Network


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

Hotspot


Dive into the research topics where Yuen Piu Chan is active.

Publication


Featured researches published by Yuen Piu Chan.


Cell Stem Cell | 2010

miR-130b Promotes CD133+ Liver Tumor-Initiating Cell Growth and Self-Renewal via Tumor Protein 53-Induced Nuclear Protein 1

Stephanie Ma; Kwan Ho Tang; Yuen Piu Chan; Terence K. Lee; Pak Shing Kwan; Antonia Castilho; Irene Ng; Kwan Man; Nathalie Wong; Ka Fai To; Bo-Jian Zheng; Paul B.S. Lai; Chung Mau Lo; Kwok Wah Chan; Xin Yuan Guan

A novel paradigm in tumor biology suggests that cancer growth is driven by stem-like cells within a tumor, called tumor-initiating cells (TICs) or cancer stem cells (CSCs). Here we describe the identification and characterization of such cells from hepatocellular carcinoma (HCC) using the marker CD133. CD133 accounts for approximately 1.3%-13.6% of the cells in the bulk tumor of human primary HCC samples. When compared with their CD133⁻ counterparts, CD133(+) cells not only possess the preferential ability to form undifferentiated tumor spheroids in vitro but also express an enhanced level of stem cell-associated genes, have a greater ability to form tumors when implanted orthotopically in immunodeficient mice, and can be serially passaged into secondary animal recipients. Xenografts resemble the original human tumor and maintain a similar percentage of tumorigenic CD133(+) cells. Quantitative PCR analysis of 41 separate HCC tissue specimens with follow-up data found that CD133(+) tumor cells were frequently detected at low quantities in HCC, and their presence was also associated with worse overall survival and higher recurrence rates. Subsequent differential microRNA expression profiling of CD133(+) and CD133⁻ cells from human HCC clinical specimens and cell lines identified an overexpression of miR-130b in CD133(+) TICs. Functional studies on miR-130b lentiviral-transduced CD133⁻ cells demonstrated superior resistance to chemotherapeutic agents, enhanced tumorigenicity in vivo, and a greater potential for self renewal. Conversely, antagonizing miR-130b in CD133(+) TICs yielded an opposing effect. The increased miR-130b paralleled the reduced TP53INP1, a known miR-130b target. Silencing TP53INP1 in CD133⁻ cells enhanced both self renewal and tumorigenicity in vivo. Collectively, miR-130b regulates CD133(+) liver TICs, in part, via silencing TP53INP1.


Hepatology | 2012

CD133 + liver tumor-initiating cells promote tumor angiogenesis, growth, and self-renewal through neurotensin/interleukin-8/CXCL1 signaling

Kwan Ho Tang; Stephanie Ma; Terence K. Lee; Yuen Piu Chan; Pak Shing Kwan; Carol Man Tong; Irene O. Ng; Kwan Man; Ka Fai To; Paul B.S. Lai; Chung Mau Lo; Xin Yuan Guan; Kwok Wah Chan

A novel theory in the field of tumor biology postulates that cancer growth is driven by a population of stem‐like cells, called tumor‐initiating cells (TICs). We previously identified a TIC population derived from hepatocellular carcinoma (HCC) that is characterized by membrane expression of CD133. Here, we describe a novel mechanism by which these cells mediate tumor growth and angiogenesis by systematic comparison of the gene expression profiles between sorted CD133 liver subpopulations through genome‐wide microarray analysis. A significantly dysregulated interleukin‐8 (IL‐8) signaling network was identified in CD133+ liver TICs obtained from HCC clinical samples and cell lines. IL‐8 was found to be overexpressed at both the genomic and proteomic levels in CD133+ cells isolated from HCC cell lines or clinical samples. Functional studies found enhanced IL‐8 secretion in CD133+ liver TICs to exhibit a greater ability to self‐renew, induce tumor angiogenesis, and initiate tumors. In further support of these observations, IL‐8 repression in CD133+ liver TICs by knockdown or neutralizing antibody abolished these effects. Subsequent studies of the IL‐8 functional network identified neurotensin (NTS) and CXCL1 to be preferentially expressed in CD133+ liver TICs. Addition of exogenous NTS resulted in concomitant up‐regulation of IL‐8 and CXCL1 with simultaneous activation of p‐ERK1/2 and RAF‐1, both key components of the mitogen‐activated protein kinase (MAPK) pathway. Enhanced IL‐8 secretion by CD133+ liver TICs can in turn activate an IL‐8‐dependent feedback loop that signals through the MAPK pathway. Further, in its role as a liver TIC marker CD133 also plays a functional part in regulating tumorigenesis of liver TICs by way of regulating NTS, IL‐8, CXCL1, and MAPK signaling. Conclusion: CD133+ liver TICs promote angiogenesis, tumorigenesis, and self‐renewal through NTS‐induced activation of the IL‐8 signaling cascade. (Hepatology 2012)


Cancer Research | 2013

A CD90+ Tumor-Initiating Cell Population with an Aggressive Signature and Metastatic Capacity in Esophageal Cancer

Kwan Ho Tang; Yong–Dong Dai; Man Tong; Yuen Piu Chan; Pak Shing Kwan; Li Fu; Yan Ru Qin; Sai Wah Tsao; Hong Lok Lung; Maria Li Lung; Daniel K. Tong; Simon Law; Kwok Wah Chan; Stephanie Ma; Xin Yuan Guan

Tumor-initiating cells (TIC), also known as cancer stem cells, are regarded widely as a specific subpopulation of cells needed for cancer initiation and progression. TICs have yet to be identified in esophageal tumors that have an increasing incidence in developed countries. Here, we report a CD90(+) cell population found in esophageal squamous cell carcinoma (ESCC), which is endowed with stem cell-like properties and high tumorigenic and metastatic potential. mRNA profiling of these cells suggested pathways through which they drive tumor growth and metastasis, with deregulation of an Ets-1/MMP signaling pathway and epithelial-mesenchymal transition figuring prominently. These cells possessed higher self-renewal activity and were sufficient for tumor growth, differentiation, metastasis, and chemotherapeutic resistance. CD90(+) TICs were isolated and characterized from ESCC clinical specimens as well as ESCC cell lines. In freshly resected clinical specimens, they represented a rare cell population, the levels of which correlated with strong family histories and lymph node metastasis. Our results prompt further study of this CD90(+) population of esophageal TICs as potential therapeutic targets.


Gastroenterology | 2012

Identification of PTK6, via RNA Sequencing Analysis, as a Suppressor of Esophageal Squamous Cell Carcinoma

Stephanie Ma; Jessie Y.J. Bao; Pak Shing Kwan; Yuen Piu Chan; Carol Man Tong; Li Fu; Na Zhang; Amy Hin Yan Tong; Yan Ru Qin; Sai Wah Tsao; Kwok Wah Chan; Si Lok; Xin Yuan Guan

BACKGROUND & AIMS Esophageal squamous cell carcinoma (ESCC) is the most commonly observed histologic subtype of esophageal cancer. ESCC is believed to develop via accumulation of numerous genetic alterations, including inactivation of tumor suppressor genes and activation of oncogenes. We searched for transcripts that were altered in human ESCC samples compared with nontumor tissues. METHODS We performed integrative transcriptome sequencing (RNA-Seq) analysis using ESCC samples from 3 patients and adjacent nontumor tissues to identify transcripts that were altered in ESCC tissue. We performed molecular and functional studies of the transcripts identified and investigated the mechanisms of alteration. RESULTS We identified protein tyrosine kinase 6 (PTK6) as a transcript that was significantly down-regulated in ESCC tissues and cell lines compared with nontumor tissues or immortalized normal esophageal cell lines. The promoter of the PTK6 gene was inactivated in ESCC tissues at least in part via hypermethylation and histone deacetylation. Knockdown of PTK6 in KYSE30 ESCC cells using small hairpin RNAs increased their ability to form foci, migrate, and invade extracellular matrix in culture and form tumors in nude mice. Overexpression of PTK6 in these cells reduced their proliferation in culture and tumor formation in mice. PTK6 reduced phosphorylation of Akt and glycogen synthase kinase (GSK)3β, leading to activation of β-catenin. CONCLUSIONS PTK6 was identified as a transcript that is down-regulated in human ESCC tissues via epigenetic modification at the PTK6 locus. Its product appears to regulate cell proliferation by reducing phosphorylation of Akt and GSK3β, leading to activation of β-catenin. Reduced levels of PTK6 promote growth of xenograft tumors in mice; it might be developed as a marker of ESCC.


International Journal of Cancer | 2012

Tumor suppressor dual-specificity phosphatase 6 (DUSP6) impairs cell invasion and epithelial-mesenchymal transition (EMT)-associated phenotype.

Victor Chun Lam Wong; Han Chen; Josephine Mun Yee Ko; Kwok Wah Chan; Yuen Piu Chan; Simon Law; Daniel Chua; Dora L.W. Kwong; Hong Lok Lung; Gopesh Srivastava; Johnny Cheuk On Tang; Sai Wah Tsao; Eugene R. Zabarovsky; Eric J. Stanbridge; Maria Li Lung

Suppressive effects of DUSP6 in tumorigenesis and EMT‐associated properties were observed. Dual‐specificity phosphatase (DUSP6) is a MAP kinase phosphatase (MKP) negatively regulating the activity of ERK, one of the major molecular switches in the MAPK signaling cascade propagating the signaling responses during malignancies. The impact of DUSP6 in EMT and its contribution to tumor dissemination has not yet been characterized. Due to differences in tumor microenvironments affecting cell signaling during cancer progression, DUSP6 may play varying roles in tumor development. We sought to examine the potential role of DUSP6‐mediated tumorigenesis and EMT‐associated properties in two aerodigestive tract cancers, namely, esophageal squamous cell carcinoma (ESCC) and nasopharyngeal carcinoma (NPC). Significant loss of DUSP6 was observed in 100% of 11 ESCC cell lines and 71% of seven NPC cell lines. DUSP6 expression was down‐regulated in 40% of 30 ESCC tumor tissues and 75% of 20 NPC tumor tissues compared to their respective normal counterparts. Suppressive effects of DUSP6 in tumor formation and cancer cell mobility are seen in in vivo tumorigenicity assay and in vitro colony formation, three‐dimensional Matrigel culture, cell migration and invasion chamber tests. Notably, overexpression of DUSP6 impairs EMT‐associated properties. Furthermore, tissue microarray analysis reveals a clinical association of DUSP6 expression with better patient survival. Taken together, our study provides a novel insight into understanding the functional impact of DUSP6 in tumorigenesis and metastasis of ESCC and NPC.


Blood | 2015

Receptor-type tyrosine-protein phosphatase κ directly targets STAT3 activation for tumor suppression in nasal NK/T-cell lymphoma

Yw Chen; Tianhuan Guo; Lijun Shen; Kai Yau Wong; Qian Tao; William W.L. Choi; Rex K.H. Au-Yeung; Yuen Piu Chan; Michelle L.Y. Wong; Johnny Cheuk On Tang; Wei Ping Liu; Gan Di Li; Norio Shimizu; Florence Loong; Eric Tse; Yok-Lam Kwong; Gopesh Srivastava

Nasal-type natural killer/T-cell lymphoma (NKTCL) is an aggressive disease characterized by frequent deletions on 6q, and constitutive activation of signal transducer and activator of transcription 3 (STAT3). Phosphorylation at Tyr705 activates STAT3, inducing dimerization, nuclear translocation, and DNA binding. In this study, we investigated whether receptor-type tyrosine-protein phosphatase κ (PTPRK), the only protein tyrosine phosphatase at 6q that contains a STAT3-specifying motif, negatively regulates STAT3 activation in NKTCL. PTPRK was highly expressed in normal NK cells but was underexpressed in 4 of 5 (80%) NKTCL cell lines and 15 of 27 (55.6%) primary tumors. Significantly, PTPRK protein expression was inversely correlated with nuclear phospho-STAT3(Tyr705) expression in NKTCL cell lines (P = .025) and tumors (P = .040). PTPRK restoration decreased nuclear phospho-STAT3(Tyr705) levels, whereas knockdown of PTPRK increased such levels in NKTCL cells. Phosphatase substrate-trapping mutant assays demonstrated the binding of PTPRK to STAT3, and phosphatase assays showed that PTPRK directly dephosphorylated phospho-STAT3(Tyr705). Restoration of PTPRK inhibited tumor cell growth and reduced the migration and invasion ability of NKTCL cells. Monoallelic deletion and promoter hypermethylation caused underexpression of PTPRK messenger RNA in NKTCL, and methylation of the PTPRK promoter significantly correlated with inferior overall survival (P = .049) in NKTCL patients treated with the steroid-dexamethasone, methotrexate, ifosfamide, l-asparaginase, and etoposide regimen. Altogether, our findings show that PTPRK underexpression leads to STAT3 activation and contributes to NKTCL pathogenesis.


International Journal of Cancer | 2011

The ECM protein LTBP-2 is a suppressor of esophageal squamous cell carcinoma tumor formation but higher tumor expression associates with poor patient outcome

Stephen Ho Kin Chan; Josephine Mun Yee Ko; Kwok Wah Chan; Yuen Piu Chan; Qian Tao; Marko Hyytiäinen; Jorma Keski-Oja; Simon Law; Gopesh Srivastava; Johnny Cheuk On Tang; Sai Wah Tsao; Han Chen; Eric J. Stanbridge; Maria Li Lung

Our previous studies of chromosome 14 transfer into tumorigenic esophageal squamous cell carcinoma (ESCC) cell line, SLMT, suggested the existence of tumor suppressor genes on chromosome 14. Gene expression profiling of microcell hybrids and the tumor segregants identified an interesting gene, LTBP‐2 (latent transforming growth factor β binding protein 2), which has been analyzed here for its role in ESCC. LTBP‐2 maps to 14q24 and encodes a secreted protein, which is a component of the extracellular matrix microfibrils. LTBP‐2 expression was downregulated in ESCC cell lines and tumor tissues. Promoter hypermethylation was found to be involved in LTBP‐2 inactivation. Functional studies indicated its tumor‐suppressive roles in ESCC. In the in vitro colony formation and Matrigel three‐dimensional culture assays, LTBP‐2 decreased the colony‐forming abilities of ESCC cell lines. LTBP‐2 expression was associated with reduction of cell migrating and invasive abilities. LTBP‐2 could also reduce the tube‐forming ability of endothelial cells. Moreover, LTBP‐2 induced tumor suppression in in vivo nude mouse assays. Tissue microarray immunohistochemical staining analysis indicated that LTBP‐2 expression is reduced in tumor tissues when compared to normal tissues, and LTBP‐2 expression correlated significantly with the survival of ESCC patients. Thus, LTBP‐2 appears to play an important role in ESCC.


International Journal of Cancer | 2009

DNA fingerprinting tags novel altered chromosomal regions and identifies the involvement of SOX5 in the progression of prostate cancer

Stephanie Ma; Yuen Piu Chan; Bruce Woolcock; Liang Hu; Ky Wong; Ming-Tat Ling; Terry C. Bainbridge; Douglas Webber; Tim Hon Man Chan; Xin Yuan Guan; Wan L. Lam; Juergen R. Vielkind; Kwok Wah Chan

Identification of genomic alterations associated with the progression of prostate cancer may facilitate the better understanding of the development of this highly variable disease. Matched normal, premalignant high‐grade prostatic intraepithelial neoplasia and invasive prostate carcinoma cells were procured by laser capture microdissection (LCM) from human radical prostatectomy specimens. From these cells, comparative DNA fingerprints were generated by a modified PCR‐based technique called scanning of microdissected archival lesion (SMAL)‐PCR. Recurrent polymorphic fingerprint fragments were used in tagging altered chromosomal regions. Altered regions were found at cytobands 1p31.3, 1q44, 2p23.1, 3p26.3, 3q22.3, 4q22.3, 4q35.2, 5q23.2, 8q22.3, 8q24.13, 9q21.3, 9q22.32, 10q11.21, 11p13, 12p12.1, 13q12.1, 16q12.2 and 18q21.31. Candidate genes in the surrounding area that may possibly harbor mutations that change normal prostatic cells to progress into their tumor stages were proposed. Of these fragments, a 420 bp alteration, absent in all 26 normal samples screened, was observed in 2 tumors. This fragment was cloned, sequenced and localized to chromosome 12p12.1. Within this region, candidate gene sex determining region Y‐box 5 (SOX5) was proposed. Further studies of SOX5 in cell lines, xenografts and human prostate specimens, at both the RNA and protein levels, found overexpression of the gene in tumors. This overexpression was then subsequently found by fluorescent in situ hybridization to be caused by amplification of the region. In conclusion, our results suggest LCM coupled with SMAL‐PCR DNA fingerprinting is a useful method for the screening and identification of chromosomal regions and genes associated with cancer development. Further, overexpression of SOX5 is associated with prostate tumor progression and early development of distant metastasis.


Cancer Research | 2014

Abstract LB-53: Regulatory role of miR-142-3p on the functional hepatic cancer stem cell marker CD133

Kai Y. Ng; Stella Chai; Man Tong; Pak Shing Kwan; Yuen Piu Chan; Terence Kin-Wah Lee; Nathalie Wong; Xin Yuan Guan; Stephanie Ma

Tumor relapse after therapy typifies hepatocellular carcinoma (HCC) and is believed to be attributable to residual cancer stem cells (CSCs) that survive treatment. We have previously identified a CSC population derived from HCC that is characterized by CD133. Despite our growing knowledge of the importance of this subset of cells in driving HCC, the regulatory mechanism of CD133 is not known. Epigenetic changes are believed to be essential in the control of cancer and stem cells. Here, we report the epigenetic regulation of CD133 by miR-142-3p. The interaction between CD133 and miR-142-3p was identified by in silico prediction and substantiated by luciferase reporter analysis. Expression of CD133 was found to be inversely correlated with miR-142-3p in HCC clinical samples as well as in cell lines. Importantly, lower miR-142-3p expression in HCC was significantly associated with worst survival. Functional studies with miR-142-3p stably transduced in HCC cells demonstrated a diminished ability to self-renew, initiate tumor growth, invade, migrate, induce angiogenesis and resist chemotherapy. Rescue experiments whereby CD133 and miR-142-3p is simultaneously overexpressed compensated the deregulated ability of the cells to confer these features. Thus, miR-142-3p directly targets CD133 to regulate its ability to confer cancer and stem cell-like features in HCC.


Cancer Research | 2013

Abstract 3023: RNA-Seq identifies neuropilin-2 as a novel oncogene in esophageal squamous cell carcinoma.

Tsun Ming Fung; Man Tong; Pak Shing Kwan; Yuen Piu Chan; Xin Yuan Guan; Stephanie Ma

Esophageal squamous cell carcinoma (ESCC), the major histologic subtype of esophageal cancer, is one of the most common and deadliest cancers in the world, with survival rates of less than 15%. Like many other solid tumors, the pathogenesis of ESCC is believed to be a multistep process with accumulation of numerous genetic alterations involving de-regulation of both tumor-suppressor genes and oncogenes. An understanding of the molecular events that result in the development and progression of the disease may lead to treatments that will increase survival rates of ESCC patients. Recent advances in sequencing technologies offer the opportunity to characterize the cancer genome at unprecedented depth and sensitivity. RNA-Seq has proved an effective means of precisely quantifying the changing expression levels of each transcript. In this study, we performed integrative RNA-Seq analysis on 3 paired patient-derived non-tumor and ESCC clinical specimens and discovered a number of commonly differentially expressed genes, of which included a significantly de-regulated neuropilin (NRP) family including neuropilin-1 (NRP1) and neuropilin-2 (NRP2). NRP is a single-spanned membrane glycoprotein. Members of the family have previously been shown to play an important role in various cancer types and to be a co-receptor of vascular endothelial growth factors. Yet to date, its role in ESCC has not been explored. Up-regulation of both NRP1 and NRP2 in ESCC was subsequently validated in the original 3 pairs of clinical samples used for RNA-Seq as well as in a larger cohort of samples (n = 40) by qPCR. Given that NRP2 de-regulation in ESCC is more significant, we chose to first focus our study on this member of the family. Immunohistochemical studies likewise found NRP2 to be up-regulated in ESCC as compared with its non-tumor counterparts in a tissue microarray. Consistently, expression studies in a panel of esophageal cell lines found NRP2 to be expressed at a significantly higher level in 7 of the 8 ESCC cell lines examined compared with immortalized normal esophageal cell lines, NE1 and NE3. Functional studies in ESCC cells, KYSE180 and KYSE140, with NRP2 stably repressed by lentiviral-based shRNA approach impaired proliferation and metastatic abilities in vitro, as demonstrated by foci formation and chemotaxis migration assays, respectively. Moreover, conditioned medium collected from NRP2 repressed cells displayed a reduced ability to induce capillary tube formation in HUVEC cells. These results suggest that NRP2 plays an important role in regulating cell proliferation, migration and angiogenesis in ESCC. Additional work on the mechanism by which NRP2 drives ESCC and the effectiveness of the use of a NRP2 neutralizing antibody against ESCC is currently being studied. We believe with further study, NRP2 may provide a prognostic biomarker for ESCC outcome prediction and a novel therapeutic target for ESCC treatment. Citation Format: Tsun Ming Fung, Man Tong, Pak Shing Kwan, Yuen Piu Chan, Xin-Yuan Guan, Stephanie Ma. RNA-Seq identifies neuropilin-2 as a novel oncogene in esophageal squamous cell carcinoma. [abstract]. In: Proceedings of the 104th Annual Meeting of the American Association for Cancer Research; 2013 Apr 6-10; Washington, DC. Philadelphia (PA): AACR; Cancer Res 2013;73(8 Suppl):Abstract nr 3023. doi:10.1158/1538-7445.AM2013-3023

Collaboration


Dive into the Yuen Piu Chan's collaboration.

Top Co-Authors

Avatar

Stephanie Ma

University of Hong Kong

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Kwan Ho Tang

University of Hong Kong

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Terence K. Lee

Hong Kong Polytechnic University

View shared research outputs
Top Co-Authors

Avatar

Man Tong

University of Hong Kong

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Nathalie Wong

The Chinese University of Hong Kong

View shared research outputs
Top Co-Authors

Avatar

Simon Law

University of Hong Kong

View shared research outputs
Researchain Logo
Decentralizing Knowledge