Network


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

Hotspot


Dive into the research topics where John R. Prensner is active.

Publication


Featured researches published by John R. Prensner.


Nature | 2010

The landscape of somatic copy-number alteration across human cancers

Rameen Beroukhim; Craig H. Mermel; Dale Porter; Guo Wei; Soumya Raychaudhuri; Jerry Donovan; Jordi Barretina; Jesse S. Boehm; Jennifer Dobson; Mitsuyoshi Urashima; Kevin T. Mc Henry; Reid M. Pinchback; Azra H. Ligon; Yoon-Jae Cho; Leila Haery; Heidi Greulich; Michael R. Reich; Wendy Winckler; Michael S. Lawrence; Barbara A. Weir; Kumiko Tanaka; Derek Y. Chiang; Adam J. Bass; Alice Loo; Carter Hoffman; John R. Prensner; Ted Liefeld; Qing Gao; Derek Yecies; Sabina Signoretti

A powerful way to discover key genes with causal roles in oncogenesis is to identify genomic regions that undergo frequent alteration in human cancers. Here we present high-resolution analyses of somatic copy-number alterations (SCNAs) from 3,131 cancer specimens, belonging largely to 26 histological types. We identify 158 regions of focal SCNA that are altered at significant frequency across several cancer types, of which 122 cannot be explained by the presence of a known cancer target gene located within these regions. Several gene families are enriched among these regions of focal SCNA, including the BCL2 family of apoptosis regulators and the NF-κΒ pathway. We show that cancer cells containing amplifications surrounding the MCL1 and BCL2L1 anti-apoptotic genes depend on the expression of these genes for survival. Finally, we demonstrate that a large majority of SCNAs identified in individual cancer types are present in several cancer types.


Nature | 2012

The mutational landscape of lethal castration-resistant prostate cancer

Catherine S. Grasso; Yi Mi Wu; Dan R. Robinson; Xuhong Cao; Saravana M. Dhanasekaran; Amjad P. Khan; Michael J. Quist; Xiaojun Jing; Robert J. Lonigro; J. Chad Brenner; Irfan A. Asangani; Bushra Ateeq; Sang Y. Chun; Javed Siddiqui; Lee Sam; Matt Anstett; Rohit Mehra; John R. Prensner; Nallasivam Palanisamy; Gregory A Ryslik; Fabio Vandin; Benjamin J. Raphael; Lakshmi P. Kunju; Daniel R. Rhodes; Kenneth J. Pienta; Arul M. Chinnaiyan; Scott A. Tomlins

Characterization of the prostate cancer transcriptome and genome has identified chromosomal rearrangements and copy number gains and losses, including ETS gene family fusions, PTEN loss and androgen receptor (AR) amplification, which drive prostate cancer development and progression to lethal, metastatic castration-resistant prostate cancer (CRPC). However, less is known about the role of mutations. Here we sequenced the exomes of 50 lethal, heavily pre-treated metastatic CRPCs obtained at rapid autopsy (including three different foci from the same patient) and 11 treatment-naive, high-grade localized prostate cancers. We identified low overall mutation rates even in heavily treated CRPCs (2.00 per megabase) and confirmed the monoclonal origin of lethal CRPC. Integrating exome copy number analysis identified disruptions of CHD1 that define a subtype of ETS gene family fusion-negative prostate cancer. Similarly, we demonstrate that ETS2, which is deleted in approximately one-third of CRPCs (commonly through TMPRSS2:ERG fusions), is also deregulated through mutation. Furthermore, we identified recurrent mutations in multiple chromatin- and histone-modifying genes, including MLL2 (mutated in 8.6% of prostate cancers), and demonstrate interaction of the MLL complex with the AR, which is required for AR-mediated signalling. We also identified novel recurrent mutations in the AR collaborating factor FOXA1, which is mutated in 5 of 147 (3.4%) prostate cancers (both untreated localized prostate cancer and CRPC), and showed that mutated FOXA1 represses androgen signalling and increases tumour growth. Proteins that physically interact with the AR, such as the ERG gene fusion product, FOXA1, MLL2, UTX (also known as KDM6A) and ASXL1 were found to be mutated in CRPC. In summary, we describe the mutational landscape of a heavily treated metastatic cancer, identify novel mechanisms of AR signalling deregulated in prostate cancer, and prioritize candidates for future study.


Proceedings of the National Academy of Sciences of the United States of America | 2007

Assessing the significance of chromosomal aberrations in cancer: Methodology and application to glioma

Rameen Beroukhim; Gad Getz; Leia Nghiemphu; Jordi Barretina; Teli Hsueh; David Linhart; Igor Vivanco; Jeffrey C. Lee; Julie H. Huang; Sethu Alexander; Jinyan Du; Tweeny R. Kau; Roman K. Thomas; Kinjal Shah; Horacio Soto; Sven Perner; John R. Prensner; Ralph DeBiasi; Francesca Demichelis; Charlie Hatton; Mark A. Rubin; Levi A. Garraway; Stan F. Nelson; Linda M. Liau; Paul S. Mischel; T. Cloughesy; Matthew Meyerson; Todd Golub; Eric S. Lander; Ingo K. Mellinghoff

Comprehensive knowledge of the genomic alterations that underlie cancer is a critical foundation for diagnostics, prognostics, and targeted therapeutics. Systematic efforts to analyze cancer genomes are underway, but the analysis is hampered by the lack of a statistical framework to distinguish meaningful events from random background aberrations. Here we describe a systematic method, called Genomic Identification of Significant Targets in Cancer (GISTIC), designed for analyzing chromosomal aberrations in cancer. We use it to study chromosomal aberrations in 141 gliomas and compare the results with two prior studies. Traditional methods highlight hundreds of altered regions with little concordance between studies. The new approach reveals a highly concordant picture involving ≈35 significant events, including 16–18 broad events near chromosome-arm size and 16–21 focal events. Approximately half of these events correspond to known cancer-related genes, only some of which have been previously tied to glioma. We also show that superimposed broad and focal events may have different biological consequences. Specifically, gliomas with broad amplification of chromosome 7 have properties different from those with overlapping focalEGFR amplification: the broad events act in part through effects on MET and its ligand HGF and correlate with MET dependence in vitro. Our results support the feasibility and utility of systematic characterization of the cancer genome.


Cancer Discovery | 2011

The emergence of lncRNAs in cancer biology

John R. Prensner; Arul M. Chinnaiyan

The discovery of numerous noncoding RNA (ncRNA) transcripts in species from yeast to mammals has dramatically altered our understanding of cell biology, especially the biology of diseases such as cancer. In humans, the identification of abundant long ncRNA (lncRNA) >200 bp has catalyzed their characterization as critical components of cancer biology. Recently, roles for lncRNAs as drivers of tumor suppressive and oncogenic functions have appeared in prevalent cancer types, such as breast and prostate cancer. In this review, we highlight the emerging impact of ncRNAs in cancer research, with a particular focus on the mechanisms and functions of lncRNAs.


Nature Genetics | 2015

The Landscape of Long Noncoding RNAs in the Human Transcriptome

Matthew K. Iyer; Yashar S. Niknafs; Rohit Malik; Udit Singhal; Anirban Sahu; Yasuyuki Hosono; Terrence R. Barrette; John R. Prensner; Joseph R. Evans; Shuang Zhao; Anton Poliakov; Xuhong Cao; Saravana M. Dhanasekaran; Yi Mi Wu; Dan R. Robinson; David G. Beer; Felix Y. Feng; Hariharan K. Iyer; Arul M. Chinnaiyan

Long noncoding RNAs (lncRNAs) are emerging as important regulators of tissue physiology and disease processes including cancer. To delineate genome-wide lncRNA expression, we curated 7,256 RNA sequencing (RNA-seq) libraries from tumors, normal tissues and cell lines comprising over 43 Tb of sequence from 25 independent studies. We applied ab initio assembly methodology to this data set, yielding a consensus human transcriptome of 91,013 expressed genes. Over 68% (58,648) of genes were classified as lncRNAs, of which 79% were previously unannotated. About 1% (597) of the lncRNAs harbored ultraconserved elements, and 7% (3,900) overlapped disease-associated SNPs. To prioritize lineage-specific, disease-associated lncRNA expression, we employed non-parametric differential expression testing and nominated 7,942 lineage- or cancer-associated lncRNA genes. The lncRNA landscape characterized here may shed light on normal biology and cancer pathogenesis and may be valuable for future biomarker development.


