1
|
Lee D, Park Y, Kim S. Towards multi-omics characterization of tumor heterogeneity: a comprehensive review of statistical and machine learning approaches. Brief Bioinform 2020; 22:5896573. [PMID: 34020548 DOI: 10.1093/bib/bbaa188] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2020] [Revised: 06/29/2020] [Accepted: 07/21/2020] [Indexed: 12/19/2022] Open
Abstract
The multi-omics molecular characterization of cancer opened a new horizon for our understanding of cancer biology and therapeutic strategies. However, a tumor biopsy comprises diverse types of cells limited not only to cancerous cells but also to tumor microenvironmental cells and adjacent normal cells. This heterogeneity is a major confounding factor that hampers a robust and reproducible bioinformatic analysis for biomarker identification using multi-omics profiles. Besides, the heterogeneity itself has been recognized over the years for its significant prognostic values in some cancer types, thus offering another promising avenue for therapeutic intervention. A number of computational approaches to unravel such heterogeneity from high-throughput molecular profiles of a tumor sample have been proposed, but most of them rely on the data from an individual omics layer. Since the heterogeneity of cells is widely distributed across multi-omics layers, methods based on an individual layer can only partially characterize the heterogeneous admixture of cells. To help facilitate further development of the methodologies that synchronously account for several multi-omics profiles, we wrote a comprehensive review of diverse approaches to characterize tumor heterogeneity based on three different omics layers: genome, epigenome and transcriptome. As a result, this review can be useful for the analysis of multi-omics profiles produced by many large-scale consortia. Contact:sunkim.bioinfo@snu.ac.kr.
Collapse
Affiliation(s)
- Dohoon Lee
- Interdisciplinary Program in Bioinformatics, Seoul National University, Seoul 08826, Korea
| | - Youngjune Park
- Department of Computer Science and Engineering, Institute of Engineering Research, Seoul National University, Seoul 08826, Korea
| | - Sun Kim
- Bioinformatics Institute, Seoul National University, Seoul 08826, Korea
| |
Collapse
|
2
|
Abstract
In the nearly 60 years since prenatal diagnosis for genetic disease was first offered, the field of prenatal diagnosis has progressed far past rudimentary uterine puncture to provide fetal material to assess gender and interpret risk. Concurrent with the improvements in invasive fetal sampling came technological advances in cytogenetics and molecular biology that widened both the scope of genetic disorders that could be diagnosed and also the resolution at which the human genome could be interrogated. Nowadays, routine blood work available to all pregnant women can determine the risk for common chromosome abnormalities; chorionic villus sampling (CVS) and amniocentesis can be used to diagnose nearly all conditions with a known genetic cause; and the genome and/or exome of a fetus with multiple anomalies can be sequenced in an attempt to determine the underlying etiology. This chapter will discuss some of the major advances in prenatal sampling and prenatal diagnostic laboratory techniques that have occurred over the past six decades.
Collapse
Affiliation(s)
- Brynn Levy
- Department of Pathology and Cell Biology, Vagelos College of Physicians and Surgeons, Columbia University Irving Medical Center, New York, NY, USA.
| | - Melissa Stosic
- Department of Obstetrics and Gynecology, Vagelos College of Physicians and Surgeons, Columbia University Irving Medical Center, New York, NY, USA
| |
Collapse
|
3
|
Chari R, Lockwood WW, Lam WL. Computational Methods for the Analysis of Array Comparative Genomic Hybridization. Cancer Inform 2017. [DOI: 10.1177/117693510600200007] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
Abstract
Array comparative genomic hybridization (array CGH) is a technique for assaying the copy number status of cancer genomes. The widespread use of this technology has lead to a rapid accumulation of high throughput data, which in turn has prompted the development of computational strategies for the analysis of array CGH data. Here we explain the principles behind array image processing, data visualization and genomic profile analysis, review currently available software packages, and raise considerations for future software development.
Collapse
Affiliation(s)
- Raj Chari
- Cancer Genetics and Developmental Biology, British Columbia Cancer Research Centre, Vancouver BC, Canada V5Z 1L3
- These authors contributed equally to this work
| | - William W. Lockwood
- Cancer Genetics and Developmental Biology, British Columbia Cancer Research Centre, Vancouver BC, Canada V5Z 1L3
- These authors contributed equally to this work
| | - Wan L. Lam
- Cancer Genetics and Developmental Biology, British Columbia Cancer Research Centre, Vancouver BC, Canada V5Z 1L3
| |
Collapse
|
4
|
Cuccaro D, De Marco EV, Cittadella R, Cavallaro S. Copy Number Variants in Alzheimer's Disease. J Alzheimers Dis 2017; 55:37-52. [PMID: 27662298 PMCID: PMC5115612 DOI: 10.3233/jad-160469] [Citation(s) in RCA: 31] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/14/2016] [Indexed: 12/18/2022]
Abstract
Alzheimer's disease (AD) is a devastating disease mainly afflicting elderly people, characterized by decreased cognition, loss of memory, and eventually death. Although risk and deterministic genes are known, major genetics research programs are underway to gain further insights into the inheritance of AD. In the last years, in particular, new developments in genome-wide scanning methodologies have enabled the association of a number of previously uncharacterized copy number variants (CNVs, gain or loss of DNA) in AD. Because of the exceedingly large number of studies performed, it has become difficult for geneticists as well as clinicians to systematically follow, evaluate, and interpret the growing number of (sometime conflicting) CNVs implicated in AD. In this review, after a brief introduction of this type of structural variation, and a description of available databases, computational analyses, and technologies involved, we provide a systematic review of all published data showing statistical and scientific significance of pathogenic CNVs and discuss the role they might play in AD.
Collapse
Affiliation(s)
- Denis Cuccaro
- Institute of Neurological Sciences, National Research Council, Section of Catania, Italy
| | | | - Rita Cittadella
- Institute of Neurological Sciences, National Research Council, Section of Mangone, Italy
| | - Sebastiano Cavallaro
- Institute of Neurological Sciences, National Research Council, Section of Catania, Italy
- Institute of Neurological Sciences, National Research Council, Section of Mangone, Italy
| |
Collapse
|
5
|
Cava C, Bertoli G, Castiglioni I. Integrating genetics and epigenetics in breast cancer: biological insights, experimental, computational methods and therapeutic potential. BMC SYSTEMS BIOLOGY 2015; 9:62. [PMID: 26391647 PMCID: PMC4578257 DOI: 10.1186/s12918-015-0211-x] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/14/2015] [Accepted: 09/15/2015] [Indexed: 12/11/2022]
Abstract
BACKGROUND Development of human cancer can proceed through the accumulation of different genetic changes affecting the structure and function of the genome. Combined analyses of molecular data at multiple levels, such as DNA copy-number alteration, mRNA and miRNA expression, can clarify biological functions and pathways deregulated in cancer. The integrative methods that are used to investigate these data involve different fields, including biology, bioinformatics, and statistics. RESULTS These methodologies are presented in this review, and their implementation in breast cancer is discussed with a focus on integration strategies. We report current applications, recent studies and interesting results leading to the identification of candidate biomarkers for diagnosis, prognosis, and therapy in breast cancer by using both individual and combined analyses. CONCLUSION This review presents a state of art of the role of different technologies in breast cancer based on the integration of genetics and epigenetics, and shares some issues related to the new opportunities and challenges offered by the application of such integrative approaches.
Collapse
Affiliation(s)
- Claudia Cava
- Institute of Molecular Bioimaging and Physiology (IBFM), National Research Council (CNR), Milan, Italy.
| | - Gloria Bertoli
- Institute of Molecular Bioimaging and Physiology (IBFM), National Research Council (CNR), Milan, Italy.
| | - Isabella Castiglioni
- Institute of Molecular Bioimaging and Physiology (IBFM), National Research Council (CNR), Milan, Italy.
| |
Collapse
|
6
|
Yang HC, Chang LC, Huggins RM, Chen CH, Mullighan CG. LOHAS: loss-of-heterozygosity analysis suite. Genet Epidemiol 2015; 35:247-60. [PMID: 21312262 DOI: 10.1002/gepi.20573] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2010] [Revised: 11/10/2010] [Accepted: 01/10/2011] [Indexed: 12/13/2022]
Abstract
Detection of loss of heterozygosity (LOH) plays an important role in genetic, genomic and cancer research. We develop computational methods to estimate the proportion of homozygous SNP calls, identify samples with structural alterations and/or unusual genotypic patterns, cluster samples with close LOH structures and map the genomic segments bearing LOH by analyzing data of genome-wide SNP arrays or customized SNP arrays. In addition to cancer genetics/genomics, we also apply the methods to study long contiguous stretches of homozygosity (LCSH) in general populations. The LCSH analysis aids in the identification of samples with complex LCSH patterns indicative of nonrandom mating and/or meiotic recombination cold spots, separation of samples with different genetic backgrounds and sex, and mapping of regions of LCSH. Affymetrix Human Mapping 500K Set SNP data from an acute lymphoblastic leukemia study containing 304 cancer patients and 50 normal controls and from the HapMap Project containing 30 African trios, 30 Caucasian trios and 90 independent Asian samples were analyzed. We identified common gene regions of LOH, e.g., ETV6 and CDKN1B, and identified frequent regions of LCSH, e.g., the region that encompasses the centromeric gene desert region of chromosome 16. Unsupervised analysis separated cancer subtypes and ethnic subpopulations by patterns of LOH/LCSH. Simulation studies considering LOH width, effect size and heterozygous interference fraction were performed, and the results show that the proposed LOH association test has good test power and controls type 1 error well. The developed algorithms are packaged into LOHAS written in R and R GUI.
Collapse
Affiliation(s)
- Hsin-Chou Yang
- Institute of Statistical Science, Academia Sinica, Nankang, Taipei, Taiwan.
| | | | | | | | | |
Collapse
|
7
|
Yang S, Cui X, Fang Z. BCRgt: a Bayesian cluster regression-based genotyping algorithm for the samples with copy number alterations. BMC Bioinformatics 2014; 15:74. [PMID: 24629125 PMCID: PMC4003822 DOI: 10.1186/1471-2105-15-74] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2013] [Accepted: 03/10/2014] [Indexed: 11/17/2022] Open
Abstract
Background Accurate genotype calling is a pre-requisite of a successful Genome-Wide Association Study (GWAS). Although most genotyping algorithms can achieve an accuracy rate greater than 99% for genotyping DNA samples without copy number alterations (CNAs), almost all of these algorithms are not designed for genotyping tumor samples that are known to have large regions of CNAs. Results This study aims to develop a statistical method that can accurately genotype tumor samples with CNAs. The proposed method adds a Bayesian layer to a cluster regression model and is termed a Bayesian Cluster Regression-based genotyping algorithm (BCRgt). We demonstrate that high concordance rates with HapMap calls can be achieved without using reference/training samples, when CNAs do not exist. By adding a training step, we have obtained higher genotyping concordance rates, without requiring large sample sizes. When CNAs exist in the samples, accuracy can be dramatically improved in regions with DNA copy loss and slightly improved in regions with copy number gain, comparing with the Bayesian Robust Linear Model with Mahalanobis distance classifier (BRLMM). Conclusions In conclusion, we have demonstrated that BCRgt can provide accurate genotyping calls for tumor samples with CNAs.
Collapse
|
8
|
Yang H, Volfovsky N, Rattray A, Chen X, Tanaka H, Strathern J. GAP-Seq: a method for identification of DNA palindromes. BMC Genomics 2014; 15:394. [PMID: 24885769 PMCID: PMC4057610 DOI: 10.1186/1471-2164-15-394] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2013] [Accepted: 04/26/2014] [Indexed: 12/13/2022] Open
Abstract
BACKGROUND Closely spaced long inverted repeats, also known as DNA palindromes, can undergo intrastrand annealing to form DNA hairpins. The ability to form these hairpins results in genome instability, difficulties in maintaining clones in Escherichia coli and major problems for most DNA sequencing approaches. Because of their role in genomic instability and gene amplification in some human cancers, it is important to develop systematic approaches to detect and characterize DNA palindromes. RESULTS We developed a new protocol to identify palindromes that couples the S1 nuclease treated Cot0 DNA (GAPF) with high-throughput sequencing (GAP-Seq). Unlike earlier protocols, it does not involve restriction enzymatic digestion prior to DNA snap-back thereby preserving longer DNA sequences. It also indicates the location of the novel junction, which can then be recovered. Using MCF-7 breast cancer cell line as the proof-of-principle analysis, we have identified 35 palindrome candidates and physically characterized the top 5 candidates and their junctions. Because this protocol eliminates many of the false positives that plague earlier techniques, we have improved palindrome identification. CONCLUSIONS The GAP-Seq approach underscores the importance of developing new tools for identifying and characterizing palindromes, and provides a new strategy to systematically assess palindromes in genomes. It will be useful for studying human cancers and other diseases associated with palindromes.
Collapse
Affiliation(s)
- Hui Yang
- />Gene Regulation and Chromosome Biology Laboratory, Frederick National Laboratory for Cancer Research, Cancer Research and Development Center, Frederick, MD 21702 USA
| | - Natalia Volfovsky
- />ABCC/ ISP, SAIC-Frederick, Inc., Frederick National Laboratory for Cancer Research, Frederick, MD 21702 USA
| | - Alison Rattray
- />Gene Regulation and Chromosome Biology Laboratory, Frederick National Laboratory for Cancer Research, Cancer Research and Development Center, Frederick, MD 21702 USA
| | - Xiongfong Chen
- />ABCC/ ISP, SAIC-Frederick, Inc., Frederick National Laboratory for Cancer Research, Frederick, MD 21702 USA
| | - Hisashi Tanaka
- />Department of Molecular Genetics, Cleveland Clinic Lerner Research Institute, Cleveland, Ohio 44195 USA
| | - Jeffrey Strathern
- />Gene Regulation and Chromosome Biology Laboratory, Frederick National Laboratory for Cancer Research, Cancer Research and Development Center, Frederick, MD 21702 USA
| |
Collapse
|
9
|
Lai LC, Tsai MH, Chen PC, Chen LH, Hsiao JH, Chen SK, Lu TP, Lee JM, Hsu CP, Hsiao CK, Chuang EY. SNP rs10248565 in HDAC9 as a novel genomic aberration biomarker of lung adenocarcinoma in non-smoking women. J Biomed Sci 2014; 21:24. [PMID: 24650256 PMCID: PMC3994426 DOI: 10.1186/1423-0127-21-24] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2014] [Accepted: 03/18/2014] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Numerous efforts have been made to elucidate the etiology and improve the treatment of lung cancer, but the overall five-year survival rate is still only 15%. Although cigarette smoking is the primary risk factor for lung cancer, only 7% of female lung cancer patients in Taiwan have a history of smoking. Since cancer results from progressive accumulation of genetic aberrations, genomic rearrangements may be early events in carcinogenesis. RESULTS In order to identify biomarkers of early-stage adenocarcinoma, the genome-wide DNA aberrations of 60 pairs of lung adenocarcinoma and adjacent normal lung tissue in non-smoking women were examined using Affymetrix Genome-Wide Human SNP 6.0 arrays. Common copy number variation (CNV) regions were identified by ≥30% of patients with copy number beyond 2 ± 0.5 of copy numbers for each single nucleotide polymorphism (SNP) and at least 100 continuous SNP variant loci. SNPs associated with lung adenocarcinoma were identified by McNemar's test. Loss of heterozygosity (LOH) SNPs were identified in ≥18% of patients with LOH in the locus. Aberration of SNP rs10248565 at HDAC9 in chromosome 7p21.1 was identified from concurrent analyses of CNVs, SNPs, and LOH. CONCLUSION The results elucidate the genetic etiology of lung adenocarcinoma by demonstrating that SNP rs10248565 may be a potential biomarker of cancer susceptibility.
Collapse
Affiliation(s)
- Liang-Chuan Lai
- Graduate Institute of Physiology, National Taiwan University, Taipei, Taiwan.
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
10
|
Nakachi I, Rice JL, Coldren CD, Edwards MG, Stearman RS, Glidewell SC, Varella-Garcia M, Franklin WA, Keith RL, Lewis MT, Gao B, Merrick DT, Miller YE, Geraci MW. Application of SNP microarrays to the genome-wide analysis of chromosomal instability in premalignant airway lesions. Cancer Prev Res (Phila) 2013; 7:255-65. [PMID: 24346345 DOI: 10.1158/1940-6207.capr-12-0485] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Chromosomal instability is central to the process of carcinogenesis. The genome-wide detection of somatic chromosomal alterations (SCA) in small premalignant lesions remains challenging because sample heterogeneity dilutes the aberrant cell information. To overcome this hurdle, we focused on the B allele frequency data from single-nucleotide polymorphism microarrays (SNP arrays). The difference of allelic fractions between paired tumor and normal samples from the same patient (delta-θ) provides a simple but sensitive detection of SCA in the affected tissue. We applied the delta-θ approach to small, heterogeneous clinical specimens, including endobronchial biopsies and brushings. Regions identified by delta-θ were validated by FISH and quantitative PCR in heterogeneous samples. Distinctive genomic variations were successfully detected across the whole genome in all invasive cancer cases (6 of 6), carcinoma in situ (3 of 3), and high-grade dysplasia (severe or moderate; 3 of 11). Not only well-described SCAs in lung squamous cell carcinoma, but also several novel chromosomal alterations were frequently found across the preinvasive dysplastic cases. Within these novel regions, losses of putative tumor suppressors (RNF20 and SSBP2) and an amplification of RASGRP3 gene with oncogenic activity were observed. Widespread sampling of the airway during bronchoscopy demonstrated that field cancerization reflected by SCAs at multiple sites was detectable. SNP arrays combined with delta-θ analysis can detect SCAs in heterogeneous clinical sample and expand our ability to assess genomic instability in the airway epithelium as a biomarker of lung cancer risk.
Collapse
Affiliation(s)
- Ichiro Nakachi
- University of Colorado, Anschutz Medical Campus, 12700, East 19th Avenue, RC2 9th Floor, Aurora, CO 80045.
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
11
|
Tabernero M, Jara-Acevedo M, Nieto AB, Caballero AR, Otero A, Sousa P, Gonçalves J, Domingues PH, Orfao A. Association between mutation of the NF2 gene and monosomy 22 in menopausal women with sporadic meningiomas. BMC MEDICAL GENETICS 2013; 14:114. [PMID: 24171707 PMCID: PMC3818970 DOI: 10.1186/1471-2350-14-114] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/28/2013] [Accepted: 10/28/2013] [Indexed: 11/10/2022]
Abstract
BACKGROUND Meningioma was the first solid tumor shown to contain a recurrent genetic alteration e.g. monosomy 22/del(22q), NF2 being the most relevant gene involved. Although monosomy 22/del(22q) is present in half of all meningiomas, and meningiomas frequently carry NF2 mutations, no study has been reported so far in which both alterations are simultaneously assessed and correlated with the features of the disease. METHODS Here, we analyzed the frequency of both copy number changes involving chromosome 22 and NF2 mutations in 20 sporadic meningiomas using high-density SNP-arrays, interphase-FISH and PCR techniques. RESULTS Our results show a significant frequency of NF2 mutations (6/20 patients, 30%), most of which (5/6) had not been previously reported in sporadic meningiomas. NF2 mutations involved five different exons and led to a truncated protein (p.Leu163CysfsX46, p.Phe62LeufsX61, p.Asp281MetfsX15, p.Phe285LeufsX11, p.Gln389ArgfsX37) and an in frame deletion of Phe119. Interestingly, all NF2 mutated cases were menopausal women with monosomy 22 but not del(22q). CONCLUSIONS These results confirm and extend on previous observations about the high frequency and heterogeneity of NF2 mutations in sporadic meningiomas and indicate they could be restricted to a well-defined cytogenetic and clinical subgroup of menopausal women. Further studies in large series of patients are required to confirm our observations.
Collapse
|
12
|
Comprehensive high-resolution genomic profiling and cytogenetics of two pediatric and one adult medulloblastoma. Pathol Res Pract 2013; 209:541-7. [PMID: 23896263 DOI: 10.1016/j.prp.2013.06.001] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Medulloblastoma (WHO grade IV) is a rare, malignant, invasive, embryonal tumor which mainly occurs in children and represents less than 1% of all adult brain tumors. Systematic comprehensive genetic analyses on medulloblastomas are rare but necessary to provide more detailed information. Therefore, we performed comprehensive cytogenetic analyses (blood and tissue) of two pediatric and one adult medulloblastoma, using trypsin-Giemsa staining, spectral karyotyping (tissues only), SNP-arrays, and gene expression analyses. We confirmed frequently detected chromosomal aberrations in medulloblastoma, such as +7q, -8p/q, -9q, -11q, -12q, and +17q and identified novel genetic events. Applying SNP-array, we identified constitutional de novo losses 5q21.1, 15q11.2, 17q21.31, 19p12 (pediatric medulloblastoma), 9p21.1, 19p12, 19q13.3, 21q11.2 (adult medulloblastoma) and gains 16p11.1-16p11.2, 18p11.32, Yq11.223-Yq11.23 (pediatric medulloblastoma), Xp22.31 (adult medulloblastoma) possibly representing inherited causal events for medulloblastoma formation. We show evidence for somatic segmental uniparental disomy in regions 1p36, 6q16.3, 6q24.1, 14q21.2, 17p13.3, and 17q22 not previously described for primary medulloblastoma. Gene expression analysis supported classification of the adult medulloblastoma to the WNT-subgroup and classification of pediatric medulloblastomas to group 3 tumors. Analyses of tumors and matched normal tissues (blood) with a combination of complementary techniques will help to further elucidate potentially causal genetic events for medulloblastomas.
Collapse
|
13
|
|
14
|
Jiang L, Jiang J, Yang J, Liu X, Wang J, Wang H, Ding X, Liu J, Zhang Q. Genome-wide detection of copy number variations using high-density SNP genotyping platforms in Holsteins. BMC Genomics 2013; 14:131. [PMID: 23442346 PMCID: PMC3639935 DOI: 10.1186/1471-2164-14-131] [Citation(s) in RCA: 71] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2012] [Accepted: 02/12/2013] [Indexed: 12/13/2022] Open
Abstract
Background Copy number variations (CNVs) are widespread in the human or animal genome and are a significant source of genetic variation, which has been demonstrated to play an important role in phenotypic diversity. Advances in technology have allowed for identification of a large number of CNVs in cattle. Comprehensive explore novel CNVs in the bovine genome would provide valuable information for functional analyses of genome structural variation and facilitating follow-up association studies between complex traits and genetic variants. Results In this study, we performed a genome-wide CNV detection based on high-density SNP genotyping data of 96 Chinese Holstein cattle. A total of 367 CNV regions (CNVRs) across the genome were identified, which cover 42.74Mb of the cattle genome and correspond to 1.61% of the genome sequence. The length of the CNVRs on autosomes range from 10.76 to 2,806.42 Kb with an average of 96.23 Kb. 218 out of these CNVRs contain 610 annotated genes, which possess a wide spectrum of molecular functions. To confirm these findings, quantitative PCR (qPCR) was performed for 17 CNVRs and 13(76.5%) of them were successfully validated. Conclusions Our study demonstrates the high density SNP array can significantly improve the accuracy and sensitivity of CNV calling. Integration of different platforms can enhance the detection of genomic structure variants. Our results provide a significant replenishment for the high resolution map of copy number variation in the bovine genome and valuable information for investigation of genomic structural variation underlying traits of interest in cattle.
Collapse
Affiliation(s)
- Li Jiang
- Key Laboratory of Animal Genetics, Breeding and Reproduction, Ministry of Agriculture, College of Animal Science and Technology, China Agricultural University, Beijing, 100193, PR China
| | | | | | | | | | | | | | | | | |
Collapse
|
15
|
Genome-wide DNA profiling of HIV-related B-cell lymphomas. Methods Mol Biol 2013; 973:213-26. [PMID: 23412793 DOI: 10.1007/978-1-62703-281-0_14] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/20/2023]
Abstract
Non-Hodgkin lymphomas represent a frequent complication of human immunodeficiency virus (HIV) infection, occurring at higher frequency than in immunocompetent individuals, and causing morbidity and mortality. Here, we present the method we have followed to analyze the genomic lesions in HIV-related and in other immunodeficiency-related lymphomas, as well in diffuse large B-cell lymphoma (DLBCL) samples derived from immunocompetent hosts. The technology we have used is represented by the GeneChip Human Mapping 250K NspI arrays (Affymetrix, Santa Clara, CA, USA), arrays based on 25mer oligonucleotides initially designed for large-scale genotyping, that is, the detection of thousands of single-nucleotide polymorphisms (SNPs), then shown to be applicable for the detection of cancer alterations. The protocol is shown in all its steps with suggestions and tips. Applications of the technology and obtained results are also briefly summarized.
Collapse
|
16
|
Jacobs S. Sample processing considerations for detecting copy number changes in formalin-fixed, paraffin-embedded tissues. Cold Spring Harb Protoc 2012; 2012:1195-1202. [PMID: 23118355 DOI: 10.1101/pdb.ip071753] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022]
Abstract
The Whole Genome Sampling Analysis (WGSA) assay in combination with Affymetrix GeneChip Mapping Arrays is used for copy number analysis of high-quality DNA samples (i.e., samples that have been collected from blood, fresh or frozen tissue, or cell lines). Formalin-fixed, paraffin-embedded (FFPE) samples, however, represent the most prevalent form of archived clinical samples, but they provide additional challenges for molecular assays. FFPE processing usually results in the degradation of FFPE DNA and in the contamination and chemical modification of these DNA samples. Because of these issues, FFPE DNA is not suitable for all molecular assays designed for high-quality DNA samples. Strategies recommended for processing FFPE DNA samples through WGSA and to the Mapping arrays are described here.
Collapse
|
17
|
Jacobs S. Data analysis considerations for detecting copy number changes in formalin-fixed, paraffin-embedded tissues. Cold Spring Harb Protoc 2012; 2012:1203-1209. [PMID: 23118356 DOI: 10.1101/pdb.ip071761] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/01/2023]
Abstract
The Whole Genome Sampling Analysis (WGSA) assay in combination with Affymetrix GeneChip Mapping Arrays is used for copy number analysis of high-quality DNA samples (i.e., samples that have been collected from blood, fresh or frozen tissue, or cell lines). Formalin-fixed, paraffin-embedded (FFPE) samples, however, represent the most prevalent form of archived clinical samples, but they provide additional challenges for molecular assays. FFPE processing usually results in the degradation of FFPE DNA and in the contamination and chemical modification of these DNA samples. In this article, we describe the steps needed to obtain reliable copy number predictions from degraded and contaminated FFPE samples.
Collapse
|
18
|
Jain D, Kumar R, Malik BK, Raina V. Copy neutral LOH of FANC F gene: implication on differential diagnosis and treatment of aplastic anemia: letter. Eur J Haematol 2012; 89:365-366. [PMID: 22775453 DOI: 10.1111/j.1600-0609.2012.01834.x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/01/2023]
|
19
|
Liu J, Huang S, Sun M, Liu S, Liu Y, Wang W, Zhang X, Wang H, Hua W. An improved allele-specific PCR primer design method for SNP marker analysis and its application. PLANT METHODS 2012; 8:34. [PMID: 22920499 PMCID: PMC3495711 DOI: 10.1186/1746-4811-8-34] [Citation(s) in RCA: 156] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/15/2012] [Accepted: 08/17/2012] [Indexed: 05/18/2023]
Abstract
BACKGROUND Although Single Nucleotide Polymorphism (SNP) marker is an invaluable tool for positional cloning, association study and evolutionary analysis, low SNP detection efficiency by Allele-Specific PCR (AS-PCR) still restricts its application as molecular marker like other markers such as Simple Sequence Repeat (SSR). To overcome this problem, primers with a single nucleotide artificial mismatch introduced within the three bases closest to the 3'end (SNP site) have been used in AS-PCR. However, for one SNP site, nine possible mismatches can be generated among the three bases and how to select the right one to increase primer specificity is still a challenge. RESULTS In this study, different from the previous reports which used a limited quantity of primers randomly (several or dozen pairs), we systematically investigated the effects of mismatch base pairs, mismatch sites and SNP types on primer specificity with 2071 primer pairs, which were designed based on SNPs from Brassica oleracea 01-88 and 02-12. According to the statistical results, we (1) found that the primers designed with SNP (A/T), in which the mismatch (CA) in the 3rd nucleotide from the 3' end, had the highest allele-specificity (81.9%). This information could be used when designing primers from a large quantity of SNP sites; (2) performed the primer design principle which forms the one and only best primer for every SNP type. This is never reported in previous studies. Additionally, we further identified its availability in rapeseed (Brassica napus L.) and sesame (Sesamum indicum). High polymorphism percent (75%) of the designed primers indicated it is a general method and can be applied in other species. CONCLUSION The method provided in this study can generate primers more effectively for every SNP site compared to other AS-PCR primer design methods. The high allele-specific efficiency of the SNP primer allows the feasibility for low- to moderate- throughput SNP analyses and is much suitable for gene mapping, map-based cloning, and marker-assisted selection in crops.
Collapse
Affiliation(s)
- Jing Liu
- Key Laboratory of Biology and Genetic Improvement of Oil Crops, Ministry of Agriculture, Oil Crops Research Institute of the Chinese Academy of Agricultural Sciences, Wuhan, 430062, People’s Republic of China
| | - Shunmou Huang
- Key Laboratory of Biology and Genetic Improvement of Oil Crops, Ministry of Agriculture, Oil Crops Research Institute of the Chinese Academy of Agricultural Sciences, Wuhan, 430062, People’s Republic of China
| | - Meiyu Sun
- Key Laboratory of Biology and Genetic Improvement of Oil Crops, Ministry of Agriculture, Oil Crops Research Institute of the Chinese Academy of Agricultural Sciences, Wuhan, 430062, People’s Republic of China
| | - Shengyi Liu
- Key Laboratory of Biology and Genetic Improvement of Oil Crops, Ministry of Agriculture, Oil Crops Research Institute of the Chinese Academy of Agricultural Sciences, Wuhan, 430062, People’s Republic of China
| | - Yumei Liu
- Institute of Vegetables and Flowers of the Chinese Academy of Agricultural Sciences, Beijing, 100081, People’s Republic of China
| | - Wanxing Wang
- Institute of Vegetables and Flowers of the Chinese Academy of Agricultural Sciences, Beijing, 100081, People’s Republic of China
| | - Xiurong Zhang
- Key Laboratory of Biology and Genetic Improvement of Oil Crops, Ministry of Agriculture, Oil Crops Research Institute of the Chinese Academy of Agricultural Sciences, Wuhan, 430062, People’s Republic of China
| | - Hanzhong Wang
- Key Laboratory of Biology and Genetic Improvement of Oil Crops, Ministry of Agriculture, Oil Crops Research Institute of the Chinese Academy of Agricultural Sciences, Wuhan, 430062, People’s Republic of China
| | - Wei Hua
- Key Laboratory of Biology and Genetic Improvement of Oil Crops, Ministry of Agriculture, Oil Crops Research Institute of the Chinese Academy of Agricultural Sciences, Wuhan, 430062, People’s Republic of China
| |
Collapse
|
20
|
Carr IM, Diggle CP, Khan K, Inglehearn C, McKibbin M, Bonthron DT, Markham AF, Anwar R, Dobbie A, Pena SDJ, Ali M. Rapid visualisation of microarray copy number data for the detection of structural variations linked to a disease phenotype. PLoS One 2012; 7:e43466. [PMID: 22912880 PMCID: PMC3422275 DOI: 10.1371/journal.pone.0043466] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2012] [Accepted: 07/20/2012] [Indexed: 12/12/2022] Open
Abstract
Whilst the majority of inherited diseases have been found to be caused by single base substitutions, small insertions or deletions (<1Kb), a significant proportion of genetic variability is due to copy number variation (CNV). The possible role of CNV in monogenic and complex diseases has recently attracted considerable interest. However, until the development of whole genome, oligonucleotide micro-arrays, designed specifically to detect the presence of copy number variation, it was not easy to screen an individual for the presence of unknown deletions or duplications with sizes below the level of sensitivity of optical microscopy (3-5 Mb). Now that currently available oligonucleotide micro-arrays have in excess of a million probes, the problem of copy number analysis has moved from one of data production to that of data analysis. We have developed CNViewer, to identify copy number variation that co-segregates with a disease phenotype in small nuclear families, from genome-wide oligonucleotide micro-array data. This freely available program should constitute a useful addition to the diagnostic armamentarium of clinical geneticists.
Collapse
Affiliation(s)
- Ian M Carr
- School of Medicine, University of Leeds, Leeds, United Kingdom.
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
21
|
Valsesia A, Stevenson BJ, Waterworth D, Mooser V, Vollenweider P, Waeber G, Jongeneel CV, Beckmann JS, Kutalik Z, Bergmann S. Identification and validation of copy number variants using SNP genotyping arrays from a large clinical cohort. BMC Genomics 2012; 13:241. [PMID: 22702538 PMCID: PMC3464625 DOI: 10.1186/1471-2164-13-241] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2011] [Accepted: 06/15/2012] [Indexed: 01/11/2023] Open
Abstract
BACKGROUND Genotypes obtained with commercial SNP arrays have been extensively used in many large case-control or population-based cohorts for SNP-based genome-wide association studies for a multitude of traits. Yet, these genotypes capture only a small fraction of the variance of the studied traits. Genomic structural variants (GSV) such as Copy Number Variation (CNV) may account for part of the missing heritability, but their comprehensive detection requires either next-generation arrays or sequencing. Sophisticated algorithms that infer CNVs by combining the intensities from SNP-probes for the two alleles can already be used to extract a partial view of such GSV from existing data sets. RESULTS Here we present several advances to facilitate the latter approach. First, we introduce a novel CNV detection method based on a Gaussian Mixture Model. Second, we propose a new algorithm, PCA merge, for combining copy-number profiles from many individuals into consensus regions. We applied both our new methods as well as existing ones to data from 5612 individuals from the CoLaus study who were genotyped on Affymetrix 500K arrays. We developed a number of procedures in order to evaluate the performance of the different methods. This includes comparison with previously published CNVs as well as using a replication sample of 239 individuals, genotyped with Illumina 550K arrays. We also established a new evaluation procedure that employs the fact that related individuals are expected to share their CNVs more frequently than randomly selected individuals. The ability to detect both rare and common CNVs provides a valuable resource that will facilitate association studies exploring potential phenotypic associations with CNVs. CONCLUSION Our new methodologies for CNV detection and their evaluation will help in extracting additional information from the large amount of SNP-genotyping data on various cohorts and use this to explore structural variants and their impact on complex traits.
Collapse
Affiliation(s)
- Armand Valsesia
- Department of Medical Genetics, University of Lausanne, Lausanne, Switzerland
| | | | | | | | | | | | | | | | | | | |
Collapse
|
22
|
Zin R, Pham K, Ashleigh M, Ravine D, Waring P, Charles A. SNP-based arrays complement classic cytogenetics in the detection of chromosomal aberrations in Wilms’ tumor. Cancer Genet 2012; 205:80-93. [DOI: 10.1016/j.cancergen.2011.12.003] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2011] [Revised: 12/09/2011] [Accepted: 12/16/2011] [Indexed: 12/11/2022]
|
23
|
Characterising chromosome rearrangements: recent technical advances in molecular cytogenetics. Heredity (Edinb) 2011; 108:75-85. [PMID: 22086080 PMCID: PMC3238113 DOI: 10.1038/hdy.2011.100] [Citation(s) in RCA: 71] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023] Open
Abstract
Genomic rearrangements can result in losses, amplifications, translocations and inversions of DNA fragments thereby modifying genome architecture, and potentially having clinical consequences. Many genomic disorders caused by structural variation have initially been uncovered by early cytogenetic methods. The last decade has seen significant progression in molecular cytogenetic techniques, allowing rapid and precise detection of structural rearrangements on a whole-genome scale. The high resolution attainable with these recently developed techniques has also uncovered the role of structural variants in normal genetic variation alongside single-nucleotide polymorphisms (SNPs). We describe how array-based comparative genomic hybridisation, SNP arrays, array painting and next-generation sequencing analytical methods (read depth, read pair and split read) allow the extensive characterisation of chromosome rearrangements in human genomes.
Collapse
|
24
|
Goldmuntz E, Paluru P, Glessner J, Hakonarson H, Biegel JA, White PS, Gai X, Shaikh TH. Microdeletions and microduplications in patients with congenital heart disease and multiple congenital anomalies. CONGENIT HEART DIS 2011; 6:592-602. [PMID: 22010865 DOI: 10.1111/j.1747-0803.2011.00582.x] [Citation(s) in RCA: 70] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
OBJECTIVE Multiple genetic syndromes are caused by recurrent chromosomal microdeletions or microduplications. The increasing use of high-resolution microarrays in clinical analysis has allowed the identification of previously undetectable submicroscopic copy number variants (CNVs) associated with genetic disorders. We hypothesized that patients with congenital heart disease and additional dysmorphic features or other anomalies would be likely to harbor previously undetected CNVs, which might identify new disease loci or disease-related genes for various cardiac defects. DESIGN Copy number analysis with single nucleotide polymorphism-based, oligonucleotide microarrays was performed on 58 patients with congenital heart disease and other dysmorphic features and/or other anomalies. The observed CNVs were validated using independent techniques and validated CNVs were further analyzed using computational algorithms and comparison with available control CNV datasets in order to assess their pathogenic potential. RESULTS Potentially pathogenic CNVs were detected in twelve of 58 patients (20.7%), ranging in size from 240 Kb to 9.6 Mb. These CNVs contained between 1 and 55 genes, including NRP1, NTRK3, MESP1, ADAM19, and HAND1, all of which are known to participate in cardiac development. CONCLUSIONS Genome-wide analysis in patients with congenital heart disease and additional phenotypes has identified potentially pathogenic CNVs affecting genes involved in cardiac development. The identified variant loci and the genes within them warrant further evaluation in similarly syndromic and nonsyndromic cardiac cohorts.
Collapse
Affiliation(s)
- Elizabeth Goldmuntz
- Divisions of Cardiology Human Genetics Oncology Center for Applied Genomics Center for Biomedical Informatics, The Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | | | | | | | | | | | | | | |
Collapse
|
25
|
Jacquemont S, Reymond A, Zufferey F, Harewood L, Walters RG, Kutalik Z, Martinet D, Shen Y, Valsesia A, Beckmann ND, Thorleifsson G, Belfiore M, Bouquillon S, Campion D, De Leeuw N, De Vries BBA, Esko T, Fernandez BA, Fernández-Aranda F, Fernández-Real JM, Gratacòs M, Guilmatre A, Hoyer J, Jarvelin MR, Kooy FR, Kurg A, Le Caignec C, Männik K, Platt OS, Sanlaville D, Van Haelst MM, Villatoro Gomez S, Walha F, Wu BL, Yu Y, Aboura A, Addor MC, Alembik Y, Antonarakis SE, Arveiler B, Barth M, Bednarek N, Béna F, Bergmann S, Beri M, Bernardini L, Blaumeiser B, Bonneau D, Bottani A, Boute O, Brunner HG, Cailley D, Callier P, Chiesa J, Chrast J, Coin L, Coutton C, Cuisset JM, Cuvellier JC, David A, De Freminville B, Delobel B, Delrue MA, Demeer B, Descamps D, Didelot G, Dieterich K, Disciglio V, Doco-Fenzy M, Drunat S, Duban-Bedu B, Dubourg C, El-Sayed Moustafa JS, Elliott P, Faas BHW, Faivre L, Faudet A, Fellmann F, Ferrarini A, Fisher R, Flori E, Forer L, Gaillard D, Gerard M, Gieger C, Gimelli S, Gimelli G, Grabe HJ, Guichet A, Guillin O, Hartikainen AL, Heron D, Hippolyte L, Holder M, Homuth G, Isidor B, Jaillard S, Jaros Z, Jiménez-Murcia S, Joly Helas G, Jonveaux P, Kaksonen S, Keren B, Kloss-Brandstätter A, Knoers NVAM, Koolen DA, Kroisel PM, Kronenberg F, Labalme A, Landais E, Lapi E, Layet V, Legallic S, Leheup B, Leube B, Lewis S, Lucas J, Macdermot KD, Magnusson P, Marshall CR, Mathieu-Dramard M, Mccarthy MI, Meitinger T, Antonietta Mencarelli M, Merla G, Moerman A, Mooser V, Morice-Picard F, Mucciolo M, Nauck M, Coumba Ndiaye N, Nordgren A, Pasquier L, Petit F, Pfundt R, Plessis G, Rajcan-Separovic E, Paolo Ramelli G, Rauch A, Ravazzolo R, Reis A, Renieri A, Richart C, Ried JS, Rieubland C, Roberts W, Roetzer KM, Rooryck C, Rossi M, Saemundsen E, Satre V, Schurmann C, Sigurdsson E, Stavropoulos DJ, Stefansson H, Tengström C, Thorsteinsdóttir U, Tinahones FJ, Touraine R, Vallée L, Van Binsbergen E, Van Der Aa N, Vincent-Delorme C, Visvikis-Siest S, Vollenweider P, Völzke H, Vulto-Van Silfhout AT, Waeber G, Wallgren-Pettersson C, Witwicki RM, Zwolinksi S, Andrieux J, Estivill X, Gusella JF, Gustafsson O, Metspalu A, Scherer SW, Stefansson K, Blakemore AIF, Beckmann JS, Froguel P. Mirror extreme BMI phenotypes associated with gene dosage at the chromosome 16p11.2 locus. Nature 2011; 478:97-102. [PMID: 21881559 PMCID: PMC3637175 DOI: 10.1038/nature10406] [Citation(s) in RCA: 326] [Impact Index Per Article: 25.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2011] [Accepted: 07/29/2011] [Indexed: 12/25/2022]
Abstract
Both obesity and being underweight have been associated with increased mortality. Underweight, defined as a body mass index (BMI) ≤ 18.5 kg per m(2) in adults and ≤ -2 standard deviations from the mean in children, is the main sign of a series of heterogeneous clinical conditions including failure to thrive, feeding and eating disorder and/or anorexia nervosa. In contrast to obesity, few genetic variants underlying these clinical conditions have been reported. We previously showed that hemizygosity of a ∼600-kilobase (kb) region on the short arm of chromosome 16 causes a highly penetrant form of obesity that is often associated with hyperphagia and intellectual disabilities. Here we show that the corresponding reciprocal duplication is associated with being underweight. We identified 138 duplication carriers (including 132 novel cases and 108 unrelated carriers) from individuals clinically referred for developmental or intellectual disabilities (DD/ID) or psychiatric disorders, or recruited from population-based cohorts. These carriers show significantly reduced postnatal weight and BMI. Half of the boys younger than five years are underweight with a probable diagnosis of failure to thrive, whereas adult duplication carriers have an 8.3-fold increased risk of being clinically underweight. We observe a trend towards increased severity in males, as well as a depletion of male carriers among non-medically ascertained cases. These features are associated with an unusually high frequency of selective and restrictive eating behaviours and a significant reduction in head circumference. Each of the observed phenotypes is the converse of one reported in carriers of deletions at this locus. The phenotypes correlate with changes in transcript levels for genes mapping within the duplication but not in flanking regions. The reciprocal impact of these 16p11.2 copy-number variants indicates that severe obesity and being underweight could have mirror aetiologies, possibly through contrasting effects on energy balance.
Collapse
Affiliation(s)
| | - Alexandre Reymond
- Centre de génomique intégrative
Université de Lausanne1015 Lausanne,CH
| | - Flore Zufferey
- Service de génétique médicale
CHU Vaudois1011 Lausanne,CH
| | - Louise Harewood
- Centre de génomique intégrative
Université de Lausanne1015 Lausanne,CH
| | - Robin G. Walters
- Department of Genomics of Common Disease
Imperial College LondonHammersmith hospital, London W12 0NN,GB
| | - Zoltán Kutalik
- Department of Medical Genetics
University of LausanneCH
- SIB, Swiss Institute of Bioinformatics
Swiss Institute of BioinformaticsQuartier Sorge - Batiment Genopode 1015 Lausanne Switzerland,CH
| | | | - Yiping Shen
- Laboratory Medicine
Children's Hospital BostonBoston, Massachusetts 02115,US
- Center for Human Genetic Research
Massachusetts General HospitalBoston, Massachusetts 02114,US
| | - Armand Valsesia
- Department of Medical Genetics
University of LausanneCH
- SIB, Swiss Institute of Bioinformatics
Swiss Institute of BioinformaticsQuartier Sorge - Batiment Genopode 1015 Lausanne Switzerland,CH
- Ludwig Institute for Cancer Research
Université de Lausanne1015 Lausanne,CH
| | | | | | - Marco Belfiore
- Service de génétique médicale
CHU Vaudois1011 Lausanne,CH
| | - Sonia Bouquillon
- Laboratoire de Génétique Médicale
Hôpital Jeanne de FlandreCHRU Lille59037 Lille Cedex,FR
| | - Dominique Campion
- Génétique médicale et fonctionnelle du cancer et des maladies neuropsychiatriques
INSERM : U614Université de RouenUFR de Medecine et de Pharmacie 22, Boulevard Gambetta 76183 Rouen cedex,FR
- Estonian Genome and Medicine
University of Tartu51010 Tartu,EE
| | - Nicole De Leeuw
- Department of human genetics
Radboud University Nijmegen Medical CentreNijmegen Centre for Molecular Life SciencesInstitute for Genetic and Metabolic Disorders6500 HB Nijmegen,NL
| | - Bert B. A. De Vries
- Department of human genetics
Radboud University Nijmegen Medical CentreNijmegen Centre for Molecular Life SciencesInstitute for Genetic and Metabolic Disorders6500 HB Nijmegen,NL
| | - Tõnu Esko
- Estonian Genome and Medicine
University of Tartu51010 Tartu,EE
- Institute of Molecular and Cell Biology
University of Tartu51010 Tartu,EE
| | - Bridget A. Fernandez
- Disciplines of Genetics and Medicine
Memorial University of NewfoundlandSt. John's Newfoundland,CA
| | - Fernando Fernández-Aranda
- IDIBELL, Department of Psychiatry
University Hospital of BellvitgeCIBERobn Fisiopatología de la Obesidad y Nutrición08907 Barcelona,ES
| | - José Manuel Fernández-Real
- Section of Diabetes, Endocrinology and Nutrition
University Hospital of GironaBiomedical Research Institute "Dr Josep Trueta"CIBERobn Fisiopatología de la Obesidad y Nutrición17007 Girona,ES
| | - Mònica Gratacòs
- CRG-UPF, Center for Genomic Regulation
CIBER de Epidemiología y Salud Pública (CIBERESP)C/ Dr. Aiguader, 88 08003 Barcelona, Catalonia, Spain,ES
| | - Audrey Guilmatre
- Génétique médicale et fonctionnelle du cancer et des maladies neuropsychiatriques
INSERM : U614Université de RouenUFR de Medecine et de Pharmacie 22, Boulevard Gambetta 76183 Rouen cedex,FR
- Estonian Genome and Medicine
University of Tartu51010 Tartu,EE
| | - Juliane Hoyer
- Institute of Human Genetics
Friedrich-Alexander University Erlangen-Nuremberg91054 Erlangen,DE
| | - Marjo-Riitta Jarvelin
- Department of child and adolescent health
National Institute for Health and WelfareUniversity of OuluInstitute of Health Sciences and Biocenter OuluBox 310, 90101 Oulu,FI
| | - Frank R. Kooy
- Department of Medical Genetics
University Hospital Antwerp2650 Edegem,BE
| | - Ants Kurg
- Institute of Molecular and Cell Biology
University of Tartu51010 Tartu,EE
| | - Cédric Le Caignec
- Service d'ORL et de Chirurgie Cervicofaciale
INSERM : U587Hôpital d'Enfants Armand-TrousseauUniversité Pierre et Marie Curie - Paris 6Paris,FR
| | - Katrin Männik
- Institute of Molecular and Cell Biology
University of Tartu51010 Tartu,EE
| | - Orah S. Platt
- Laboratory Medicine
Children's Hospital BostonBoston, Massachusetts 02115,US
| | - Damien Sanlaville
- Service de cytogénétique constitutionnelle
Hospices Civils de LyonCHU de LyonCentre Neuroscience et Recherche69000 Lyon,FR
| | - Mieke M. Van Haelst
- Department of Genomics of Common Disease
Imperial College LondonHammersmith hospital, London W12 0NN,GB
- Department of Medical Genetics
University Medical Center Utrecht3584 EA Utrecht,NL
| | - Sergi Villatoro Gomez
- CRG-UPF, Center for Genomic Regulation
CIBER de Epidemiología y Salud Pública (CIBERESP)C/ Dr. Aiguader, 88 08003 Barcelona, Catalonia, Spain,ES
| | - Faida Walha
- Centre de génomique intégrative
Université de Lausanne1015 Lausanne,CH
| | - Bai-Lin Wu
- Laboratory Medicine
Children's Hospital BostonBoston, Massachusetts 02115,US
- Institutes of Biomedical Science
Fudan UniversityChildren's Hospital200032 Shanghai,CN
| | - Yongguo Yu
- Laboratory Medicine
Children's Hospital BostonBoston, Massachusetts 02115,US
- Shanghai Children's Medical Center
Shanghai Children's Medical Center200127 Shanghai,CN
| | - Azzedine Aboura
- Département de génétique
Assistance publique - Hôpitaux de Paris (AP-HP)Hôpital Robert DebréUniversité Paris VII - Paris Diderot48, boulevard Sérurier 75935 Paris cedex 19,FR
| | | | - Yves Alembik
- Service de cytogénétique
CHU StrasbourgHôpital de Hautepierre1 Av Moliere 67098 Strasbourg Cedex,FR
| | | | - Benoît Arveiler
- MRGM, Maladies Rares - Génétique et Métabolisme
Hôpital PellegrinService de Génétique Médicale du CHU de BordeauxUniversité Victor Segalen - Bordeaux II : EA4576146 rue Léo-Saignat - 33076 Bordeaux Cedex,FR
- Service de génétique médicale
CHU BordeauxGroupe hospitalier PellegrinUniversité de BordeauxBordeaux,FR
| | - Magalie Barth
- Service de génétique [Angers]
CHU AngersUniversité d'Angersrue Larrey, 49100 Angers,FR
| | - Nathalie Bednarek
- URCA, Université de Reims Champagne-Ardenne
Ministère de l'Enseignement Supérieur et de la Recherche Scientifique9 boulevard Paix - 51097 Reims cedex,FR
| | - Frédérique Béna
- Génétique médicale
Hôpitaux Universitaires de Genève1205 Geneva,CH
| | - Sven Bergmann
- Department of Medical Genetics
University of LausanneCH
- SIB, Swiss Institute of Bioinformatics
Swiss Institute of BioinformaticsQuartier Sorge - Batiment Genopode 1015 Lausanne Switzerland,CH
- Department of Molecular Genetics
Weizmann Institute of ScienceRehovot,IL
| | - Mylène Beri
- Laboratoire de Génétique
CHU NancyVandoeuvre les Nancy,FR
| | - Laura Bernardini
- Mendel Laboratory
IRCCS Casa Sollievo della Sofferenza Hospital71013 San Giovanni Rotondo,IT
| | - Bettina Blaumeiser
- Department of Medical Genetics
University Hospital Antwerp2650 Edegem,BE
| | - Dominique Bonneau
- Service de génétique [Angers]
CHU AngersUniversité d'Angersrue Larrey, 49100 Angers,FR
| | - Armand Bottani
- Génétique médicale
Hôpitaux Universitaires de Genève1205 Geneva,CH
| | - Odile Boute
- Service de Génétique clinique
Hôpital Jeanne de FlandreCHRU Lille2 avenue Oscar Lambret, 59000 Lille,FR
| | - Han G. Brunner
- Department of human genetics
Radboud University Nijmegen Medical CentreNijmegen Centre for Molecular Life SciencesInstitute for Genetic and Metabolic Disorders6500 HB Nijmegen,NL
| | - Dorothée Cailley
- Service de génétique médicale
CHU BordeauxGroupe hospitalier PellegrinUniversité de BordeauxBordeaux,FR
| | | | - Jean Chiesa
- Laboratoire de Cytogénétique
CHU Nîmes30029 Nimes,FR
| | - Jacqueline Chrast
- Centre de génomique intégrative
Université de Lausanne1015 Lausanne,CH
| | - Lachlan Coin
- Department of Genomics of Common Disease
Imperial College LondonHammersmith hospital, London W12 0NN,GB
| | - Charles Coutton
- Département de génétique et procréation
CHU GrenobleUniversité Joseph Fourier - Grenoble Ifaculté de médecine-pharmacieDomaine de la Merci, 38706 Grenoble,FR
- AGIM, AGeing and IMagery, CNRS FRE3405
Université Joseph Fourier - Grenoble IEcole Pratique des Hautes EtudesCNRS : UMR5525Faculté de médecine de Grenoble, 38700 La Tronche,FR
- Laboratoire de biochimie et génétique moléculaire
CHU Grenoble38043 Grenoble,FR
| | - Jean-Marie Cuisset
- Service de Neuropédiatrie
CHRU LilleHôpital Roger Salengro59037 Lille,FR
| | | | - Albert David
- Service d'ORL et de Chirurgie Cervicofaciale
INSERM : U587Hôpital d'Enfants Armand-TrousseauUniversité Pierre et Marie Curie - Paris 6Paris,FR
| | | | - Bruno Delobel
- Centre de Génétique Chromosomique
GHICLHôpital Saint Vincent de PaulBoulevard de Belfort B.P. 387 59020 LILLE CEDEX,FR
| | - Marie-Ange Delrue
- MRGM, Maladies Rares - Génétique et Métabolisme
Hôpital PellegrinService de Génétique Médicale du CHU de BordeauxUniversité Victor Segalen - Bordeaux II : EA4576146 rue Léo-Saignat - 33076 Bordeaux Cedex,FR
- Service de génétique médicale
CHU BordeauxGroupe hospitalier PellegrinUniversité de BordeauxBordeaux,FR
| | - Bénédicte Demeer
- Service de génétique médicale
CHU AMIENSPlace Victor Pauchet, 80054 Amiens Cedex 1,FR
| | - Dominique Descamps
- Centre hospitalier de Béthune
Centre hospitalier de Béthune62408 Bethune,FR
| | - Gérard Didelot
- Centre de génomique intégrative
Université de Lausanne1015 Lausanne,CH
| | | | - Vittoria Disciglio
- Department of Biotechnology
Università degli studi di SienaMedical Genetics53100 Siena,IT
| | - Martine Doco-Fenzy
- Service de Génétique
CHU ReimsHôpital Maison BlancheIFR 5351092 Reims,FR
| | - Séverine Drunat
- Département de génétique
Assistance publique - Hôpitaux de Paris (AP-HP)Hôpital Robert DebréUniversité Paris VII - Paris Diderot48, boulevard Sérurier 75935 Paris cedex 19,FR
| | - Bénédicte Duban-Bedu
- Centre de Génétique Chromosomique
GHICLHôpital Saint Vincent de PaulBoulevard de Belfort B.P. 387 59020 LILLE CEDEX,FR
| | - Christèle Dubourg
- IGDR, Institut de Génétique et Développement de Rennes
CNRS : UMR6061Université de Rennes 1IFR140Faculté de Médecine - CS 34317 2 Av du Professeur Léon Bernard 35043 RENNES CEDEX,FR
| | | | - Paul Elliott
- Department of Epidemiology and Public Health
Imperial College LondonSt Mary's Campus, Norfolk Place, London W2 1PG,GB
| | - Brigitte H. W. Faas
- Department of human genetics
Radboud University Nijmegen Medical CentreNijmegen Centre for Molecular Life SciencesInstitute for Genetic and Metabolic Disorders6500 HB Nijmegen,NL
- Department of Human Genetics, Radboud University Medical Centre, PO Box 9101, 6500 HB Nijmegen
Department of Human Genetics, Radboud University Medical Centre, PO Box 9101, 6500 HB NijmegenNL
| | - Laurence Faivre
- Department of Experimental Cardiology
Heart Failure Research Center (HFRC)Academic Medical Center (AMC)Meibergdreef 9, PO Box 22660, 1100 DD Amsterdam,NL
| | - Anne Faudet
- Département de Génétique Cytogénétique et Embryologie
Assistance publique - Hôpitaux de Paris (AP-HP)Hôpital Pitié-SalpêtrièreUniversité Paris VI - Pierre et Marie Curie47-83, boulevard de l'Hôpital 75651 PARIS Cedex 13,FR
| | | | | | - Richard Fisher
- Institute of human genetics
International Centre for LifeNewcastle Upon Tyne NE1 4EP,GB
| | - Elisabeth Flori
- Service de cytogénétique
CHU StrasbourgHôpital de Hautepierre1 Av Moliere 67098 Strasbourg Cedex,FR
| | - Lukas Forer
- Division of genetic epidemiology
Innsbruck Medical UniversityDepartment of Medical GeneticsMolecular and Clinical Pharmacology6020 Innsbruck,AT
| | - Dominique Gaillard
- Service de Génétique
CHU ReimsHôpital Maison BlancheIFR 5351092 Reims,FR
| | - Marion Gerard
- Département de génétique
Assistance publique - Hôpitaux de Paris (AP-HP)Hôpital Robert DebréUniversité Paris VII - Paris Diderot48, boulevard Sérurier 75935 Paris cedex 19,FR
| | - Christian Gieger
- Institute of Experimental Medicine
Academy of Sciences of the Czech RepublicVídeÅ�ská 1083 142 20 Prague,CZ
| | - Stefania Gimelli
- Génétique médicale
Hôpitaux Universitaires de Genève1205 Geneva,CH
- Department of Obstetrics and Gynecology
Institute of Clinical MedicineUniversity of Oulu90570 Oulu,FI
| | - Giorgio Gimelli
- Laboratorio di citogenetica
G. Gaslini Institute16147 Genova,IT
| | - Hans J. Grabe
- Department of Psychiatry and Psychotherapy
Ernst-Moritz-Arndt University Greifswald17475 Greifswald and D-18437 Stralsund,DE
| | - Agnès Guichet
- Service de génétique [Angers]
CHU AngersUniversité d'Angersrue Larrey, 49100 Angers,FR
| | - Olivier Guillin
- Génétique médicale et fonctionnelle du cancer et des maladies neuropsychiatriques
INSERM : U614Université de RouenUFR de Medecine et de Pharmacie 22, Boulevard Gambetta 76183 Rouen cedex,FR
| | - Anna-Liisa Hartikainen
- Department of Obstetrics and Gynecology
Institute of Clinical MedicineUniversity of Oulu90570 Oulu,FI
| | - Délphine Heron
- Département de Génétique Cytogénétique et Embryologie
Assistance publique - Hôpitaux de Paris (AP-HP)Hôpital Pitié-SalpêtrièreUniversité Paris VI - Pierre et Marie Curie47-83, boulevard de l'Hôpital 75651 PARIS Cedex 13,FR
| | | | - Muriel Holder
- Service de Génétique clinique
Hôpital Jeanne de FlandreCHRU Lille2 avenue Oscar Lambret, 59000 Lille,FR
| | - Georg Homuth
- Interfaculty Institute for Genetics and Functional Genomics
Ernst-Moritz-Arndt University GreifswaldD-17487 Greifswald,DE
| | - Bertrand Isidor
- Service d'ORL et de Chirurgie Cervicofaciale
INSERM : U587Hôpital d'Enfants Armand-TrousseauUniversité Pierre et Marie Curie - Paris 6Paris,FR
| | - Sylvie Jaillard
- IGDR, Institut de Génétique et Développement de Rennes
CNRS : UMR6061Université de Rennes 1IFR140Faculté de Médecine - CS 34317 2 Av du Professeur Léon Bernard 35043 RENNES CEDEX,FR
| | - Zdenek Jaros
- Abteilung für Kinder und Jugendheilkunde
Landesklinikum Waldviertel Zwettl3910 Zwettl,AT
| | - Susana Jiménez-Murcia
- IDIBELL, Department of Psychiatry
University Hospital of BellvitgeCIBERobn Fisiopatología de la Obesidad y Nutrición08907 Barcelona,ES
| | | | | | - Satu Kaksonen
- The Habilitation Unit of Folkhalsan
The Habilitation Unit of FolkhalsanFolkhalsan, SF 00250 Helsinki,FI
| | - Boris Keren
- Département de Génétique Cytogénétique et Embryologie
Assistance publique - Hôpitaux de Paris (AP-HP)Hôpital Pitié-SalpêtrièreUniversité Paris VI - Pierre et Marie Curie47-83, boulevard de l'Hôpital 75651 PARIS Cedex 13,FR
| | - Anita Kloss-Brandstätter
- Division of genetic epidemiology
Innsbruck Medical UniversityDepartment of Medical GeneticsMolecular and Clinical Pharmacology6020 Innsbruck,AT
| | - Nine V. A. M. Knoers
- Department of Medical Genetics
University Medical Center Utrecht3584 EA Utrecht,NL
| | - David A. Koolen
- Department of human genetics
Radboud University Nijmegen Medical CentreNijmegen Centre for Molecular Life SciencesInstitute for Genetic and Metabolic Disorders6500 HB Nijmegen,NL
| | | | - Florian Kronenberg
- Division of genetic epidemiology
Innsbruck Medical UniversityDepartment of Medical GeneticsMolecular and Clinical Pharmacology6020 Innsbruck,AT
| | - Audrey Labalme
- Service de cytogénétique constitutionnelle
Hospices Civils de LyonCHU de LyonCentre Neuroscience et Recherche69000 Lyon,FR
| | - Emilie Landais
- Service de Génétique
CHU ReimsHôpital Maison BlancheIFR 5351092 Reims,FR
| | - Elisabetta Lapi
- Medical Genetics Unit
Children's Hospital Anna Meyer50139 Firenze,IT
| | - Valérie Layet
- Unité de Cytogénétique et Génétique Médicale
Hôpital Gustave FlaubertGroupe Hospitalier du Havre76600 Le Havre,FR
| | - Solenn Legallic
- Génétique médicale et fonctionnelle du cancer et des maladies neuropsychiatriques
INSERM : U614Université de RouenUFR de Medecine et de Pharmacie 22, Boulevard Gambetta 76183 Rouen cedex,FR
| | - Bruno Leheup
- Service de médecine infantile III et génétique clinique
CHU NancyUniversité Henri Poincaré - Nancy IPRES de l'université de Lorraine54511 Vandoeuvre les Nancy,FR
| | - Barbara Leube
- Institute of Human Genetics and Anthropology
Heinrich-Heine University Hospital DuesseldorfD-40001 Duesseldorf,DE
| | - Suzanne Lewis
- Department of Medical Genetics
University of British ColumbiaChild and Family Research InstituteVancouver V6H 3N1,CA
| | - Josette Lucas
- IGDR, Institut de Génétique et Développement de Rennes
CNRS : UMR6061Université de Rennes 1IFR140Faculté de Médecine - CS 34317 2 Av du Professeur Léon Bernard 35043 RENNES CEDEX,FR
| | - Kay D. Macdermot
- North West Thames Regional Genetics Service
Northwick Park & St Marks HospitalHarrow HA1 3UJ,GB
| | - Pall Magnusson
- Child and Adolescent Psychiatry
Landspitali University HospitalIS-105 Reykjavík,IS
| | - Christian R. Marshall
- The Centre for Applied Genomics and Program in Genetics and Genomic Biology
The Hospital for Sick ChildrenToronto, Ontario, M5G 1L7,CA
| | | | - Mark I. Mccarthy
- OCDEM, Oxford Centre for Diabetes, Endocrinology and Metabolism
University of OxfordChurchill Hospital Oxford OX3 7LJ,GB
- Wellcome Trust Centre for Human Genetics
University of OxfordOxford,GB
| | - Thomas Meitinger
- Institute of Human Genetics
HelmholtzZentrum MünchenTechnische Universität München (TUM)German Research Center for Environmental Health85764 Neuherberg,DE
| | | | - Giuseppe Merla
- Medical Genetics Unit
IRCCS Casa Sollievo della Sofferenza Hospital71013 San Giovanni Rotondo,IT
| | - Alexandre Moerman
- Service de Génétique clinique
Hôpital Jeanne de FlandreCHRU Lille2 avenue Oscar Lambret, 59000 Lille,FR
| | - Vincent Mooser
- Genetics, GlaxoSmithKline R&D
GlaxoSmithKline720 Swedeland Road, King of Prussia, Pennsylvania 19406,US
| | - Fanny Morice-Picard
- MRGM, Maladies Rares - Génétique et Métabolisme
Hôpital PellegrinService de Génétique Médicale du CHU de BordeauxUniversité Victor Segalen - Bordeaux II : EA4576146 rue Léo-Saignat - 33076 Bordeaux Cedex,FR
- Service de génétique médicale
CHU BordeauxGroupe hospitalier PellegrinUniversité de BordeauxBordeaux,FR
| | - Mafalda Mucciolo
- Department of Biotechnology
Università degli studi di SienaMedical Genetics53100 Siena,IT
| | - Matthias Nauck
- Institute of Clinical Chemistry and Laboratory Medicine
Ernst-Moritz-Arndt University GreifswaldD-17475 Greifswald,DE
| | - Ndeye Coumba Ndiaye
- Génétique cardiovasculaire
Université Henri Poincaré - Nancy I : EA437354000 Nancy,FR
| | - Ann Nordgren
- Department of Molecular Medicine and Surgery
Karolinska InstitutetSE
| | - Laurent Pasquier
- IGDR, Institut de Génétique et Développement de Rennes
CNRS : UMR6061Université de Rennes 1IFR140Faculté de Médecine - CS 34317 2 Av du Professeur Léon Bernard 35043 RENNES CEDEX,FR
| | - Florence Petit
- Service de Génétique clinique
Hôpital Jeanne de FlandreCHRU Lille2 avenue Oscar Lambret, 59000 Lille,FR
| | - Rolph Pfundt
- Department of human genetics
Radboud University Nijmegen Medical CentreNijmegen Centre for Molecular Life SciencesInstitute for Genetic and Metabolic Disorders6500 HB Nijmegen,NL
| | - Ghislaine Plessis
- Service de génétique
CHU CaenHôpital ClémenceauAvenue Georges Clémenceau, Caen,FR
| | - Evica Rajcan-Separovic
- Department of Pathology
University of British ColumbiaChild and Family Research InstituteVancouver, British Columbia V5Z 4H4,CA
| | | | - Anita Rauch
- Institute of Medical Genetics
University of Zurich8603 Schwerzenbach,CH
| | - Roberto Ravazzolo
- Department of pediatrics and CEBR
University of GenovaG. Gaslini Institute16126 Genova,IT
| | - Andre Reis
- Institute of Human Genetics
Friedrich-Alexander University Erlangen-Nuremberg91054 Erlangen,DE
| | - Alessandra Renieri
- Department of Biotechnology
Università degli studi di SienaMedical Genetics53100 Siena,IT
| | - Cristobal Richart
- Department of Internal Medicine
University Hospital Juan XXIIIUniversitat Rovira y VirgiliCiber Fisiopatologia Obesidad y Nutricion (CIBEROBN)Instituto Salud Carlos III43005 Tarragona,ES
| | - Janina S. Ried
- Institute of Experimental Medicine
Academy of Sciences of the Czech RepublicVídeÅ�ská 1083 142 20 Prague,CZ
| | - Claudine Rieubland
- Division of Human Genetics
University of BernDepartment of Paediatrics, Inselspital3010 Bern,CH
| | - Wendy Roberts
- Autism Research Unit
The Hospital for Sick Children and Bloorview Kids RehabilitationUniversity of TorontoToronto, Ontario, M5G 1Z8,CA
| | | | - Caroline Rooryck
- MRGM, Maladies Rares - Génétique et Métabolisme
Hôpital PellegrinService de Génétique Médicale du CHU de BordeauxUniversité Victor Segalen - Bordeaux II : EA4576146 rue Léo-Saignat - 33076 Bordeaux Cedex,FR
- Service de génétique médicale
CHU BordeauxGroupe hospitalier PellegrinUniversité de BordeauxBordeaux,FR
| | - Massimiliano Rossi
- Service de cytogénétique constitutionnelle
Hospices Civils de LyonCHU de LyonCentre Neuroscience et Recherche69000 Lyon,FR
| | | | - Véronique Satre
- Département de génétique et procréation
CHU GrenobleUniversité Joseph Fourier - Grenoble Ifaculté de médecine-pharmacieDomaine de la Merci, 38706 Grenoble,FR
- AGIM, AGeing and IMagery, CNRS FRE3405
Université Joseph Fourier - Grenoble IEcole Pratique des Hautes EtudesCNRS : UMR5525Faculté de médecine de Grenoble, 38700 La Tronche,FR
| | - Claudia Schurmann
- Interfaculty Institute for Genetics and Functional Genomics
Ernst-Moritz-Arndt University GreifswaldD-17487 Greifswald,DE
| | - Engilbert Sigurdsson
- University of Iceland
University of IcelandDepartment of Electrical and Computer Engineering, University of Iceland, Hjardarhaga 2-6, 107 Reykjavik, Iceland;,IS
| | - Dimitri J. Stavropoulos
- Department of Pediatric Laboratory Medicine
Hospital for Sick ChildrenToronto, Ontario M5G 1X8,CA
| | | | - Carola Tengström
- Genetic Services
Rinnekoti Research FoundationKumputie 1, SF-02980 Espoo,FI
| | | | - Francisco J. Tinahones
- Department of Endocrinology and Nutrition
Clinic Hospital of Virgen de la VictoriaCiber Fisiopatologia y Nutricion (CIBEROBN)Instituto Salud Carlos III29010 Malaga,ES
| | - Renaud Touraine
- Service de génétique
CHU Saint-EtienneHôpital nord42055 St Etienne,FR
| | - Louis Vallée
- Service de Neuropédiatrie
CHRU LilleHôpital Roger Salengro59037 Lille,FR
| | - Ellen Van Binsbergen
- Department of Medical Genetics
University Medical Center Utrecht3584 EA Utrecht,NL
| | | | - Catherine Vincent-Delorme
- Centre de Maladies Rares
Anomalies du Développement Nord de FranceCH Arras - CHRU Lille59000 Arras,FR
| | - Sophie Visvikis-Siest
- Génétique cardiovasculaire
Université Henri Poincaré - Nancy I : EA437354000 Nancy,FR
| | - Peter Vollenweider
- Department of Internal Medicine
Centre Hospitalier Universitaire Vaudois1011 Lausanne,CH
| | - Henry Völzke
- Institute for Community Medicine
Ernst-Moritz-Arndt University GreifswaldD-17475 Greifswald,DE
| | - Anneke T. Vulto-Van Silfhout
- Department of human genetics
Radboud University Nijmegen Medical CentreNijmegen Centre for Molecular Life SciencesInstitute for Genetic and Metabolic Disorders6500 HB Nijmegen,NL
| | - Gérard Waeber
- Department of Internal Medicine
Centre Hospitalier Universitaire Vaudois1011 Lausanne,CH
| | - Carina Wallgren-Pettersson
- Department of Medical Genetics
University of HelsinskiFolkhälsan Insitute of GeneticsHaartman Institute00251 Helsinki,FI
| | | | - Simon Zwolinksi
- Institute of human genetics
International Centre for LifeNewcastle Upon Tyne NE1 4EP,GB
| | - Joris Andrieux
- Laboratoire de Génétique Médicale
Hôpital Jeanne de FlandreCHRU Lille59037 Lille Cedex,FR
| | - Xavier Estivill
- CRG-UPF, Center for Genomic Regulation
CIBER de Epidemiología y Salud Pública (CIBERESP)C/ Dr. Aiguader, 88 08003 Barcelona, Catalonia, Spain,ES
| | - James F. Gusella
- Center for Human Genetic Research
Massachusetts General HospitalBoston, Massachusetts 02114,US
| | | | - Andres Metspalu
- Estonian Genome and Medicine
University of Tartu51010 Tartu,EE
- Institute of Molecular and Cell Biology
University of Tartu51010 Tartu,EE
| | - Stephen W. Scherer
- The Centre for Applied Genomics
The Hospital for Sick ChildrenMcLaughlin CentreDepartment of Molecular GeneticsUniversity of TorontoToronto, Ontario, Canada M5G 1L7,CA
| | | | - Alexandra I. F. Blakemore
- Department of Genomics of Common Disease
Imperial College LondonHammersmith hospital, London W12 0NN,GB
| | - Jacques S. Beckmann
- Service de génétique médicale
CHU Vaudois1011 Lausanne,CH
- Department of Medical Genetics
University of LausanneCH
| | - Philippe Froguel
- Department of Genomics of Common Disease
Imperial College LondonHammersmith hospital, London W12 0NN,GB
- IBLI, Institut de biologie de Lille - IBL
Institut Pasteur de LilleCNRS : UMR8090Université Lille I - Sciences et technologiesUniversité Lille II - Droit et santéInstitut de Biologie de Lille 1 Rue du Professeur Calmette - 447 59021 LILLE CEDEX,FR
| |
Collapse
|
26
|
Liu Y, Lee YF, Ng MK. SNP and gene networks construction and analysis from classification of copy number variations data. BMC Bioinformatics 2011; 12 Suppl 5:S4. [PMID: 21989070 PMCID: PMC3226254 DOI: 10.1186/1471-2105-12-s5-s4] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/01/2023] Open
Abstract
BACKGROUND Detection of genomic DNA copy number variations (CNVs) can provide a complete and more comprehensive view of human disease. It is interesting to identify and represent relevant CNVs from a genome-wide data due to high data volume and the complexity of interactions. RESULTS In this paper, we incorporate the DNA copy number variation data derived from SNP arrays into a computational shrunken model and formalize the detection of copy number variations as a case-control classification problem. More than 80% accuracy can be obtained using our classification model and by shrinkage, the number of relevant CNVs to disease can be determined. In order to understand relevant CNVs, we study their corresponding SNPs in the genome and a statistical software PLINK is employed to compute the pair-wise SNP-SNP interactions, and identify SNP networks based on their P-values. Our selected SNP networks are statistically significant compared with random SNP networks and play a role in the biological process. For the unique genes that those SNPs are located in, a gene-gene similarity value is computed using GOSemSim and gene pairs that have similarity values being greater than a threshold are selected to construct gene networks. A gene enrichment analysis show that our gene networks are functionally important.Experimental results demonstrate that our selected SNP and gene networks based on the selected CNVs contain some functional relationships directly or indirectly to disease study. CONCLUSIONS Two datasets are given to demonstrate the effectiveness of the introduced method. Some statistical and biological analysis show that this shrunken classification model is effective in identifying CNVs from genome-wide data and our proposed framework has a potential to become a useful analysis tool for SNP data sets.
Collapse
Affiliation(s)
- Yang Liu
- Department of Mathematics, Hong Kong Baptist University, Kowloon Tong, Hong Kong
| | | | | |
Collapse
|
27
|
Wang HY, Greenawalt D, Cui X, Tereshchenko IV, Luo M, Yang Q, Azaro MA, Hu G, Chu Y, Li JY, Shen L, Lin Y, Zhang L, Li H. Identification of possible genetic alterations in the breast cancer cell line MCF-7 using high-density SNP genotyping microarray. J Carcinog 2011; 8:6. [PMID: 19439911 PMCID: PMC2687141 DOI: 10.4103/1477-3163.50886] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/24/2023] Open
Abstract
Context: Cancer cell lines are used extensively in various research. Knowledge of genetic alterations in these lines is important for understanding mechanisms underlying their biology. However, since paired normal tissues are usually unavailable for comparison, precisely determining genetic alterations in cancer cell lines is difficult. To address this issue, a highly efficient and reliable method is developed. Aims: Establishing a highly efficient and reliable experimental system for genetic profiling of cell lines. Materials and Methods: A widely used breast cancer cell line, MCF-7, was genetically profiled with 4,396 single nucleotide polymorphisms (SNPs) spanning 11 whole chromosomes and two other small regions using a newly developed high-throughput multiplex genotyping approach. Results: The fractions of homozygous SNPs in MCF-7 (13.3%) were significantly lower than those in the control cell line and in 24 normal human individuals (25.1% and 27.4%, respectively). Homozygous SNPs in MCF-7 were found in clusters. The sizes of these clusters were significantly larger than the expected based on random allelic combination. Fourteen such regions were found on chromosomes 1p, 1q, 2q, 6q, 13, 15q, 16q, 17q and 18p in MCF-7 and two in the small regions. Conclusions: These results are generally concordant with those obtained using different approaches but are better in defining their chromosomal positions. The used approach provides a reliable way to detecting possible genetic alterations in cancer cell lines without paired normal tissues.
Collapse
Affiliation(s)
- Hui-Yun Wang
- Department of Molecular Genetics, Microbiology and Immunology/The Cancer Institute of New Jersey, Piscataway, New Jersey, 08854, USA
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
28
|
Mao X, Young BD, Lu YJ. The application of single nucleotide polymorphism microarrays in cancer research. Curr Genomics 2011; 8:219-28. [PMID: 18645599 DOI: 10.2174/138920207781386924] [Citation(s) in RCA: 48] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2006] [Revised: 01/22/2007] [Accepted: 03/02/2007] [Indexed: 01/21/2023] Open
Abstract
The development of microarray technology has had a significant impact on the genetic analysis of human disease. The recently developed single nucleotide polymorphism (SNP) array can be used to measure both DNA polymorphism and dosage changes. Our laboratory has applied SNP microarray analysis to uncover frequent uniparental disomies and sub-microscopic genomic copy number gains and losses in different cancers. This review will focus on the wide range of applications of SNP microarray analysis to cancer research. SNP array genotyping can determine loss of heterozygosity, genomic copy number changes and DNA methylation alterations of cancer cells. The same technology can also be used to investigate allelic association in cancers. Therefore, it can be applied to the identification of cancer predisposition genes, oncogenes and tumor suppressor genes in specific types of tumors. As a consequence, they have potential in cancer risk assessment, diagnosis, prognosis and treatment selection.
Collapse
Affiliation(s)
- Xueying Mao
- Medical Oncology Centre, Cancer Institute, Barts and London School of Medicine and Dentistry, Queen Mary, University of London, Charterhouse Square, London, UK
| | | | | |
Collapse
|
29
|
Abstract
Acute myeloid leukemia (AML) can develop as a secondary malignancy following radiotherapy, but also following low-dose environmental or occupational radiation exposure. Therapy-related AML frequently carries deletions of chromosome 5q and/or 7, but for low-dose exposure associated AML this has not been described. For the present study we performed genome-wide screens for loss-of-heterozygosity (LOH) in a set of 19 AML cases that developed after radiation-exposure following the Chernobyl accident. Using Affymetrix SNP arrays we found large regions of LOH in 16 of the cases. Eight cases (42%) demonstrated LOH at 5q and/or 7, which is a known marker of complex karyotypic changes and poor prognosis. We could show here for the first time that exposure to low-dose ionizing radiation induces AML with molecular alterations similar to those seen in therapy-related cases.
Collapse
|
30
|
Pinto D, Darvishi K, Shi X, Rajan D, Rigler D, Fitzgerald T, Lionel AC, Thiruvahindrapuram B, Macdonald JR, Mills R, Prasad A, Noonan K, Gribble S, Prigmore E, Donahoe PK, Smith RS, Park JH, Hurles ME, Carter NP, Lee C, Scherer SW, Feuk L. Comprehensive assessment of array-based platforms and calling algorithms for detection of copy number variants. Nat Biotechnol 2011; 29:512-20. [PMID: 21552272 PMCID: PMC3270583 DOI: 10.1038/nbt.1852] [Citation(s) in RCA: 325] [Impact Index Per Article: 25.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2010] [Accepted: 03/22/2011] [Indexed: 11/09/2022]
Abstract
We have systematically compared copy number variant (CNV) detection on eleven microarrays to evaluate data quality and CNV calling, reproducibility, concordance across array platforms and laboratory sites, breakpoint accuracy and analysis tool variability. Different analytic tools applied to the same raw data typically yield CNV calls with <50% concordance. Moreover, reproducibility in replicate experiments is <70% for most platforms. Nevertheless, these findings should not preclude detection of large CNVs for clinical diagnostic purposes because large CNVs with poor reproducibility are found primarily in complex genomic regions and would typically be removed by standard clinical data curation. The striking differences between CNV calls from different platforms and analytic tools highlight the importance of careful assessment of experimental design in discovery and association studies and of strict data curation and filtering in diagnostics. The CNV resource presented here allows independent data evaluation and provides a means to benchmark new algorithms.
Collapse
Affiliation(s)
- Dalila Pinto
- The Centre for Applied Genomics, The Hospital for Sick Children, Toronto, Ontario, Canada
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
31
|
SNP array-based karyotyping: differences and similarities between aplastic anemia and hypocellular myelodysplastic syndromes. Blood 2011; 117:6876-84. [PMID: 21527527 DOI: 10.1182/blood-2010-11-314393] [Citation(s) in RCA: 102] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
In aplastic anemia (AA), contraction of the stem cell pool may result in oligoclonality, while in myelodysplastic syndromes (MDS) a single hematopoietic clone often characterized by chromosomal aberrations expands and outcompetes normal stem cells. We analyzed patients with AA (N = 93) and hypocellular MDS (hMDS, N = 24) using single nucleotide polymorphism arrays (SNP-A) complementing routine cytogenetics. We hypothesized that clinically important cryptic clonal aberrations may exist in some patients with BM failure. Combined metaphase and SNP-A karyotyping improved detection of chromosomal lesions: 19% and 54% of AA and hMDS cases harbored clonal abnormalities including copy-neutral loss of heterozygosity (UPD, 7%). Remarkably, lesions involving the HLA locus suggestive of clonal immune escape were found in 3 of 93 patients with AA. In hMDS, additional clonal lesions were detected in 5 (36%) of 14 patients with normal/noninformative routine cytogenetics. In a subset of AA patients studied at presentation, persistent chromosomal genomic lesions were found in 10 of 33, suggesting that the initial diagnosis may have been hMDS. Similarly, using SNP-A, earlier clonal evolution was found in 4 of 7 AA patients followed serially. In sum, our results indicate that SNP-A identify cryptic clonal genomic aberrations in AA and hMDS leading to improved distinction of these disease entities.
Collapse
|
32
|
Yang HC, Lin HC, Kang M, Chen CH, Lin CW, Li LH, Wu JY, Chen YT, Pan WH. SAQC: SNP array quality control. BMC Bioinformatics 2011; 12:100. [PMID: 21501472 PMCID: PMC3101186 DOI: 10.1186/1471-2105-12-100] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2010] [Accepted: 04/18/2011] [Indexed: 12/21/2022] Open
Abstract
BACKGROUND Genome-wide single-nucleotide polymorphism (SNP) arrays containing hundreds of thousands of SNPs from the human genome have proven useful for studying important human genome questions. Data quality of SNP arrays plays a key role in the accuracy and precision of downstream data analyses. However, good indices for assessing data quality of SNP arrays have not yet been developed. RESULTS We developed new quality indices to measure the quality of SNP arrays and/or DNA samples and investigated their statistical properties. The indices quantify a departure of estimated individual-level allele frequencies (AFs) from expected frequencies via standardized distances. The proposed quality indices followed lognormal distributions in several large genomic studies that we empirically evaluated. AF reference data and quality index reference data for different SNP array platforms were established based on samples from various reference populations. Furthermore, a confidence interval method based on the underlying empirical distributions of quality indices was developed to identify poor-quality SNP arrays and/or DNA samples. Analyses of authentic biological data and simulated data show that this new method is sensitive and specific for the detection of poor-quality SNP arrays and/or DNA samples. CONCLUSIONS This study introduces new quality indices, establishes references for AFs and quality indices, and develops a detection method for poor-quality SNP arrays and/or DNA samples. We have developed a new computer program that utilizes these methods called SNP Array Quality Control (SAQC). SAQC software is written in R and R-GUI and was developed as a user-friendly tool for the visualization and evaluation of data quality of genome-wide SNP arrays. The program is available online (http://www.stat.sinica.edu.tw/hsinchou/genetics/quality/SAQC.htm).
Collapse
Affiliation(s)
- Hsin-Chou Yang
- Institute of Statistical Science, Academia Sinica, Taipei, Taiwan.
| | | | | | | | | | | | | | | | | |
Collapse
|
33
|
Meadows KL, Andrews DMK, Xu Z, Carswell GK, Laughlin SK, Baird DD, Taylor JA. Genome-wide analysis of loss of heterozygosity and copy number amplification in uterine leiomyomas using the 100K single nucleotide polymorphism array. Exp Mol Pathol 2011; 91:434-9. [PMID: 21497600 DOI: 10.1016/j.yexmp.2011.03.007] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2011] [Accepted: 03/29/2011] [Indexed: 10/18/2022]
Abstract
PURPOSE Uterine leiomyomas (fibroids) are benign smooth muscle tumors commonly found among reproductive-aged women. Though benign, these tumors are the leading indication for hysterectomies in the United States and cause significant morbidity. Despite the importance of this tumor in women's health, relatively little is known about the molecular etiology. METHODS In this study, we used the Affymetrix 100K single nucleotide polymorphism (SNP) chip to assess whether the pattern and frequency of genome-wide loss of heterozygosity (LOH) and copy number amplifications is associated with clinical heterogeneity. RESULTS Thirty-seven tumors with varying sizes and histology from eleven patients were analyzed. LOH was observed in 4/37 tumors (10.8%) and significantly associated with large-sized tumors (p<0.0014). Two tumors revealed hemizygosity on chromosome 7q, a region that has been consistently reported to have LOH. Additionally, we detected one novel region of LOH, 16p13.11 in one tumor (2.7%). Copy number amplifications were observed on all chromosomes; however, most were low-level amplifications and only detected in a single tumor. One region of amplification at 3p26.3 was detected in four tumors. CONCLUSIONS Despite the use of a high-density SNP platform, our results suggest that genome-wide LOH and copy number amplifications are infrequent events and generally do not determine clinical and histologic characteristics of this disease.
Collapse
Affiliation(s)
- Kellen L Meadows
- Laboratory of Molecular Carcinogenesis, National Institute of Environmental Health Sciences, Research Triangle Park, NC 27709, USA
| | | | | | | | | | | | | |
Collapse
|
34
|
Zhang D, Qian Y, Akula N, Alliey-Rodriguez N, Tang J, Gershon ES, Liu C. Accuracy of CNV Detection from GWAS Data. PLoS One 2011; 6:e14511. [PMID: 21249187 PMCID: PMC3020939 DOI: 10.1371/journal.pone.0014511] [Citation(s) in RCA: 57] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2010] [Accepted: 12/19/2010] [Indexed: 12/01/2022] Open
Abstract
Several computer programs are available for detecting copy number variants (CNVs) using genome-wide SNP arrays. We evaluated the performance of four CNV detection software suites—Birdsuite, Partek, HelixTree, and PennCNV-Affy—in the identification of both rare and common CNVs. Each program's performance was assessed in two ways. The first was its recovery rate, i.e., its ability to call 893 CNVs previously identified in eight HapMap samples by paired-end sequencing of whole-genome fosmid clones, and 51,440 CNVs identified by array Comparative Genome Hybridization (aCGH) followed by validation procedures, in 90 HapMap CEU samples. The second evaluation was program performance calling rare and common CNVs in the Bipolar Genome Study (BiGS) data set (1001 bipolar cases and 1033 controls, all of European ancestry) as measured by the Affymetrix SNP 6.0 array. Accuracy in calling rare CNVs was assessed by positive predictive value, based on the proportion of rare CNVs validated by quantitative real-time PCR (qPCR), while accuracy in calling common CNVs was assessed by false positive/false negative rates based on qPCR validation results from a subset of common CNVs. Birdsuite recovered the highest percentages of known HapMap CNVs containing >20 markers in two reference CNV datasets. The recovery rate increased with decreased CNV frequency. In the tested rare CNV data, Birdsuite and Partek had higher positive predictive values than the other software suites. In a test of three common CNVs in the BiGS dataset, Birdsuite's call was 98.8% consistent with qPCR quantification in one CNV region, but the other two regions showed an unacceptable degree of accuracy. We found relatively poor consistency between the two “gold standards,” the sequence data of Kidd et al., and aCGH data of Conrad et al. Algorithms for calling CNVs especially common ones need substantial improvement, and a “gold standard” for detection of CNVs remains to be established.
Collapse
Affiliation(s)
- Dandan Zhang
- Department of Pathology, School of Medicine, Zhejiang University, Hangzhou, People's Republic of China
- Department of Psychiatry and Behavioral Neuroscience, The University of Chicago, Chicago, Illinois, United States of America
| | - Yudong Qian
- Department of Psychiatry and Behavioral Neuroscience, The University of Chicago, Chicago, Illinois, United States of America
| | - Nirmala Akula
- Intramural Research Programs, The National Institute of Mental Health, Bethesda, Maryland, United States of America
| | - Ney Alliey-Rodriguez
- Department of Psychiatry and Behavioral Neuroscience, The University of Chicago, Chicago, Illinois, United States of America
| | - Jinsong Tang
- Institute of Mental Health, The Second Xiangya Hospital, Central South University, Changsha, People's Republic of China
| | | | - Elliot S. Gershon
- Department of Psychiatry and Behavioral Neuroscience, The University of Chicago, Chicago, Illinois, United States of America
- * E-mail: (CL); (ESG)
| | - Chunyu Liu
- Department of Psychiatry and Behavioral Neuroscience, The University of Chicago, Chicago, Illinois, United States of America
- * E-mail: (CL); (ESG)
| |
Collapse
|
35
|
de Leeuw N, Hehir-Kwa JY, Simons A, Geurts van Kessel A, Smeets DF, Faas BHW, Pfundt R. SNP Array Analysis in Constitutional and Cancer Genome Diagnostics – Copy Number Variants, Genotyping and Quality Control. Cytogenet Genome Res 2011; 135:212-21. [PMID: 21934286 DOI: 10.1159/000331273] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022] Open
Affiliation(s)
- N de Leeuw
- Department of Human Genetics, Radboud University Nijmegen Medical Centre, Nijmegen, The Netherlands.
| | | | | | | | | | | | | |
Collapse
|
36
|
Tsukasa K, Nobuharu Y, Takeshi O, Hiroki B, Takashi Y, Akira K, Nobuo T, Takahiko S. Analysis of copy number abnormality (CNA) and loss of heterozygosity (LOH) in the whole genome using single nucleotide polymorphism (SNP) genotyping arrays in tongue squamous cell carcinoma. J Korean Assoc Oral Maxillofac Surg 2011. [DOI: 10.5125/jkaoms.2011.37.6.550] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2022] Open
Affiliation(s)
- Kuroiwa Tsukasa
- Department of Oral and Maxillofacial Surgery, Tokyo Dental College, Japan
| | - Yamamoto Nobuharu
- Department of Oral and Maxillofacial Surgery, Tokyo Dental College, Japan
| | - Onda Takeshi
- Department of Oral and Maxillofacial Surgery, Tokyo Dental College, Japan
| | - Bessyo Hiroki
- Department of Oral and Maxillofacial Surgery, Tokyo Dental College, Japan
| | - Yakushiji Takashi
- Department of Oral and Maxillofacial Surgery, Tokyo Dental College, Japan
| | - Katakura Akira
- Department of Oral and Maxillofacial Surgery, Tokyo Dental College, Japan
| | - Takano Nobuo
- Department of Oral and Maxillofacial Surgery, Tokyo Dental College, Japan
| | - Shibahara Takahiko
- Department of Oral and Maxillofacial Surgery, Tokyo Dental College, Japan
| |
Collapse
|
37
|
Mullighan CG. Single nucleotide polymorphism microarray analysis of genetic alterations in cancer. Methods Mol Biol 2011; 730:235-58. [PMID: 21431646 DOI: 10.1007/978-1-61779-074-4_17] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023]
Abstract
The identification of structural genetic alterations, including DNA amplifications, deletions, and loss of heterozygosity (LOH), using single nucleotide polymorphism (SNP) microarrays has provided important insights into the pathogenesis of a number of hematologic malignancies. Currently available SNP arrays comprise over a million SNP and copy number oligonucleotide probes that interrogate the genome at sub-kilobase resolution. The accurate detection of DNA copy number abnormalities and LOH is critically dependent on the use of high-quality DNA, the use of matched reference samples wherever possible, optimal normalization of raw microarray data, and computational algorithms to detect copy number alterations sensitively and robustly. This chapter provides methods and guidelines for preparing samples, processing and analyzing data, and validation of novel lesions. Specific examples are provided for Affymetrix SNP arrays in acute lymphoblastic leukemia.
Collapse
Affiliation(s)
- Charles G Mullighan
- Department of Pathology, St. Jude Children's Research Hospital, Memphis, TN, USA.
| |
Collapse
|
38
|
Guo B, Villagran A, Vannucci M, Wang J, Davis C, Man TK, Lau C, Guerra R. Bayesian estimation of genomic copy number with single nucleotide polymorphism genotyping arrays. BMC Res Notes 2010; 3:350. [PMID: 21192799 PMCID: PMC3023756 DOI: 10.1186/1756-0500-3-350] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2010] [Accepted: 12/30/2010] [Indexed: 11/19/2022] Open
Abstract
Background The identification of copy number aberration in the human genome is an important area in cancer research. We develop a model for determining genomic copy numbers using high-density single nucleotide polymorphism genotyping microarrays. The method is based on a Bayesian spatial normal mixture model with an unknown number of components corresponding to true copy numbers. A reversible jump Markov chain Monte Carlo algorithm is used to implement the model and perform posterior inference. Results The performance of the algorithm is examined on both simulated and real cancer data, and it is compared with the popular CNAG algorithm for copy number detection. Conclusions We demonstrate that our Bayesian mixture model performs at least as well as the hidden Markov model based CNAG algorithm and in certain cases does better. One of the added advantages of our method is the flexibility of modeling normal cell contamination in tumor samples.
Collapse
Affiliation(s)
- Beibei Guo
- Department of Statistics, Rice University, 6100 Main, Houston, TX 77005-1827, USA.
| | | | | | | | | | | | | | | |
Collapse
|
39
|
Hu N, Clifford RJ, Yang HH, Wang C, Goldstein AM, Ding T, Taylor PR, Lee MP. Genome wide analysis of DNA copy number neutral loss of heterozygosity (CNNLOH) and its relation to gene expression in esophageal squamous cell carcinoma. BMC Genomics 2010; 11:576. [PMID: 20955586 PMCID: PMC3091724 DOI: 10.1186/1471-2164-11-576] [Citation(s) in RCA: 137] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2010] [Accepted: 10/18/2010] [Indexed: 12/14/2022] Open
Abstract
Background Genomic instability plays an important role in human cancers. We previously characterized genomic instability in esophageal squamous cell carcinomas (ESCC) in terms of loss of heterozygosity (LOH) and copy number (CN) changes in tumors using the Affymetrix GeneChip Human Mapping 500K array in 30 cases from a high-risk region of China. In the current study we focused on copy number neutral (CN = 2) LOH (CNNLOH) and its relation to gene expression in ESCC. Results Overall we found that 70% of all LOH observed was CNNLOH. Ninety percent of ESCCs showed CNNLOH (median frequency in cases = 60%) and this was the most common type of LOH in two-thirds of cases. CNNLOH occurred on all 39 autosomal chromosome arms, with highest frequencies on 19p (100%), 5p (96%), 2p (95%), and 20q (95%). In contrast, LOH with CN loss represented 19% of all LOH, occurred in just half of ESCCs (median frequency in cases = 0%), and was most frequent on 3p (56%), 5q (47%), and 21q (41%). LOH with CN gain was 11% of all LOH, occurred in 93% of ESCCs (median frequency in cases = 13%), and was most common on 20p (82%), 8q (74%), and 3q (42%). To examine the effect of genomic instability on gene expression, we evaluated RNA profiles from 17 pairs of matched normal and tumor samples (a subset of the 30 ESCCs) using Affymetrix U133A 2.0 arrays. In CN neutral regions, expression of 168 genes (containing 1976 SNPs) differed significantly in tumors with LOH versus tumors without LOH, including 101 genes that were up-regulated and 67 that were down-regulated. Conclusion Our results indicate that CNNLOH has a profound impact on gene expression in ESCC, which in turn may affect tumor development.
Collapse
Affiliation(s)
- Nan Hu
- Genetic Epidemiology Branch, Division of Cancer Epidemiology and Genetics, National Cancer Institute, NIH, DHHS, Bethesda, Maryland, USA
| | | | | | | | | | | | | | | |
Collapse
|
40
|
Onvani S, Etame AB, Smith CA, Rutka JT. Genetics of medulloblastoma: clues for novel therapies. Expert Rev Neurother 2010; 10:811-23. [PMID: 20420498 DOI: 10.1586/ern.10.31] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
Medulloblastoma is the most common malignant brain tumor in children. Current medulloblastoma therapy entails surgery, radiation and chemotherapy. The 5-year survival rate for patients ranges from 40 to 70%, with most survivors suffering from serious long-term treatment-related sequelae. Additional research on the molecular biology and genetics of medulloblastoma is needed to identify robust prognostic markers for disease-risk stratification, to improve current treatment regimes and to discover novel and more effective molecular-targeted therapies. Recent advances in molecular biology have led to the development of powerful tools for the study of medulloblastoma tumorigenesis, which have revealed new insights into the molecular underpinnings of this disease. Here we discuss the signaling pathway alterations implicated in medulloblastoma pathogenesis, the techniques used in molecular profiling of these tumors and recent molecular subclassification schemes. Particular emphasis is given to the identification of novel molecular targets for less toxic, patient-tailored therapeutic approaches.
Collapse
Affiliation(s)
- Sara Onvani
- The Hospital for Sick Children, Ontario, Canada
| | | | | | | |
Collapse
|
41
|
Christensen BC, Houseman EA, Poage GM, Godleski JJ, Bueno R, Sugarbaker DJ, Wiencke JK, Nelson HH, Marsit CJ, Kelsey KT. Integrated profiling reveals a global correlation between epigenetic and genetic alterations in mesothelioma. Cancer Res 2010; 70:5686-94. [PMID: 20587528 DOI: 10.1158/0008-5472.can-10-0190] [Citation(s) in RCA: 45] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Development of mesothelioma is linked mainly to asbestos exposure, but the combined contributions of genetic and epigenetic alterations are unclear. We investigated the potential relationships between gene copy number (CN) alterations and DNA methylation profiles in a case series of pleural mesotheliomas (n = 23). There were no instances of significantly correlated CN alteration and methylation at probed loci, whereas averaging loci over their associated genes revealed only two genes with significantly correlated CN and methylation alterations. In contrast to the lack of discrete correlations, the overall extent of tumor CN alteration was significantly associated with DNA methylation profile when comparing CN alteration extent among methylation profile classes. Further, there was evidence that this association was partially attributable to prevalent allele loss at the DNA methyltransferase gene DNMT1. Our findings define a strong association between global genetic and global epigenetic dysregulation in mesothelioma, rather than a discrete, local coordination of gene inactivation.
Collapse
Affiliation(s)
- Brock C Christensen
- Departments of Pathology and Laboratory Medicine, Community Health, Center for Environmental Health and Technology, and Molecular Pharmacology and Physiology, Brown University, Providence, Rhode Island 02903, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
42
|
Rancoita PMV, Hutter M, Bertoni F, Kwee I. An integrated Bayesian analysis of LOH and copy number data. BMC Bioinformatics 2010; 11:321. [PMID: 20550648 PMCID: PMC2912301 DOI: 10.1186/1471-2105-11-321] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2009] [Accepted: 06/15/2010] [Indexed: 12/17/2022] Open
Abstract
Background Cancer and other disorders are due to genomic lesions. SNP-microarrays are able to measure simultaneously both genotype and copy number (CN) at several Single Nucleotide Polymorphisms (SNPs) along the genome. CN is defined as the number of DNA copies, and the normal is two, since we have two copies of each chromosome. The genotype of a SNP is the status given by the nucleotides (alleles) which are present on the two copies of DNA. It is defined homozygous or heterozygous if the two alleles are the same or if they differ, respectively. Loss of heterozygosity (LOH) is the loss of the heterozygous status due to genomic events. Combining CN and LOH data, it is possible to better identify different types of genomic aberrations. For example, a long sequence of homozygous SNPs might be caused by either the physical loss of one copy or a uniparental disomy event (UPD), i.e. each SNP has two identical nucleotides both derived from only one parent. In this situation, the knowledge of the CN can help in distinguishing between these two events. Results To better identify genomic aberrations, we propose a method (called gBPCR) which infers the type of aberration occurred, taking into account all the possible influence in the microarray detection of the homozygosity status of the SNPs, resulting from an altered CN level. Namely, we model the distributions of the detected genotype, given a specific genomic alteration and we estimate the parameters involved on public reference datasets. The estimation is performed similarly to the modified Bayesian Piecewise Constant Regression, but with improved estimators for the detection of the breakpoints. Using artificial and real data, we evaluate the quality of the estimation of gBPCR and we also show that it outperforms other well-known methods for LOH estimation. Conclusions We propose a method (gBPCR) for the estimation of both LOH and CN aberrations, improving their estimation by integrating both types of data and accounting for their relationships. Moreover, gBPCR performed very well in comparison with other methods for LOH estimation and the estimated CN lesions on real data have been validated with another technique.
Collapse
Affiliation(s)
- Paola M V Rancoita
- Istituto Dalle Molle di Studi sull'Intelligenza Artificiale, Manno-Lugano, Switzerland.
| | | | | | | |
Collapse
|
43
|
Sui Y, Zhao X, Speed TP, Wu Z. Background adjustment for DNA microarrays using a database of microarray experiments. J Comput Biol 2010; 16:1501-15. [PMID: 19958080 DOI: 10.1089/cmb.2009.0063] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
DNA microarrays have become an indispensable technique in biomedical research. The raw measurements from microarrays undergo a number of preprocessing steps before the data are converted to the genomic level for further analysis. Background adjustment is an important step in preprocessing. Estimating background noise has been challenging because background levels vary a lot from probe to probe, yet there are limited observations on each probe. Most current methods have used the empirical Bayes approach to borrow information across probes on the same array. These approaches shrink the background estimate for either the entire sample or probes sharing similar sequence structures. In this article, we present a solution that is truly probe specific by using a database of large number of microarray experiments. Information is borrowed across samples and background noise is estimated for each probe individually. The ability to obtain probe specific background distributions allows us to extend the dynamic range of gene expression levels. We illustrate the improvement in detecting gene expression variation on two datasets: a Latin Square spike-in experiment from Affymetrix and an Estrogen Receptor experiment with biological replicates. An R package dbRMA implementing our method can be obtained from the authors.
Collapse
Affiliation(s)
- Yunxia Sui
- Department of Community Health, Brown University, Providence, Rhode Island 02912, USA
| | | | | | | |
Collapse
|
44
|
Poage GM, Christensen BC, Houseman EA, McClean MD, Wiencke JK, Posner MR, Clark JR, Nelson HH, Marsit CJ, Kelsey KT. Genetic and epigenetic somatic alterations in head and neck squamous cell carcinomas are globally coordinated but not locally targeted. PLoS One 2010; 5:e9651. [PMID: 20300172 PMCID: PMC2836370 DOI: 10.1371/journal.pone.0009651] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2010] [Accepted: 02/17/2010] [Indexed: 01/22/2023] Open
Abstract
Background Solid tumors, including head and neck squamous cell carcinomas (HNSCC), arise as a result of genetic and epigenetic alterations in a sustained stress environment. Little work has been done that simultaneously examines the spectrum of both types of changes in human tumors on a genome-wide scale and results so far have been limited and mixed. Since it has been hypothesized that epigenetic alterations may act by providing the second carcinogenic hit in gene silencing, we sought to identify genome-wide DNA copy number alterations and CpG dinucleotide methylation events and examine the global/local relationships between these types of alterations in HNSCC. Methodology/Principal Findings We have extended a prior analysis of 1,413 cancer-associated loci for epigenetic changes in HNSCC by integrating DNA copy number alterations, measured at 500,000 polymorphic loci, in a case series of 19 primary HNSCC tumors. We have previously demonstrated that local copy number does not bias methylation measurements in this array platform. Importantly, we found that the global pattern of copy number alterations in these tumors was significantly associated with tumor methylation profiles (p<0.002). However at the local level, gene promoter regions did not exhibit a correlation between copy number and methylation (lowest q = 0.3), and the spectrum of genes affected by each type of alteration was unique. Conclusion/Significance This work, using a novel and robust statistical approach demonstrates that, although a “second hit” mechanism is not likely the predominant mode of action for epigenetic dysregulation in cancer, the patterns of methylation events are associated with the patterns of allele loss. Our work further highlights the utility of integrative genomics approaches in exploring the driving somatic alterations in solid tumors.
Collapse
Affiliation(s)
- Graham M Poage
- Departments of Molecular Pharmacology and Physiology, Brown University, Providence, Rhode Island, United States of America
| | | | | | | | | | | | | | | | | | | |
Collapse
|
45
|
Walters RG, Jacquemont S, Valsesia A, de Smith AJ, Martinet D, Andersson J, Falchi M, Chen F, Andrieux J, Lobbens S, Delobel B, Stutzmann F, Moustafa JSES, Chèvre JC, Lecoeur C, Vatin V, Bouquillon S, Buxton JL, Boute O, Holder-Espinasse M, Cuisset JM, Lemaitre MP, Ambresin AE, Brioshi A, Gaillard M, Giusti V, Fellmann F, Ferrarini A, Hadjikhani N, Campion D, Guilmatre A, Goldenberg A, Calmels N, Mandel JL, Le Caignec C, David A, Isidor B, Cordier MP, Dupuis-Girod S, Labalme A, Sanlaville D, Béri-Deixheimer M, Jonveaux P, Leheup B, Õunap K, Bochukova EG, Henning E, Keogh J, Ellis RJ, MacDermot KD, Vincent-Delorme C, Plessis G, Touraine R, Philippe A, Malan V, Mathieu-Dramard M, Chiesa J, Blaumeiser B, Kooy RF, Caiazzo R, Pigeyre M, Balkau B, Sladek R, Bergmann S, Mooser V, Waterworth D, Reymond A, Vollenweider P, Waeber G, Kurg A, Palta P, Esko T, Metspalu A, Nelis M, Elliott P, Hartikainen AL, McCarthy MI, Peltonen L, Carlsson L, Jacobson P, Sjöström L, Huang N, Hurles ME, O’Rahilly S, Farooqi IS, Männik K, Jarvelin MR, Pattou F, Meyre D, Walley AJ, Coin LJM, Blakemore AIF, Froguel P, Beckmann JS. A new highly penetrant form of obesity due to deletions on chromosome 16p11.2. Nature 2010; 463:671-5. [PMID: 20130649 PMCID: PMC2880448 DOI: 10.1038/nature08727] [Citation(s) in RCA: 345] [Impact Index Per Article: 24.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2009] [Accepted: 12/01/2009] [Indexed: 01/04/2023]
Abstract
Obesity has become a major worldwide challenge to public health, owing to an interaction between the Western 'obesogenic' environment and a strong genetic contribution. Recent extensive genome-wide association studies (GWASs) have identified numerous single nucleotide polymorphisms associated with obesity, but these loci together account for only a small fraction of the known heritable component. Thus, the 'common disease, common variant' hypothesis is increasingly coming under challenge. Here we report a highly penetrant form of obesity, initially observed in 31 subjects who were heterozygous for deletions of at least 593 kilobases at 16p11.2 and whose ascertainment included cognitive deficits. Nineteen similar deletions were identified from GWAS data in 16,053 individuals from eight European cohorts. These deletions were absent from healthy non-obese controls and accounted for 0.7% of our morbid obesity cases (body mass index (BMI) >or= 40 kg m(-2) or BMI standard deviation score >or= 4; P = 6.4 x 10(-8), odds ratio 43.0), demonstrating the potential importance in common disease of rare variants with strong effects. This highlights a promising strategy for identifying missing heritability in obesity and other complex traits: cohorts with extreme phenotypes are likely to be enriched for rare variants, thereby improving power for their discovery. Subsequent analysis of the loci so identified may well reveal additional rare variants that further contribute to the missing heritability, as recently reported for SIM1 (ref. 3). The most productive approach may therefore be to combine the 'power of the extreme' in small, well-phenotyped cohorts, with targeted follow-up in case-control and population cohorts.
Collapse
Affiliation(s)
- R. G. Walters
- Section of Genomic Medicine, Imperial College London, London, UK
- Department of Epidemiology and Public Health, Imperial College London, London, UK
| | - S. Jacquemont
- Service de Génétique Médicale, Centre Hospitalier Universitaire Vaudois, Lausanne, Switzerland
| | - A. Valsesia
- Departement de Génétique Médicale, Université de Lausanne, Lausanne, Switzerland
- Swiss Institute of Bioinformatics, Lausanne, Switzerland
| | - A. J. de Smith
- Section of Genomic Medicine, Imperial College London, London, UK
| | - D. Martinet
- Service de Génétique Médicale, Centre Hospitalier Universitaire Vaudois, Lausanne, Switzerland
| | - J. Andersson
- Section of Genomic Medicine, Imperial College London, London, UK
| | - M. Falchi
- Section of Genomic Medicine, Imperial College London, London, UK
| | - F. Chen
- Brain Mind Institute, Ecole Polytechnique Fédérale de Lausanne, Lausanne, Switzerland
| | - J. Andrieux
- Laboratoire de Génétique Médicale, Centre Hospitalier Régional Universitaire, Lille, France
| | - S. Lobbens
- CNRS 8090-Institute of Biology, Pasteur Institute, Lille, France
| | - B. Delobel
- Centre de Génétique Chromosomique, Hôpital Saint-Vincent de Paul, GHICL, Lille, France
| | - F. Stutzmann
- CNRS 8090-Institute of Biology, Pasteur Institute, Lille, France
| | | | - J.-C. Chèvre
- CNRS 8090-Institute of Biology, Pasteur Institute, Lille, France
| | - C. Lecoeur
- CNRS 8090-Institute of Biology, Pasteur Institute, Lille, France
| | - V. Vatin
- CNRS 8090-Institute of Biology, Pasteur Institute, Lille, France
| | - S. Bouquillon
- Laboratoire de Génétique Médicale, Centre Hospitalier Régional Universitaire, Lille, France
| | - J. L. Buxton
- Section of Genomic Medicine, Imperial College London, London, UK
| | - O. Boute
- Service de Génétique Clinique, Hôpital Jeanne de Flandre, Centre Hospitalier Universitaire de Lille, Lille, France
| | - M. Holder-Espinasse
- Service de Génétique Clinique, Hôpital Jeanne de Flandre, Centre Hospitalier Universitaire de Lille, Lille, France
| | - J.-M. Cuisset
- Service de Neuropédiatrie, Centre Hospitalier Régional Universitaire, Lille, France
| | - M.-P. Lemaitre
- Service de Neuropédiatrie, Centre Hospitalier Régional Universitaire, Lille, France
| | - A.-E. Ambresin
- Unité Multidisciplinaire de Santé des Adolescents, Centre Hospitalier Universitaire Vaudois, Lausanne, Switzerland
| | - A. Brioshi
- Service de Neuropsychologie et de Neuroréhabilitation, Centre Hospitalier Universitaire Vaudois, Lausanne, Switzerland
| | - M. Gaillard
- Service de Génétique Médicale, Centre Hospitalier Universitaire Vaudois, Lausanne, Switzerland
| | - V. Giusti
- Service d’Endocrinologie, Centre Hospitalier Universitaire Vaudois, Lausanne, Switzerland
| | - F. Fellmann
- Service de Génétique Médicale, Centre Hospitalier Universitaire Vaudois, Lausanne, Switzerland
| | - A. Ferrarini
- Service de Génétique Médicale, Centre Hospitalier Universitaire Vaudois, Lausanne, Switzerland
| | - N. Hadjikhani
- Brain Mind Institute, Ecole Polytechnique Fédérale de Lausanne, Lausanne, Switzerland
- Athinoula A Martinos Center for Biomedical Imaging, Massachusetts General Hospital, Harvard Medical School, Charlestown MA, USA
| | - D. Campion
- INSERM, U614, Faculté de Médecine, Rouen, France
| | - A. Guilmatre
- INSERM, U614, Faculté de Médecine, Rouen, France
| | - A. Goldenberg
- Service de Génétique, Centre Hospitalier Universitaire de Rouen, Rouen, France
| | - N. Calmels
- Laboratoire de Diagnostic Génétique, Nouvel hôpital civil, Strasbourg, France
| | - J.-L. Mandel
- Laboratoire de Diagnostic Génétique, Nouvel hôpital civil, Strasbourg, France
| | - C. Le Caignec
- Centre Hospitalier Universitaire Nantes, Service de Génétique Médicale, Nantes, France
- INSERM, UMR915, L’Institut du Thorax, Nantes, France
| | - A. David
- Centre Hospitalier Universitaire Nantes, Service de Génétique Médicale, Nantes, France
| | - B. Isidor
- Centre Hospitalier Universitaire Nantes, Service de Génétique Médicale, Nantes, France
| | - M.-P. Cordier
- Service de Génétique, Hospices Civils de Lyon, Hôpital de l’Hotel Dieu, Lyon, France
| | - S. Dupuis-Girod
- Service de Génétique, Hospices Civils de Lyon, Hôpital de l’Hotel Dieu, Lyon, France
| | - A. Labalme
- Service de Génétique, Hospices Civils de Lyon, Hôpital de l’Hotel Dieu, Lyon, France
| | - D. Sanlaville
- Service de Génétique, Hospices Civils de Lyon, Hôpital de l’Hotel Dieu, Lyon, France
- EA 4171, Université Claude Bernard, Lyon, France
| | - M. Béri-Deixheimer
- Laboratoire de Génétique, Centre Hospitalier Universitaire, Nancy University, Nancy, France
| | - P. Jonveaux
- Laboratoire de Génétique, Centre Hospitalier Universitaire, Nancy University, Nancy, France
| | - B. Leheup
- Laboratoire de Génétique, Centre Hospitalier Universitaire, Nancy University, Nancy, France
- EA4368 Medical School Nancy, Université Henri Poincaré, Nancy, France
| | - K. Õunap
- Department of Genetics, United Laboratories,Tartu University Children’s Hospital, Tartu, Estonia
| | - E. G. Bochukova
- University of Cambridge Metabolic Research Laboratories, Institute of Metabolic Science, Addenbrooke’s Hospital, Cambridge, UK
| | - E. Henning
- University of Cambridge Metabolic Research Laboratories, Institute of Metabolic Science, Addenbrooke’s Hospital, Cambridge, UK
| | - J. Keogh
- University of Cambridge Metabolic Research Laboratories, Institute of Metabolic Science, Addenbrooke’s Hospital, Cambridge, UK
| | - R. J. Ellis
- North West Thames Regional Genetics Service, Northwick Park & St Marks Hospital, Harrow, UK
| | - K. D. MacDermot
- North West Thames Regional Genetics Service, Northwick Park & St Marks Hospital, Harrow, UK
| | | | - G. Plessis
- Service de Génétique Médicale, Centre Hospitalier Universitaire Clemenceau, Caen, France
| | - R. Touraine
- Centre Hospitalier Universitaire–Hôpital Nord, Service de Génétique, Saint Etienne, France
| | - A. Philippe
- Département de Génétique et INSERM U781, Université Paris Descartes, Hôpital Necker-Enfants Malades, Paris, France
| | - V. Malan
- Département de Génétique et INSERM U781, Université Paris Descartes, Hôpital Necker-Enfants Malades, Paris, France
| | - M. Mathieu-Dramard
- Service de Génétique Clinique, Centre Hospitalier Universitaire, Amiens, France
| | - J. Chiesa
- Laboratoire de Cytogénétique, Centre Hospitalier Universitaire Caremeau, Nîmes, France
| | - B. Blaumeiser
- Department of Medical Genetics, University Hospital & University of Antwerp, Antwerp, Belgium
| | - R. F. Kooy
- Department of Medical Genetics, University Hospital & University of Antwerp, Antwerp, Belgium
| | - R. Caiazzo
- INSERM U859, Biotherapies for Diabetes, Lille, France
- University Lille Nord de France, Centre Hospitalier Universitaire Lille, France
| | - M. Pigeyre
- University Lille Nord de France, Centre Hospitalier Universitaire Lille, France
| | - B. Balkau
- INSERM U780-IFR69, Villejuif, France
| | - R. Sladek
- Genome Quebec Innovation Centre, Montreal, Canada
- Department of Medicine and Human Genetics, McGill University, Montreal, Canada
| | - S. Bergmann
- Departement de Génétique Médicale, Université de Lausanne, Lausanne, Switzerland
- Swiss Institute of Bioinformatics, Lausanne, Switzerland
| | - V. Mooser
- Division of Genetics, GlaxoSmithKline, Philadelphia PA, USA
| | - D. Waterworth
- Division of Genetics, GlaxoSmithKline, Philadelphia PA, USA
| | - A. Reymond
- The Center for Integrated Genomics, University of Lausanne, Lausanne, Switzerland
| | - P. Vollenweider
- Department of Medicine, Centre Hospitalier Universitaire Vaudois, Lausanne, Switzerland
| | - G. Waeber
- Department of Medicine, Centre Hospitalier Universitaire Vaudois, Lausanne, Switzerland
| | - A. Kurg
- Institute of Molecular and Cell Biology, University of Tartu, Tartu, Estonia
| | - P. Palta
- Institute of Molecular and Cell Biology, University of Tartu, Tartu, Estonia
| | - T. Esko
- Estonian Genome Project, University of Tartu, Tartu, Estonia
- Estonian Biocentre, Tartu, Estonia
| | - A. Metspalu
- Estonian Genome Project, University of Tartu, Tartu, Estonia
- Estonian Biocentre, Tartu, Estonia
| | - M. Nelis
- Estonian Genome Project, University of Tartu, Tartu, Estonia
- Estonian Biocentre, Tartu, Estonia
| | - P. Elliott
- Department of Epidemiology and Public Health, Imperial College London, London, UK
| | - A.-L. Hartikainen
- Department of Obstetrics and Gynaecology, University of Oulu, Oulu, Finland
| | - M. I. McCarthy
- Oxford Centre for Diabetes, Endocrinology and Metabolism, University of Oxford, Oxford, UK
- Wellcome Trust Centre for Human Genetics, University of Oxford, Oxford, UK
| | - L. Peltonen
- Wellcome Trust Sanger Institute, Hinxton, Cambridgeshire, UK
- Massachusetts Institute of Technology, The Broad Institute, Cambridge MA, USA
| | - L. Carlsson
- Department of Molecular and Clinical Medicine and Center for Cardiovascular and Metabolic Research, The Sahlgrenska Academy, Göteborg, Sweden
| | - P. Jacobson
- Department of Molecular and Clinical Medicine and Center for Cardiovascular and Metabolic Research, The Sahlgrenska Academy, Göteborg, Sweden
| | - L. Sjöström
- Department of Molecular and Clinical Medicine and Center for Cardiovascular and Metabolic Research, The Sahlgrenska Academy, Göteborg, Sweden
| | - N. Huang
- Wellcome Trust Sanger Institute, Hinxton, Cambridgeshire, UK
| | - M. E. Hurles
- Wellcome Trust Sanger Institute, Hinxton, Cambridgeshire, UK
| | - S. O’Rahilly
- University of Cambridge Metabolic Research Laboratories, Institute of Metabolic Science, Addenbrooke’s Hospital, Cambridge, UK
| | - I. S. Farooqi
- University of Cambridge Metabolic Research Laboratories, Institute of Metabolic Science, Addenbrooke’s Hospital, Cambridge, UK
| | - K. Männik
- Institute of Molecular and Cell Biology, University of Tartu, Tartu, Estonia
| | - M.-R. Jarvelin
- Department of Epidemiology and Public Health, Imperial College London, London, UK
- Department of Child and Adolescent Health, National Public Health Institute, Oulu, Finland
- Institute of Health Sciences and Biocenter Oulu, University of Oulu, Oulu Finland
| | - F. Pattou
- INSERM U859, Biotherapies for Diabetes, Lille, France
- University Lille Nord de France, Centre Hospitalier Universitaire Lille, France
| | - D. Meyre
- CNRS 8090-Institute of Biology, Pasteur Institute, Lille, France
| | - A. J. Walley
- Section of Genomic Medicine, Imperial College London, London, UK
| | - L. J. M. Coin
- Department of Epidemiology and Public Health, Imperial College London, London, UK
| | | | - P. Froguel
- Section of Genomic Medicine, Imperial College London, London, UK
- CNRS 8090-Institute of Biology, Pasteur Institute, Lille, France
| | - J. S. Beckmann
- Service de Génétique Médicale, Centre Hospitalier Universitaire Vaudois, Lausanne, Switzerland
- Departement de Génétique Médicale, Université de Lausanne, Lausanne, Switzerland
| |
Collapse
|
46
|
Hagenkord JM, Monzon FA, Kash SF, Lilleberg S, Xie Q, Kant JA. Array-based karyotyping for prognostic assessment in chronic lymphocytic leukemia: performance comparison of Affymetrix 10K2.0, 250K Nsp, and SNP6.0 arrays. J Mol Diagn 2010; 12:184-96. [PMID: 20075210 DOI: 10.2353/jmoldx.2010.090118] [Citation(s) in RCA: 47] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Specific chromosomal alterations are recognized as important prognostic factors in chronic lymphocytic leukemia (CLL). Array-based karyotyping is gaining acceptance as an alternative to the standard fluorescence in situ hybridization (FISH) panel for detecting these aberrations. This study explores the optimum single nucleotide polymorphism (SNP) array probe density for routine clinical use, presents clinical validation results for the 250K Nsp Affymetrix SNP array, and highlights clinically actionable genetic lesions missed by FISH and conventional cytogenetics. CLL samples were processed on low (10K2.0), medium (250K Nsp), and high (SNP6.0) probe density Affymetrix SNP arrays. Break point definition and detection rates for clinically relevant genetic lesions were compared. The 250K Nsp array was subsequently validated for routine clinical use and demonstrated 98.5% concordance with the standard CLL FISH panel. SNP array karyotyping detected genomic complexity and/or acquired uniparental disomy not detected by the FISH panel. In particular, a region of acquired uniparental disomy on 17p was shown to harbor two mutated copies of TP53 that would have gone undetected by FISH, conventional cytogenetics, or array comparative genomic hybridization. SNP array karyotyping allows genome-wide, high resolution detection of copy number and uniparental disomy at genomic regions with established prognostic significance in CLL, detects lesions missed by FISH, and provides insight into gene dosage at these loci.
Collapse
Affiliation(s)
- Jill M Hagenkord
- Molecular Pathology and Clinical Genomics, Creighton University Medical Center, Department of Pathology, 601 N. 30 Street, Suite 2400, Omaha, NE 68131-2197, USA.
| | | | | | | | | | | |
Collapse
|
47
|
Increased expression of cellular retinol-binding protein 1 in laryngeal squamous cell carcinoma. J Cancer Res Clin Oncol 2010; 136:931-8. [PMID: 20054560 DOI: 10.1007/s00432-009-0735-9] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2009] [Accepted: 11/16/2009] [Indexed: 01/24/2023]
Abstract
PURPOSE To investigate the genomic alterations in larynx carcinomas (LaCa) tissues and its prognostics values in predicting survival. METHODS To analyse the aberrations in the genome of LaCa patients, we used array comparative genomic hybridization in 19 human laryngeal tumour samples. DNA samples were also subjected to detect human papillomavirus (HPV) sequences by polymerase chain reaction (PCR). Copy number gain was confirmed by real-time PCR. The cellular retinol-binding protein 1 (CRBP-1) gene expression was also confirmed by immunohistochemistry assay on LaCa tissues. To identify prognostic feature, CRBP-1 gene gain was correlated to patient survival. RESULTS The most common gains were detected for CRBP-1 and EGFR genes, while DNA lost in RAF-1 gene. Immunohistochemistry assay was revealed strong expression of CRBP1 protein in those cases with CRBP-1 gene gain. The CRBP-1 gene gain and its expression correlated significantly with survival (P = 0.003). Cox regression analysis indicated that CRBP-1 expression level was a factor of survival (P = 0.008). HPV sequences were detected in 42% of the samples, and did not show any relationship with specific gene alterations. CONCLUSION Our data shows that CRBP-1 gene gain can be determined by immunohistochemistry on routinely processed tissue specimens, and could support as a potential novel marker for long-term survival in laryngeal squamous cell carcinoma.
Collapse
|
48
|
|
49
|
Northcott PA, Rutka JT, Taylor MD. Genomics of medulloblastoma: from Giemsa-banding to next-generation sequencing in 20 years. Neurosurg Focus 2010; 28:E6. [DOI: 10.3171/2009.10.focus09218] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
Advances in the field of genomics have recently enabled the unprecedented characterization of the cancer genome, providing novel insight into the molecular mechanisms underlying malignancies in humans. The application of high-resolution microarray platforms to the study of medulloblastoma has revealed new oncogenes and tumor suppressors and has implicated changes in DNA copy number, gene expression, and methylation state in its etiology. Additionally, the integration of medulloblastoma genomics with patient clinical data has confirmed molecular markers of prognostic significance and highlighted the potential utility of molecular disease stratification. The advent of next-generation sequencing technologies promises to greatly transform our understanding of medulloblastoma pathogenesis in the next few years, permitting comprehensive analyses of all aspects of the genome and increasing the likelihood that genomic medicine will become part of the routine diagnosis and treatment of medulloblastoma.
Collapse
Affiliation(s)
- Paul A. Northcott
- 1Division of Neurosurgery, Arthur and Sonia Labatt Brain Tumour Research Centre
- 2Program in Developmental and Stem Cell Biology, The Hospital for Sick Children; and
- 3Department of Laboratory Medicine and Pathobiology, University of Toronto, Ontario, Canada
| | - James T. Rutka
- 1Division of Neurosurgery, Arthur and Sonia Labatt Brain Tumour Research Centre
- 3Department of Laboratory Medicine and Pathobiology, University of Toronto, Ontario, Canada
| | - Michael D. Taylor
- 1Division of Neurosurgery, Arthur and Sonia Labatt Brain Tumour Research Centre
- 2Program in Developmental and Stem Cell Biology, The Hospital for Sick Children; and
- 3Department of Laboratory Medicine and Pathobiology, University of Toronto, Ontario, Canada
| |
Collapse
|
50
|
Jaiswal BS, Janakiraman V, Kljavin NM, Chaudhuri S, Stern HM, Wang W, Kan Z, Dbouk HA, Peters BA, Waring P, Vega TD, Kenski DM, Bowman K, Lorenzo M, Li H, Wu J, Modrusan Z, Stinson J, Eby M, Yue P, Kaminker J, de Sauvage FJ, Backer JM, Seshagiri S. Somatic mutations in p85alpha promote tumorigenesis through class IA PI3K activation. Cancer Cell 2009; 16:463-74. [PMID: 19962665 PMCID: PMC2804903 DOI: 10.1016/j.ccr.2009.10.016] [Citation(s) in RCA: 248] [Impact Index Per Article: 16.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/28/2009] [Revised: 08/18/2009] [Accepted: 10/19/2009] [Indexed: 12/19/2022]
Abstract
Members of the mammalian phosphoinositide-3-OH kinase (PI3K) family of proteins are critical regulators of various cellular process including cell survival, growth, proliferation, and motility. Oncogenic activating mutations in the p110alpha catalytic subunit of the heterodimeric p110/p85 PI3K enzyme are frequent in human cancers. Here we show the presence of frequent mutations in p85alpha in colon cancer, a majority of which occurs in the inter-Src homology-2 (iSH2) domain. These mutations uncouple and retain p85alpha's p110-stabilizing activity, while abrogating its p110-inhibitory activity. The p85alpha mutants promote cell survival, AKT activation, anchorage-independent cell growth, and oncogenesis in a p110-dependent manner.
Collapse
Affiliation(s)
- Bijay S. Jaiswal
- Department of Molecular Biology, Genentech Inc., 1 DNA WAY, South San Francisco, CA 94080
| | | | - Noelyn M. Kljavin
- Department of Molecular Biology, Genentech Inc., 1 DNA WAY, South San Francisco, CA 94080
| | - Subhra Chaudhuri
- Department of Molecular Biology, Genentech Inc., 1 DNA WAY, South San Francisco, CA 94080
| | - Howard M. Stern
- Department of Pathology, Genentech Inc., 1 DNA WAY, South San Francisco, CA 94080
| | - Weiru Wang
- Department of Protein Engineering, Genentech Inc., 1 DNA WAY, South San Francisco, CA 94080
| | - Zhengyan Kan
- Department of Molecular Biology, Genentech Inc., 1 DNA WAY, South San Francisco, CA 94080
| | - Hashem A. Dbouk
- Department of Molecular Pharmacology, Albert Einstein College of Medicine, Bronx, NY 10461, USA
| | - Brock A. Peters
- Department of Molecular Biology, Genentech Inc., 1 DNA WAY, South San Francisco, CA 94080
| | - Paul Waring
- Department of Pathology, Genentech Inc., 1 DNA WAY, South San Francisco, CA 94080
| | - Trisha Dela Vega
- Department of Protein Engineering, Genentech Inc., 1 DNA WAY, South San Francisco, CA 94080
| | - Denise M. Kenski
- Department of Molecular Biology, Genentech Inc., 1 DNA WAY, South San Francisco, CA 94080
| | - Krista Bowman
- Department of Protein Engineering, Genentech Inc., 1 DNA WAY, South San Francisco, CA 94080
| | - Maria Lorenzo
- Department of Protein Chemistry, Genentech Inc., 1 DNA WAY, South San Francisco, CA 94080
| | - Hong Li
- Department of Protein Chemistry, Genentech Inc., 1 DNA WAY, South San Francisco, CA 94080
| | - Jiansheng Wu
- Department of Protein Chemistry, Genentech Inc., 1 DNA WAY, South San Francisco, CA 94080
| | - Zora Modrusan
- Department of Molecular Biology, Genentech Inc., 1 DNA WAY, South San Francisco, CA 94080
| | - Jeremy Stinson
- Department of Molecular Biology, Genentech Inc., 1 DNA WAY, South San Francisco, CA 94080
| | - Michael Eby
- Department of Translational Oncology, Genentech Inc., 1 DNA WAY, South San Francisco, CA 94080
| | - Peng Yue
- Department of Bioinformatics, Genentech Inc., 1 DNA WAY, South San Francisco, CA 94080
| | - Josh Kaminker
- Department of Bioinformatics, Genentech Inc., 1 DNA WAY, South San Francisco, CA 94080
| | - Frederic J. de Sauvage
- Department of Molecular Biology, Genentech Inc., 1 DNA WAY, South San Francisco, CA 94080
| | - Jonathan M. Backer
- Department of Molecular Pharmacology, Albert Einstein College of Medicine, Bronx, NY 10461, USA
| | - Somasekar Seshagiri
- Department of Molecular Biology, Genentech Inc., 1 DNA WAY, South San Francisco, CA 94080
- Correspondence: ; phone: 650-225-1000; fax: 650-225-1762
| |
Collapse
|