Network


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

Hotspot


Dive into the research topics where Maria Li Lung is active.

Publication


Featured researches published by Maria Li Lung.


Oncogene | 2005

THY1 is a candidate tumour suppressor gene with decreased expression in metastatic nasopharyngeal carcinoma

Hong Lok Lung; Dhinoth Kumar Bangarusamy; Dan Xie; Arthur Kwok Leung Cheung; Yue Cheng; Mande Kuppusamy Kumaran; Lance D. Miller; Edison T. Liu; Xin Yuan Guan; Jonathan S. T. Sham; Yan Fang; LiQiong Li; Nancy Wang; A. I. Protopopov; Eugene R. Zabarovsky; Sai Wah Tsao; Eric J. Stanbridge; Maria Li Lung

Using oligonucleotide microarray analysis, THY1, mapping close to a previously defined 11q22–23 nasopharyngeal carcinoma (NPC) critical region was identified as showing consistent downregulated expression in the tumour segregants, as compared to their parental tumour-suppressing microcell hybrids (MCHs). Gene expression and protein analyses show that THY1 was not expressed in the NPC HONE1 recipient cells, tumour segregants, and other NPC cell lines; THY1 was exclusively expressed in the non-tumourigenic MCHs. The mechanism of THY1 gene inactivation in these cell lines was attributed to hypermethylation. Clinical study showed that in 65% of NPC specimens there was either downregulation or loss of THY1 gene expression. Using a tissue microarray and immunohistochemical staining, 44% of the NPC cases showed downregulated expression of THY1 and 9% lost THY1 expression. The frequency of THY1 downregulated expression in lymph node metastatic NPC was 63%, which was significantly higher than in the primary tumour (33%). After transfection of THY1 gene into HONE1 cells, a dramatic reduction of colony formation ability was observed. These findings suggest that THY1 is a good candidate tumour suppressor gene in NPC, which is significantly associated with lymph node metastases.


Cancer Research | 2006

TSLC1 Is a Tumor Suppressor Gene Associated with Metastasis in Nasopharyngeal Carcinoma

Hong Lok Lung; Arthur Kwok Leung Cheung; Dan Xie; Yue Cheng; Fung Mei Kwong; Yoshinori Murakami; Xin Yuan Guan; Jonathan S. T. Sham; Daniel Chua; A. I. Protopopov; Eugene R. Zabarovsky; Sai Wah Tsao; Eric J. Stanbridge; Maria Li Lung

In up to 87% of nasopharyngeal carcinoma (NPC) clinical tumor specimens, there was either down-regulation or loss of TSLC1 gene expression. Using a tissue microarray and immunohistochemical staining, the frequency of down-regulated or loss of expression of TSLC1 in metastatic lymph node NPC was 83% and the frequency of loss of expression of TSLC1 was 35%, which was significantly higher than that in primary NPC (12%). To examine the possible growth-suppressive activity of TSLC1 in NPC, three NPC cell lines, HONE1, HNE1, and CNE2, were transfected with the wild-type TSLC1 gene cloned into the pCR3.1 expression vector; a reduction of colony formation ability was observed for all three cell lines. A tetracycline-inducible expression vector, pETE-Bsd, was also used to obtain stable transfectants of TSLC1. There was a dramatic difference between colony formation ability in the presence or absence of doxycycline when the gene is shut off or expressed, respectively, with the tetracycline-inducible system. Tumorigenicity assay results show that the activation of TSLC1 suppresses tumor formation in nude mice and functional inactivation of this gene is observed in all the tumors derived from tumorigenic transfectants. Further studies indicate that expression of TSLC1 inhibits HONE1 cell growth in vitro by arresting cells in G(0)-G(1) phase in normal culture conditions, whereas in the absence of serum, TSLC1 induced apoptosis. These findings suggest that TSLC1 is a tumor suppressor gene in NPC, which is significantly associated with lymph node metastases.


Oncogene | 2007

Epigenetic identification of ADAMTS18 as a novel 16q23.1 tumor suppressor frequently silenced in esophageal, nasopharyngeal and multiple other carcinomas

Hongchuan Jin; Xian Wang; Jianming Ying; Ada Ho Yan Wong; Hongyu Li; Kwan Yeung Lee; Gopesh Srivastava; Anthony Tc Chan; Winnie Yeo; B. Ma; Thomas Choudary Putti; Maria Li Lung; Zhong-Ying Shen; Li-Yan Xu; Cordelia Langford; Qian Tao

Tumor suppressor genes (TSGs) often locate at chromosomal regions with frequent deletions in tumors. Loss of 16q23 occurs frequently in multiple tumors, indicating the presence of critical TSGs at this locus, such as the well-studied WWOX. Herein, we found that ADAMTS18, located next to WWOX, was significantly downregulated in multiple carcinoma cell lines. No deletion of ADAMTS18 was detected with multiplex differential DNA-PCR or high-resolution 1-Mb array-based comparative genomic hybridization (CGH) analysis. Instead, methylation of the ADAMTS18 promoter CpG Island was frequently detected with methylation-specific PCR and bisulfite genome sequencing in multiple carcinoma cell lines and primary carcinomas, but not in any nontumor cell line and normal epithelial tissue. Both pharmacological and genetic demethylation dramatically induced the ADAMTS18 expression, indicating that CpG methylation directly contributes to the tumor-specific silencing of ADAMTS18. Ectopic ADAMTS18 expression led to significant inhibition of both anchorage-dependent and -independent growth of carcinoma cells lacking the expression. Thus, through functional epigenetics, we identified ADAMTS18 as a novel functional tumor suppressor, being frequently inactivated epigenetically in multiple carcinomas.


Virology | 1992

Transcription of BamHI-A region of the EBV genome in NPC tissues and B cells.

Honglin Chen; Maria Li Lung; Jonathan S.T. Sham; D. Choy; B.E. Griffin; M. H. Ng

Analysis by Northern blotting and sequencing of cDNA clones from a transcription library of a tumor biopsy from a nasopharyngeal carcinoma (NPC) patient showed that the BamHI-A region of the Epstein-Barr virus genome is abundantly and regularly transcribed in tumor tissues from NPC patients. The transcription occurred in a rightward direction terminating between coordinates 160,965 and 160,995, where two polyadenylation sites are located. Rightward transcription of this region also occurred in B lymphoid cells harboring the viral genome, albeit at a lower level than in the tumor tissues. Differential splicing yields a family of related transcripts displaying at least four splicing patterns. Different promoters may be utilized, further contributing to the diversity of this family of transcripts. A 2.8-kb unspliced transcript present in B95-8 cells was probably initiated from a TATA box located in position 158,204, while the other transcripts may utilize other promoters localized to other regions. All the transcripts encompass a putative open reading frame, BARFO, which is predicted to encode a basic protein of about 20 kDa. It shares 40% colinear amino acid sequence homology with the DNA binding region of a transcription factor, ICP4, specified by herpes simplex virus.


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.


Oncogene | 2007

Identification of a tumor suppressive critical region mapping to 3p14.2 in esophageal squamous cell carcinoma and studies of a candidate tumor suppressor gene, ADAMTS9.

Paulisally Hau Yi Lo; Alfred Chi Chung Leung; C Y C Kwok; W S Y Cheung; Josephine Mun Yee Ko; Lichun Yang; Simon Law; Li Dong Wang; Li J; Eric J. Stanbridge; G Srivastava; Johnny Cheuk On Tang; S. W. Tsao; Maria Li Lung

A gene critical to esophageal cancer has been identified. Functional studies using microcell-mediated chromosome transfer of intact and truncated donor chromosomes 3 into an esophageal cancer cell line and nude mouse tumorigenicity assays were used to identify a 1.61 Mb tumor suppressive critical region (CR) mapping to chromosome 3p14.2. This CR is bounded by D3S1600 and D3S1285 microsatellite markers. One candidate tumor suppressor gene, ADAMTS9, maps to this CR. Further studies showed normal expression levels of this gene in tumor-suppressed microcell hybrids, levels that were much higher than observed in the recipient cells. Complete loss or downregulation of ADAMTS9 gene expression was found in 15 out of 16 esophageal carcinoma cell lines. Promoter hypermethylation was detected in the cell lines that do not express this gene. Re-expression of ADAMTS9 was observed after demethylation drug treatment, confirming that hypermethylation is involved in gene downregulation. Downregulation of ADAMTS9 was also found in 43.5 and 47.6% of primary esophageal tumor tissues from Hong Kong and from the high-risk region of Henan, respectively. Thus, this study identifies and provides functional evidence for a CR associated with tumor suppression on 3p14.2 and provides the first evidence that ADAMTS9, mapping to this region, may contribute to esophageal cancer development.


Proceedings of the National Academy of Sciences of the United States of America | 2012

Cyclin D1 overexpression supports stable EBV infection in nasopharyngeal epithelial cells

Chi Man Tsang; Yim Ling Yip; Kwok Wai Lo; Wen Deng; Ka Fai To; Pok Man Hau; Victoria Ming Yi Lau; Kenzo Takada; Vivian Wai Yan Lui; Maria Li Lung; Honglin Chen; Mu Sheng Zeng; Jaap M. Middeldorp; Annie Lai Man Cheung; Sai Wah Tsao

Undifferentiated nasopharyngeal carcinomas (NPCs) are commonly present with latent EBV infection. However, events regulating EBV infection at early stages of the disease and the role of EBV in disease pathogenesis are largely undefined. Genetic alterations leading to activation of cyclin D1 signaling in premalignant nasopharyngeal epithelial (NPE) cells have been postulated to predispose cells to EBV infection. We previously reported that loss of p16, a negative regulator of cyclin D1 signaling, is a frequent feature of NPC tumors. Here, we report that early premalignant lesions of nasopharyngeal epithelium overexpress cyclin D1. Furthermore, overexpression of cyclin D1 is closely associated with EBV infection. Therefore we investigated the potential role of cyclin D1 overexpression in dysplastic NPE cells in vitro. In human telomerase reverse transcriptase-immortalized NPE cells, overexpression of cyclin D1 or a p16-resistant form of CDK4 (CDK4R24C) suppressed differentiation. This suppression may have implications for the close association of EBV infection with undifferentiated NPC. In these in vitro models, we found that cellular growth arrest and senescence occurred in EBV-infected cell populations immediately after infection. Nevertheless, overexpression of cyclin D1 or a p16-resistant form of CDK4 or knockdown of p16 in the human telomerase reverse transcriptase-immortalized NPE cell lines could counteract the EBV-induced growth arrest and senescence. We conclude that dysregulated expression of cyclin D1 in NPE cells may contribute to NPC pathogenesis by enabling persistent infection of EBV.


The Lancet | 1985

EVIDENCE THAT RESPIRATORY TRACT IS MAJOR RESERVOIR FOR EPSTEIN-BARR VIRUS

Maria Li Lung; S. Y. So; King-Pan Chan; Wah-Kit Lam; Wai Pang Lam; M. H. Ng

Exfoliated cells harvested from bronchial washings of 53 patients with suspected bronchogenic carcinoma were tested by means of DNA dot hybridisation using the cloned large internal repeat (IR) sequence of Epstein-Barr virus (EBV) genome as a probe. 25 of these patients gave positive results. Since the patients had diseases that were not related to the virus, this finding suggests that the lower respiratory tract is a major reservoir for EBV. Attempts at cellular localisation of the virus revealed only an occasional cell which harboured the viral genome or expressed viral capsid antigens. These cells could not account for the quantity of the viral DNA detected in bronchial washings. Moreover, patients had similar profiles of serum EBV antibodies whether they were positive or negative for EBV DNA by dot hybridisation. These findings are compatible with a state of viral latency in which cells harbour a low copy number of the viral genome. Viral expression rarely occurs in these cells, which seem to elicit a minimum host immune response. If it is assumed that each latently infected cell harbours a maximum of approximately 30 EBV genomes (which is the lower limit of detection by the in-situ hybridisation method used in this study), the findings suggest that a considerable proportion of the exfoliative cells from the lower respiratory tract, of the order of 0.1-16%, harbour latent EBV.


Cancer Research | 2010

Extracellular protease ADAMTS9 suppresses esophageal and nasopharyngeal carcinoma tumor formation by inhibiting angiogenesis

Paulisally Hau Yi Lo; Hong Lok Lung; Arthur Kwok Leung Cheung; Suneel S. Apte; Kwok Wah Chan; Fung Mei Kwong; Josephine Mun Yee Ko; Yue Cheng; Simon Law; Gopesh Srivastava; Eugene R. Zabarovsky; Sai Wah Tsao; Johnny Cheuk On Tang; Eric J. Stanbridge; Maria Li Lung

ADAMTS metalloprotease family member ADAMTS9 maps to 3p14.2 and shows significant associations with the aerodigestive tract cancers esophageal squamous cell carcinoma (ESCC) and nasopharyngeal carcinoma (NPC). However, the functional impact of ADAMTS9 on cancer development has not been explored. In this study, we evaluated the hypothesized antiangiogenic and tumor-suppressive functions of ADAMTS9 in ESCC and NPC, in stringent tumorigenicity and Matrigel plug angiogenesis assays. ADAMTS9 activation suppressed tumor formation in nude mice. Conversely, knockdown of ADAMTS9 resulted in clones reverting to the tumorigenic phenotype of parental cells. In vivo angiogenesis assays revealed a reduction in microvessel numbers in gel plugs injected with tumor-suppressive cell transfectants. Similarly, conditioned medium from cell transfectants dramatically reduced the tube-forming capacity of human umbilical vein endothelial cells. These activities were associated with a reduction in expression levels of the proangiogenic factors MMP9 and VEGFA, which were consistently reduced in ADAMTS9 transfectants derived from both cancers. Taken together, our results indicate that ADAMTS9 contributes an important function in the tumor microenvironment that acts to inhibit angiogenesis and tumor growth in both ESCC and NPC.


Cancer Epidemiology, Biomarkers & Prevention | 2008

CYP1A1, GSTM1, and GSTT1 Polymorphisms, Smoking, and Lung Cancer Risk in a Pooled Analysis among Asian Populations

Kyoung Mu Lee; Daehee Kang; Margie L. Clapper; Magnus Ingelman-Sundberg; Masko Ono-Kihara; Chikako Kiyohara; Shen Min; Qing Lan; Loic Le Marchand; Pinpin Lin; Maria Li Lung; Hatice Pinarbasi; Paola Pisani; Petcharin Srivatanakul; Adeline Seow; Haruhiko Sugimura; Shinkan Tokudome; Jun Yokota; Emanuela Taioli

To evaluate the roles of CYP1A1 polymorphisms [Ile462Val and T6235C (MspI)] and deletion of GSTM1 and GSTT1 in lung cancer development in Asian populations, a pooled analysis was conducted on 13 existing studies included in Genetic Susceptibility to Environmental Carcinogenesis database. This pooled analysis included 1,971 cases and 2,130 controls. Lung cancer risk was estimated as odds ratios (OR) and 95% confidence intervals (95% CI) using unconditional logistic regression model adjusting for age, sex, and pack-year. The CYP1A1 6235C variant was associated with squamous cell lung cancer (TC versus TT: OR, 1.42; 95% CI, 0.96-2.09; CC versus TT: OR, 1.97; 95% CI, 1.26-3.07; Ptrend = 0.003). In haplotype analysis, 462Val-6235T and Ile-C haplotypes were associated with lung cancer risk with reference to the Ile-T haplotype (OR, 3.41; 95% CI, 1.78-6.53 and OR, 1.39; 95% CI, 1.12-1.71, respectively). The GSTM1-null genotype increased squamous cell lung cancer risk (OR, 1.36; 95% CI, 1.05-1.77). When the interaction was evaluated with smoking, increasing trend of lung cancer risk as pack-year increased was stronger among those with the CYP1A1 6235 TC/CC genotype compared with those with TT genotype (Pinteraction = 0.001) and with the GSTM1-null genotype compared with the present type (Pinteraction = 0.08, when no genotype effect with no exposure was assumed). These results suggest that genetic polymorphisms in CYP1A1 and GSTM1 are associated with lung cancer risk in Asian populations. However, further investigation is warranted considering the relatively small sample size when subgroup analyses were done and the lack of environmental exposure data other than smoking. (Cancer Epidemiol Biomarkers Prev 2008;17(5):1120–6)

Collaboration


Dive into the Maria Li Lung's collaboration.

Top Co-Authors

Avatar

Josephine Mun Yee Ko

Hong Kong University of Science and Technology

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Yue Cheng

Hong Kong University of Science and Technology

View shared research outputs
Top Co-Authors

Avatar

Simon Law

University of Hong Kong

View shared research outputs
Top Co-Authors

Avatar

Wei Dai

University of Hong Kong

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge