Network


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

Hotspot


Dive into the research topics where Wennuan Liu is active.

Publication


Featured researches published by Wennuan Liu.


The New England Journal of Medicine | 2008

Cumulative Association of Five Genetic Variants with Prostate Cancer

S. Lilly Zheng; Jielin Sun; Fredrik Wiklund; Shelly Smith; Pär Stattin; Ge Li; Hans-Olov Adami; Fang-Chi Hsu; Yi Zhu; Katarina Bälter; A. Karim Kader; Aubrey R. Turner; Wennuan Liu; Eugene R. Bleecker; Deborah A. Meyers; David Duggan; John D. Carpten; Bao Li Chang; William B. Isaacs; Jianfeng Xu; Henrik Grönberg

BACKGROUND Single-nucleotide polymorphisms (SNPs) in five chromosomal regions--three at 8q24 and one each at 17q12 and 17q24.3--have been associated with prostate cancer. Each SNP has only a moderate association, but when SNPs are combined, the association may be stronger. METHODS We evaluated 16 SNPs from five chromosomal regions in a Swedish population (2893 subjects with prostate cancer and 1781 control subjects) and assessed the individual and combined association of the SNPs with prostate cancer. RESULTS Multiple SNPs in each of the five regions were associated with prostate cancer in single SNP analysis. When the most significant SNP from each of the five regions was selected and included in a multivariate analysis, each SNP remained significant after adjustment for other SNPs and family history. Together, the five SNPs and family history were estimated to account for 46% of the cases of prostate cancer in the Swedish men we studied. The five SNPs plus family history had a cumulative association with prostate cancer (P for trend, 3.93x10(-28)). In men who had any five or more of these factors associated with prostate cancer, the odds ratio for prostate cancer was 9.46 (P=1.29x10(-8)), as compared with men without any of the factors. The cumulative effect of these variants and family history was independent of serum levels of prostate-specific antigen at diagnosis. CONCLUSIONS SNPs in five chromosomal regions plus a family history of prostate cancer have a cumulative and significant association with prostate cancer.


Nature Medicine | 2009

Copy Number Analysis Indicates Monoclonal Origin of Lethal Metastatic Prostate Cancer

Wennuan Liu; Sari Laitinen; Sofia Khan; Mauno Vihinen; Jeanne Kowalski; Guoqiang Yu; Li Chen; Charles M. Ewing; Mario A. Eisenberger; Michael A. Carducci; William G. Nelson; Srinivasan Yegnasubramanian; Jun Luo; Yue Wang; Jianfeng Xu; William B. Isaacs; Tapio Visakorpi; G. Steven Bova

Many studies have shown that primary prostate cancers are multifocal and are composed of multiple genetically distinct cancer cell clones. Whether or not multiclonal primary prostate cancers typically give rise to multiclonal or monoclonal prostate cancer metastases is largely unknown, although studies at single chromosomal loci are consistent with the latter case. Here we show through a high-resolution genome-wide single nucleotide polymorphism and copy number survey that most, if not all, metastatic prostate cancers have monoclonal origins and maintain a unique signature copy number pattern of the parent cancer cell while also accumulating a variable number of separate subclonally sustained changes. We find no relationship between anatomic site of metastasis and genomic copy number change pattern. Taken together with past animal and cytogenetic studies of metastasis and recent single-locus genetic data in prostate and other metastatic cancers, these data indicate that despite common genomic heterogeneity in primary cancers, most metastatic cancers arise from a single precursor cancer cell. This study establishes that genomic archeology of multiple anatomically separate metastatic cancers in individuals can be used to define the salient genomic features of a parent cancer clone of proven lethal metastatic phenotype.


Nature Genetics | 2010

Androgen-induced TOP2B-mediated double-strand breaks and prostate cancer gene rearrangements

Michael C. Haffner; Martin J. Aryee; Antoun Toubaji; David Esopi; Roula Albadine; Bora Gurel; William B. Isaacs; G. Steven Bova; Wennuan Liu; Jianfeng Xu; Alan K. Meeker; George J. Netto; Angelo M. De Marzo; William G. Nelson; Srinivasan Yegnasubramanian

DNA double-strand breaks (DSBs) can lead to the development of genomic rearrangements, which are hallmarks of cancer. Fusions between TMPRSS2, encoding the transmembrane serine protease isoform 2, and ERG, encoding the v-ets erythroblastosis virus E26 oncogene homolog, are among the most common oncogenic rearrangements observed in human cancer. We show that androgen signaling promotes co-recruitment of androgen receptor and topoisomerase II beta (TOP2B) to sites of TMPRSS2-ERG genomic breakpoints, triggering recombinogenic TOP2B-mediated DSBs. Furthermore, androgen stimulation resulted in de novo production of TMPRSS2-ERG fusion transcripts in a process that required TOP2B and components of the DSB repair machinery. Finally, unlike normal prostate epithelium, prostatic intraepithelial neoplasia cells showed strong coexpression of androgen receptor and TOP2B. These findings implicate androgen-induced TOP2B-mediated DSBs in generating TMPRSS2-ERG rearrangements.


Clinical Cancer Research | 2011

PTEN Protein Loss by Immunostaining: Analytic Validation and Prognostic Indicator for a High Risk Surgical Cohort of Prostate Cancer Patients

Tamara L. Lotan; Bora Gurel; Siobhan Sutcliffe; David Esopi; Wennuan Liu; Jianfeng Xu; Jessica Hicks; Ben Ho Park; Elizabeth B. Humphreys; Alan W. Partin; Misop Han; George J. Netto; William B. Isaacs; Angelo M. De Marzo

Purpose: Analytically validated assays to interrogate biomarker status in clinical samples are crucial for personalized medicine. PTEN is a tumor suppressor commonly inactivated in prostate cancer that has been mechanistically linked to disease aggressiveness. Though deletion of PTEN, as detected by cumbersome FISH spot counting assays, is associated with poor prognosis, few studies have validated immunohistochemistry (IHC) assays to determine whether loss of PTEN protein is associated with unfavorable disease. Experimental Design: PTEN IHC was validated by employing formalin fixed and paraffin-embedded isogenic human cell lines containing or lacking intact PTEN alleles. PTEN IHC was 100% sensitive and 97.8% specific for detecting genomic alterations in 58 additional cell lines. PTEN protein loss was then assessed on 376 prostate tumor samples, and PTEN FISH or high resolution single nucleotide polymorphism microarray analysis was done on a subset of these cases. Results: PTEN protein loss, as assessed as a dichotomous IHC variable, was highly reproducible, correlated strongly with adverse pathologic features (e.g., Gleason score and pathologic stage), detected between 75% and 86% of cases with PTEN genomic loss, and was found at times in the absence of apparent genomic loss. In a cohort of 217 high risk surgically treated patients, PTEN protein loss was associated with decreased time to metastasis. Conclusion: These studies validate a simple method to interrogate PTEN status in clinical specimens and support the utility of this test in future multicenter studies, clinical trials, and ultimately perhaps for routine clinical care. Clin Cancer Res; 17(20); 6563–73. ©2011 AACR.


Nature Genetics | 2008

Evidence for two independent prostate cancer risk-associated loci in the HNF1B gene at 17q12.

Jielin Sun; Siqun Lilly Zheng; Fredrik Wiklund; Sarah D. Isaacs; Lina Purcell; Zhengrong Gao; Fang-Chi Hsu; Seong Tae Kim; Wennuan Liu; Yi Zhu; Pär Stattin; Hans-Olov Adami; Kathleen E. Wiley; Latchezar Dimitrov; Jishan Sun; Tao Li; Aubrey R. Turner; Tamara S. Adams; Jan Adolfsson; Jan-Erik Johansson; James Lowey; Bruce J. Trock; Alan W. Partin; Patrick C. Walsh; Jeffrey M. Trent; David Duggan; John D. Carpten; Bao Li Chang; Henrik Grönberg; William B. Isaacs

We carried out a fine-mapping study in the HNF1B gene at 17q12 in two study populations and identified a second locus associated with prostate cancer risk, ∼26 kb centromeric to the first known locus (rs4430796); these loci are separated by a recombination hot spot. We confirmed the association with a SNP in the second locus (rs11649743) in five additional populations, with P = 1.7 × 10−9 for an allelic test of the seven studies combined. The association at each SNP remained significant after adjustment for the other SNP.


Science Translational Medicine | 2013

DNA Methylation Alterations Exhibit Intraindividual Stability and Interindividual Heterogeneity in Prostate Cancer Metastases

Martin J. Aryee; Wennuan Liu; Julia C. Engelmann; Philipp Nuhn; Meltem Gürel; Michael C. Haffner; David Esopi; Rafael A. Irizarry; Robert H. Getzenberg; William G. Nelson; Jun Luo; Jianfeng Xu; William B. Isaacs; G. Steven Bova; Srinivasan Yegnasubramanian

Prostate tumors develop a unique epigenetic DNA methylation signature that is clonally maintained in disseminated metastases. Surveying the DNA Methylation “Cityscape” of Prostate Cancer Alterations in DNA methylation are a hallmark of human cancers, including prostate cancer. Understanding which of these alterations “drive” cancer initiation, progression, and metastasis, and which of these are merely “passengers” not involved in the chain of causation, is a major translational challenge. To tackle this challenge in the context of metastatic prostate cancer, Aryee et al. carried out genome-scale analyses of DNA methylation alterations in multiple metastases from each of 13 men who had died of metastatic prostate cancer. To visualize both the frequency of each methylation alteration in the metastases and the consistency with which each alteration was maintained across all metastases from an individual, the authors created DNA methylation “cityscape” plots. These analyses revealed that each individual developed a unique DNA methylation signature that was largely maintained across all metastases within that individual. Additionally, a set of DNA “hypermethylation” alterations, defined as regions that were normally unmethylated but acquired cancer-specific DNA methylation, were enriched for prostate cancer “drivers.” Such DNA hypermethylation alterations are attractive potential targets for development of longitudinal markers and therapeutic strategies for prostate cancer management. Human cancers almost ubiquitously harbor epigenetic alterations. Although such alterations in epigenetic marks, including DNA methylation, are potentially heritable, they can also be dynamically altered. Given this potential for plasticity, the degree to which epigenetic changes can be subject to selection and act as drivers of neoplasia has been questioned. We carried out genome-scale analyses of DNA methylation alterations in lethal metastatic prostate cancer and created DNA methylation “cityscape” plots to visualize these complex data. We show that somatic DNA methylation alterations, despite showing marked interindividual heterogeneity among men with lethal metastatic prostate cancer, were maintained across all metastases within the same individual. The overall extent of maintenance in DNA methylation changes was comparable to that of genetic copy number alterations. Regions that were frequently hypermethylated across individuals were markedly enriched for cancer- and development/differentiation-related genes. Additionally, regions exhibiting high consistency of hypermethylation across metastases within individuals, even if variably hypermethylated across individuals, showed enrichment for cancer-related genes. Whereas some regions showed intraindividual metastatic tumor heterogeneity in promoter methylation, such methylation alterations were generally not correlated with gene expression. This was despite a general tendency for promoter methylation patterns to be strongly correlated with gene expression, particularly at regions that were variably methylated across individuals. These findings suggest that DNA methylation alterations have the potential for producing selectable driver events in carcinogenesis and disease progression and highlight the possibility of targeting such epigenome alterations for development of longitudinal markers and therapeutic strategies.


Nature Genetics | 2012

Genome-wide association study in Chinese men identifies two new prostate cancer risk loci at 9q31.2 and 19q13.4

Jianfeng Xu; Zengnan Mo; Dingwei Ye; Meilin Wang; Fang Liu; Guangfu Jin; Chuanliang Xu; Xiang Wang; Qiang Shao; Zhiwen Chen; Zhihua Tao; Jun Qi; Fangjian Zhou; Zhong Wang; Yaowen Fu; Dalin He; Qiang Wei; Jianming Guo; Denglong Wu; Xin Gao; Jianlin Yuan; Gongxian Wang; Yong Xu; Guozeng Wang; Haijun Yao; Pei Dong; Yang Jiao; Mo Shen; Jin Yang; Jun OuYang

Prostate cancer risk–associated variants have been reported in populations of European descent, African-Americans and Japanese using genome-wide association studies (GWAS). To systematically investigate prostate cancer risk–associated variants in Chinese men, we performed the first GWAS in Han Chinese. In addition to confirming several associations reported in other ancestry groups, this study identified two new risk-associated loci for prostate cancer on chromosomes 9q31.2 (rs817826, P = 5.45 × 10−14) and 19q13.4 (rs103294, P = 5.34 × 10−16) in 4,484 prostate cancer cases and 8,934 controls. The rs103294 marker at 19q13.4 is in strong linkage equilibrium with a 6.7-kb germline deletion that removes the first six of seven exons in LILRA3, a gene regulating inflammatory response, and was significantly associated with the mRNA expression of LILRA3 in T cells (P < 1 × 10−4). These findings may advance the understanding of genetic susceptibility to prostate cancer.


European Urology | 2013

Exome sequencing of prostate cancer supports the hypothesis of independent tumour origins.

Johan Lindberg; Daniel Klevebring; Wennuan Liu; Mårten Neiman; Jianfeng Xu; Peter Wiklund; Fredrik Wiklund; Ian G. Mills; Lars Egevad; Henrik Grönberg

BACKGROUND Prostate cancer (PCa) is a clinically and pathologically heterogeneous disease. The rapid development of sequencing technology has the potential to deliver new biomarkers with emphasis on aggressive disease and to revolutionise personalised cancer treatment. However, a prostate harbouring cancer commonly contains multiple separate tumour foci, with the potential to aggravate tumour sampling. The level of intraprostatic tumour heterogeneity remains to be determined. OBJECTIVE To determine the level of intraprostatic tumour heterogeneity through genome-wide, high-resolution profiling of multiple tumour samples from the same individual. DESIGN, SETTINGS, AND PARTICIPANTS Multiple tumour samples were obtained from four individuals following radical prostatectomy. One individual (SWE-1) contained >70% cancer cells in all tumour samples, whereas the other three (SWE-2 to SWE-4) required the use of laser capture microdissection for tumour cell enrichment. Subsequently, DNA was extracted from all tissue samples, and exome sequencing was performed. All tumour foci of SWE-1 were also profiled using a high-resolution array for the identification of copy number alterations (CNA). OUTCOME MEASUREMENTS AND STATISTICAL ANALYSIS Shared somatic high-frequency single nucleotide variants (SNV) and CNAs were used to infer the level of intraprostatic tumour heterogeneity. RESULTS AND LIMITATIONS No high-frequency mutations, common for the three tumour samples of SWE-1, were identified. Ten randomly chosen positions were validated with Sanger sequencing in all foci, which verified the exome data. The high level of intraprostatic heterogeneity was consistent in all individuals. In total, three out of four individuals harboured tumours without an apparent common somatic denominator. Although we cannot exclude the presence of common structural rearrangements, a high-density array was used for the detection of deletions and amplifications in SWE-1, which agreed with the exome data. CONCLUSIONS We present evidence for the presence of somatically independent tumours within the same prostate. This finding will have implications for personalised cancer treatment and biomarker discovery.


International Journal of Cancer | 2006

Genetic variation in the COX-2 gene and the association with prostate cancer risk.

K. Shahedi; Sara Lindström; Siqun Zheng; Fredrik Wiklund; Jan Adolfsson; Jishan Sun; Katarina Augustsson-Bälter; Bao-Li Chang; Hans-Olov Adami; Wennuan Liu; Henrik Grönberg; Jianfeng Xu

COX‐2 is a key enzyme in the conversion of arachidonic acid to prostaglandins. The prostaglandins produced by COX‐2 are involved in inflammation and pain response in different tissues in the body. Accumulating evidence from epidemiologic studies, chemical carcinogen‐induced rodent models and clinical trials indicate that COX‐2 plays a role in human carcinogenesis and is overexpressed in prostate cancer tissue. We examined whether sequence variants in the COX‐2 gene are associated with prostate cancer risk. We analyzed a large population‐based case–control study, cancer prostate in Sweden (CAPS) consisting of 1,378 cases and 782 controls. We evaluated 16 single nucleotide polymorphisms (SNPs) spanning the entire COX‐2 gene in 94 subjects of the control group. Five SNPs had a minor allele frequency of more than 5% in our study population and these were genotyped in all case patients and control subjects and gene‐specific haplotypes were constructed. A statistically significant difference in allele frequency between cases and controls was observed for 2 of the SNPs (+3100 T/G and +8365 C/T), with an odds ratio of 0.78 (95% CI = 0.64–0.96) and 0.65 (95% CI = 0.45–0.94) respectively. In the haplotype analysis, 1 haplotype carrying the variant allele from both +3100 T/G and +8365 C/T, with a population frequency of 3%, was also significantly associated with decreased risk of prostate cancer (p = 0.036, global simulated p‐value = 0.046). This study supports the hypothesis that inflammation is involved in prostate carcinogenesis and that sequence variation within the COX‐2 gene influence the risk of prostate cancer.


Genes, Chromosomes and Cancer | 2006

Comprehensive assessment of DNA copy number alterations in human prostate cancers using Affymetrix 100K SNP mapping array

Wennuan Liu; Bao-Li Chang; Jurga Sauvageot; Latchezar Dimitrov; Marta Gielzak; Tao Li; Guifang Yan; Jishan Sun; Jielin Sun; Tamara S. Adams; Aubrey R. Turner; Jin Woo Kim; Deborah A. Meyers; Siqun Lilly Zheng; William B. Isaacs; Jianfeng Xu

Although multiple recurrent chromosomal alterations have been identified in prostate cancer cells, the specific genes driving the apparent selection of these changes remain largely unknown. In part, this uncertainty is due to the limited resolution of the techniques used to detect these alterations. In this study, we applied a high‐resolution genome‐wide method, Affymetrix 100K SNP mapping array, to screen for somatic DNA copy number (CN) alterations among 22 pairs of samples from primary prostate cancers and matched nonmalignant tissues. We detected 355 recurrent deletions and 223 recurrent gains, many of which were novel. As expected, the sizes of novel alterations tend to be smaller. Importantly, among tumors with increasing grade, Gleason sum 6, 7, and 8, we found a significant trend of larger number of alterations in the tumors with higher grade. Overall, gains are significantly more likely to occur within genes (74%) than are deletions (49%). However, when we looked at the most frequent CN alterations, defined as those in ≥4 subjects, we observed that both gains (85%) and deletions (57%) occur preferentially within genes. An example of a novel, recurrent alteration observed in this study was a deletion between the ERG and TMPRSS2 genes on chromosome 21, presumably related to the recently identified fusion transcripts from these two genes. Results from this study provide a basis for a systematic and comprehensive cataloging of CN alterations associated with grades of prostate cancer, and the subsequent identification of specific genes that associated with initiation and progression of the disease. This article contains supplementary material available via the Internet at http://www.interscience.wiley.com/jpages/1045‐2257/suppmat.

Collaboration


Dive into the Wennuan Liu's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar

William B. Isaacs

Johns Hopkins University School of Medicine

View shared research outputs
Top Co-Authors

Avatar

Jielin Sun

Wake Forest University

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Jin Woo Kim

Wake Forest University

View shared research outputs
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge