Network


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

Hotspot


Dive into the research topics where Edward Wang is active.

Publication


Featured researches published by Edward Wang.


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

Aurora B kinase phosphorylates and instigates degradation of p53

Chris Gully; Guermarie Velazquez-Torres; Ji Hyun Shin; Enrique Fuentes-Mattei; Edward Wang; Colin Carlock; Jian Chen; Daniel Rothenberg; Henry P. Adams; Hyun Ho Choi; Sergei Guma; Liem Phan; Ping Chieh Chou; Chun Hui Su; Fanmao Zhang; Jiun Sheng Chen; Tsung Ying Yang; Sai Ching J. Yeung; Mong Hong Lee

Aurora B is a mitotic checkpoint kinase that plays a pivotal role in the cell cycle, ensuring correct chromosome segregation and normal progression through mitosis. Aurora B is overexpressed in many types of human cancers, which has made it an attractive target for cancer therapies. Tumor suppressor p53 is a genome guardian and important negative regulator of the cell cycle. Whether Aurora B and p53 are coordinately regulated during the cell cycle is not known. We report that Aurora B directly interacts with p53 at different subcellular localizations and during different phases of the cell cycle (for instance, at the nucleus in interphase and the centromeres in prometaphase of mitosis). We show that Aurora B phosphorylates p53 at S183, T211, and S215 to accelerate the degradation of p53 through the polyubiquitination–proteasome pathway, thus functionally suppressing the expression of p53 target genes involved in cell cycle inhibition and apoptosis (e.g., p21 and PUMA). Pharmacologic inhibition of Aurora B in cancer cells with WT p53 increased p53 protein level and expression of p53 target genes to inhibit tumor growth. Together, these results define a mechanism of p53 inactivation during the cell cycle and imply that oncogenic hyperactivation or overexpression of Aurora B may compromise the tumor suppressor function of p53. We have elucidated the antineoplastic mechanism for Aurora B kinase inhibitors in cancer cells with WT p53.


Nature Communications | 2015

The cell cycle regulator 14-3-3σ opposes and reverses cancer metabolic reprogramming

Liem Phan; Ping Chieh Chou; Guermarie Velazquez-Torres; Ismael Samudio; Kenneth Parreno; Yaling Huang; Chieh Tseng; Thuy Vu; Chris Gully; Chun Hui Su; Edward Wang; Jian Chen; Hyun Ho Choi; Enrique Fuentes-Mattei; Ji-Hyun Shin; Christine Y. Shiang; Brian C. Grabiner; Marzenna Blonska; Stephen Skerl; Yiping Shao; Dianna Cody; Jorge Delacerda; Charles Kingsley; Douglas Webb; Colin Carlock; Zhongguo Zhou; Yun Chih Hsieh; Jae-Hyuk Lee; Andrew M. Elliott; Marc S. Ramirez

Summary Extensive reprogramming of cellular energy metabolism is a hallmark of cancer. Despite its importance, the molecular mechanism controlling this tumour metabolic shift remains not fully understood. Here we show that 14-3-3σ regulates cancer metabolic reprogramming and protects cells from tumourigenic transformation. 14-3-3σ opposes tumour-promoting metabolic programs by enhancing c-Myc poly-ubiquitination and subsequent degradation. 14-3-3σ demonstrates the suppressive impact on cancer glycolysis, glutaminolysis, mitochondrial biogenesis and other major metabolic processes of tumours. Importantly, 14-3-3σ expression levels predict overall and recurrence-free survival rates, tumour glucose uptake and metabolic gene expression in breast cancer patients. Thus, these results highlight that 14-3-3σ is an important regulator of tumour metabolism, and loss of 14-3-3σ expression is critical for cancer metabolic reprogramming. We anticipate that pharmacologically elevating the function of 14-3-3σ in tumours could be a promising direction for targeted anti-cancer metabolism therapy development in future.


Nature Communications | 2014

CSN6 drives carcinogenesis by positively regulating Myc stability

Jian Chen; Ji Hyun Shin; Ruiying Zhao; Liem Phan; Hua Wang; Yuwen Xue; Sean M. Post; Hyun Ho Choi; Jiun Sheng Chen; Edward Wang; Zhongguo Zhou; Chieh Tseng; Christopher Gully; Guermarie Velazquez-Torres; Enrique Fuentes-Mattei; Giselle Yeung; Yi Qiao; Ping Chieh Chou; Chun Hui Su; Yun Chih Hsieh; Shih Lan Hsu; Kazufumi Ohshiro; Tattym Shaikenov; Huamin Wang; Sai Ching Jim Yeung; Mong Hong Lee

Summary Cullin-RING ubiquitin ligases (CRL) are critical in ubiquitinating Myc, while COP9 signalosome (CSN) controls neddylation of Cullin in CRL. The mechanistic link between Cullin neddylation and Myc ubiquitination/degradation is unclear. Here we show that Myc is a target of the CSN subunit 6 (CSN6)–Cullin signaling axis and that CSN6 is a positive regulator of Myc. CSN6 enhanced neddylation of Cullin-1 and facilitated auto-ubiquitination/degradation of Fbxw7, a component of CRL involved in Myc ubiquitination, thereby stabilizing Myc. Csn6 haplo-insufficiency decreased Cullin-1 neddylation but increased Fbxw7 stability to compromise Myc stability and activity in an Eµ-Myc mouse model, resulting in decelerated lymphomagenesis. We found that CSN6 overexpression, which leads to aberrant expression of Myc target genes, is frequent in human cancers. Together, these results define a mechanism for the regulation of Myc stability through the CSN-Cullin-Fbxw7 axis and provide insights into the correlation of CSN6 overexpression with Myc stabilization/activation during tumorigenesis.


Cell Cycle | 2015

Nuclear PTEN tumor-suppressor functions through maintaining heterochromatin structure.

Lili Gong; Jeane M. Govan; Elizabeth B. Evans; Hui Dai; Edward Wang; Szu Wei Lee; Hui Kuan Lin; Alexander J. Lazar; Gordon B. Mills; Shiaw Yih Lin

The tumor suppressor, PTEN, is one of the most commonly mutated genes in cancer. Recently, PTEN has been shown to localize in the nucleus and is required to maintain genomic stability. Here, we show that nuclear PTEN, independent of its phosphatase activity, is essential for maintaining heterochromatin structure. Depletion of PTEN leads to loss of heterochromatic foci, decreased chromatin compaction, overexpression of heterochromatic genes, and reduced protein stability of heterochromatin protein 1 α. We found that the C-terminus of PTEN is required to maintain heterochromatin structure. Additionally, cancer-associated PTEN mutants lost their tumor-suppressor function when their heterochromatin structure was compromised. We propose that this novel role of PTEN accounts for its function in guarding genomic stability and suppressing tumor development.


Cancer Research | 2015

TUSC4 functions as a tumor suppressor by regulating BRCA1 stability.

Yang Peng; Hui Dai; Edward Wang; Curtis Chun Jen Lin; Wei Mo; Guang Peng; Shiaw Yih Lin

BRCA1 expression is lost frequently in breast cancers in which it promotes malignant development. In the present study, we performed a global expression analysis of breast cancer cells in which the tumor-suppressor candidate gene TUSC4 was silenced to gain insights into its function. TUSC4 silencing affected genes involved in cell cycle and cell death, which have broad reaching influence on cancer development. Most importantly, we found a cluster pattern of gene-expression profiles in TUSC4-silenced cells that defined a homologous recombination (HR) repair defect signature. Mechanistic investigations indicated that TUSC4 protein could physically interact with the E3 ligase Herc2, which prevents BRCA1 degradation through the ubiquitination pathway. TUSC4 silencing enhanced BRCA1 polyubiquitination, leading to its degradation and a marked reduction in HR repair efficiency. Notably, ectopic expression of TUSC4 suppressed the proliferation, invasion, and colony formation of breast cancer cells in vitro and tumorigenesis in vivo. Furthermore, TUSC4 silencing was sufficient to transform normal mammary epithelial cells and to enhance sensitivity to PARP inhibitors. Our results provide a set of genetic and biologic proofs that TUSC4 functions as a bona fide tumor suppressor by regulating the protein stability and function of BRCA1 in breast cancer.


