Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 29
Filter
1.
Public Health Genomics ; 19(6): 315-324, 2016.
Article in English | MEDLINE | ID: mdl-27898412

ABSTRACT

Beginning in 2009, the advent of exome sequencing has contributed significantly towards new discoveries of heritable germline mutations and de novo mutations for rare Mendelian disorders with hitherto unknown genetic aetiologies. Exome sequencing is an efficient tool to identify disease mutations without the need of a multi-generational pedigree. Sequencing a single proband or multiple affected individuals has been shown to be successful in identifying disease mutations, but parents would be required in the case of de novo mutations. In addition to heritable germline and de novo mutations, exome sequencing has also succeeded in unravelling somatic driver mutations for a wide range of cancers through individual studies or international collaborative effort such as the Cancer Genome International Consortium. By contrast, the application of exome sequencing in complex diseases is relatively limited; probably it would be too expensive were it applied to thousands of samples to achieve the statistical power for rare or low frequency variants (<1%). On top of research discoveries, the application of exome sequencing as a diagnostic tool is also increasingly evident. In this article, we summarize and discuss the progress that has been made in these areas during almost a decade.


Subject(s)
Exome/genetics , Mutation , Sequence Analysis, DNA/trends , Genetic Diseases, Inborn/diagnosis , Genetic Diseases, Inborn/genetics , Germ-Line Mutation , High-Throughput Nucleotide Sequencing/methods , Humans , Neoplasms/genetics , Sequence Analysis, DNA/methods
3.
World J Gastroenterol ; 19(13): 2011-8, 2013 Apr 07.
Article in English | MEDLINE | ID: mdl-23599619

ABSTRACT

The arrival of both high-throughput and bench-top next-generation sequencing technologies and sequence enrichment methods has revolutionized our approach to dissecting the genetic basis of cancer. These technologies have been almost invariably employed in whole-genome sequencing (WGS) and whole-exome sequencing (WES) studies. Both WGS and WES approaches have been widely applied to interrogate the somatic mutational landscape of sporadic cancers and identify novel germline mutations underlying familial cancer syndromes. The clinical implications of cancer genome sequencing have become increasingly clear, for example in diagnostics. In this editorial, we present these advances in the context of research discovery and discuss both the clinical relevance of cancer genome sequencing and the challenges associated with the adoption of these genomic technologies in a clinical setting.


Subject(s)
Neoplasms/genetics , DNA Mutational Analysis , Databases, Genetic , Exome , Genetic Predisposition to Disease , Genome, Human , High-Throughput Nucleotide Sequencing , Humans , International Cooperation , Mutation , Neoplasms/diagnosis , Neoplasms/drug therapy
4.
J Med Genet ; 50(4): 203-11, 2013 Apr.
Article in English | MEDLINE | ID: mdl-23396985

ABSTRACT

Human germline mutations arise anew during meiosis in every generation. Such spontaneously occurring genetic variants are termed de novo mutations. Although the introduction of microarray based approaches led to the discovery of numerous de novo copy number variants underlying a range of human genetic conditions, de novo single nucleotide variants (SNVs) remained refractory to analysis at the whole genome level until the advent of next generation sequencing technologies such as whole genome sequencing and whole exome sequencing. These approaches have recently allowed the estimation of the mutation rate of de novo SNVs and greatly increased our understanding of their contribution to human genetic disease. Indeed, de novo SNVs have been found to underlie various common human neurodevelopmental conditions such as schizophrenia, autism and intellectual disability, as well as sporadic cases of rare Mendelian disorders. In many cases, however, confirmation of the pathogenicity of identified de novo SNVs remains a major challenge.


Subject(s)
DNA Copy Number Variations , Genetic Diseases, Inborn , Genetic Predisposition to Disease , Germ-Line Mutation , Autistic Disorder/genetics , Genome, Human , High-Throughput Nucleotide Sequencing , Humans , Intellectual Disability/genetics , Meiosis , Schizophrenia/genetics
8.
Bioinformatics ; 28(21): 2711-8, 2012 Nov 01.
Article in English | MEDLINE | ID: mdl-22942022

ABSTRACT

MOTIVATION: Analysing next-generation sequencing (NGS) data for copy number variations (CNVs) detection is a relatively new and challenging field, with no accepted standard protocols or quality control measures so far. There are by now several algorithms developed for each of the four broad methods for CNV detection using NGS, namely the depth of coverage (DOC), read-pair, split-read and assembly-based methods. However, because of the complexity of the genome and the short read lengths from NGS technology, there are still many challenges associated with the analysis of NGS data for CNVs, no matter which method or algorithm is used. RESULTS: In this review, we describe and discuss areas of potential biases in CNV detection for each of the four methods. In particular, we focus on issues pertaining to (i) mappability, (ii) GC-content bias, (iii) quality control measures of reads and (iv) difficulty in identifying duplications. To gain insights to some of the issues discussed, we also download real data from the 1000 Genomes Project and analyse its DOC data. We show examples of how reads in repeated regions can affect CNV detection, demonstrate current GC-correction algorithms, investigate sensitivity of DOC algorithm before and after quality control of reads and discuss reasons for which duplications are harder to detect than deletions.


Subject(s)
Algorithms , Chromosome Mapping/statistics & numerical data , DNA Copy Number Variations , Sequence Analysis, DNA/statistics & numerical data , Base Composition , Base Sequence , Gene Deletion , Gene Duplication , Quality Control , Selection Bias , Sequence Analysis, DNA/methods
9.
Ann Surg Oncol ; 19(12): 3646-50, 2012 Nov.
Article in English | MEDLINE | ID: mdl-22851046

ABSTRACT

Revolutionary sequencing technologies have changed biomedical research and life science exponentially. Revealing the whole landscape of causal somatic and inherited mutations underlying individual patient's cancer sample by whole-genome sequencing (WGS) and whole-exome sequencing (WES) can lead to not only a new mutations-based taxonomy of solid tumors (Stratton, Science 331:1553-1558, 2011). But also shapes a roadmap for precision medicine (Roychowdhury et al., Sci Transl Med 3:111ra121, 2011; Roukos, Expert Rev Mol Diagn 12:215-218, 2012; Mirnezami et al., N Engl J Med 366:489-491, 2012). This inevitable approach for personalized diagnostics in concert with free-falling genome sequencing costs raises now the question of applying next-generation sequencing (NGS) technology in the clinic. In the pragmatic clinical world and in contrast to innovative research, is NGS-based clinical evidence sufficient for decision-making on tailoring the best available treatment to the individual cancer patient?


Subject(s)
Genome, Human , Molecular Targeted Therapy , Neoplasms/therapy , Precision Medicine , Humans , Neoplasms/genetics
12.
Expert Rev Mol Diagn ; 12(3): 241-51, 2012 Apr.
Article in English | MEDLINE | ID: mdl-22468815

ABSTRACT

The advent of next-generation sequencing technologies has revolutionized the study of genetic variation in the human genome. Whole-genome sequencing currently represents the most comprehensive strategy for variant detection genome-wide but is costly for large sample sizes, and variants detected in noncoding regions remain largely uninterpretable. By contrast, whole-exome sequencing has been widely applied in the identification of germline mutations underlying Mendelian disorders, somatic mutations in various cancers and de novo mutations in neurodevelopmental disorders. Since whole-exome sequencing focuses upon the entire set of exons in the genome (the exome), it requires additional exome-enrichment steps compared with whole-genome sequencing. Although the availability of multiple commercial exome-enrichment kits has made whole-exome sequencing technically feasible, it has also added to the overall cost. This has led to the emergence of transcriptome (or RNA) sequencing as a potential alternative approach to variant detection within protein coding regions, since the transcriptome of a given tissue represents a quasi-complete set of transcribed genes (mRNAs) and other noncoding RNAs. A further advantage of this approach is that it bypasses the need for exome enrichment. Here we discuss the relative merits and limitations of these approaches as they are applied in the context of variant detection within gene coding regions.


Subject(s)
Exome/genetics , Genetic Variation , High-Throughput Nucleotide Sequencing/methods , Transcriptome/genetics , Base Sequence , Genome, Human , Genome-Wide Association Study , Humans , RNA, Messenger/analysis , Sequence Analysis, DNA , Sequence Analysis, RNA
13.
Mod Pathol ; 25(8): 1055-68, 2012 Aug.
Article in English | MEDLINE | ID: mdl-22522846

ABSTRACT

Recent advances in genotyping and sequencing technologies have provided powerful tools with which to explore the genetic basis of both Mendelian (monogenic) and sporadic (polygenic) diseases. Several hundred genome-wide association studies have so far been performed to explore the genetics of various polygenic or complex diseases including those cancers with a genetic predisposition. Exome sequencing has also proven very successful in elucidating the etiology of a range of hitherto poorly understood Mendelian disorders caused by high-penetrance mutations. Despite such progress, the genetic etiology of several familial cancers, such as familial colorectal cancer type X, has remained elusive. Familial colorectal cancer type X and Lynch syndrome are similar in terms of their fulfilling certain clinical criteria, but the former group is not characterized by germline mutations in DNA mismatch-repair genes. On the other hand, the genetics of sporadic colorectal cancer have been investigated by genome-wide association studies, leading to the identification of multiple new susceptibility loci. In addition, there is increasing evidence to suggest that familial and sporadic cancers exhibit similarities in terms of their genetic etiologies. In this review, we have summarized our current knowledge of familial colorectal cancer type X, discussed current approaches to probing its genetic etiology through the application of new sequencing technologies and the recruitment of the results of colorectal cancer genome-wide association studies, and explore the challenges that remain to be overcome given the uncertainty of the current genetic model (ie, monogenic vs polygenic) of familial colorectal cancer type X.


Subject(s)
Colorectal Neoplasms, Hereditary Nonpolyposis/genetics , Colorectal Neoplasms/genetics , Exome/genetics , Genetic Predisposition to Disease , Genome-Wide Association Study , Multifactorial Inheritance , Colorectal Neoplasms, Hereditary Nonpolyposis/pathology , Humans , Mutation , Sequence Analysis, DNA
14.
Expert Rev Mol Diagn ; 12(2): 159-73, 2012 Mar.
Article in English | MEDLINE | ID: mdl-22369376

ABSTRACT

The potential applications of next-generation sequencing technologies in diagnostic laboratories have become increasingly evident despite the various technical challenges that still need to be overcome to potentiate its widespread adoption in a clinical setting. Whole-genome sequencing is now both technically feasible and 'cost effective' using next-generation sequencing techniques. However, this approach is still considered to be 'expensive' for a diagnostic test. Although the goal of the US$1000 genome is fast approaching, neither the analytical hurdles nor the ethical issues involved are trivial. In addition, the cost of data analysis and storage has been much higher than initially expected. As a result, it is widely perceived that targeted sequencing and whole-exome sequencing are more likely to be adopted as diagnostic tools in the foreseeable future. However, the information-generating power of whole-exome sequencing has also sparked considerable debate in relation to its deployment in genetic diagnostics, particularly with reference to the revelation of incidental findings. In this review, we focus on the targeted sequencing approach and its potential as a genetic diagnostic tool.


Subject(s)
Germ-Line Mutation , Molecular Diagnostic Techniques/methods , Sequence Analysis, DNA/methods , Exome , High-Throughput Nucleotide Sequencing , Humans , Multiplex Polymerase Chain Reaction , Polymorphism, Genetic
15.
Ann Neurol ; 71(1): 5-14, 2012 Jan.
Article in English | MEDLINE | ID: mdl-22275248

ABSTRACT

Recent developments in high-throughput sequence capture methods and next-generation sequencing technologies have now made exome sequencing a viable approach to elucidate the genetic basis of Mendelian disorders with hitherto unknown etiology. In addition, exome sequencing is increasingly being employed as a diagnostic tool for specific genetic diseases, particularly in the context of those disorders characterized by significant genetic and phenotypic heterogeneity, for example, Charcot-Marie-Tooth disease and congenital disorders of glycosylation. Such disorders are challenging to interrogate with conventional polymerase chain reaction-Sanger sequencing methods, because of the inherent difficulty in prioritizing candidate genes for diagnostic testing. Here, we explore the value of exome sequencing as a diagnostic tool and discuss whether exome sequencing can come to serve a dual role in diagnosis and discovery. We summarize the current status of exome sequencing, the technical challenges facing it, and its adaptation to diagnostics, and make recommendations for the use of exome sequencing as a routine diagnostic tool. Finally, we discuss pertinent ethical concerns, such as the use of exome sequencing data, originally generated in a diagnostic context, in research investigations.


Subject(s)
Exome/genetics , Gene Targeting/methods , Gene Targeting/trends , Sequence Analysis, DNA/methods , Sequence Analysis, DNA/trends , Animals , Charcot-Marie-Tooth Disease/diagnosis , Charcot-Marie-Tooth Disease/genetics , Gene Targeting/ethics , Genome, Human/genetics , Humans , Sequence Analysis, DNA/ethics
16.
J Hum Genet ; 57(2): 101-8, 2012 Feb.
Article in English | MEDLINE | ID: mdl-22129560

ABSTRACT

The genomes of outbred populations were first shown in 2006 to contain regions of homozygosity (ROHs) of several megabases. Further studies have also investigated the characteristics of ROHs in healthy individuals in various populations but there are no studies on Singapore populations to date. This study aims to identify and investigate the characteristics of ROHs in three Singapore populations. A total of 268 samples (96 Chinese, 89 Malays and 83 Indians) are genotyped on Illumina Human 1 M Beadchip and Affymetrix Genome-Wide Human SNP Array 6.0. We use the PennCNV algorithm to detect ROHs. We report an abundance of ROHs (≥500 kb), with an average of more than one hundred regions per individual. On average, the Indian population has the lowest number of ROHs and smallest total length of ROHs per individual compared with the Chinese and Malay populations. We further investigate the relationship between the occurrence of ROHs and haplotype frequency, regional linkage disequilibrium (LD) and positive selection. Based on the results of this data set, we find that the frequency of occurrence of ROHs is positively associated with haplotype frequency and regional LD. The majority of regions detected for recent positive selection and regions with differential LD between populations overlap with the ROH loci. When we consider both the location of the ROHs and the allelic form of the ROHs, we are able to separate the populations by principal component analysis, demonstrating that ROHs contain information on population structure and the demographic history of a population.


Subject(s)
Genetic Loci , Homozygote , Asian People , Gene Frequency , Haplotypes , Humans , Linkage Disequilibrium , Principal Component Analysis , Sequence Analysis, DNA , Singapore , White People
17.
Hum Genomics ; 5(6): 577-622, 2011 Oct.
Article in English | MEDLINE | ID: mdl-22155605

ABSTRACT

Substantial progress has been made in human genetics and genomics research over the past ten years since the publication of the draft sequence of the human genome in 2001. Findings emanating directly from the Human Genome Project, together with those from follow-on studies, have had an enormous impact on our understanding of the architecture and function of the human genome. Major developments have been made in cataloguing genetic variation, the International HapMap Project, and with respect to advances in genotyping technologies. These developments are vital for the emergence of genome-wide association studies in the investigation of complex diseases and traits. In parallel, the advent of high-throughput sequencing technologies has ushered in the 'personal genome sequencing' era for both normal and cancer genomes, and made possible large-scale genome sequencing studies such as the 1000 Genomes Project and the International Cancer Genome Consortium. The high-throughput sequencing and sequence-capture technologies are also providing new opportunities to study Mendelian disorders through exome sequencing and whole-genome sequencing. This paper reviews these major developments in human genetics and genomics over the past decade.


Subject(s)
Genetic Variation , Genome, Human , Genomics , Human Genome Project , Genome-Wide Association Study , Humans , Time Factors
18.
J Med Genet ; 48(11): 721-30, 2011 Nov.
Article in English | MEDLINE | ID: mdl-21825079

ABSTRACT

The advances in next generation sequencing (NGS) technologies have had a significant impact on epigenomic research. The arrival of NGS technologies has enabled a more powerful sequencing based method--that is, ChIP-Seq--to interrogate whole genome histone modifications, improving on the conventional microarray based method (ChIP-chip). Similarly, the first human DNA methylome was mapped using NGS technologies. More importantly, studies of DNA methylation and histone modification using NGS technologies have yielded new discoveries and improved our knowledge of human biology and diseases. The concept that cytosine methylation was restricted to CpG dinucleotides has only been recently challenged by new data generated from sequencing the DNA methylome. Approximately 25% of all cytosine methylation identified in stem cells was in a non-CG context. The non-CG methylation was more enriched in gene bodies and depleted in protein binding sites and enhancers. The recent developments of third generation sequencing technologies have shown promising results of directly sequencing methylated nucleotides and having the ability to differentiate between 5-methylcytosine and 5-hydroxymethylcytosine. The importance of 5-hydroxymethylcytosine remains largely unknown, but it has been found in various tissues. 5-hydroxymethylcytosine was particularly enriched at promoters and in intragenic regions (gene bodies) but was largely absent from non-gene regions in DNA from human brain frontal lobe tissue. The presence of 5-hydroxymethylcytosine in gene bodies was more positively correlated with gene expression levels. The importance of studying 5-methylcytosine and 5-hydroxymethylcytosine separately for their biological roles will become clearer when more efficient methods to distinguish them are available.


Subject(s)
Cytosine/analogs & derivatives , DNA/chemistry , Epigenesis, Genetic , Epigenomics/methods , Genome , High-Throughput Nucleotide Sequencing/methods , Histones/metabolism , 5-Methylcytosine/metabolism , Animals , Base Composition , Cytosine/metabolism , DNA/genetics , DNA Methylation , DNA, Intergenic , Epigenomics/instrumentation , Gene Expression , Genome-Wide Association Study , High-Throughput Nucleotide Sequencing/instrumentation , Histones/genetics , Humans , Mice , Oligonucleotide Array Sequence Analysis/methods , Promoter Regions, Genetic , Quantitative Trait Loci , Quantitative Trait, Heritable
19.
J Hum Genet ; 56(7): 524-33, 2011 Jul.
Article in English | MEDLINE | ID: mdl-21633363

ABSTRACT

The abundance of copy number variants (CNVs) and regions of homozygosity (ROHs) have been well documented in previous studies. In addition, their roles in complex diseases and traits have since been increasingly appreciated. However, only a limited amount of CNV and ROH data is currently available for the Swedish population. We conducted a population-based study to detect and characterize CNVs and ROHs in 87 randomly selected healthy Swedish individuals using the Affymetrix SNP Array 6.0. More than 600 CNV loci were detected in the population using two different CNV-detection algorithms (PennCNV and Birdsuite). A total of 196 loci were consistently identified by both algorithms, suggesting their reliability. Numerous disease-associated and pharmacogenetics-related genes were found to be overlapping with common CNV loci such as CFHR1/R3, LCE3B/3C, UGT2B17 and GSTT1. Correlation analysis between copy number polymorphisms (CNPs) and genome-wide association studies-identified single-nucleotide polymorphisms also indicates the potential roles of several CNPs as causal variants for diseases and traits such as body mass index, Crohn's disease and multiple sclerosis. In addition, we also identified a total of 14 815 ROHs 500 kb or 2814 ROHs 1M in the Swedish individuals with an average of 170 and 32 regions detected per individual respectively. Approximately 141 Mb or 4.92% of the genome is homozygous in each individual of the Swedish population. This is the first population-based study to investigate the population characteristics of CNVs and ROHs in the Swedish population. This study found many CNV loci that warrant further investigation, and also highlighted the abundance and importance of investigating ROHs for their associations with complex diseases and traits.


Subject(s)
DNA Copy Number Variations , Homozygote , Algorithms , Databases, Genetic , Genetics, Population , Genome, Human , Genome-Wide Association Study , Humans , Sweden
20.
J Hum Genet ; 56(8): 552-60, 2011 Aug.
Article in English | MEDLINE | ID: mdl-21677662

ABSTRACT

Copy number variations can be identified using newer genotyping arrays with higher single nucleotide polymorphisms (SNPs) density and copy number probes accompanied by newer algorithms. McCarroll et al. (2008) applied these to the HapMap II samples and identified 1316 copy number polymorphisms (CNPs). In our study, we applied the same approach to 859 samples from three Singapore populations and seven HapMap III populations. Approximately 50% of the 1291 autosomal CNPs were found to be polymorphic only in populations of non-African ancestry. Pairwise comparisons among the 10 populations showed substantial differences in the CNPs frequencies. Additionally, 698 CNPs showed significant differences with false discovery rate (FDR)<0.01 among the 10 populations and these loci overlap with known disease-associated or pharmacogenetic-related genes such as CFHR3 and CFHR1 (age related macular degeneration), GSTTI (metabolism of various carcinogenic compounds and cancers) and UGT2B17 (prostate cancer and graft-versus-host disease). The correlations between CNPs and genome-wide association studies-SNPs were investigated and several loci, which were previously unreported, that may potentially be implicated in complex diseases and traits were found; for example, childhood acute lymphoblastic leukaemia, age-related macular degeneration, breast cancer, response to antipsychotic treatment, rheumatoid arthritis and type-1 diabetes. Additionally, we also found 5014 novel copy number loci that have not been reported previously by McCarroll et al. (2008) in the 10 populations.


Subject(s)
DNA Copy Number Variations , Genome-Wide Association Study/methods , Haplotypes/genetics , Polymorphism, Single Nucleotide , Blood Proteins/genetics , China/ethnology , Complement C3b Inactivator Proteins/genetics , Gene Frequency , Genetic Predisposition to Disease/ethnology , Genetic Predisposition to Disease/genetics , Genetics, Population , Genotype , Glucuronosyltransferase/genetics , Glutathione Transferase/genetics , HapMap Project , Humans , India/ethnology , Malaysia/ethnology , Minor Histocompatibility Antigens , Reproducibility of Results , Singapore
SELECTION OF CITATIONS
SEARCH DETAIL
...