephen R.T. Evans; Bibhuti Mishra; Lopa Mishra; M. Blair Marshall


Archive | 2008

Peptides and proteins for early liver development and anitibodies thereto, and their use in therapeutic diagnosis and treatment

Lopa Mishra; Yi Tang; Kri Kitisin; Eugene A. Volpe; Jonathan Mendelson; Wilma Jogunoori; Cuiling Li; Chuxia Deng; Susette C. Mueller; Habtom W. Ressom; Asif Rashid; Aiwu Ruthhe; Anton Sidaway; Stephen R.T. Evans; Kirti Shetty; Michael Zasloff; Bibhuti Mishra; E. P. Reddy; Johnson Lynt


Cancer Research | 2008

CDK4 is an important target for hepatocellular cancer with inactivation of TGF-beta signaling through Smad and ELF

Michael J. Pishvaian; Geeta Upadhyay; Zhixing Yao; Young D. Kim; Anan Said; Jon Mendelson; Krit Kitisin; Eugene A. Volpe; Tiffany Blake; Sang Kim; Hae Baek; Anthony D. Kang; Bibhuti Mishra; E. Premkumar Reddy; Lopa Mishra


Gastroenterology | 2008

M2040 Expression of Proteins Involved in TGF-Beta Signaling Pathway in Human Pancreatic Cancer Cell Lines

Eugene A. Volpe; Bibhutibushan Mishra; Lopa Mishra; Stephen R.T. Evans


Journal of The American College of Surgeons | 2007

Hepatocellular cancer loses tgf-beta signaling progenitor/stem cell niche, and is suppressed by crosstalk between TGF-beta and IL-6 signaling pathways

Krit Kitisin; Yi Tang; Varalakshmi Katuri; Eugene A. Volpe; Jonathan Mendelson; Wilma Jogunoori; Kirti Shetty; Stephen R.T. Evans; Bibhuti Mishra; Lopa Mishra; Lynt B. Johnson

Collaboration


Dive into the Eugene A. Volpe's collaboration.

Top Co-Authors

Avatar

Lopa Mishra

George Washington University

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Yi Tang

Georgetown University

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Lynt B. Johnson

MedStar Georgetown University Hospital

View shared research outputs
Top Co-Authors

Avatar

Kirti Shetty

Johns Hopkins University

View shared research outputs
Top Co-Authors

Avatar

Asif Rashid

University of Texas MD Anderson Cancer Center

View shared research outputs
Researchain Logo
Decentralizing Knowledge