Cell Cycle | 2013

Zinc finger protein 668 interacts with Tip60 to promote H2AX acetylation after DNA damage

Ruozhen Hu; Edward Wang; Guang Peng; Hui Dai; Shiaw Yih Lin

Many tumor suppressors play an important role in the DNA damage pathway. Zinc finger protein 668 (ZNF668) has recently been identified as one of the potential tumor suppressors in breast cancer, but its function in DNA damage response is unknown. Herein, we report that ZNF668 is a regulator of DNA repair. ZNF668 knockdown impairs cell survival after DNA damage without affecting the ATM/ATR DNA-damage signaling cascade. However, recruitment of repair proteins to DNA lesions is decreased. In response to IR, ZNF668 knockdown reduces Tip60-H2AX interaction and impairs IR-induced histone H2AX hyperacetylation, thus impairing chromatin relaxation. Impaired chromatin relaxation causes decreased recruitment of repair proteins to DNA lesions, defective homologous recombination (HR) repair and impaired cell survival after IR. In addition, ZNF668 knockdown decreased RPA phosphorylation and its recruitment to DNA damage foci in response to UV. In both IR and UV damage responses, chromatin relaxation counteracted the impaired loading of repair proteins and DNA repair defects in ZNF668-deficient U2OS cells, indicating that impeded chromatin accessibility at sites of DNA breaks caused the DNA repair defects observed in the absence of ZNF668. Our findings suggest that ZNF668 is a key molecule that links chromatin relaxation with DNA damage response in DNA repair control.


Carcinogenesis | 2013

BRIT1 regulates p53 stability and functions as a tumor suppressor in breast cancer.

Bo Zhang; Edward Wang; Hui Dai; Ruozhen Hu; Yulong Liang; Kaiyi Li; Guobin Wang; Guang Peng; Shiaw Yih Lin

In humans, the gene encoding the BRCA1 C terminus-repeat inhibitor of human telomerase expression 1 (BRIT1) protein is located on chromosome 8p23.1, a region implicated in the development of several malignancies, including breast cancer. Previous studies by our group and others suggested that BRIT1 might function as a novel tumor suppressor. Thus, identifying the molecular mechanisms that underlie BRIT1s tumor suppressive function is important to understand cancer etiology and to identify effective therapeutic strategies for BRIT1-deficient tumors. We thus investigated the role of BRIT1 as a tumor suppressor in breast cancer by using genetic approaches. We discovered that BRIT1 functions as a post-transcriptional regulator of p53 expression. BRIT1 regulates p53 protein stability through blocking murine double minute 2-mediated p53 ubiquitination. To fully demonstrate the role of BRIT1 as a tumor suppressor, we depleted BRIT1 in normal breast epithelial cells. We found that knockdown of BRIT1 caused the oncogenic transformation of normal mammary epithelial cells. Furthermore, ectopic expression of BRIT1 effectively suppressed breast cancer cell proliferation and colony formation in vitro and tumor growth in vivo. Taken together, our study provides new insights into the biological functions of BRIT1 as a tumor suppressor in human breast cancer.


Journal of Biological Chemistry | 2014

Phosphorylation of the BRCA1 C terminus (BRCT) repeat inhibitor of hTERT (BRIT1) protein coordinates TopBP1 protein recruitment and amplifies ataxia telangiectasia-mutated and Rad3-related (ATR) Signaling.

Bo Zhang; Edward Wang; Hui Dai; Jianfeng Shen; Hui Ju Hsieh; Xiongbin Lu; Guang Peng

Background: ATR signaling depends on TopBP1 recruitment to replication stress sites. Results: Interaction between BRIT1 and TopBP1 is required for sustained TopBP1 recruitment to sites of replication stress, which maintains ATR substrate activation. Conclusion: ATR-mediated phosphorylation of BRIT1 and BRIT1 interaction with TopBP1 provide mechanisms of TopBP1 recruitment to promote ATR signaling. Significance: We further the understanding of how ATR signaling is maintained through TopBP1. The ataxia telangiectasia-mutated and Rad3-related (ATR) kinase functions as a central node in the DNA damage response signaling network. The mechanisms by which ATR activity is amplified and/or maintained are not understood. Here we demonstrate that BRIT1/microcephalin (MCPH1), a human disease-related protein, is dispensable for the initiation but essential for the amplification of ATR signaling. BRIT1 interacts with and recruits topoisomerase-binding protein 1 (TopBP1), a key activator of ATR signaling, to the sites of DNA damage. Notably, replication stress-induced ataxia telangiectasia-mutated or ATR-dependent BRIT1 phosphorylation at Ser-322 facilitates efficient TopBP1 recruitment. These results reveal a mechanism that ensures the continuation of ATR-initiated DNA damage signaling. Our study uncovers a previously unknown regulatory axis of ATR signaling in maintaining genomic integrity, which may provide mechanistic insights into the perturbation of ATR signaling in human diseases such as neurodevelopmental defects and cancer.


Archive | 2013

Chromatin Remodeling in DNA Damage Response and Human Aging

Lili Gong; Edward Wang; Shiaw-Yih Lin

Chromatin consists of the DNA and all proteins involved in organizing and regulating DNA structure. The building block of chromatin is the nucleosome, which is composed of 146 base pairs of DNA and a core histone octamer. The core histone oactomer is composed of two heterodimers of histone H2A and histone H2B and a tetramer of histone H3 and histone H4 [1]. The overall chromatin structure is very dynamic in response to diverse biological events. Regulation of chromatin structure is achieved by two major mechanisms. The first is post‐ translational modification (PTM) of histones and other chromatin proteins via phosphoryla‐ tion, methylation, acetylation, ubiquitination and sumoylation [2, 3]. The second is through ATP-dependent nucleosome structure alteration. Cooperation between histone PTMs and chromatin remodelers allows chromatin remodeling to regulate diverse biological events including transcription, chromosome segregation, DNA replication, and DNA repair. In this chapter, we summarize how chromatin structure is regulated during DNA damage response (DDR), focusing particularly on three PTMs: phosphorylation, Poly(ADP-ribosyl)ation (PARylation) and sumoylation. We discuss the DDR in a highly compacted chromatic structure, heterochromatin, as well as the interplay between chromatin remodeling, DNA damage and human aging.


BIO-PROTOCOL | 2016

Ubiquitination Assay for Mammalian Cells

Yang Peng; Edward Wang; Guang Peng; Shiaw-Yih Lin

Collaboration


Dive into the Edward Wang's collaboration.

Top Co-Authors

Avatar

Guang Peng

University of Texas Health Science Center at Houston

View shared research outputs
Top Co-Authors

Avatar

Hui Dai

University of Texas MD Anderson Cancer Center

View shared research outputs
Top Co-Authors

Avatar

Shiaw Yih Lin

University of Texas MD Anderson Cancer Center

View shared research outputs
Top Co-Authors

Avatar

Chun Hui Su

University of Texas MD Anderson Cancer Center

View shared research outputs
Top Co-Authors

Avatar

Enrique Fuentes-Mattei

University of Texas MD Anderson Cancer Center

View shared research outputs
Top Co-Authors

Avatar

Guermarie Velazquez-Torres

University of Texas MD Anderson Cancer Center

View shared research outputs
Top Co-Authors

Avatar

Hyun Ho Choi

University of Texas MD Anderson Cancer Center

View shared research outputs
Top Co-Authors

Avatar

Jian Chen

University of Texas MD Anderson Cancer Center

View shared research outputs
Top Co-Authors

Avatar

Liem Phan

University of Texas MD Anderson Cancer Center

View shared research outputs
Top Co-Authors

Avatar

Ping Chieh Chou

University of Texas MD Anderson Cancer Center

View shared research outputs
Researchain Logo
Decentralizing Knowledge