Nature Biotechnology | 2011

Transcriptome sequencing across a prostate cancer cohort identifies PCAT-1 , an unannotated lincRNA implicated in disease progression

John R. Prensner; Matthew K. Iyer; O. Alejandro Balbin; Saravana M. Dhanasekaran; Qi Cao; J. Chad Brenner; Bharathi Laxman; Irfan A. Asangani; Catherine S. Grasso; Hal D. Kominsky; Xuhong Cao; Xiaojun Jing; Xiaoju Wang; Javed Siddiqui; John T. Wei; Dan R. Robinson; Hari Iyer; Nallasivam Palanisamy; Christopher A. Maher; Arul M. Chinnaiyan

Noncoding RNAs (ncRNAs) are emerging as key molecules in human cancer, with the potential to serve as novel markers of disease and to reveal uncharacterized aspects of tumor biology. Here we discover 121 unannotated prostate cancer–associated ncRNA transcripts (PCATs) by ab initio assembly of high-throughput sequencing of polyA+ RNA (RNA-Seq) from a cohort of 102 prostate tissues and cells lines. We characterized one ncRNA, PCAT-1, as a prostate-specific regulator of cell proliferation and show that it is a target of the Polycomb Repressive Complex 2 (PRC2). We further found that patterns of PCAT-1 and PRC2 expression stratified patient tissues into molecular subtypes distinguished by expression signatures of PCAT-1–repressed target genes. Taken together, our findings suggest that PCAT-1 is a transcriptional repressor implicated in a subset of prostate cancer patients. These findings establish the utility of RNA-Seq to identify disease-associated ncRNAs that may improve the stratification of cancer subtypes.High-throughput sequencing of polyA+ RNA (RNA-Seq) in human cancer shows remarkable potential to identify both novel markers of disease and uncharacterized aspects of tumor biology, particularly non-coding RNA (ncRNA) species. We employed RNA-Seq on a cohort of 102 prostate tissues and cells lines and performed ab initio transcriptome assembly to discover unannotated ncRNAs. We nominated 121 such Prostate Cancer Associated Transcripts (PCATs) with cancer-specific expression patterns. Among these, we characterized PCAT-1 as a novel prostate-specific regulator of cell proliferation and target of the Polycomb Repressive Complex 2 (PRC2). We further found that high PCAT-1 and PRC2 expression stratified patient tissues into molecular subtypes distinguished by expression signatures of PCAT-1-repressed target genes. Taken together, the findings presented herein identify PCAT-1 as a novel transcriptional repressor implicated in subset of prostate cancer patients. These findings establish the utility of RNA-Seq to identify disease-associated ncRNAs that may improve the stratification of cancer subtypes.


Nature Genetics | 2013

The long noncoding RNA SChLAP1 promotes aggressive prostate cancer and antagonizes the SWI/SNF complex

John R. Prensner; Matthew K. Iyer; Anirban Sahu; Irfan A. Asangani; Qi Cao; Lalit Patel; Ismael A. Vergara; Elai Davicioni; Nicholas Erho; Mercedeh Ghadessi; Robert B. Jenkins; Timothy J. Triche; Rohit Malik; Rachel Bedenis; Natalie McGregor; Teng Ma; Wei Chen; Sumin Han; Xiaojun Jing; Xuhong Cao; Xiaoju Wang; Benjamin Chandler; Wei Yan; Javed Siddiqui; Lakshmi P. Kunju; Saravana M. Dhanasekaran; Kenneth J. Pienta; Felix Y. Feng; Arul M. Chinnaiyan

Prostate cancers remain indolent in the majority of individuals but behave aggressively in a minority. The molecular basis for this clinical heterogeneity remains incompletely understood. Here we characterize a long noncoding RNA termed SChLAP1 (second chromosome locus associated with prostate-1; also called LINC00913) that is overexpressed in a subset of prostate cancers. SChLAP1 levels independently predict poor outcomes, including metastasis and prostate cancer–specific mortality. In vitro and in vivo gain-of-function and loss-of-function experiments indicate that SChLAP1 is critical for cancer cell invasiveness and metastasis. Mechanistically, SChLAP1 antagonizes the genome-wide localization and regulatory functions of the SWI/SNF chromatin-modifying complex. These results suggest that SChLAP1 contributes to the development of lethal cancer at least in part by antagonizing the tumor-suppressive functions of the SWI/SNF complex.


Science Translational Medicine | 2012

Beyond PSA: The Next Generation of Prostate Cancer Biomarkers

John R. Prensner; Mark A. Rubin; John T. Wei; Arul M. Chinnaiyan

Prostate cancer biomarkers that improve upon the PSA test should address unmet clinical needs. Next-Gen Biomarkers For decades, the prostate-specific antigen (PSA) test has been used to assist in the diagnosis of prostate cancer. It has also become more prevalent in early cancer screening. Although the number of deaths related to prostate cancer has decreased, it is not clear that the PSA test has been the major factor. As Prensner and co-authors point out, the PSA test may “do more harm than good.” This Review focuses on new ways to address unmet clinical needs in the area of biomarkers, to hopefully add to the armamentarium of diagnostic, prognostic, and predictive tools for prostate cancer, including PSA. Since the introduction of serum prostate-specific antigen (PSA) screening 25 years ago, prostate cancer diagnosis and management have been guided by this biomarker. Yet, PSA has proven controversial as a screening assay owing to several inherent limitations. The next wave of prostate cancer biomarkers has emerged, introducing new assays in serum and urine that may supplement or, in time, replace PSA because of their higher cancer specificity. This expanding universe of biomarkers has been facilitated, in large part, by new genomic technologies that have enabled an unbiased look at cancer biology. Such efforts have produced several notable success stories that involve rapidly moving biomarkers from the bench to the clinic. However, biomarker research has centered on disease diagnostics, rather than prognosis and prediction, which would address disease management. The development of biomarkers to stratify risk of prostate cancer aggressiveness at the time of screening remains the greatest unmet clinical need in prostate cancer. We review the current state of prostate cancer biomarker research, including the PSA revolution, its impact on early cancer detection, the recent advances in biomarker discovery, and the future efforts that promise to improve clinical management of this disease.


Cancer Cell | 2011

Coordinated Regulation of Polycomb Group Complexes through microRNAs in Cancer

Qi Cao; Ram Shankar Mani; Bushra Ateeq; Saravana M. Dhanasekaran; Irfan A. Asangani; John R. Prensner; Jung Kim; J. Chad Brenner; Xiaojun Jing; Xuhong Cao; Rui Wang; Yong Li; Arun Dahiya; Lei Wang; Mithil Pandhi; Robert J. Lonigro; Yi Mi Wu; Scott A. Tomlins; Nallasivam Palanisamy; Zhaohui S. Qin; Jindan Yu; Christopher A. Maher; Sooryanarayana Varambally; Arul M. Chinnaiyan

Polycomb Repressive Complexes (PRC1 and PRC2)-mediated epigenetic regulation is critical for maintaining cellular homeostasis. Members of Polycomb Group (PcG) proteins including EZH2, a PRC2 component, are upregulated in various cancer types, implicating their role in tumorigenesis. Here, we have identified several microRNAs (miRNAs) that are repressed by EZH2. These miRNAs, in turn, regulate the expression of PRC1 proteins BMI1 and RING2. We found that ectopic overexpression of EZH2-regulated miRNAs attenuated cancer cell growth and invasiveness, and abrogated cancer stem cell properties. Importantly, expression analysis revealed an inverse correlation between miRNA and PRC protein levels in cell culture and prostate cancer tissues. Taken together, our data have uncovered a coordinate regulation of PRC1 and PRC2 activities that is mediated by miRNAs.


Cancer Research | 2012

PARP-1 inhibition as a targeted strategy to treat Ewing's sarcoma

J. Chad Brenner; Felix Y. Feng; Sumin Han; Sonam Patel; Siddharth V. Goyal; Laura M. Bou-Maroun; Meilan Liu; Robert J. Lonigro; John R. Prensner; Scott A. Tomlins; Arul M. Chinnaiyan

Ewings sarcoma family of tumors (ESFT) refers to aggressive malignancies which frequently harbor characteristic EWS-FLI1 or EWS-ERG genomic fusions. Here, we report that these fusion products interact with the DNA damage response protein and transcriptional coregulator PARP-1. ESFT cells, primary tumor xenografts, and tumor metastases were all highly sensitive to PARP1 inhibition. Addition of a PARP1 inhibitor to the second-line chemotherapeutic agent temozolamide resulted in complete responses of all treated tumors in an EWS-FLI1-driven mouse xenograft model of ESFT. Mechanistic investigations revealed that DNA damage induced by expression of EWS-FLI1 or EWS-ERG fusion genes was potentiated by PARP1 inhibition in ESFT cell lines. Notably, EWS-FLI1 fusion genes acted in a positive feedback loop to maintain the expression of PARP1, which was required for EWS-FLI-mediated transcription, thereby enforcing oncogene-dependent sensitivity to PARP-1 inhibition. Together, our findings offer a strong preclinical rationale to target the EWS-FLI1:PARP1 intersection as a therapeutic strategy to improve the treatment of ESFTs.

Collaboration


Dive into the John R. Prensner's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar

Xuhong Cao

University of Michigan

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Felix Y. Feng

University of California

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Qi Cao

University of Michigan

View shared research outputs
Top Co-Authors

Avatar

Rohit Malik

University of Michigan

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Rohit Mehra

University of Michigan

View shared research outputs
Researchain Logo
Decentralizing Knowledge