Network


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

Hotspot


Dive into the research topics where Ruliang Xu is active.

Publication


Featured researches published by Ruliang Xu.


American Journal of Pathology | 2012

Co-Expression of XIAP and Cyclin D1 Complex Correlates with a Poor Prognosis in Patients with Hepatocellular Carcinoma

Yufang Che; Fei Ye; Ruliang Xu; Haitao Qing; Xinying Wang; Fei Yin; Miao Cui; David E. Burstein; Bo Jiang; David Y. Zhang

Hepatocellular carcinoma (HCC) is one of the most common malignant tumors in the world. Despite improved diagnosis and treatment, the prognosis for HCC patients remains poor. The goal of this study was to identify key regulatory proteins and signaling pathways important for cell apoptosis and proliferation as biomarkers for prognostication and targeted therapy. Protein Pathway Array was applied to screen 38 signaling proteins and phosphoproteins in 12 paired HCC tumors and surrounding benign tissues and found that 20 of them, including XIAP, CDK4, CDK6, and Cyclin D1, were overexpressed in HCC tissues. Immunostaining results of XIAP, CDK4, and Cyclin D1 in an additional 59 HCC tissues showed that the expression of XIAP correlated with the expression of CDK4/Cyclin D1, and that the increased expression of these proteins correlated with poor overall survival in these patients. Further studies using the HCC Huh7 cell line transfected with XIAP siRNA or expression vector demonstrated that XIAP regulated the expression of CDK4, CDK6, and Cyclin D1 via NF-êB and PTEN pathways. Finally, inhibition of XIAP using embelin, a XIAP-specific small molecule, leads to an increased apoptosis and decreased cell proliferation via arrest at G1 phase. Taken together, XIAP is a central modulator regulating cell apoptosis and cell cycle progression. Therefore, XIAP together with cell cycle regulatory proteins can be used as prognostic markers and therapeutic targets.


PLOS ONE | 2011

Expression and function of androgen receptor coactivator p44/MEP50/WDR77 in ovarian cancer

Martin Ligr; Ruzeen Rohintan Patwa; Garrett Daniels; Lorraine Pan; Xinyu Wu; Yirong Li; Liantian Tian; Zhenxing Wang; Ruliang Xu; Jingjing Wu; Fan Chen; Jinsong Liu; Jian Jun Wei; Peng Lee

Hormones, including estrogen and progesterone, and their receptors play an important role in the development and progression of ovarian carcinoma. Androgen, its receptor and coactivators have also been implicated in these processes. p44/Mep50/WDR77 was identified as a subunit of the methylosome complex and lately characterized as a steroid receptor coactivator that enhances androgen receptor as well as estrogen receptor-mediated transcriptional activity in a ligand-dependent manner. We previously described distinct expression and function of p44 in prostate, testis, and breast cancers. In this report, we examined the expression and function of p44 in ovarian cancer. In contrast to findings in prostate and testicular cancer and similar to breast cancer, p44 shows strong cytoplasmic localization in morphologically normal ovarian surface and fallopian tube epithelia, while nuclear p44 is observed in invasive ovarian carcinoma. We observed that p44 can serve as a coactivator of both androgen receptor (AR) and estrogen receptor (ER) in ovarian cells. Further, overexpression of nuclear-localized p44 stimulates proliferation and invasion in ovarian cancer cells in the presence of estrogen or androgen. These findings strongly suggest that p44 plays a role in mediating the effects of hormones during ovarian tumorigenesis.


Journal of Cellular and Molecular Medicine | 2010

Androgen receptor coactivator p44/Mep50 in breast cancer growth and invasion

Yi Peng; Yirong Li; Lan Lin Gellert; Xuanyi Zou; Jun Wang; Baljit Singh; Ruliang Xu; Luis Chiriboga; Garrett Daniels; Ruimin Pan; David Y. Zhang; Michael J. Garabedian; Robert J. Schneider; Zhengxin Wang; Peng Lee

Hormones and their receptors play an important role in the development and progression of breast carcinoma. Although the primary focus has been on oestrogen and oestrogen receptor (ER), androgen, androgen receptor (AR) and its coactivator(s) have been implicated in tumorigenesis of breast carcinoma and warrant further investigation. AR coactivator p44/Mep50 is identified as a subunit of methylosome complex and lately characterized as an AR coactivator that enhances AR mediated transcription activity in a ligand dependent manner. In prostate cancer, p44 is expressed in the nucleus of benign epithelia and translocated into the cytoplasm in cancer cells. Furthermore, nuclear expression of p44 inhibits prostate cancer growth. In this report, we examined the expression and function of p44 in breast cancer. In addition to being an AR coactivator, p44 also functions as an ER coactivator. In contrast to findings in prostate cancer, the expression of p44 shows strong cytoplasmic expression in morphologically normal terminal ductal lobular units, while nuclear p44 is observed in both ductal carcinoma in situ and invasive carcinoma. Further, overexpression of nuclear‐localized p44 stimulates proliferation and invasion in MCF7 breast cancer cells in the presence of oestrogen and the process is ERα dependent. These findings strongly suggest that p44 plays a role in mediating the effects of hormones during tumorigenesis in breast.


