Network


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

Hotspot


Dive into the research topics where Alireza Heravi-Moussavi is active.

Publication


Featured researches published by Alireza Heravi-Moussavi.


Nature | 2012

The clonal and mutational evolution spectrum of primary triple-negative breast cancers.

Sohrab P. Shah; Andrew Roth; Rodrigo Goya; Arusha Oloumi; Gavin Ha; Yongjun Zhao; Gulisa Turashvili; Jiarui Ding; Kane Tse; Gholamreza Haffari; Ali Bashashati; Leah M Prentice; Jaswinder Khattra; Angela Burleigh; Damian Yap; Virginie Bernard; Andrew McPherson; Karey Shumansky; Anamaria Crisan; Ryan Giuliany; Alireza Heravi-Moussavi; Jamie Rosner; Daniel Lai; Inanc Birol; Richard Varhol; Angela Tam; Noreen Dhalla; Thomas Zeng; Kevin Ma; Simon K. Chan

Primary triple-negative breast cancers (TNBCs), a tumour type defined by lack of oestrogen receptor, progesterone receptor and ERBB2 gene amplification, represent approximately 16% of all breast cancers. Here we show in 104 TNBC cases that at the time of diagnosis these cancers exhibit a wide and continuous spectrum of genomic evolution, with some having only a handful of coding somatic aberrations in a few pathways, whereas others contain hundreds of coding somatic mutations. High-throughput RNA sequencing (RNA-seq) revealed that only approximately 36% of mutations are expressed. Using deep re-sequencing measurements of allelic abundance for 2,414 somatic mutations, we determine for the first time—to our knowledge—in an epithelial tumour subtype, the relative abundance of clonal frequencies among cases representative of the population. We show that TNBCs vary widely in their clonal frequencies at the time of diagnosis, with the basal subtype of TNBC showing more variation than non-basal TNBC. Although p53 (also known as TP53), PIK3CA and PTEN somatic mutations seem to be clonally dominant compared to other genes, in some tumours their clonal frequencies are incompatible with founder status. Mutations in cytoskeletal, cell shape and motility proteins occurred at lower clonal frequencies, suggesting that they occurred later during tumour progression. Taken together, our results show that understanding the biology and therapeutic responses of patients with TNBC will require the determination of individual tumour clonal genotypes.


The New England Journal of Medicine | 2010

ARID1A Mutations in Endometriosis-Associated Ovarian Carcinomas

Kimberly C. Wiegand; Sohrab P. Shah; Osama M. Al-Agha; Yongjun Zhao; Kane Tse; Thomas Zeng; Janine Senz; Melissa K. McConechy; Michael S. Anglesio; Steve E. Kalloger; Winnie Yang; Alireza Heravi-Moussavi; Ryan Giuliany; Christine Chow; John Fee; Abdalnasser Zayed; Leah M Prentice; Nataliya Melnyk; Gulisa Turashvili; Allen Delaney; Jason Madore; Stephen Yip; Andrew McPherson; Gavin Ha; Lynda Bell; Sian Fereday; Angela Tam; Laura Galletta; Patricia N. Tonin; Diane Provencher

BACKGROUND Ovarian clear-cell and endometrioid carcinomas may arise from endometriosis, but the molecular events involved in this transformation have not been described. METHODS We sequenced the whole transcriptomes of 18 ovarian clear-cell carcinomas and 1 ovarian clear-cell carcinoma cell line and found somatic mutations in ARID1A (the AT-rich interactive domain 1A [SWI-like] gene) in 6 of the samples. ARID1A encodes BAF250a, a key component of the SWI–SNF chromatin remodeling complex. We sequenced ARID1A in an additional 210 ovarian carcinomas and a second ovarian clear-cell carcinoma cell line and measured BAF250a expression by means of immunohistochemical analysis in an additional 455 ovarian carcinomas. RESULTS ARID1A mutations were seen in 55 of 119 ovarian clear-cell carcinomas (46%), 10 of 33 endometrioid carcinomas (30%), and none of the 76 high-grade serous ovarian carcinomas. Seventeen carcinomas had two somatic mutations each. Loss of the BAF250a protein correlated strongly with the ovarian clear-cell carcinoma and endometrioid carcinoma subtypes and the presence of ARID1A mutations. In two patients, ARID1A mutations and loss of BAF250a expression were evident in the tumor and contiguous atypical endometriosis but not in distant endometriotic lesions. CONCLUSIONS These data implicate ARID1A as a tumor-suppressor gene frequently disrupted in ovarian clear-cell and endometrioid carcinomas. Since ARID1A mutation and loss of BAF250a can be seen in the preneoplastic lesions, we speculate that this is an early event in the transformation of endometriosis into cancer. (Funded by the British Columbia Cancer Foundation and the Vancouver General Hospital–University of British Columbia Hospital Foundation.).


The New England Journal of Medicine | 2012

Recurrent Somatic DICER1 Mutations in Nonepithelial Ovarian Cancers

Alireza Heravi-Moussavi; Michael S. Anglesio; S.-W. Grace Cheng; Janine Senz; Winnie Yang; Leah M Prentice; Anthony P. Fejes; Christine Chow; Alicia A. Tone; Steve E. Kalloger; Nancy Hamel; Andrew Roth; Gavin Ha; Adrian Wan; Sarah Maines-Bandiera; Clara Salamanca; Barbara Pasini; Blaise Clarke; Anna F. Lee; Cheng-Han Lee; Chengquan Zhao; Robert H. Young; Samuel Aparicio; Poul H. Sorensen; Michelle Woo; Niki Boyd; Steven J.M. Jones; Martin Hirst; Marco A. Marra; Blake Gilks

BACKGROUND Germline truncating mutations in DICER1, an endoribonuclease in the RNase III family that is essential for processing microRNAs, have been observed in families with the pleuropulmonary blastoma-family tumor and dysplasia syndrome. Mutation carriers are at risk for nonepithelial ovarian tumors, notably sex cord-stromal tumors. METHODS We sequenced the whole transcriptomes or exomes of 14 nonepithelial ovarian tumors and noted closely clustered mutations in the region of DICER1 encoding the RNase IIIb domain of DICER1 in four samples. We then sequenced this region of DICER1 in additional ovarian tumors and in certain other tumors and queried the effect of the mutations on the enzymatic activity of DICER1 using in vitro RNA cleavage assays. RESULTS DICER1 mutations in the RNase IIIb domain were found in 30 of 102 nonepithelial ovarian tumors (29%), predominantly in Sertoli-Leydig cell tumors (26 of 43, or 60%), including 4 tumors with additional germline DICER1 mutations. These mutations were restricted to codons encoding metal-binding sites within the RNase IIIb catalytic centers, which are critical for microRNA interaction and cleavage, and were somatic in all 16 samples in which germline DNA was available for testing. We also detected mutations in 1 of 14 nonseminomatous testicular germ-cell tumors, in 2 of 5 embryonal rhabdomyosarcomas, and in 1 of 266 epithelial ovarian and endometrial carcinomas. The mutant DICER1 proteins had reduced RNase IIIb activity but retained RNase IIIa activity. CONCLUSIONS Somatic missense mutations affecting the RNase IIIb domain of DICER1 are common in nonepithelial ovarian tumors. These mutations do not obliterate DICER1 function but alter it in specific cell types, a novel mechanism through which perturbation of microRNA processing may be oncogenic. (Funded by the Terry Fox Research Institute and others.).


The Journal of Pathology | 2011

Subtype-specific mutation of PPP2R1A in endometrial and ovarian carcinomas

Melissa K. McConechy; Michael S. Anglesio; Steve E. Kalloger; Winnie Yang; Janine Senz; Christine Chow; Alireza Heravi-Moussavi; Gregg B. Morin; Anne-Marie Mes-Masson; Mark S. Carey; Jessica N. McAlpine; Janice S. Kwon; Leah M Prentice; Niki Boyd; Sohrab P. Shah; C. Blake Gilks; David Huntsman

PPP2R1A mutations have recently been described in 3/42 (7%) of clear cell carcinomas of the ovary. PPP2R1A encodes the α‐isoform of the scaffolding subunit of the serine/threonine protein phosphatase 2A (PP2A) holoenzyme. This putative tumour suppressor complex is involved in growth and survival pathways. Through targeted sequencing of PPP2R1A, we identified somatic missense mutations in 40.8% (20/49) of high‐grade serous endometrial tumours, and 5.0% (3/60) of endometrial endometrioid carcinomas. Mutations were also identified in ovarian tumours at lower frequencies: 12.2% (5/41) of endometrioid and 4.1% (2/49) of clear cell carcinomas. No mutations were found in 50 high‐grade and 12 low‐grade serous carcinomas. Amino acid residues affected by these mutations are highly conserved across species and are involved in direct interactions with regulatory B‐subunits of the PP2A holoenzyme. PPP2R1A mutations in endometrial high‐grade serous carcinomas are a frequent and potentially targetable feature of this disease. The finding of frequent PPP2R1A mutations in high‐grade serous carcinoma of the endometrium but not in high‐grade serous carcinoma of the ovary provides clear genetic evidence that these are distinct diseases. Copyright


PLOS ONE | 2009

The Specificity of the FOXL2 c.402C>G Somatic Mutation: A Survey of Solid Tumors

Kasmintan A. Schrader; Bella O. Gorbatcheva; Janine Senz; Alireza Heravi-Moussavi; Nataliya Melnyk; Clara Salamanca; Sarah Maines-Bandiera; Susanna L. Cooke; Peter C. K. Leung; James D. Brenton; C. Blake Gilks; John E. Monahan; David Huntsman

Background A somatic mutation in the FOXL2 gene is reported to be present in almost all (97%; 86/89) morphologically defined, adult-type, granulosa-cell tumors (A-GCTs). This FOXL2 c.402C>G mutation changes a highly conserved cysteine residue to a tryptophan (p.C134W). It was also found in a minority of other ovarian malignant stromal tumors, but not in benign ovarian stromal tumors or unrelated ovarian tumors or breast cancers. Methodology/Principal Findings Herein we studied other cancers and cell lines for the presence of this mutation. We screened DNA from 752 tumors of epithelial and mesenchymal origin and 28 ovarian cancer cell lines and 52 other cancer cell lines of varied origin. We found the FOXL2 c.402C>G mutation in an unreported A-GCT case and the A-GCT-derived cell line KGN. All other tumors and cell lines analyzed were mutation negative. Conclusions/Significance In addition to proving that the KGN cell line is a useful model to study A-GCTs, these data show that the c.402C>G mutation in FOXL2 is not commonly found in a wide variety of other cancers and therefore it is likely pathognomonic for A-GCTs and closely related tumors.


Cancer Cell | 2016

Genome-Wide Profiles of Extra-cranial Malignant Rhabdoid Tumors Reveal Heterogeneity and Dysregulated Developmental Pathways

Hye Jung E Chun; Emilia L. Lim; Alireza Heravi-Moussavi; Saeed Saberi; Karen Mungall; Mikhail Bilenky; Annaick Carles; Kane Tse; Inna Shlafman; Kelsey Zhu; Jenny Q. Qian; Diana L. Palmquist; An He; William Long; Rodrigo Goya; Michelle Ng; Veronique G. LeBlanc; Erin Pleasance; Nina Thiessen; Tina Wong; Eric Chuah; Yong Jun Zhao; Jacquie Schein; Daniela S. Gerhard; Michael D. Taylor; Andrew J. Mungall; Richard A. Moore; Yussanne Ma; Steven J.M. Jones; Elizabeth J. Perlman

Malignant rhabdoid tumors (MRTs) are rare lethal tumors of childhood that most commonly occur in the kidney and brain. MRTs are driven by SMARCB1 loss, but the molecular consequences of SMARCB1 loss in extra-cranial tumors have not been comprehensively described and genomic resources for analyses of extra-cranial MRT are limited. To provide such data, we used whole-genome sequencing, whole-genome bisulfite sequencing, whole transcriptome (RNA-seq) and microRNA sequencing (miRNA-seq), and histone modification profiling to characterize extra-cranial MRTs. Our analyses revealed gene expression and methylation subgroups and focused on dysregulated pathways, including those involved in neural crest development.


Nature Communications | 2015

Epigenetic and transcriptional determinants of the human breast

Philippe Gascard; Misha Bilenky; Mahvash Sigaroudinia; Jianxin Zhao; Luolan Li; Annaick Carles; Allen Delaney; Angela Tam; Baljit Kamoh; Stephanie Cho; Malachi Griffith; Andy Chu; Gordon Robertson; Dorothy Cheung; Irene Li; Alireza Heravi-Moussavi; Michelle Moksa; Matthew Mingay; Angela Hussainkhel; Brad H. Davis; Raman P. Nagarajan; Chibo Hong; Lorigail Echipare; Henriette O'Geen; Matthew J. Hangauer; Jeffrey B. Cheng; Dana S. Neel; Donglei Hu; Michael T. McManus; Richard A. Moore

While significant effort has been dedicated to the characterization of epigenetic changes associated with prenatal differentiation, relatively little is known about the epigenetic changes that accompany post-natal differentiation where fully functional differentiated cell types with limited lifespans arise. Here we sought to address this gap by generating epigenomic and transcriptional profiles from primary human breast cell types isolated from disease-free human subjects. From these data we define a comprehensive human breast transcriptional network, including a set of myoepithelial- and luminal epithelial-specific intronic retention events. Intersection of epigenetic states with RNA expression from distinct breast epithelium lineages demonstrates that mCpG provides a stable record of exonic and intronic usage, whereas H3K36me3 is dynamic. We find a striking asymmetry in epigenomic reprogramming between luminal and myoepithelial cell types, with the genomes of luminal cells harbouring more than twice the number of hypomethylated enhancer elements compared with myoepithelial cells.


The Journal of Pathology | 2011

Using next-generation sequencing for the diagnosis of rare disorders: a family with retinitis pigmentosa and skeletal abnormalities†

Kasmintan A. Schrader; Alireza Heravi-Moussavi; Paula J. Waters; Janine Senz; James Whelan; Gavin Ha; Patrice Eydoux; Torsten O. Nielsen; Barry Gallagher; Arusha Oloumi; Niki Boyd; Bridget A. Fernandez; Terry-Lynn Young; Steven J.M. Jones; Martin Hirst; Sohrab P. Shah; Marco A. Marra; Jane Green; David Huntsman

Linkage analysis with subsequent candidate gene sequencing is typically used to diagnose novel inherited syndromes. It is now possible to expedite diagnosis through the sequencing of all coding regions of the genome (the exome) or full genomes. We sequenced the exomes of four members of a family presenting with spondylo‐epiphyseal dysplasia and retinitis pigmentosa and identified a six‐base‐pair (6‐bp) deletion in GNPTG, the gene implicated in mucolipidosis type IIIγ. The diagnosis was confirmed by biochemical studies and both broadens the mucolipidosis type III phenotype and demonstrates the clinical utility of next‐generation sequencing to diagnose rare genetic diseases. Copyright


Cell Reports | 2016

Analysis of Normal Human Mammary Epigenomes Reveals Cell-Specific Active Enhancer States and Associated Transcription Factor Networks

Davide Pellacani; Misha Bilenky; Nagarajan Kannan; Alireza Heravi-Moussavi; David J.H.F. Knapp; Sitanshu Gakkhar; Michelle Moksa; Annaick Carles; Richard A. Moore; Andrew J. Mungall; Marco A. Marra; Steven J.M. Jones; Samuel Aparicio; Martin Hirst; Connie J. Eaves

The normal adult human mammary gland is a continuous bilayered epithelial system. Bipotent and myoepithelial progenitors are prominent and unique components of the outer (basal) layer. The inner (luminal) layer includes both luminal-restricted progenitors and a phenotypically separable fraction that lacks progenitor activity. We now report an epigenomic comparison of these three subsets with one another, with their associated stromal cells, and with three immortalized, non-tumorigenic human mammary cell lines. Each genome-wide analysis contains profiles for six histone marks, methylated DNA, and RNA transcripts. Analysis of these datasets shows that each cell type has unique features, primarily within genomic regulatory regions, and that the cell lines group together. Analyses of the promoter and enhancer profiles place the luminal progenitors in between the basal cells and the non-progenitor luminal subset. Integrative analysis reveals networks of subset-specific transcription factors.


Bioinformatics | 2014

ALEA: a toolbox for allele-specific epigenomics analysis

Hamid Younesy; Torsten Möller; Alireza Heravi-Moussavi; Jeffrey B. Cheng; Joseph F. Costello; Matthew C. Lorincz; Mohammad M. Karimi; Steven J.M. Jones

The assessment of expression and epigenomic status using sequencing based methods provides an unprecedented opportunity to identify and correlate allelic differences with epigenomic status. We present ALEA, a computational toolbox for allele-specific epigenomics analysis, which incorporates allelic variation data within existing resources, allowing for the identification of significant associations between epigenetic modifications and specific allelic variants in human and mouse cells. ALEA provides a customizable pipeline of command line tools for allele-specific analysis of next-generation sequencing data (ChIP-seq, RNA-seq, etc.) that takes the raw sequencing data and produces separate allelic tracks ready to be viewed on genome browsers. The pipeline has been validated using human and hybrid mouse ChIP-seq and RNA-seq data. AVAILABILITY The package, test data and usage instructions are available online at http://www.bcgsc.ca/platform/bioinfo/software/alea CONTACT: : [email protected] or [email protected] Supplementary information: Supplementary data are available at Bioinformatics online.

Collaboration


Dive into the Alireza Heravi-Moussavi's collaboration.

Top Co-Authors

Avatar

Steven J.M. Jones

University of British Columbia

View shared research outputs
Top Co-Authors

Avatar

Martin Hirst

University of British Columbia

View shared research outputs
Top Co-Authors

Avatar

Annaick Carles

University of British Columbia

View shared research outputs
Top Co-Authors

Avatar

Janine Senz

University of British Columbia

View shared research outputs
Top Co-Authors

Avatar

Angela Tam

University of British Columbia

View shared research outputs
Top Co-Authors

Avatar

David Huntsman

University of British Columbia

View shared research outputs
Top Co-Authors

Avatar

Marco A. Marra

University of British Columbia

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Leah M Prentice

University of British Columbia

View shared research outputs
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge