Network


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

Hotspot


Dive into the research topics where Xiaobo Zhou is active.

Publication


Featured researches published by Xiaobo Zhou.


Journal of Virology | 2007

Human Papillomavirus Type 16 E7 Oncoprotein Associates with the Cullin 2 Ubiquitin Ligase Complex, Which Contributes to Degradation of the Retinoblastoma Tumor Suppressor

Kyung-Won Huh; Xiaobo Zhou; Hiroyuki Hayakawa; Je Yoel Cho; Towia A. Libermann; Jianping Jin; J. Wade Harper; Karl Münger

ABSTRACT Human papillomavirus type 16 (HPV16) and other high-risk HPVs are etiologically linked to the development of cervical carcinomas and contribute to a number of other tumors of the anogenital tract, as well as oral cancers. The high-risk HPV E6 and E7 oncoproteins are consistently expressed in cervical cancer cells and are necessary for the induction and maintenance of the transformed phenotype. An important aspect of HPV16 E7s oncogenic activities is destabilization of the retinoblastoma tumor suppressor (pRB) through a ubiquitin/proteasome-dependent mechanism, although the exact molecular mechanism is unknown. Here, we report that HPV16 E7 is associated with an enzymatically active cullin 2 ubiquitin ligase complex and that the HPV16 E7/pRB complex contains cullin 2. Depletion of cullin 2 by RNA interference causes increased steady-state levels and stability of pRB in HPV16 E7-expressing cells, and ectopic expression of HPV16 E7 and the cullin 2 complex leads to pRB ubiquitination in vivo. Hence, we propose that the HPV16 E7-associated cullin 2 ubiquitin ligase complex contributes to aberrant degradation of the pRB tumor suppressor in HPV16 E7-expressing cells.


The Lancet Respiratory Medicine | 2014

Risk loci for chronic obstructive pulmonary disease: a genome-wide association study and meta-analysis

Michael H. Cho; Merry-Lynn N. McDonald; Xiaobo Zhou; Manuel Mattheisen; Peter J. Castaldi; Craig P. Hersh; Dawn L. DeMeo; Jody S. Sylvia; John Ziniti; Nan M. Laird; Christoph Lange; Augusto A. Litonjua; David Sparrow; Richard Casaburi; R. Graham Barr; Elizabeth A. Regan; Barry J. Make; John E. Hokanson; Sharon M. Lutz; Tanda Murray Dudenkov; Homayoon Farzadegan; Jacqueline B. Hetmanski; Ruth Tal-Singer; David A. Lomas; Per Bakke; Amund Gulsvik; James D. Crapo; Edwin K. Silverman; Terri H. Beaty

BACKGROUND The genetic risk factors for susceptibility to chronic obstructive pulmonary disease (COPD) are still largely unknown. Additional genetic variants are likely to be identified by genome-wide association studies in larger cohorts or specific subgroups. We sought to identify risk loci for moderate to severe and severe COPD with data from several cohort studies. METHODS We combined genome-wide association analysis data from participants in the COPDGene study (non-Hispanic white and African-American ethnic origin) and the ECLIPSE, NETT/NAS, and Norway GenKOLS studies (self-described white ethnic origin). We did analyses comparing control individuals with individuals with moderate to severe COPD and with a subset of individuals with severe COPD. Single nucleotide polymorphisms yielding a p value of less than 5 × 10(-7) in the meta-analysis at loci not previously described were genotyped in individuals from the family-based ICGN study. We combined results in a joint meta-analysis (threshold for significance p<5 × 10(-8)). FINDINGS Analysis of 6633 individuals with moderate to severe COPD and 5704 control individuals confirmed association at three known loci: CHRNA3 (p=6·38 × 10(-14)), FAM13A (p=1·12 × 10(-14)), and HHIP (p=1·57 × 10(-12)). We also showed significant evidence of association at a novel locus near RIN3 (p=5·25 × 10(-9)). In the overall meta-analysis (ie, including data from 2859 ICGN participants), the association with RIN3 remained significant (p=5·4 × 10(-9)). 3497 individuals were included in our analysis of severe COPD. The effect estimates for the loci near HHIP and CHRNA3 were significantly stronger in severe disease than in moderate to severe disease (p<0·01). We also identified associations at two additional loci: MMP12 (overall joint meta-analysis p=2·6 × 10(-9)) and TGFB2 (overall joint meta-analysis p=8·3 × 10(-9)). INTERPRETATION We have confirmed associations with COPD at three known loci and identified three new genome-wide significant associations. Genetic variants other than in α-1 antitrypsin increase the risk of COPD. FUNDING US National Heart, Lung, and Blood Institute; the Alpha-1 Foundation; the COPD Foundation through contributions from AstraZeneca, Boehringer Ingelheim, Novartis, and Sepracor; GlaxoSmithKline; Centers for Medicare and Medicaid Services; Agency for Healthcare Research and Quality; and US Department of Veterans Affairs.


Breast Cancer Research | 2005

Knockdown of c-Myc expression by RNAi inhibits MCF-7 breast tumor cells growth in vitro and in vivo

Yihua Wang; Shuang Liu; Guo Zhang; Cuiqi Zhou; Hongxia Zhu; Xiaobo Zhou; Lanping Quan; Jinfeng Bai; Ningzhi Xu

IntroductionBreast cancer is the leading cause of cancer death in women worldwide. Elevated expression of c-Myc is a frequent genetic abnormality seen in this malignancy. For a better understanding of its role in maintaining the malignant phenotype, we used RNA interference (RNAi) directed against c-Myc in our study. RNAi provides a new, reliable method to investigate gene function and has the potential for gene therapy. The aim of the study was to examine the anti-tumor effects elicited by a decrease in the protein level of c-Myc by RNAi and its possible mechanism of effects in MCF-7 cells.MethodA plasmid-based polymerase III promoter system was used to deliver and express short interfering RNA (siRNA) targeting c-myc to reduce its expression in MCF-7 cells. Western blot analysis was used to measure the protein level of c-Myc. We assessed the effects of c-Myc silencing on tumor growth by a growth curve, by soft agar assay and by nude mice experiments in vivo. Standard fluorescence-activated cell sorter analysis and TdT-mediated dUTP nick end labelling assay were used to determine apoptosis of the cells.ResultsOur data showed that plasmids expressing siRNA against c-myc markedly and durably reduced its expression in MCF-7 cells by up to 80%, decreased the growth rate of MCF-7 cells, inhibited colony formation in soft agar and significantly reduced tumor growth in nude mice. We also found that depletion of c-Myc in this manner promoted apoptosis of MCF-7 cells upon serum withdrawal.Conclusionc-Myc has a pivotal function in the development of breast cancer. Our data show that decreasing the c-Myc protein level in MCF-7 cells by RNAi could significantly inhibit tumor growth both in vitro and in vivo, and imply the therapeutic potential of RNAi on the treatment of breast cancer by targeting overexpression oncogenes such as c-myc, and c-myc might be a potential therapeutic target for human breast cancer.


The New England Journal of Medicine | 2016

Patterns of Growth and Decline in Lung Function in Persistent Childhood Asthma.

Michael J. McGeachie; Katherine P. Yates; Xiaobo Zhou; Feng Guo; Alice L. Sternberg; Mark L. Van Natta; Robert A. Wise; Stanley J. Szefler; Sunita Sharma; Alvin T. Kho; Michael H. Cho; Damien C. Croteau-Chonka; Peter J. Castaldi; Gaurav Jain; Amartya Sanyal; Ye Zhan; Bryan R. Lajoie; Job Dekker; John A. Stamatoyannopoulos; Ronina A. Covar; Robert S. Zeiger; N. Franklin Adkinson; Paul T. Williams; H. William Kelly; Hartmut Grasemann; Judith M. Vonk; Gerard H. Koppelman; Dirkje S. Postma; Benjamin A. Raby; Isaac Houston

BACKGROUND Tracking longitudinal measurements of growth and decline in lung function in patients with persistent childhood asthma may reveal links between asthma and subsequent chronic airflow obstruction. METHODS We classified children with asthma according to four characteristic patterns of lung-function growth and decline on the basis of graphs showing forced expiratory volume in 1 second (FEV1), representing spirometric measurements performed from childhood into adulthood. Risk factors associated with abnormal patterns were also examined. To define normal values, we used FEV1 values from participants in the National Health and Nutrition Examination Survey who did not have asthma. RESULTS Of the 684 study participants, 170 (25%) had a normal pattern of lung-function growth without early decline, and 514 (75%) had abnormal patterns: 176 (26%) had reduced growth and an early decline, 160 (23%) had reduced growth only, and 178 (26%) had normal growth and an early decline. Lower baseline values for FEV1, smaller bronchodilator response, airway hyperresponsiveness at baseline, and male sex were associated with reduced growth (P<0.001 for all comparisons). At the last spirometric measurement (mean [±SD] age, 26.0±1.8 years), 73 participants (11%) met Global Initiative for Chronic Obstructive Lung Disease spirometric criteria for lung-function impairment that was consistent with chronic obstructive pulmonary disease (COPD); these participants were more likely to have a reduced pattern of growth than a normal pattern (18% vs. 3%, P<0.001). CONCLUSIONS Childhood impairment of lung function and male sex were the most significant predictors of abnormal longitudinal patterns of lung-function growth and decline. Children with persistent asthma and reduced growth of lung function are at increased risk for fixed airflow obstruction and possibly COPD in early adulthood. (Funded by the Parker B. Francis Foundation and others; ClinicalTrials.gov number, NCT00000575.).


Human Molecular Genetics | 2012

Identification of a chronic obstructive pulmonary disease genetic determinant that regulates HHIP

Xiaobo Zhou; Rebecca M. Baron; Megan Hardin; Michael H. Cho; Jan Zieliński; Iwona Hawryłkiewicz; Pawel Sliwinski; Craig P. Hersh; John D. Mancini; Ke Lu; Derek Thibault; Amy L. Donahue; Barbara J. Klanderman; Bernard Rosner; Benjamin A. Raby; Quan Lu; Adriana Miele Geldart; Matthew D. Layne; Mark A. Perrella; Scott T. Weiss; Augustine M. K. Choi; Edwin K. Silverman

Multiple intergenic single-nucleotide polymorphisms (SNPs) near hedgehog interacting protein (HHIP) on chromosome 4q31 have been strongly associated with pulmonary function levels and moderate-to-severe chronic obstructive pulmonary disease (COPD). However, whether the effects of variants in this region are related to HHIP or another gene has not been proven. We confirmed genetic association of SNPs in the 4q31 COPD genome-wide association study (GWAS) region in a Polish cohort containing severe COPD cases and healthy smoking controls (P = 0.001 to 0.002). We found that HHIP expression at both mRNA and protein levels is reduced in COPD lung tissues. We identified a genomic region located ∼85 kb upstream of HHIP which contains a subset of associated SNPs, interacts with the HHIP promoter through a chromatin loop and functions as an HHIP enhancer. The COPD risk haplotype of two SNPs within this enhancer region (rs6537296A and rs1542725C) was associated with statistically significant reductions in HHIP promoter activity. Moreover, rs1542725 demonstrates differential binding to the transcription factor Sp3; the COPD-associated allele exhibits increased Sp3 binding, which is consistent with Sp3s usual function as a transcriptional repressor. Thus, increased Sp3 binding at a functional SNP within the chromosome 4q31 COPD GWAS locus leads to reduced HHIP expression and increased susceptibility to COPD through distal transcriptional regulation. Together, our findings reveal one mechanism through which SNPs upstream of the HHIP gene modulate the expression of HHIP and functionally implicate reduced HHIP gene expression in the pathogenesis of COPD.


International Journal of Cancer | 2005

Overexpression of human pituitary tumor transforming gene (hPTTG), is regulated by β‐catenin /TCF pathway in human esophageal squamous cell carcinoma

Cuiqi Zhou; Shuang Liu; Xiaobo Zhou; Liyan Xue; Lanping Quan; Ning Lu; Guo Zhang; Jinfeng Bai; Yihua Wang; Zhihua Liu; Qimin Zhan; Hongxia Zhu; Ningzhi Xu

Overexpression of human pituitary tumor transforming gene (PTTG) is wildly detected in many tumors, including esophageal cancer. Besides overexpression of PTTG in esophageal squamous cell carcinoma (ESCC) tissues and cells, we detected accumulation of cytoplasmic β‐catenin in ESCC. In our study, a putative TCF4‐binding element (TBE) was identified in PTTG promoter region. The activity of PTTG promoter containing the TBE was activated by S37Aβ‐catenin and inhibited by dominant‐negative TCF. Furthermore, the activation by S37Aβ‐catenin was mostly abrogated among PTTG promoter region without the TBE or with a mutant one. By using biotin‐streptavidin pull‐down assay, we also found that the TBE among PTTG promoter bound to TCF‐4 protein. Moreover, levels of PTTG mRNA and protein were increased by S37Aβ‐catenin. Finally, it is noticeable that we detected a correlation between β‐catenin localization and PTTG expression in 69 primary ESCC (p<0.01). In brief, our study shows that overexpression of PTTG in ESCC is likely due to the activation of β‐catenin/WNT signaling. As a target gene of β‐catenin/TCF pathway, PTTG may play an important role in tumorigenesis of human ESCC.


Oncogene | 2005

Overexpression of EB1 in human esophageal squamous cell carcinoma (ESCC) may promote cellular growth by activating β -catenin/TCF pathway

Yihua Wang; Xiaobo Zhou; Hongxia Zhu; Shuang Liu; Cuiqi Zhou; Guo Zhang; Liyan Xue; Ning Lu; Lanping Quan; Jinfeng Bai; Qimin Zhan; Ningzhi Xu

Esophageal squamous cell carcinoma (ESCC) has a multifactorial etiology involving environmental and/or genetic factors. End-binding protein 1 (EB1), which was cloned as an interacting partner of the adenomatous polyposis coli (APC) tumor suppressor protein, was previously found overexpressed in ESCC. However, the precise role of EB1 in the development of this malignancy has not yet been elucidated. In this study, we analysed freshly resected ESCC specimens and demonstrated that EB1 was overexpressed in approximately 63% of tumor samples compared to matched normal tissue. We report that overexpression of EB1 in the ESCC line EC9706 significantly promotes cell growth, whereas suppression of EB1 protein level by RNA interference significantly inhibited growth of esophageal tumor cells. In addition, EB1 overexpression induced nuclear accumulation of β-catenin and promoted the transcriptional activity of β-catenin/T-cell factor (TCF). These effects were partially or completely abolished by coexpression of APC or ΔN TCF4, respectively. Also, we found that EB1 affected the interaction between β-catenin and APC. Furthermore, EB1 overexpression was correlated with cytoplasmic/nuclear accumulation of β-catenin in primary human ESCC. Taken together, these results support the novel hypothesis that EB1 overexpression may play a role in the development of ESCC by affecting APC function and activating the β-catenin/TCF pathway.


Human Molecular Genetics | 2015

A disease module in the interactome explains disease heterogeneity, drug response and captures novel pathways and genes in asthma

Amitabh Sharma; Jörg Menche; C. Chris Huang; Tatiana Ort; Xiaobo Zhou; Maksim Kitsak; Nidhi Sahni; Derek Thibault; Linh Voung; Feng Guo; Susan Dina Ghiassian; Natali Gulbahce; Frédéric Baribaud; Joel Tocker; Radu Dobrin; Elliot S. Barnathan; Hao Liu; Reynold A. Panettieri; Kelan G. Tantisira; Weiliang Qiu; Benjamin A. Raby; Edwin K. Silverman; Marc Vidal; Scott T. Weiss; Albert-László Barabási

Recent advances in genetics have spurred rapid progress towards the systematic identification of genes involved in complex diseases. Still, the detailed understanding of the molecular and physiological mechanisms through which these genes affect disease phenotypes remains a major challenge. Here, we identify the asthma disease module, i.e. the local neighborhood of the interactome whose perturbation is associated with asthma, and validate it for functional and pathophysiological relevance, using both computational and experimental approaches. We find that the asthma disease module is enriched with modest GWAS P-values against the background of random variation, and with differentially expressed genes from normal and asthmatic fibroblast cells treated with an asthma-specific drug. The asthma module also contains immune response mechanisms that are shared with other immune-related disease modules. Further, using diverse omics (genomics, gene-expression, drug response) data, we identify the GAB1 signaling pathway as an important novel modulator in asthma. The wiring diagram of the uncovered asthma module suggests a relatively close link between GAB1 and glucocorticoids (GCs), which we experimentally validate, observing an increase in the level of GAB1 after GC treatment in BEAS-2B bronchial epithelial cells. The siRNA knockdown of GAB1 in the BEAS-2B cell line resulted in a decrease in the NFkB level, suggesting a novel regulatory path of the pro-inflammatory factor NFkB by GAB1 in asthma.


Journal of Biological Chemistry | 2012

Caveolin-1 Inhibits Expression of Antioxidant Enzymes through Direct Interaction with Nuclear Erythroid 2 p45-related Factor-2 (Nrf2)

Wen Li; Hui Liu; Jie-Sen Zhou; Jiao-Fei Cao; Xiaobo Zhou; Augustine M. K. Choi; Zhihua Chen; Huahao Shen

Background: Caveolin-1 regulates cellular antioxidant capacity, but mechanisms remain unknown. Results: Caveolin-1 interacts with Nrf2 and suppresses its transcriptional activity and down-regulates cellular antioxidant enzymes. Conclusion: Caveolin-1 regulates cellular antioxidant capacity through interaction with Nrf2. Significance: Clarifying how caveolin-1 regulates cellular antioxidant capacity and identifying a novel target to establish the contribution of oxidative stress to human pathologies. The Nrf2 (nuclear erythroid 2 p45-related factor-2) signaling pathway is known to play a pivotal role in a variety of oxidative stress-related human disorders. It has been reported recently that the plasma membrane resident protein caveolin-1 (Cav-1) can regulate expression of certain antioxidant enzymes and involves in the pathogenesis of oxidative lung injury, but the detailed molecular mechanisms remain incompletely understood. Here, we demonstrated that Cav-1 inhibited the expression of antioxidant enzymes through direct interaction with Nrf2 and subsequent suppression of its transcriptional activity in lung epithelial Beas-2B cells. Cav-1 deficiency cells exhibited higher levels of antioxidant enzymes and were more resistant to oxidative stress induced cytotoxicity, whereas overexpression of Cav-1 suppressed the induction of these enzymes and further augmented the oxidative cell death. Cav-1 constitutively interacted with Nrf2 in both cytosol and nucleus. Stimulation of 4-hydroxynonenol increased the Cav-1-Nrf2 interaction in cytosol but disrupted their association in the nucleus. Knockdown of Cav-1 also disassociated the interaction between Nrf2 and its cytoplasmic inhibitor Keap1 (Kelch-like ECH-associated protein 1) and increased the Nrf2 transcription activity. Mutation of the resembling Cav-1 binding motif on Nrf2 effectively attenuated their interaction, which exhibited higher transcription activity and induced higher levels of antioxidant enzymes relative to the wild-type control. Altogether, these studies clearly demonstrate that Cav-1 inhibits cellular antioxidant capacity through direct interaction with Nrf2 and subsequent suppression of its activity, thereby implicating in certain oxidative stress-related human pathologies.


Human Molecular Genetics | 2015

Genetic control of gene expression at novel and established chronic obstructive pulmonary disease loci

Peter J. Castaldi; Michael H. Cho; Xiaobo Zhou; Weiliang Qiu; Michael J. McGeachie; Bartolome R. Celli; Per Bakke; Amund Gulsvik; David A. Lomas; James D. Crapo; Terri H. Beaty; Stephen I. Rennard; Benjamin J. Harshfield; Christoph Lange; Dave Singh; Ruth Tal-Singer; John H. Riley; John Quackenbush; Benjamin A. Raby; Vincent J. Carey; Edwin K. Silverman; Craig P. Hersh

Genetic risk loci have been identified for a wide range of diseases through genome-wide association studies (GWAS), but the relevant functional mechanisms have been identified for only a small proportion of these GWAS-identified loci. By integrating results from the largest current GWAS of chronic obstructive disease (COPD) with expression quantitative trait locus (eQTL) analysis in whole blood and sputum from 121 subjects with COPD from the ECLIPSE Study, this analysis identifies loci that are simultaneously associated with COPD and the expression of nearby genes (COPD eQTLs). After integrative analysis, 19 COPD eQTLs were identified, including all four previously identified genome-wide significant loci near HHIP, FAM13A, and the 15q25 and 19q13 loci. For each COPD eQTL, fine mapping and colocalization analysis to identify causal shared eQTL and GWAS variants identified a subset of sites with moderate-to-strong evidence of harboring at least one shared variant responsible for both the eQTL and GWAS signals. Transcription factor binding site (TFBS) analysis confirms that multiple COPD eQTL lead SNPs disrupt TFBS, and enhancer enrichment analysis for loci with the strongest colocalization signals showed enrichment for blood-related cell types (CD3 and CD4+ T cells, lymphoblastoid cell lines). In summary, integrative eQTL and GWAS analysis confirms that genetic control of gene expression plays a key role in the genetic architecture of COPD and identifies specific blood-related cell types as likely participants in the functional pathway from GWAS-associated variant to disease phenotype.

Collaboration


Dive into the Xiaobo Zhou's collaboration.

Top Co-Authors

Avatar

Edwin K. Silverman

Brigham and Women's Hospital

View shared research outputs
Top Co-Authors

Avatar

Weiliang Qiu

Brigham and Women's Hospital

View shared research outputs
Top Co-Authors

Avatar

Lanping Quan

Peking Union Medical College

View shared research outputs
Top Co-Authors

Avatar

Ningzhi Xu

Peking Union Medical College

View shared research outputs
Top Co-Authors

Avatar

Craig P. Hersh

Brigham and Women's Hospital

View shared research outputs
Top Co-Authors

Avatar

Michael H. Cho

Brigham and Women's Hospital

View shared research outputs
Top Co-Authors

Avatar

Benjamin A. Raby

Brigham and Women's Hospital

View shared research outputs
Top Co-Authors

Avatar

Taotao Lao

Brigham and Women's Hospital

View shared research outputs
Top Co-Authors

Avatar

Hongxia Zhu

Peking Union Medical College

View shared research outputs
Top Co-Authors

Avatar

John D. Mancini

Brigham and Women's Hospital

View shared research outputs
Researchain Logo
Decentralizing Knowledge