PLOS ONE | 2013

Functional Domains of Androgen Receptor Coactivator p44/Mep50/WDR77and Its Interaction with Smad1

Yirong Li; Liantian Tian; Martin Ligr; Garrett Daniels; Yi Peng; Xinyu Wu; Mandeep Singh; Jian Jun Wei; Yongzhao Shao; Herbert Lepor; Ruliang Xu; Zhijie Chang; Zhengxin Wang; Peng Lee

p44/MEP50/WDR77 has been identified as a coactivator of androgen receptor (AR), with distinct growth suppression and promotion function in gender specific endocrine organs and their malignancies. We dissected the functional domains of p44 for protein interaction with transcription factors, transcriptional activation, as well as the functional domains in p44 related to its growth inhibition in prostate cancer. Using a yeast two-hybrid screen, we identified a novel transcription complex AR-p44-Smad1, confirmed for physical interaction by co-immunoprecipitaion and functional interaction with luciferase assays in human prostate cancer cells. Yeast two-hybrid assay revealed that the N-terminal region of p44, instead of the traditional WD40 domain at the C-terminus, mediates the interaction among p44, N-terminus of AR and full length Smad1. Although both N and C terminal domains of p44 are necessary for maximum AR transcriptional activation, the N terminal fragment of p44 alone maintains the basic effect on AR transcriptional activation. Cell proliferation assays with N- and C- terminal deletion mutations indicated that the central portion of p44 is required for nuclear p44 mediated prostate cancer growth inhibition.


Breast Cancer Research and Treatment | 2011

Distinct function of androgen receptor coactivator ARA70α and ARA70β in mammary gland development, and in breast cancer

Xinyu Wu; Fei Chen; Aysegul A. Sahin; Constance Albarracin; Zhiheng Pei; Xuanyi Zou; Baljit Singh; Ruliang Xu; Garrett Daniels; Yirong Li; Jian Jun Wei; Marvin Blake; Robert J. Schneider; Pamela Cowin; Peng Lee

Steroid receptor coactivators are important in regulating the function of the receptors in endocrine organ development and in cancers, including breast. Androgen receptor (AR) coactivator ARA70, was first identified as a gene fused to the ret oncogene and later characterized as an AR coactivator. We previously reported that the full length ARA70α functions as a tumor suppressor gene and that ARA70β functions as an oncogene in prostate cancer. Here we show that both ARA70α and ARA70β function as AR and estrogen receptor (ER) coactivators in breast cancer cells. However, ARA70α and ARA70β serve different functions in mammary gland development and breast cancer tumorigenesis. We observed hypoplastic development of mammary glands in MMTV driven ARA70α transgenic mice and overgrowth of mammary glands in ARA70β transgenic mice at virgin and pregnant stages. We determined that ARA70α inhibited cell proliferation, and that ARA70β promotes proliferation in MCF7 breast cancer cells. These effects were observed in hormone-free media, or in media with androgen or estrogen, though to varying degrees. Additionally, we observed that ARA70β strongly enhanced the invasive ability of MCF7 breast cancer cells in in vitro Matrigel assays. Significantly, decreased ARA70α expression is associated with increased tendency of breast cancer metastasis. In summary, ARA70α and ARA70β have distinct effects in mammary gland development and in the progression of breast cancer.


Annals of Surgical Oncology | 2012

Race and the Molecular Origins of Breast Cancer in Chinese Women

Margaret Chen; Ruliang Xu; James W. Turner; Michael Warhol; Phyllis August; Peng Lee

Although there is considerable controversy regarding the role of race in the etiology of human disease, evidence suggests that breast cancers are racially distinct diseases. Clinical features and genetic alterations are different in Chinese women with breast cancer compared with white women. These differences are significant and may influence clinical care. In this review, we summarize the literature addressing genetic heterogeneity in Chinese women with breast cancer. Data support important variations in genes involved in tumorigenic pathways of DNA repair, steroid synthesis and receptor expression, apoptosis, immunity, inflammation, cell cycle control, cancer growth and metastasis, and growth receptor signaling. These genetic differences contribute to our understanding of the molecular origins of breast cancer and may accelerate the development of personalized disease prevention strategies.


Cancer Epidemiology, Biomarkers & Prevention | 2012

Abstract B63: Receptor status in early-onset breast cancer across races.

Mandeep Singh; Yi Ding; Liying Zhang; Susan Lee; Dong Song; Shruti Grover; Dinesh Chandra Doval; Dara S. Ross; Charles Shao; Changcheng Zhu; Ruliang Xu; Zili He; David Zhang; Jinhua Wang; Victor Chang; Margaret Chen; Phyllis August; Peng Lee

Breast cancer is a heterogeneous disease and, racial disparity of breast cancer has been known to influence the age of occurrence, prognosis and response to the treatment. The objective of this study is to analyze the variations in the expression of the receptor proteins- estrogen receptor (ER), progesterone receptor (PR) and HER-2/neu, in early onset breast cancer cross races. Data was collected by reviewing patient charts obtained from the participating institutions and was stratified according to age (Younger group: 40 year old and younger women; older age group: greater than 40 year old women), races (Caucasian, African American, Hispanic, Chinese and Indian), receptor status (ER, PR and HER-2) according to Luminal A, B, HER-2 and Triple Negative Breast Cancer (TNBC) subtypes, grade and stage. Results show that while African American women have higher percentage of TNBC than the Caucasian, there are evident variations in receptor status among other races. In Chinese woman of younger age group, 18.2% presented with TNBC subtype. However, in the same group, the HER-2 overexpression (ER-/PR-, HER-2+), luminal A (ER+/PR+, HER2-) and B (ER+/PR+, HER2+) subtypes were 9.1%, 45.5% and 9.0% respectively. In Indian women, TNBC has been found to be about 1.5 folds higher in younger age group (34 %) than older group (23 %) while HER-2 overexpression and the luminal A and B subgroups were 14.3%, 42.2% and 9.5%, respectively, somewhat similar to those in Chinese women. Our preliminary findings demonstrated that the expression of receptor proteins (ER, PR and Her-2/neu) in early onset breast cancer differs among different ethnic or racial women, suggesting a biological basis of distinct prognosis or therapeutic response related to racial disparities. This study will help to develop optimal personalized management strategies for breast cancer cross the races. Citation Format: Mandeep Singh,, Yi Ding, Liying Zhang, Susan Lee, Dong Song, Shruti Grover, Dinesh Chandra Doval, Dara S. Ross, Charles Shao, Changcheng Zhu, Ruliang Xu, Zili He, David Zhang, Jinhua Wang, Victor Chang, Margaret Chen, Phyllis August, Peng Lee,,. Receptor status in early-onset breast cancer across races. [abstract]. In: Proceedings of the Fifth AACR Conference on the Science of Cancer Health Disparities in Racial/Ethnic Minorities and the Medically Underserved; 2012 Oct 27-30; San Diego, CA. Philadelphia (PA): AACR; Cancer Epidemiol Biomarkers Prev 2012;21(10 Suppl):Abstract nr B63.


American Journal of Cancer Research | 2014

Distinct breast cancer subtypes in women with early-onset disease across races.

Mandeep Singh; Yi Ding; Zhang Ly; Song D; Gong Y; Adams S; Ross Ds; Wang Jh; Grover S; Doval Dc; Shao C; He Zl; Chang; Chin Ww; Fang-Ming Deng; Baljit Singh; Zhang D; Ruliang Xu; Peng Lee


American Journal of Cancer Research | 2014

Imbalanced expression of Tif1γ inhibits pancreatic ductal epithelial cell growth.

Martin Ligr; Xinyu Wu; Garrett Daniels; David Y. Zhang; Huamin Wang; Cristina H. Hajdu; Jinhua Wang; Ruimin Pan; Zhiheng Pei; Lanjing Zhang; Marcovalerio Melis; Matthew R. Pincus; John K. Saunders; Peng Lee; Ruliang Xu


Archive | 2008

Diagnostic methodology and technology

Josephine Wu; Tao Feng; Ruliang Xu; Fei Ye; Bruce Petersen; Liang Cheng; David Y. Zhang

Collaboration


Dive into the Ruliang Xu's collaboration.

Top Co-Authors

Avatar

Peng Lee

University of Texas MD Anderson Cancer Center

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

David Y. Zhang

Icahn School of Medicine at Mount Sinai

View shared research outputs
Top Co-Authors

Avatar

Jian Jun Wei

Northwestern University

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge