Network


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

Hotspot


Dive into the research topics where Wun Chey Sin is active.

Publication


Featured researches published by Wun Chey Sin.


Glia | 2007

Connexin43 enhances glioma invasion by a mechanism involving the carboxy terminus

Dave C. Bates; Wun Chey Sin; Q. Aftab; Christian C. Naus

Gliomas are particularly difficult to cure owing largely to their invasive nature. The neoplastic changes of astrocytes which give rise to these tumors frequently include a reduction of connexin43 (Cx43), the most abundant connexin isoform expressed in astrocytes. Cx43 is a subunit of gap junctions (GJ), intercellular channels which directly link the cytosol of adjacent cells and allow the regulated passage of ions and small molecules. To examine the role of Cx43 in glioma motility, we identified two variant C6 cell lines which endogenously express high (C6‐H) or low (C6‐L) levels of Cx43. In wound healing and transwell assays, C6‐H cells were more motile than C6‐L cells. To deduce whether Cx43 mediated these differences, assays were conducted on C6‐H cells retrovirally transduced with Cx43 shRNA. Coincident with the stable knockdown of endogenous Cx43, a decrease in motility and invasion was observed. Gap junctional intercellular communication was also decreased, however motility assays conducted in the presence of GJ inhibitors did not reveal significant differences in cell motility. C6 cells transfected with full length or C‐terminal truncated Cx43 (Cx43ΔCT) were subjected to the aforementioned motility assays to expose alternate mechanisms of Cx43‐mediated motility. Cells expressing full length Cx43 exhibited increased motility while cells expressing Cx43ΔCT did not. This report, the first in which RNAi has been employed to reduce Cx43 expression in gliomas, indicates that the downregulation of Cx43 decreases motility of C6 cells. Furthermore, it is the first report to suggest that the Cx43 CT plays an important role in glioma motility.


Journal of Biological Chemistry | 2014

Gap junction intercellular communication mediated by connexin43 in astrocytes is essential for their resistance to oxidative stress

Hoa T. Le; Wun Chey Sin; Shannon Lozinsky; John F. Bechberger; José L. Vega; Xu Qiu Guo; Juan C. Sáez; Christian C. Naus

Background: The gap junction protein Cx43 is implicated in maintaining anti-oxidative defense in astrocytes. Results: In contrast to hypoxia/reoxygenation, oxidative stress induced by H2O2 triggers more astrocytic death in the absence of Cx43 channels. Conclusion: Gap junction intercellular communication is required for Cx43-mediated resistance to H2O2. Significance: An altered Cx43 phosphorylation state in response to cellular stress may be critical for Cx43-mediated cell death or recovery. Oxidative stress induced by reactive oxygen species (ROS) is associated with various neurological disorders including aging, neurodegenerative diseases, as well as traumatic and ischemic insults. Astrocytes have an important role in the anti-oxidative defense in the brain. The gap junction protein connexin43 (Cx43) forms intercellular channels as well as hemichannels in astrocytes. In the present study, we investigated the contribution of Cx43 to astrocytic death induced by the ROS hydrogen peroxide (H2O2) and the mechanism by which Cx43 exerts its effects. Lack of Cx43 expression or blockage of Cx43 channels resulted in increased ROS-induced astrocytic death, supporting a cell protective effect of functional Cx43 channels. H2O2 transiently increased hemichannel activity, but reduced gap junction intercellular communication (GJIC). GJIC in wild-type astrocytes recovered after 7 h, but was absent in Cx43 knock-out astrocytes. Blockage of Cx43 hemichannels incompletely inhibited H2O2-induced hemichannel activity, indicating the presence of other hemichannel proteins. Panx1, which is predicted to be a major hemichannel contributor in astrocytes, did not appear to have any cell protective effect from H2O2 insults. Our data suggest that GJIC is important for Cx43-mediated ROS resistance. In contrast to hypoxia/reoxygenation, H2O2 treatment decreased the ratio of the hypophosphorylated isoform to total Cx43 level. Cx43 has been reported to promote astrocytic death induced by hypoxia/reoxygenation. We therefore speculate the increase in Cx43 dephosphorylation may account for the facilitation of astrocytic death. Our findings suggest that the role of Cx43 in response to cellular stress is dependent on the activation of signaling pathways leading to alteration of Cx43 phosphorylation states.


Journal of Cellular Biochemistry | 2008

Dose-Dependent Differential Upregulation of CCN1 /Cyr61 and CCN3/NOV by the Gap Junction Protein Connexin43 in Glioma Cells

Wun Chey Sin; John F. Bechberger; Walter J. Rushlow; Christian C. Naus

Gap junctions form channels that allow exchange of materials between cells and are composed of transmembrane protein subunits called connexins. While connexins are believed to mediate cellular signaling by permitting intercellular communication to occur, there is also increasing evidence that suggest connexins may mediate growth control via a junction‐independent mechanism. Connexin43 (Cx43) is the most abundant gap junction protein found in astrocytes, and gliomas exhibit reduced Cx43 expression. We have previously observed that restoration of Cx43 levels in glioma cells led to increased expression of CCN3 (NOV) proteins. We now report that overexpression of Cx43 in C6‐glioma cells (C6‐Cx43) also upregulates the expression of CCN1 (Cyr61). Both CCN1 and CCN3 belong to the Cyr61/Connective tissue growth factor/Nephroblastoma‐overexpressed (CCN) family of secretory proteins. The CCN proteins are tightly associated with the extracellular matrix and have important roles in cell proliferation and migration. CCN1 promotes growth in glioma cells, as shown by the increased proliferation rate of CCN1‐overexpressing C6 cells. In addition to its effect on cell growth, CCN1 also increased the motility of glioma cells in the presence of extracellular substrates such as fibronectin. Gliomas expressing high levels of Cx43 preferentially upregulated CCN3 which resulted in reduced growth rate. CCN3 could also be observed in Cx43 gap junction plaques in confluent C6‐Cx43H culture at the stationary phase of their growth. Our results suggest that the dissimilar growth characteristics between high and low Cx43 expressors may be due to differential regulation of CCN3 by varying levels of Cx43. J. Cell. Biochem. 103: 1772–1782, 2007.


Journal of Biological Chemistry | 2009

Matricellular protein CCN3 (NOV) regulates actin cytoskeleton reorganization

Wun Chey Sin; Mimi Tse; Nathalie Planque; Bernard Perbal; Paul D. Lampe; Christian C. Naus

CCN3 (NOV), a putative ligand for integrin receptors, is tightly associated with the extracellular matrix and mediates diverse cellular functions, including cell adhesion and proliferation. CCN3 has been shown to negatively regulate growth although it promotes migration in a cell type-specific manner. In this study, overexpression of CCN3 reduces growth and increases intercellular adhesion of breast cancer cells. Interestingly, CCN3 overexpression also led to the formation of multiple pseudopodia that are enriched in actin, CCN3, and vinculin. Breast cancer cells preincubated with exogenous CCN3 protein also induced the same phenotype, indicating that secreted CCN3 is sufficient to induce changes in cell morphology. Surprisingly, extracellular CCN3 is internalized to the early endosomes but not to the membrane protrusions, suggesting pseudopodia-enriched CCN3 may derive from a different source. The presence of an intracellular variant of CCN3 will be consistent with our finding that the cytoplasmic tail of the gap junction protein connexin43 (Cx43) associates with CCN3. Cx43 is a channel protein permitting intercellular communication to occur. However, neither the channel properties nor the protein levels of Cx43 are affected by the CCN3 protein. In contrast, CCN3 proteins are down-regulated in the absence of Cx43. Finally, we showed that overexpression of CCN3 increases the activity of the small GTPase Rac1, thereby revealing a pathway that links Cx43 directly to actin reorganization.


Journal of Neuropathology and Experimental Neurology | 2015

Podoplanin: a marker for reactive gliosis in gliomas and brain injury.

Kushal Kolar; Moises Freitas-Andrade; John F. Bechberger; Harini Krishnan; Gary S. Goldberg; Christian C. Naus; Wun Chey Sin

Abstract Reactive astrogliosis is associated with many pathologic processes in the central nervous system, including gliomas. The glycoprotein podoplanin (PDPN) is upregulated in malignant gliomas. Using a syngeneic intracranial glioma mouse model, we show that PDPN is highly expressed in a subset of glial fibrillary acidic protein–positive astrocytes within and adjacent to gliomas. The expression of PDPN in tumor-associated reactive astrocytes was confirmed by its colocalization with the astrocytic marker S100&bgr; and with connexin43, a major astrocytic gap junction protein. To determine whether the increase in PDPN is a general feature of gliosis, we used 2 mouse models in which astrogliosis was induced either by a needle injury or ischemia and observed similar upregulation of PDPN in reactive astrocytes in both models. Astrocytic PDPN was also found to be coexpressed with nestin, an intermediate filament marker for neural stem/progenitor cells. Our findings confirm that expression of PDPN is part of the normal host response to brain injury and gliomas, and suggest that it may be a novel cell surface marker for a specific population of reactive astrocytes in the vicinity of gliomas and nonneoplastic brain lesions. The findings also highlight the heterogeneity of glial fibrillary acidic protein–positive astrocytes in reactive gliosis.


Current Opinion in Pharmacology | 2018

Novel approach to temozolomide resistance in malignant glioma: connexin43-directed therapeutics

Christina L. Grek; Zhi Sheng; Christian C. Naus; Wun Chey Sin; Robert G. Gourdie; Gautam G Ghatnekar

HighlightsCx43 regulates invasiveness, proliferation, and resistance of malignant glioma.Cx43 provides a permissive niche for the invasion and channel‐mediated TMZ resistance.Targeting Cx43 with C‐terminal peptidomimetics can sensitize glioblastoma to TMZ.Mechanism of action for peptidomimetics may be channel‐dependent or channel‐independent. &NA; Resistance of malignant glioma, including glioblastoma (GBM), to the chemotherapeutic temozolomide (TMZ) remains a key obstacle in treatment strategies. The gap junction protein connexin43 (Cx43) has complex roles in the establishment, progression, and persistence of malignant glioma. Recent findings demonstrate that connexins play an important role in the microenvironment of malignant glioma and that Cx43 is capable of conferring chemotherapeutic resistance to GBM cells. Carboxyl‐terminal Cx43 peptidomimetics show therapeutic promise in overcoming TMZ resistance via mechanisms that may include modulating junctional activity between tumor cells and peritumoral cells and/or downstream molecular signaling events mediated by Cx43 protein binding. High levels of intra‐tumor and inter‐tumor heterogeneity make it difficult to clearly define specific populations for Cx43‐targeted therapy; hence, development of in vitro models that better mimic the microenvironment of malignant glioma, and the incorporation of patient‐derived stem cells, could provide opportunities for patient‐specific drug screening. This review summarizes recent advances in understanding the roles of Cx43 in malignant glioma, with a special focus on tumor microenvironment, TMZ resistance, and therapeutic opportunity offered by Cx43 peptidomimetics.


Neuropharmacology | 2013

Connexin43 confers Temozolomide resistance in human glioma cells by modulating the mitochondrial apoptosis pathway.

Paul R. Gielen; Qurratulain Aftab; Noreen Ma; Vincent C. Chen; Xiaoting Hong; Shannon Lozinsky; Christian C. Naus; Wun Chey Sin


Seminars in Cell & Developmental Biology | 2016

Common mechanisms linking connexin43 to neural progenitor cell migration and glioma invasion.

Christian C. Naus; Qurratulain Aftab; Wun Chey Sin


Cancer Letters | 2007

Tissue microarray analysis of connexin expression and its prognostic significance in human breast cancer

Chris M.J. Conklin; David Huntsman; Erika Yorida; Nikita Makretsov; Dmitry Turbin; John F. Bechberger; Wun Chey Sin; Christian C. Naus


Neuropharmacology | 2013

Cerebral ischemic injury is enhanced in a model of oculodentodigital dysplasia.

Michael G. Kozoriz; Simon Lai; José L. Vega; Juan C. Sáez; Wun Chey Sin; John F. Bechberger; Christian C. Naus

Collaboration


Dive into the Wun Chey Sin's collaboration.

Top Co-Authors

Avatar

Christian C. Naus

University of British Columbia

View shared research outputs
Top Co-Authors

Avatar

John F. Bechberger

University of British Columbia

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Paul D. Lampe

Fred Hutchinson Cancer Research Center

View shared research outputs
Top Co-Authors

Avatar

Moises Freitas-Andrade

University of British Columbia

View shared research outputs
Top Co-Authors

Avatar

Qurratulain Aftab

University of British Columbia

View shared research outputs
Top Co-Authors

Avatar

Shannon Lozinsky

University of British Columbia

View shared research outputs
Top Co-Authors

Avatar

José L. Vega

University of Antofagasta

View shared research outputs
Top Co-Authors

Avatar

Juan C. Sáez

Pontifical Catholic University of Chile

View shared research outputs
Top Co-Authors

Avatar

Christina L. Grek

Medical University of South Carolina

View shared research outputs
Researchain Logo
Decentralizing Knowledge