Network


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

Hotspot


Dive into the research topics where Adam Kiezun is active.

Publication


Featured researches published by Adam Kiezun.


Nature | 2016

Analysis of protein-coding genetic variation in 60,706 humans

Monkol Lek; Konrad J. Karczewski; Eric Vallabh Minikel; Kaitlin E. Samocha; Eric Banks; Timothy Fennell; Anne H. O’Donnell-Luria; James S. Ware; Andrew Hill; Beryl B. Cummings; Taru Tukiainen; Daniel P. Birnbaum; Jack A. Kosmicki; Laramie Duncan; Karol Estrada; Fengmei Zhao; James Zou; Emma Pierce-Hoffman; Joanne Berghout; David Neil Cooper; Nicole Deflaux; Mark A. DePristo; Ron Do; Jason Flannick; Menachem Fromer; Laura Gauthier; Jackie Goldstein; Namrata Gupta; Daniel P. Howrigan; Adam Kiezun

Large-scale reference data sets of human genetic variation are critical for the medical and functional interpretation of DNA sequence changes. Here we describe the aggregation and analysis of high-quality exome (protein-coding region) DNA sequence data for 60,706 individuals of diverse ancestries generated as part of the Exome Aggregation Consortium (ExAC). This catalogue of human genetic diversity contains an average of one variant every eight bases of the exome, and provides direct evidence for the presence of widespread mutational recurrence. We have used this catalogue to calculate objective metrics of pathogenicity for sequence variants, and to identify genes subject to strong selection against various classes of mutation; identifying 3,230 genes with near-complete depletion of predicted protein-truncating variants, with 72% of these genes having no currently established human disease phenotype. Finally, we demonstrate that these data can be used for the efficient filtering of candidate disease-causing variants, and for the discovery of human ‘knockout’ variants in protein-coding genes.


Nature | 2013

Mutational heterogeneity in cancer and the search for new cancer-associated genes.

Michael S. Lawrence; Petar Stojanov; Paz Polak; Gregory V. Kryukov; Kristian Cibulskis; Andrey Sivachenko; Scott L. Carter; Chip Stewart; Craig H. Mermel; Steven A. Roberts; Adam Kiezun; Peter S. Hammerman; Aaron McKenna; Yotam Drier; Lihua Zou; Alex H. Ramos; Trevor J. Pugh; Nicolas Stransky; Elena Helman; Jaegil Kim; Carrie Sougnez; Lauren Ambrogio; Elizabeth Nickerson; Erica Shefler; Maria L. Cortes; Daniel Auclair; Gordon Saksena; Douglas Voet; Michael S. Noble; Daniel DiCara

Major international projects are underway that are aimed at creating a comprehensive catalogue of all the genes responsible for the initiation and progression of cancer. These studies involve the sequencing of matched tumour–normal samples followed by mathematical analysis to identify those genes in which mutations occur more frequently than expected by random chance. Here we describe a fundamental problem with cancer genome studies: as the sample size increases, the list of putatively significant genes produced by current analytical methods burgeons into the hundreds. The list includes many implausible genes (such as those encoding olfactory receptors and the muscle protein titin), suggesting extensive false-positive findings that overshadow true driver events. We show that this problem stems largely from mutational heterogeneity and provide a novel analytical methodology, MutSigCV, for resolving the problem. We apply MutSigCV to exome sequences from 3,083 tumour–normal pairs and discover extraordinary variation in mutation frequency and spectrum within cancer types, which sheds light on mutational processes and disease aetiology, and in mutation frequency across the genome, which is strongly correlated with DNA replication timing and also with transcriptional activity. By incorporating mutational heterogeneity into the analyses, MutSigCV is able to eliminate most of the apparent artefactual findings and enable the identification of genes truly associated with cancer.


Nature | 2015

Exome sequencing identifies rare LDLR and APOA5 alleles conferring risk for myocardial infarction

Ron Do; Nathan O. Stitziel; Hong-Hee Won; Anders Jørgensen; Stefano Duga; Pier Angelica Merlini; Adam Kiezun; Martin Farrall; Anuj Goel; Or Zuk; Illaria Guella; Rosanna Asselta; Leslie A. Lange; Gina M. Peloso; Paul L. Auer; Domenico Girelli; Nicola Martinelli; Deborah N. Farlow; Mark A. DePristo; Robert Roberts; Alex Stewart; Danish Saleheen; John Danesh; Stephen E. Epstein; Suthesh Sivapalaratnam; G. Kees Hovingh; John J. P. Kastelein; Nilesh J. Samani; Heribert Schunkert; Jeanette Erdmann

Summary Myocardial infarction (MI), a leading cause of death around the world, displays a complex pattern of inheritance1,2. When MI occurs early in life, the role of inheritance is substantially greater1. Previously, rare mutations in low-density lipoprotein (LDL) genes have been shown to contribute to MI risk in individual families3–8 whereas common variants at more than 45 loci have been associated with MI risk in the population9–15. Here, we evaluate the contribution of rare mutations to MI risk in the population. We sequenced the protein-coding regions of 9,793 genomes from patients with MI at an early age (≤50 years in males and ≤60 years in females) along with MI-free controls. We identified two genes where rare coding-sequence mutations were more frequent in cases versus controls at exome-wide significance. At low-density lipoprotein receptor (LDLR), carriers of rare, damaging mutations (3.1% of cases versus 1.3% of controls) were at 2.4-fold increased risk for MI; carriers of null alleles at LDLR were at even higher risk (13-fold difference). This sequence-based estimate of the proportion of early MI cases due to LDLR mutations is remarkably similar to an estimate made more than 40 years ago using total cholesterol16. At apolipoprotein A-V (APOA5), carriers of rare nonsynonymous mutations (1.4% of cases versus 0.6% of controls) were at 2.2-fold increased risk for MI. When compared with non-carriers, LDLR mutation carriers had higher plasma LDL cholesterol whereas APOA5 mutation carriers had higher plasma triglycerides. Recent evidence has connected MI risk with coding sequence mutations at two genes functionally related to APOA5, namely lipoprotein lipase15,17 and apolipoprotein C318,19. When combined, these observations suggest that, beyond LDL cholesterol, disordered metabolism of triglyceride-rich lipoproteins contributes to MI risk.


Nature Genetics | 2013

Somatic mutation of CDKN1B in small intestine neuroendocrine tumors

Joshua M. Francis; Adam Kiezun; Alex H. Ramos; Stefano Serra; Chandra Sekhar Pedamallu; Zhi Rong Qian; Michaela S. Banck; Rahul Kanwar; Amit A. Kulkarni; Anna Karpathakis; Veronica E. Manzo; Tanupriya Contractor; Juliet Philips; Elizabeth Nickerson; Nam H. Pho; Susanne M. Hooshmand; Lauren K. Brais; Michael S. Lawrence; Trevor J. Pugh; Aaron McKenna; Andrey Sivachenko; Kristian Cibulskis; Scott L. Carter; Akinyemi I. Ojesina; Samuel S. Freeman; Robert T. Jones; Douglas Voet; Gordon Saksena; Daniel Auclair; Robert C. Onofrio

The diagnosed incidence of small intestine neuroendocrine tumors (SI-NETs) is increasing, and the underlying genomic mechanisms have not yet been defined. Using exome- and genome-sequence analysis of SI-NETs, we identified recurrent somatic mutations and deletions in CDKN1B, the cyclin-dependent kinase inhibitor gene, which encodes p27. We observed frameshift mutations of CDKN1B in 14 of 180 SI-NETs, and we detected hemizygous deletions encompassing CDKN1B in 7 out of 50 SI-NETs, nominating p27 as a tumor suppressor and implicating cell cycle dysregulation in the etiology of SI-NETs.


Cancer Discovery | 2014

The Genomic Landscape of Pediatric Ewing Sarcoma

Brian D. Crompton; Chip Stewart; Amaro Taylor-Weiner; Gabriela Alexe; Kurek Kc; Monica L. Calicchio; Adam Kiezun; Scott L. Carter; Sachet A. Shukla; Swapnil Mehta; Aaron R. Thorner; de Torres C; Cinzia Lavarino; Mariona Suñol; Aaron McKenna; Andrey Sivachenko; Kristian Cibulskis; Michael S. Lawrence; Petar Stojanov; Mara Rosenberg; Lauren Ambrogio; Daniel Auclair; Sara Seepo; Brendan Blumenstiel; Matthew DeFelice; Ivan Imaz-Rosshandler; Miguel Rivera; Carlos Rodriguez-Galindo; Fleming; Todd R. Golub

UNLABELLED Pediatric Ewing sarcoma is characterized by the expression of chimeric fusions of EWS and ETS family transcription factors, representing a paradigm for studying cancers driven by transcription factor rearrangements. In this study, we describe the somatic landscape of pediatric Ewing sarcoma. These tumors are among the most genetically normal cancers characterized to date, with only EWS-ETS rearrangements identified in the majority of tumors. STAG2 loss, however, is present in more than 15% of Ewing sarcoma tumors; occurs by point mutation, rearrangement, and likely nongenetic mechanisms; and is associated with disease dissemination. Perhaps the most striking finding is the paucity of mutations in immediately targetable signal transduction pathways, highlighting the need for new therapeutic approaches to target EWS-ETS fusions in this disease. SIGNIFICANCE We performed next-generation sequencing of Ewing sarcoma, a pediatric cancer involving bone, characterized by expression of EWS-ETS fusions. We found remarkably few mutations. However, we discovered that loss of STAG2 expression occurs in 15% of tumors and is associated with metastatic disease, suggesting a potential genetic vulnerability in Ewing sarcoma.


Proceedings of the National Academy of Sciences of the United States of America | 2014

Complementary genomic approaches highlight the PI3K/mTOR pathway as a common vulnerability in osteosarcoma

Jennifer A. Perry; Adam Kiezun; Peter Tonzi; Eliezer M. Van Allen; Scott L. Carter; Sylvan C. Baca; Glenn S. Cowley; Ami S. Bhatt; Esther Rheinbay; Chandra Sekhar Pedamallu; Elena Helman; Amaro Taylor-Weiner; Aaron McKenna; David S. DeLuca; Michael S. Lawrence; Lauren Ambrogio; Carrie Sougnez; Andrey Sivachenko; Loren D. Walensky; Nikhil Wagle; Jaume Mora; Carmen Torres; Cinzia Lavarino; Simone dos Santos Aguiar; José Andrés Yunes; Silvia Regina Brandalise; Gabriela Elisa Mercado-Celis; Jorge Melendez-Zajgla; Rocio Cardenas-Cardos; Liliana Velasco-Hidalgo

Significance We present, to our knowledge, the first comprehensive next-generation sequencing of osteosarcoma in combination with a functional genomic screen in a genetically defined mouse model of osteosarcoma. Our data provide a strong rationale for targeting the phosphatidylinositol 3-kinase/mammalian target of rapamycin pathway in osteosarcoma and a foundation for rational clinical trial design. These findings present an immediate clinical opportunity because multiple inhibitors of this pathway are currently in clinical trials. Osteosarcoma is the most common primary bone tumor, yet there have been no substantial advances in treatment or survival in three decades. We examined 59 tumor/normal pairs by whole-exome, whole-genome, and RNA-sequencing. Only the TP53 gene was mutated at significant frequency across all samples. The mean nonsilent somatic mutation rate was 1.2 mutations per megabase, and there was a median of 230 somatic rearrangements per tumor. Complex chains of rearrangements and localized hypermutation were detected in almost all cases. Given the intertumor heterogeneity, the extent of genomic instability, and the difficulty in acquiring a large sample size in a rare tumor, we used several methods to identify genomic events contributing to osteosarcoma survival. Pathway analysis, a heuristic analytic algorithm, a comparative oncology approach, and an shRNA screen converged on the phosphatidylinositol 3-kinase/mammalian target of rapamycin (PI3K/mTOR) pathway as a central vulnerability for therapeutic exploitation in osteosarcoma. Osteosarcoma cell lines are responsive to pharmacologic and genetic inhibition of the PI3K/mTOR pathway both in vitro and in vivo.


PLOS ONE | 2015

Efficient Differentiation of Steroidogenic and Germ-Like Cells from Epigenetically-Related iPSCs Derived from Ovarian Granulosa Cells

Raymond M. Anchan; Behzad Gerami-Naini; Jennifer S. Lindsey; Joshua W. K. Ho; Adam Kiezun; Shane Lipskind; Nicholas Ng; Joseph A. LiCausi; Chloe S. Kim; P.R. Brezina; Thomas Tuschl; Richard L. Maas; W.G. Kearns; Zev Williams

To explore restoration of ovarian function using epigenetically-related, induced pluripotent stem cells (iPSCs), we functionally evaluated the epigenetic memory of novel iPSC lines, derived from mouse and human ovarian granulosa cells (GCs) using c-Myc, Klf4, Sox2 and Oct4 retroviral vectors. The stem cell identity of the mouse and human GC-derived iPSCs (mGriPSCs, hGriPSCs) was verified by demonstrating embryonic stem cell (ESC) antigen expression using immunocytochemistry and RT-PCR analysis, as well as formation of embryoid bodies (EBs) and teratomas that are capable of differentiating into cells from all three germ layers. GriPSCs’ gene expression profiles associate more closely with those of ESCs than of the originating GCs as demonstrated by genome-wide analysis of mRNA and microRNA. A comparative analysis of EBs generated from three different mouse cell lines (mGriPSCs; fibroblast-derived iPSC, mFiPSCs; G4 embryonic stem cells, G4 mESCs) revealed that differentiated mGriPSC-EBs synthesize 10-fold more estradiol (E2) than either differentiated FiPSC- or mESC-EBs under identical culture conditions. By contrast, mESC-EBs primarily synthesize progesterone (P4) and FiPSC-EBs produce neither E2 nor P4. Differentiated mGriPSC-EBs also express ovarian markers (AMHR, FSHR, Cyp19a1, ER and Inha) as well as markers of early gametogenesis (Mvh, Dazl, Gdf9, Boule and Zp1) more frequently than EBs of the other cell lines. These results provide evidence of preferential homotypic differentiation of mGriPSCs into ovarian cell types. Collectively, our data support the hypothesis that generating iPSCs from the desired tissue type may prove advantageous due to the iPSCs’ epigenetic memory.


Cancer Research | 2013

Abstract 3806: Progressive biallelic loss of TP53 is associated with progression of pleuropulmonary blastoma initiated by germline loss and somatic mutation of DICER1.

Trevor J. Pugh; Weiying Yu; Jiandong Yang; Lauren Ambrogio; Scott L. Carter; Adam Kiezun; Aaron McKenna; Petros Giannikopoulos; Gad Getz; Yoav Messinger; Matthew Meyerson; D. Ashley Hill

Proceedings: AACR 104th Annual Meeting 2013; Apr 6-10, 2013; Washington, DC Pleuropulmonary blastoma (PPB) is a rare, aggressive childhood cancer arising from lung mesenchymal cells. In early stages, these cells reside in the walls of epithelial-lined cysts and may remain dormant or progress to thickened cysts and then highly malignant masses over several years. Familial germline loss-of-function DICER1 variants have been described, although carriers may not be affected. Therefore, we sought to uncover additional genetic events underlying tumor development through whole exome sequencing of 15 tumor/normal pairs. In addition to known loss-of-function germline variants found throughout DICER1, this analysis identified somatic missense mutations clustered in the DICER1 RNase IIIb domain in nearly every case. While the somatic mutations impact novel amino acid residues, expression analysis demonstrated depletion of 5p-derived miRNAs similar to that described in other DICER1-mutant tumors. While mutation rates were relatively high compared to other pediatric solid tumors, most mutations, except those in DICER1 and TP53, appeared to be subclonal. Early stage tumors (PPB Type II) had predominantly single-copy deletion of TP53 whereas late stage tumors (PPB Type III) had secondary TP53 mutations. Several late stage tumors also had activating mutations in RAS pathway members, including NRAS and BRAF. Therefore, compound DICER1 loss appears to be initiating and synergistic with incremental loss of TP53 and acquisition of additional driver mutations. We subsequently characterized this molecular pattern using deep sequencing and immunohistochemistry in an extension cohort of 45 additional tumors. This step-wise series of germline and somatic alterations illustrates a genetic mechanism consistent with the progressive nature of pleuropulmonary blastoma. Citation Format: Trevor J. Pugh, Weiying Yu, Jiandong Yang, Lauren Ambrogio, Scott L. Carter, Adam Kiezun, Aaron McKenna, Petros Giannikopoulos, Gad Getz, Yoav Messinger, Matthew Meyerson, D. Ashley Hill. Progressive biallelic loss of TP53 is associated with progression of pleuropulmonary blastoma initiated by germline loss and somatic mutation of DICER1. [abstract]. In: Proceedings of the 104th Annual Meeting of the American Association for Cancer Research; 2013 Apr 6-10; Washington, DC. Philadelphia (PA): AACR; Cancer Res 2013;73(8 Suppl):Abstract nr 3806. doi:10.1158/1538-7445.AM2013-3806


Reproductive Sciences | 2018

An Embryonic and Induced Pluripotent Stem Cell Model for Ovarian Granulosa Cell Development and Steroidogenesis

Shane Lipskind; Jennifer S. Lindsey; Behzad Gerami-Naini; Jennifer L. Eaton; Daniel J. O’Connell; Adam Kiezun; Joshua W. K. Ho; Nicholas Ng; Parveen Parasar; Michelle Ng; Michael Nickerson; Utkan Demirci; Richard L. Maas; Raymond M. Anchan

Embryoid bodies (EBs) can serve as a system for evaluating pluripotency, cellular differentiation, and tissue morphogenesis. In this study, we use EBs derived from mouse embryonic stem cells (mESCs) and human amniocyte–derived induced pluripotent stem cells (hAdiPSCs) as a model for ovarian granulosa cell (GC) development and steroidogenic cell commitment. We demonstrated that spontaneously differentiated murine EBs (mEBs) and human EBs (hEBs) displayed ovarian GC markers, such as aromatase (CYP19A1), FOXL2, AMHR2, FSHR, and GJA1. Comparative microarray analysis identified both shared and unique gene expression between mEBs and the maturing mouse ovary. Gene sets related to gonadogenesis, lipid metabolism, and ovarian development were significantly overrepresented in EBs. Of the 29 genes, 15 that were differentially regulated in steroidogenic mEBs displayed temporal expression changes between embryonic, postnatal, and mature ovarian tissues by polymerase chain reaction. Importantly, both mEBs and hEBs were capable of gonadotropin-responsive estradiol (E2) synthesis in vitro (217-759 pg/mL). Live fluorescence-activated cell sorting–sorted AMHR2+ granulosa-like cells from mEBs continued to produce E2 after purification (15.3 pg/mL) and secreted significantly more E2 than AMHR2− cells (8.6 pg/mL, P < .05). We conclude that spontaneously differentiated EBs of both mESC and hAdiPSC origin can serve as a biologically relevant model for ovarian GC differentiation and steroidogenic cell commitment. These cells should be further investigated for therapeutic uses, such as stem cell–based hormone replacement therapy and in vitro maturation of oocytes.


bioRxiv | 2017

A Strategy for Large-Scale Systematic Pan-Cancer Germline Rare Variation Analysis

Mykyta Artomov; Vijai Joseph; Grace Tiao; Tinu Thomas; Kasmintan A. Schrader; Robert J. Klein; Adam Kiezun; Namrata Gupta; Lauren Margolin; Alexander J. Stratigos; Ivana K. Kim; Kristen Shannon; Leif W. Ellisen; Daniel A. Haber; Gad Getz; Hensin Tsao; Steven M. Lipkin; David Altshuler; Kenneth Offit; Mark J. Daly

Traditionally, genetic studies in cancer are focused on somatic mutations found in tumors and absent from the normal tissue. However, this approach omits inherited component of the cancer risk. We assembled exome sequences from about 2,000 patients with different types of cancers: breast cancer, colon cancer and cutaneous and ocular melanomas matched to more than 7,000 non-cancer controls. Using this dataset, we described germline variation in the known cancer genes grouped by inheritance mode or inclusion in a known cancer pathway. According to our observations, protein-truncating singleton variants in loss-of-function tolerant genes following autosomal dominant inheritance mode are driving the association signal in both genetically enriched and unselected cancer cases. We also performed separate gene-based association analysis for individual phenotypes and proposed a list of new cancer risk gene candidates. Taken together, these results extend existing knowledge of germline variation contribution to cancer onset and provide a strategy for novel gene discovery.Traditionally, genetic studies in cancer are focused on somatic mutations found in tumors and absent from the normal tissue. Identification of shared attributes in germline variation could aid discrimination of high-risk from likely benign mutations and narrow the search space for new cancer predisposing genes. Extraordinary progress made in analysis of common variation with GWAS methodology does not provide sufficient resolution to understand rare variation. To fulfill missing classification for rare germline variation we assembled datasets of whole exome sequences from >2,000 patients with different types of cancers: breast cancer, colon cancer and cutaneous and ocular melanomas matched to more than 7,000 non-cancer controls and analyzed germline variation in known cancer predisposing genes to identify common properties of disease associated mutations and new candidate cancer susceptibility genes. Lists of all cancer predisposing genes were divided into subclasses according to the mode of inheritance of the related cancer syndrome or contribution to known major cancer pathways. Out of all subclasses only genes linked to dominant syndromes presented significant rare germline variants enrichment in cases. Separate analysis of protein-truncating and missense variation in this subclass of genes confirmed significant prevalence of protein-truncating variants in cases only in loss-of-function tolerant genes (pLI 0.9). Taken together, our findings provide insights into the distribution and types of mutations underlying inherited cancer predisposition.

Collaboration


Dive into the Adam Kiezun's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Aaron McKenna

University of Washington

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
Researchain Logo
Decentralizing Knowledge