Network


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

Hotspot


Dive into the research topics where Belamy B. Cheung is active.

Publication


Featured researches published by Belamy B. Cheung.


Cell Death & Differentiation | 2013

The histone deacetylase SIRT2 stabilizes Myc oncoproteins.

Pei Yan Liu; Ning Xu; Alena Malyukova; Christopher J. Scarlett; Yuting Sun; Xu Dong Zhang; Dora Ling; Shih-Ping Su; Charlotte Nelson; David K. Chang; Jessica Koach; Andrew E. Tee; Michelle Haber; Norris; Christopher Toon; Ilse Rooman; Chengyuan Xue; Belamy B. Cheung; Sharad Kumar; Glenn M. Marshall; Andrew V. Biankin; Tao Liu

Myc oncoproteins are commonly upregulated in human cancers of different organ origins, stabilized by Aurora A, degraded through ubiquitin–proteasome pathway-mediated proteolysis, and exert oncogenic effects by modulating gene and protein expression. Histone deacetylases are emerging as targets for cancer therapy. Here we demonstrated that the class III histone deacetylase SIRT2 was upregulated by N-Myc in neuroblastoma cells and by c-Myc in pancreatic cancer cells, and that SIRT2 enhanced N-Myc and c-Myc protein stability and promoted cancer cell proliferation. Affymetrix gene array studies revealed that the gene most significantly repressed by SIRT2 was the ubiquitin–protein ligase NEDD4. Consistent with this finding, SIRT2 repressed NEDD4 gene expression by directly binding to the NEDD4 gene core promoter and deacetylating histone H4 lysine 16. Importantly, NEDD4 directly bound to Myc oncoproteins and targeted Myc oncoproteins for ubiquitination and degradation, and small-molecule SIRT2 inhibitors reactivated NEDD4 gene expression, reduced N-Myc and c-Myc protein expression, and suppressed neuroblastoma and pancreatic cancer cell proliferation. Additionally, SIRT2 upregulated and small-molecule SIRT2 inhibitors decreased Aurora A expression. Our data reveal a novel pathway critical for Myc oncoprotein stability, and provide important evidences for potential application of SIRT2 inhibitors for the prevention and therapy of Myc-induced malignancies.


Nature Reviews Cancer | 2014

The prenatal origins of cancer

Glenn M. Marshall; Daniel Carter; Belamy B. Cheung; Tao Liu; Marion K. Mateos; Justin G. Meyerowitz; William A. Weiss

The concept that some childhood malignancies arise from postnatally persistent embryonal cells has a long history. Recent research has strengthened the links between driver mutations and embryonal and early postnatal development. This evidence, coupled with much greater detail on the cell of origin and the initial steps in embryonal cancer initiation, has identified important therapeutic targets and provided renewed interest in strategies for the early detection and prevention of childhood cancer.


Oncogene | 1998

Favorable prognostic significance of high-level retinoic acid receptor β expression in neuroblastoma mediated by effects on cell cycle regulation

Belamy B. Cheung; Jayne E. Hocker; Stewart A. Smith; Murray D. Norris; Michelle Haber; Glenn M. Marshall

We have previously shown that ectopic overexpression of retinoic acid receptor (RAR) subtypes α, β and γ in human neuroblastoma cells had different effects on growth and retinoid sensitivity. Only overexpressed RARβ induced profound growth inhibition in the absence of additional retinoid, and increased retinoid sensitivity. In this study, we measured mRNA expression levels of RARα, β, and γ in 50 primary neuroblastoma tumor samples, and found a strong correlation between favorable patient prognosis and high-level RARβ expression. Human neuroblastoma cells transfected with a vector expressing RARβ demonstrated irreversible growth arrest following a 1 week exposure to all-trans-retinoic acid, whereas control cells continued to proliferate. In the absence of additional retinoid, RARβ transfectants demonstrated a higher proportion of cells in the G0/G1 phase of the cell cycle, increased p21WAF1/CIP1 expression and specific binding to a retinoic acid response element. These were changes which we also observed in control neuroblastoma cells following retinoid treatment. Our data indicate that RARβ is an important factor mediating the growth inhibitory effects of retinoids in neuroblastoma cells. The favorable effect of high-level RARβ expression on prognosis in primary tumor tissue may occur through RARβ effects on p21 expression and consequent G0/G1 cell cycle arrest.


PLOS ONE | 2012

TRIM16 acts as an E3 ubiquitin ligase and can heterodimerize with other TRIM family members.

Jessica L. Bell; Alena Malyukova; Jessica K. Holien; Jessica Koach; Michael W. Parker; Maria Kavallaris; Glenn M. Marshall; Belamy B. Cheung

The TRIM family of proteins is distinguished by its tripartite motif (TRIM). Typically, TRIM proteins contain a RING finger domain, one or two B-box domains, a coiled-coil domain and the more variable C-terminal domains. TRIM16 does not have a RING domain but does harbour two B-box domains. Here we showed that TRIM16 homodimerized through its coiled-coil domain and heterodimerized with other TRIM family members; TRIM24, Promyelocytic leukaemia (PML) protein and Midline-1 (MID1). Although, TRIM16 has no classic RING domain, three-dimensional modelling of TRIM16 suggested that its B-box domains adopts RING-like folds leading to the hypothesis that TRIM16 acts as an ubiquitin ligase. Consistent with this hypothesis, we demonstrated that TRIM16, devoid of a classical RING domain had auto-polyubiquitination activity and acted as an E3 ubiquitin ligase in vivo and in vitro assays. Thus via its unique structure, TRIM16 possesses both heterodimerization function with other TRIM proteins and also has E3 ubiquitin ligase activity.


Oncogene | 2013

Direct effects of Bmi1 on p53 protein stability inactivates oncoprotein stress responses in embryonal cancer precursor cells at tumor initiation

M Calao; Eric Sekyere; Hongjuan Cui; Belamy B. Cheung; W D Thomas; Joanna Keating; J Chen; Anna Raif; K Jankowski; N P Davies; M V Bekkum; B Chen; Owen Tan; Tammy Ellis; Murray D. Norris; Michelle Haber; E S Kim; Jason M. Shohet; Toby Trahair; Tao Liu; Brandon J. Wainwright; Han Fei Ding; Glenn M. Marshall

Embryonal cancer can arise from postnatally persistent embryonal remnant or rest cells, which are uniquely characterized by the absence of p53 mutations. Perinatal overexpression of the MycN oncoprotein in embryonal cancer precursor cells causes postnatal rests, and later tumor formation through unknown mechanisms. However, overexpression of Myc in adult tissues normally activates apoptosis and/or senescence signals as an organismal defense mechanism against cancer. Here, we show that perinatal neuroblastoma precursor cells exhibited a transiently diminished p53 response to MycN oncoprotein stress and resistance to trophic factor withdrawal, compared with their adult counterpart cells from the TH-MYCN+/+ transgenic mouse model of neuroblastoma. The adult stem cell maintenance factor and Polycomb group protein, Bmi1 (B-cell-specific Moloney murine leukemia virus integration site), had a critical role at neuroblastoma initiation in the model, by repressing p53 responses in precursor cells. We further show in neuroblastoma tumor cells that Bmi1 could directly bind p53 in a complex with other Polycomb complex proteins, Ring1A or Ring1B, leading to increased p53 ubiquitination and degradation. Repressed p53 signal responses were also seen in precursor cells for other embryonal cancer types, medulloblastoma and acute lymphoblastic leukemia. Collectively, these date indicate a general mechanism for p53 inactivation in some embryonal cell types and consequent susceptibility to MycN oncogenesis at the point of embryonal tumor initiation.


Journal of the National Cancer Institute | 2014

Effects of a Novel Long Noncoding RNA, lncUSMycN, on N-Myc Expression and Neuroblastoma Progression

Pei Y. Liu; Daniela Erriquez; Glenn M. Marshall; Andrew E. Tee; Patsie Polly; Mathew Wong; Bing Liu; Jessica L. Bell; Xu D. Zhang; Giorgio Milazzo; Belamy B. Cheung; Archa H. Fox; Alexander Swarbrick; Stefan Hüttelmaier; Maria Kavallaris; Giovanni Perini; John S. Mattick; Marcel E. Dinger; Tao Liu

BACKGROUND Patients with neuroblastoma due to the amplification of a 130-kb genomic DNA region containing the MYCN oncogene have poor prognoses. METHODS Bioinformatics data were used to discover a novel long noncoding RNA, lncUSMycN, at the 130-kb amplicon. RNA-protein pull-down assays were used to identify proteins bound to lncUSMycN RNA. Kaplan-Meier survival analysis, multivariable Cox regression, and two-sided log-rank test were used to examine the prognostic value of lncUSMycN and NonO expression in three cohorts of neuroblastoma patients (n = 47, 88, and 476, respectively). Neuroblastoma-bearing mice were treated with antisense oligonucleotides targeting lncUSMycN (n = 12) or mismatch sequence (n = 13), and results were analyzed by multiple comparison two-way analysis of variance. All statistical tests were two-sided. RESULTS Bioinformatics data predicted lncUSMycN gene and RNA, and reverse-transcription polymerase chain reaction confirmed its three exons and two introns. The lncUSMycN gene was coamplified with MYCN in 88 of 341 human neuroblastoma tissues. lncUSMycN RNA bound to the RNA-binding protein NonO, leading to N-Myc RNA upregulation and neuroblastoma cell proliferation. High levels of lncUSMycN and NonO expression in human neuroblastoma tissues independently predicted poor patient prognoses (lncUSMycN: hazard ratio [HR] = 1.87, 95% confidence interval [CI] = 1.06 to 3.28, P = .03; NonO: HR = 2.48, 95% CI = 1.34 to 4.57, P = .004). Treatment with antisense oligonucleotides targeting lncUSMycN in neuroblastoma-bearing mice statistically significantly hindered tumor progression (P < .001). CONCLUSIONS Our data demonstrate the important roles of lncUSMycN and NonO in regulating N-Myc expression and neuroblastoma oncogenesis and provide the first evidence that amplification of long noncoding RNA genes can contribute to tumorigenesis.


Oncogene | 2009

Patched1 deletion increases N-Myc protein stability as a mechanism of medulloblastoma initiation and progression

Wayne Thomas; J Chen; Y R Gao; Belamy B. Cheung; Jessica Koach; Eric Sekyere; Murray D. Norris; Michelle Haber; Tammy Ellis; Brandon J. Wainwright; Glenn M. Marshall

Medulloblastoma tumorigenesis caused by inactivating mutations in the PATCHED1 (PTCH1) gene is initiated by persistently activated Sonic Hedgehog (Shh) signaling in granule neuron precursors (GNPs) during the late stages of cerebellar development. Both normal cerebellar development and Shh-driven medulloblastoma tumorigenesis require N-Myc expression. However, the mechanisms by which N-Myc affects the stages of medulloblastoma initiation and progression are unknown. Here we used a mouse model of Ptch1 heterozygosity and medulloblastoma to show that increased N-Myc expression characterized the earliest selection of focal GNP hyperplasia destined for later tumor progression. Step-wise loss of Ptch1 expression, from tumor initiation to progression, led to incremental increases in N-Myc protein, rather than mRNA, expression. Increased N-Myc resulted in enhanced proliferation and death resistance of perinatal GNPs at tumor initiation. Sequential N-Myc protein phosphorylation at serine-62 and serine-62/threonine-58 characterized the early and late stages of medulloblastoma tumorigenesis, respectively. Shh pathway activation led to increased Myc protein stability and reduced expression of key regulatory factors. Taken together our data identify N-Myc protein stability as the result of loss of Ptch1, which distinguishes normal cerebellar development from medulloblastoma tumorigenesis.


British Journal of Cancer | 2005

The retinoid anticancer signal: mechanisms of target gene regulation

Tao Liu; A Bohlken; Selena Kuljaca; Michelle Lee; Tue Nguyen; Stewart A. Smith; Belamy B. Cheung; Murray D. Norris; Michelle Haber; A J Holloway; David Bowtell; Glenn M. Marshall

Retinoids induce growth arrest, differentiation, and cell death in many cancer cell types. One factor determining the sensitivity or resistance to the retinoid anticancer signal is the transcriptional response of retinoid-regulated target genes in cancer cells. We used cDNA microarray to identify 31 retinoid-regulated target genes shared by two retinoid-sensitive neuroblastoma cell lines, and then sought to determine the relevance of the target gene responses to the retinoid anticancer signal. The pattern of retinoid responsiveness for six of 13 target genes (RARβ2, CYP26A1, CRBP1, RGS16, DUSP6, EGR1) correlated with phenotypic retinoid sensitivity, across a panel of retinoid-sensitive or -resistant lung and breast cancer cell lines. Retinoid treatment of MYCN transgenic mice bearing neuroblastoma altered the expression of five of nine target genes examined (RARβ2, CYP26A1, CRBP1, DUSP6, PLAT) in neuroblastoma tumour tissue in vivo. In retinoid-sensitive neuroblastoma, lung and breast cancer cell lines, direct inhibition of retinoid-induced RARβ2 expression blocked induction of only one of eight retinoid target genes (CYP26A1). DNA demethylation, histone acetylation, and exogenous overexpression of RARβ2 partially restored retinoid-responsive CYP26A1 expression in RA-resistant MDA-MB-231 breast, but not SK-MES-1 lung, cancer cells. Combined, rather than individual, inhibition of DUSP6 and RGS16 was required to block retinoid-induced growth inhibition in neuroblastoma cells, through phosphorylation of extracellular-signal-regulated kinase. In conclusion, sensitivity to the retinoid anticancer signal is determined in part by the transcriptional response of key retinoid-regulated target genes, such as RARβ2, DUSP6, and RGS16.


Oncogene | 2010

TRIM16 acts as a tumour suppressor by inhibitory effects on cytoplasmic vimentin and nuclear E2F1 in neuroblastoma cells

Glenn M. Marshall; Jessica L. Bell; Jessica Koach; Owen Tan; Patrick Y. Kim; Alena Malyukova; Wayne Thomas; Eric Sekyere; Tao Liu; Anne M. Cunningham; Vivienne Tobias; Murray D. Norris; Michelle Haber; Maria Kavallaris; Belamy B. Cheung

The family of tripartite-motif (TRIM) proteins are involved in diverse cellular processes, but are often characterized by critical protein–protein interactions necessary for their function. TRIM16 is induced in different cancer types, when the cancer cell is forced to proceed down a differentiation pathway. We have identified TRIM16 as a DNA-binding protein with histone acetylase activity, which is required for the retinoic acid receptor β2 transcriptional response in retinoid-treated cancer cells. In this study, we show that overexpressed TRIM16 reduced neuroblastoma cell growth, enhanced retinoid-induced differentiation and reduced tumourigenicity in vivo. TRIM16 was only expressed in the differentiated ganglion cell component of primary human neuroblastoma tumour tissues. TRIM16 bound directly to cytoplasmic vimentin and nuclear E2F1 in neuroblastoma cells. TRIM16 reduced cell motility and this required downregulation of vimentin. Retinoid treatment and enforced overexpression caused TRIM16 to translocate to the nucleus, and bind to and downregulate nuclear E2F1, required for cell replication. This study, for the first time, demonstrates that TRIM16 acts as a tumour suppressor, affecting neuritic differentiation, cell migration and replication through interactions with cytoplasmic vimentin and nuclear E2F1 in neuroblastoma cells.


Oncotarget | 2016

The long noncoding RNA MALAT1 promotes tumor-driven angiogenesis by up-regulating pro-angiogenic gene expression

Andrew E. Tee; Bing Liu; Renhua Song; Jinyan Li; Eddy Pasquier; Belamy B. Cheung; Cizhong Jiang; Glenn M. Marshall; Michelle Haber; Murray D. Norris; Jamie I. Fletcher; Marcel E. Dinger; Tao Liu

Neuroblastoma is the most common solid tumor during early childhood. One of the key features of neuroblastoma is extensive tumor-driven angiogenesis due to hypoxia. However, the mechanism through which neuroblastoma cells drive angiogenesis is poorly understood. Here we show that the long noncoding RNA MALAT1 was upregulated in human neuroblastoma cell lines under hypoxic conditions. Conditioned media from neuroblastoma cells transfected with small interfering RNAs (siRNA) targeting MALAT1, compared with conditioned media from neuroblastoma cells transfected with control siRNAs, induced significantly less endothelial cell migration, invasion and vasculature formation. Microarray-based differential gene expression analysis showed that one of the genes most significantly down-regulated following MALAT1 suppression in human neuroblastoma cells under hypoxic conditions was fibroblast growth factor 2 (FGF2). RT-PCR and immunoblot analyses confirmed that MALAT1 suppression reduced FGF2 expression, and Enzyme-Linked Immunosorbent Assays revealed that transfection with MALAT1 siRNAs reduced FGF2 protein secretion from neuroblastoma cells. Importantly, addition of recombinant FGF2 protein to the cell culture media reversed the effects of MALAT1 siRNA on vasculature formation. Taken together, our data suggest that up-regulation of MALAT1 expression in human neuroblastoma cells under hypoxic conditions increases FGF2 expression and promotes vasculature formation, and therefore plays an important role in tumor-driven angiogenesis.

Collaboration


Dive into the Belamy B. Cheung's collaboration.

Top Co-Authors

Avatar

Glenn M. Marshall

Boston Children's Hospital

View shared research outputs
Top Co-Authors

Avatar

Michelle Haber

University of New South Wales

View shared research outputs
Top Co-Authors

Avatar

Murray D. Norris

University of New South Wales

View shared research outputs
Top Co-Authors

Avatar

Tao Liu

University of New South Wales

View shared research outputs
Top Co-Authors

Avatar

Daniel Carter

University of New South Wales

View shared research outputs
Top Co-Authors

Avatar

Jessica Koach

University of New South Wales

View shared research outputs
Top Co-Authors

Avatar

Bing Liu

University of New South Wales

View shared research outputs
Top Co-Authors

Avatar

Maria Kavallaris

University of New South Wales

View shared research outputs
Top Co-Authors

Avatar

Owen Tan

University of New South Wales

View shared research outputs
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge