Network


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

Hotspot


Dive into the research topics where Wenguo Yao is active.

Publication


Featured researches published by Wenguo Yao.


Gastroenterology | 2010

220 Targeting CEA Rescues Proliferation Inhibiting Effects of TGF-β Signaling Pathway in Colorectal Cancers

Hong Cao; Ying Li; Zhongxian Jiao; Wenguo Yao; Wilma Jogunoori; Arun Thenappan; Lopa Mishra

Carcinoembryonic antigen (CEA) is a tumor marker for the clinical management of colorectal cancer (CRC). The elevated blood levels of CEA are associated with metastasis and poor prognosis in CRC. There is mounting evidence that CEA enhances the metastatic potential of cancer cells. CEA increases the ability of weakly metastatic CRC to colonize the liver and to develop spontaneous hematogeneous liver and lung metastases. CEA expression has also been related with resistance to cytotoxic chemotherapy and to anoikis, a form of apoptosis caused by detachment from cell matrix. Yet the mechanism of CEA mediated metastasis is only partially understood. The TGF-β (transforming growth factor beta) signaling pathway contributes to tumorigenesis by controlling several biological processes, including cell proliferation, differentiation, migration and apoptosis. It has been reported that TGF-β regulates CEA transcription and secretion, however, little is known about the effects of CEA on TGFβ signaling. Aims: Based on the above facts, we focused on the influence of CEA on the TGF-β signaling in both normal cells and colorectal cancer cells. Results: Our preliminary data showed that CEA directly interacted with TGF-β receptors. Overexpression of CEA blocked TGF-β induced SMAD3 phosphorylation, SMAD3 translocation to nuclear and the downregulation of c-myc transcription. Targeting CEA with anti-CEA antibody rescued TGFβ response in CRC cell lines with elevated CEA expression, thereby restoring the inhibitory effects of TGF-β on the proliferation of these cancer cells. Finally, in animal experiment, we found that CEA enhanced survival of colorectal cancer cell in both local colonization and liver metastasis. Conclusion: Since CEA is a well-characterized tumor-associated antigen that is frequently overexpressed in tumors, specific antibodies targeting CEA have been developed as a novel therapeutic approach for treatment of tumors expressing CEA on their surface. Based on our study, it may be helpful to combine CEA antibody and TGF-β to inhibit cancer cell proliferation and metastasis in some cases.


Cancer Research | 2010

Abstract 4258: Role of β2SP in asymmetric division and differentiation of intestine epithelial cells

Ying Li; Wilma Jogunoori; Wenguo Yao; Hong Cao; Bibhuti Mishra; Lopa Mishra

Proceedings: AACR 101st Annual Meeting 2010‐‐ Apr 17‐21, 2010; Washington, DC The adult intestinal epithelium of the gastrointestinal tract has a well-defined organizational structure. The epithelium can be divided into two regions, a functional region that houses differentiated cells (villi) and a proliferative region (crypts of Lieberkuhn) that represents the epithelial stem cell niche. In the small intestine, finger-like projections protrude from the floor of the intestine, forming villi. Multiple proliferative crypts surround each villus base. All epithelial cells that populate the villus originate from these crypts. Multipotent epithelial stem cells reside in the crypts and give rise to four principal epithelial lineages: absorptive enterocytes, mucin secreting-goblet cells, peptide hormone secreting enteroendocrine cells, and Paneth cells. Three of these cell lineages differentiate as they migrate up and out of the crypts onto the adjacent villus. The fourth lineage, Paneth cells, differentiates with a downward migration to reside at the base of the crypt. It is believed that these different cell types are generated by asymmetric cell divisions of stem/progenitor cells. However, the molecular mechanisms of how stem cells differentiate into four different cell types are only partially understood. Aim: in the current study, we evaluated the role of β2SP (β2 spectrin) in the differentiation and asymmetric division of stem/progenitor cells in normal intestine epithelium. Results: Our preliminary data showed that the number of secreting-goblet cells and peptide hormone secreting enteroendocrine cells was reduced in β2SP+/− mice compared to the wt mice. The number of cells undergo asymmetric division was also 3 folds lower in β2SP+/− mice than in wt mice. In in vitro studies, polarization of Par3 and PKCζ was impaired in β2SP−/− MEF cells. The coimmunoprecipitation assay demonstrated that β2SP directly bound to Par3 and actin in normal intestine epithelial cells, indicating that β2SP was involved in the spindle orientation and positioning during the asymmetric division of intestine epithelial cells. Conclusion: β2SP interacts with Par3 and regulates spindle orientation and polarization in the process of cell division, thereby contributing to the differentiation and asymmetric division of intestinal stem/progenitor cells. Citation Format: {Authors}. {Abstract title} [abstract]. In: Proceedings of the 101st Annual Meeting of the American Association for Cancer Research; 2010 Apr 17-21; Washington, DC. Philadelphia (PA): AACR; Cancer Res 2010;70(8 Suppl):Abstract nr 4258.


Cancer Research | 2010

Abstract 3079: Loss of β2-spectrin may lead to the activation of telomerase reverse transcriptase (TERT) and hepatocellular cancer formation

Zhongxian Jiao; Sanjeev Shukla; Gyanendra Kumar; Wenguo Yao; Hong Cao; Wilma Jogunoori; Zhixing Yao; Lopa Mishra; Bibhuti Mishra

Proceedings: AACR 101st Annual Meeting 2010‐‐ Apr 17‐21, 2010; Washington, DC Background and Aims: Telomerase is a specialized reverse transcriptase (TERT) that synthesizes repeat DNA sequences at telomeres- the specialized ends of chromosomes is suppressed in normal cells by multiple proteins that include TGF-β and Smad3 resulting in the progressive shortening of telomeres with each cell division, ultimately leading to chromosomal instability and cellular replicative senescence or growth arrest. Emerging evidence indicates that β2-spectrin (β2SP), a Smad3/Smad4 adaptor protein required for TGF-β signaling, is a powerful tumor suppressor. β2SP+/−, β2SP+/−/Smad3+/− and β2SP+/−/Smad4+/− mice dramatically develop Hepatic cancers. Because TERT is markedly activated in β2SP+/−/Smad3+/− hepatocellular cancer tissues compared to Smad3−/− tissues, we hypothesized that β2SP interaction could be involved in the interaction with Smad3 to suppress TERT expression in normal tissues, and the loss of β2SP may lead to telomerase gene activation resulting in HCC. Results: We tested human TERT (hTERT) expression levels in several HCC cell lines that have different levels of β2SP. hTERT expression levels negatively correlate with β2SP by Western Blot analysis. Loss of β2SP in β2SP+/− and β2SP−/− mouse embryonic fibroblasts (MEFs) significantly activated mTERT expression. Co-transfection of a β2SP expression plasmid and hTERT promoter-luciferase construct significantly inhibited the hTERT promoter in β2SP-deficient SNU-398 cells. Depletion of Smad3 by in vitro RNAi and the expression of hTERT-luciferase in Smad3 depleted cells upon co-transfection with Smad3 and/or β2SP expression plasmids suggest that both Smad3 and β2SP are required for the suppression of hTERT. Furthermore, chromatin immunoprecipitation (ChIP) assay suggests binding of Smad3/β2SP protein to hTERT promoter. Conclusions: Taken together our data suggest that divergent pathways converge on β2SP and Smad3 for the regulation of TERT. Activation of the TGF-β signaling pathway with suppression of TERT provides a strong strategy for generating targeted therapeutics to these lethal human cancers. Citation Format: {Authors}. {Abstract title} [abstract]. In: Proceedings of the 101st Annual Meeting of the American Association for Cancer Research; 2010 Apr 17-21; Washington, DC. Philadelphia (PA): AACR; Cancer Res 2010;70(8 Suppl):Abstract nr 3079.


Cancer Research | 2010

Abstract 4912: The role of long-range enhancer blocker CTCF in TGF-β/β-2 spectrin signaling: A human cancer stem cell disorder, Beckwith-Wiedemann syndrome

Zhixing Yao; Wilma Jogunoori; Sanjeev Shukla; Wenguo Yao; Bibhuti Mishra; Lopa Mishra

Hereditary cancer syndromes provide powerful insights into dysfunctional signaling pathways that lead to sporadic cancers. Beckwith-Wiedemann syndrome (BWS) is a hereditary human cancer stem cell syndrome currently linked to deregulated imprinting at chromosome 11p15 and uniparental disomy (UPD) and is characterized by overgrowth and is associated with an 800-fold increased risk of embryonal neoplasms of childhood that include Wilms’ tumors, hepatoblastomas, pancreatoblastoma, neuroblastoma, rhabdomyosarcoma, and adrenocortical carcinomas. The transcriptional insulator CTCF (CCCTC-binding factor), a highly conserved zinc finger protein, has been implicated in BWS and has diverse regulatory functions, including transcriptional activation/repression, insulation, imprinting, and X chromosome inactivation However, causal molecular defects and genetic models have remained elusive to date in the majority of cases. TGF-β employs Smad proteins as intracellular mediators of signaling to regulate the target gene expression by activating or repressing gene transcription. The non-pleckstrin homology (PH) domain β-general-spectrin (β2SP), a scaffolding protein, functions as a potent TGF-β signaling Smad3/4 adaptor in tumor suppression and development. Yet, the role of the β2SP/TGF-β pathway in human tumor syndromes remains unclear, Our previous data have shown that β2SP is an important effector of TGF-β tumor suppressor function. Deletion of β2SP results in a dramatic and spontaneous formation of liver, gastrointestinal (GI) and pancreatic cancers. Aims: In this study, we investigated the relationship between epigenetic silencing of β-2 Spectrin/TGF-β[[Unsupported Character - ]] Signaling and loss of CTCF protein stabilization in a Human Cancer Stem Cell Disorder: Beckwith-Wiedemann Syndrome. Results show that: 1) β2SP is silenced at its promoter by DNA Methylation in human BWS non-tumor tissues and cell lines. 2) Loss of β2SP, mis-localization of Smad3 and disruption of TGF-β signaling are observed in human Beckwith-Wiedemann non-tumor tissues and cell lines 3) Ectopic expression of β2SP in human BWS cells rescues Smad3 localization and TGF-β target gene activation 4) Increased IGF2 expression in β2SP+/− and β2SP+/−/Smad3+/− mice is similar to that observed in human BWS. 5) CTCF protein levels are markedly decreased in β2SP+/− and β2SP+/−/Smad3+/− tissues. Conclusions: Our results suggest that epigenetic regulation of the TGF-β pathway results in the loss of β2SP and its crucial Smad3 adaptor function. The loss of an adaptor protein β2SP affecting CTCF stabilization, is causally related to the genesis and progression of human BWS. In addition, β2SP+/− and β2SP+/−/Smad3+/− mice provide an important animal model to study BWS. Citation Format: {Authors}. {Abstract title} [abstract]. In: Proceedings of the 101st Annual Meeting of the American Association for Cancer Research; 2010 Apr 17-21; Washington, DC. Philadelphia (PA): AACR; Cancer Res 2010;70(8 Suppl):Abstract nr 4912.


Cancer Research | 2010

Abstract 1491: CEA interacts with TGF-β receptor and inhibits TGF-β signaling in colorectal cancers

Hong Cao; Ying Li; Wenguo Yao; Wilma Jogunoori; Arun Thenappan; Lopa Mishra

Carcinoembryonic antigen (CEA) is a tumor marker for the clinical management of colorectal cancer (CRC). The elevated blood levels of CEA are associated with metastasis and poor prognosis in CRC. There is mounting evidence that CEA enhances the metastatic potential of cancer cells. CEA increases the ability of weakly metastatic CRC to colonize the liver and to develop spontaneous hematogeneous liver and lung metastases. CEA expression has also been related with resistance to cytotoxic chemotherapy and to anoikis, a form of apoptosis caused by detachment from cell matrix. Yet the mechanism of CEA mediated metastasis is only partially understood. The TGF-β (transforming growth factor beta) signaling pathway contributes to tumorigenesis by controlling several biological processes, including cell proliferation, differentiation, migration and apoptosis. It has been reported that TGF-β regulates CEA transcription and secretion, however, little is known about the effects of CEA on TGF-β signaling. Aims: Based on the above facts, we focused on the influence of CEA on the TGF-β signaling in both normal cells and colorectal cancer cells. Results: Our preliminary data showed that CEA directly interacted with TGF-β receptors. Overexpression of CEA blocked TGF-β induced SMAD3 phosphorylation, SMAD3 translocation to nuclear and the downregulation of c-myc transcription. Targeting CEA with anti-CEA antibody rescued TGF-β response in CRC cell lines with elevated CEA expression, thereby restoring the inhibitory effects of TGF-β on the proliferation of these cancer cells. Finally, in animal experiment, we found that CEA enhanced survival of colorectal cancer cell in both local colonization and liver metastasis. Conclusion: Since CEA is a well-characterized tumor-associated antigen that is frequently overexpressed in tumors, specific antibodies targeting CEA have been developed as a novel therapeutic approach for treatment of tumors expressing CEA on their surface. Based on our study, it may be helpful to combine CEA antibody and TGF-β to inhibit cancer cell proliferation and metastasis in some cases. Citation Format: {Authors}. {Abstract title} [abstract]. In: Proceedings of the 101st Annual Meeting of the American Association for Cancer Research; 2010 Apr 17-21; Washington, DC. Philadelphia (PA): AACR; Cancer Res 2010;70(8 Suppl):Abstract nr 1491.


Archive | 2010

EpigeneticSilencingof-Spectrin,aTGF-Signaling/Scaffolding Protein in a Human Cancer Stem Cell Disorder

Zhixing Yao; Wilma Jogunoori; Sanaa Choufani; Asif Rashid; Tiffany Blake; Wenguo Yao; Peter Kreishman; Rupen Amin; Anton A. Sidawy; Stephen R.T. Evans; Milton J. Finegold; E. Premkumar Reddy; Bibhuti Mishra; Rosanna Weksberg; Rakesh Kumar; Lopa Mishra


Gastroenterology | 2010

S1670 The β2 Spectrin Suppresses Telomerase Reverse Transcriptase in Normal Cells. Its Loss May Lead to Hepatocellular Cancer Progression

Zhongxian Jiao; Sanjeev Shukla; Gyanendra Kumar; Zhixing Yao; Wilma Jogunoori; Wenguo Yao; Lopa Mishra; Bibhuti Mishra


Gastroenterology | 2010

225 Epigenetic Silencing of TGF-β/β-2 Spectrin Signaling in a Human Cancer Stem Cell Disorder: Beckwith-Wiedemann Syndrome-Implication for GI Cancers

Zhixing Yao; Wilma Jogunoori; Sanjeev Shukla; Wenguo Yao; Bibhuti Mishra; Lopa Mishra


Gastroenterology | 2010

T1726 β2 Spectrin Mediates Asymmetric Division and Differentiation of Intestine Epithelial Cells by Regulating Spindle Polarization

Ying Li; Wenguo Yao; Wilma Jogunoori; Hong Cao; Bibhuti Mishra; Lopa Mishra


Gastroenterology | 2009

764 Causal Relationship Between Loss of β-Spectrin (ELF), Its TGF-β Signaling Adaptor Function and a Hereditary Human Gastrointestinal Cancer Syndrome: Beckwith Wiedemann Syndrome (BWS)

Zhixing Yao; Wilma Jogunoori; Geeta Upadhyay; Wenguo Yao; Bibhuti Mishra; Lopa Mishra

Collaboration


Dive into the Wenguo Yao's collaboration.

Top Co-Authors

Avatar

Lopa Mishra

George Washington University

View shared research outputs
Top Co-Authors

Avatar

Wilma Jogunoori

George Washington University

View shared research outputs
Top Co-Authors

Avatar

Bibhuti Mishra

National Institutes of Health

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Hong Cao

Georgetown University

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Ying Li

Georgetown University

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge