Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 12 de 12
Filter
Add more filters










Publication year range
1.
Hum Mutat ; 37(12): 1318-1328, 2016 12.
Article in English | MEDLINE | ID: mdl-27633797

ABSTRACT

As next-generation sequencing increases access to human genetic variation, the challenge of determining clinical significance of variants becomes ever more acute. Germline variants in the BRCA1 and BRCA2 genes can confer substantial lifetime risk of breast and ovarian cancer. Assessment of variant pathogenicity is a vital part of clinical genetic testing for these genes. A database of clinical observations of BRCA variants is a critical resource in that process. This article describes BRCA Share™, a database created by a unique international alliance of academic centers and commercial testing laboratories. By integrating the content of the Universal Mutation Database generated by the French Unicancer Genetic Group with the testing results of two large commercial laboratories, Quest Diagnostics and Laboratory Corporation of America (LabCorp), BRCA Share™ has assembled one of the largest publicly accessible collections of BRCA variants currently available. Although access is available to academic researchers without charge, commercial participants in the project are required to pay a support fee and contribute their data. The fees fund the ongoing curation effort, as well as planned experiments to functionally characterize variants of uncertain significance. BRCA Share™ databases can therefore be considered as models of successful data sharing between private companies and the academic world.


Subject(s)
BRCA1 Protein/genetics , BRCA2 Protein/genetics , Breast Neoplasms/genetics , Databases, Factual , Ovarian Neoplasms/genetics , Data Curation , Databases, Factual/economics , Female , Genetic Predisposition to Disease , Humans , Mutation
2.
Hum Mutat ; 37(1): 127-34, 2016 Jan.
Article in English | MEDLINE | ID: mdl-26467025

ABSTRACT

We developed a rules-based scoring system to classify DNA variants into five categories including pathogenic, likely pathogenic, variant of uncertain significance (VUS), likely benign, and benign. Over 16,500 pathogenicity assessments on 11,894 variants from 338 genes were analyzed for pathogenicity based on prediction tools, population frequency, co-occurrence, segregation, and functional studies collected from internal and external sources. Scores were calculated by trained scientists using a quantitative framework that assigned differential weighting to these five types of data. We performed descriptive and comparative statistics on the dataset and tested interobserver concordance among the trained scientists. Private variants defined as variants found within single families (n = 5,182), were either VUS (80.5%; n = 4,169) or likely pathogenic (19.5%; n = 1,013). The remaining variants (n = 6,712) were VUS (38.4%; n = 2,577) or likely benign/benign (34.7%; n = 2,327) or likely pathogenic/pathogenic (26.9%, n = 1,808). Exact agreement between the trained scientists on the final variant score was 98.5% [95% confidence interval (CI) (98.0, 98.9)] with an interobserver consistency of 97% [95% CI (91.5, 99.4)]. Variant scores were stable and showed increasing odds of being in agreement with new data when re-evaluated periodically. This carefully curated, standardized variant pathogenicity scoring system provides reliable pathogenicity scores for DNA variants encountered in a clinical laboratory setting.


Subject(s)
Computational Biology/methods , Genetic Predisposition to Disease , Genetic Variation , Genomics/methods , Software , Humans , Observer Variation , Reproducibility of Results , Web Browser
3.
Mol Genet Genomic Med ; 2(6): 522-9, 2014 Nov.
Article in English | MEDLINE | ID: mdl-25614874

ABSTRACT

We report the frequency, positive rate, and type of mutations in 14 genes (PMP22, GJB1, MPZ, MFN2, SH3TC2, GDAP1, NEFL, LITAF, GARS, HSPB1, FIG4, EGR2, PRX, and RAB7A) associated with Charcot-Marie-Tooth disease (CMT) in a cohort of 17,880 individuals referred to a commercial genetic testing laboratory. Deidentified results from sequencing assays and multiplex ligation-dependent probe amplification (MLPA) were analyzed including 100,102 Sanger sequencing, 2338 next-generation sequencing (NGS), and 21,990 MLPA assays. Genetic abnormalities were identified in 18.5% (n = 3312) of all individuals. Testing by Sanger and MLPA (n = 3216) showed that duplications (dup) (56.7%) or deletions (del) (21.9%) in the PMP22 gene accounted for the majority of positive findings followed by mutations in the GJB1 (6.7%), MPZ (5.3%), and MFN2 (4.3%) genes. GJB1 del and mutations in the remaining genes explained 5.3% of the abnormalities. Pathogenic mutations were distributed as follows: missense (70.6%), nonsense (14.3%), frameshift (8.7%), splicing (3.3%), in-frame deletions/insertions (1.8%), initiator methionine mutations (0.8%), and nonstop changes (0.5%). Mutation frequencies, positive rates, and the types of mutations were similar between tests performed by either Sanger (n = 17,377) or NGS (n = 503). Among patients with a positive genetic finding in a CMT-related gene, 94.9% were positive in one of four genes (PMP22, GJB1, MPZ, or MFN2).

4.
Neurogenetics ; 12(3): 169-73, 2011 Aug.
Article in English | MEDLINE | ID: mdl-21630033

ABSTRACT

The rate of DNA variation discovery has accelerated the need to collate, store and interpret the data in a standardised coherent way and is becoming a critical step in maximising the impact of discovery on the understanding and treatment of human disease. This particularly applies to the field of neurology as neurological function is impaired in many human disorders. Furthermore, the field of neurogenetics has been proven to show remarkably complex genotype-to-phenotype relationships. To facilitate the collection of DNA sequence variation pertaining to neurogenetic disorders, we have initiated the "Neurogenetics Consortium" under the umbrella of the Human Variome Project. The Consortium's founding group consisted of basic researchers, clinicians, informaticians and database creators. This report outlines the strategic aims established at the preliminary meetings of the Neurogenetics Consortium and calls for the involvement of the wider neurogenetic community in enabling the development of this important resource.


Subject(s)
Databases, Genetic/standards , Genetic Variation , Genetics, Medical/organization & administration , International Cooperation , Nervous System/metabolism , Algorithms , Congresses as Topic , Genetic Variation/physiology , Genetics, Medical/standards , Human Genome Project/organization & administration , Humans , Nervous System Diseases/genetics , Nervous System Diseases/metabolism , Research Report
5.
Am J Med Genet A ; 143A(15): 1775-7, 2007 Aug 01.
Article in English | MEDLINE | ID: mdl-17593543

ABSTRACT

Ataxia with oculomotor apraxia type 2 (AOA2) is an autosomal recessive disorder associated with mutations in the Senataxin (SETX) gene. Clinical manifestations (ataxia, peripheral neuropathy, oculomotor apraxia) of this disease have previously been limited to the nervous system. We describe a patient homozygous for a novel mutation of SETX who manifested not only ataxia but also ovarian failure.


Subject(s)
Apraxia, Ideomotor/genetics , Mutation , Primary Ovarian Insufficiency/complications , RNA Helicases/genetics , Adult , Apraxia, Ideomotor/complications , DNA/blood , DNA/genetics , DNA/isolation & purification , DNA Helicases , Female , Homozygote , Humans , Multifunctional Enzymes , Primary Ovarian Insufficiency/diagnostic imaging , Primary Ovarian Insufficiency/genetics , Radiography , alpha-Fetoproteins/metabolism
6.
J Biol Chem ; 280(45): 37400-7, 2005 Nov 11.
Article in English | MEDLINE | ID: mdl-16131487

ABSTRACT

The replication of eukaryotic genomes necessitates the coordination of histone biosynthesis with DNA replication at the onset of S phase. The multiple histone H4 genes encode identical proteins, but their regulatory sequences differ. The contributions of these individual genes to histone H4 mRNA expression have not been described. We have determined, by real-time quantitative PCR and RNase protection, that the human histone H4 genes are not equally expressed and that a subset contributes disproportionately to the total pool of H4 mRNA. Differences in histone H4 gene expression can be attributed to observed unequal activities of the H4 gene promoters, which exhibit variations in gene regulatory elements. The overall expression pattern of the histone H4 gene complement is similar in normal and cancer cells. However, H4 genes that are moderately expressed in normal cells are sporadically silenced in tumor cells with compensation of expression by other H4 gene copies. Chromatin immunoprecipitation analyses and in vitro DNA binding assays indicated that 11 of the 15 histone H4 genes interact with the cell cycle regulatory histone nuclear factor P, which forms a complex with the cyclin E/CDK2-responsive co-regulator p220(NPAT). These 11 H4 genes account for 95% of the histone H4 mRNA pool. We conclude that the cyclin E/CDK2/p220(NPAT)/histone nuclear factor P signaling pathway is the principal regulator of histone H4 biosynthesis.


Subject(s)
Gene Expression Regulation, Neoplastic , Gene Expression Regulation , Histones/genetics , S Phase/genetics , S Phase/physiology , Animals , Base Sequence , Cell Line , Consensus Sequence , DNA Replication , Gene Expression Profiling , Humans , Promoter Regions, Genetic , RNA, Messenger/metabolism , Signal Transduction
7.
Mol Cell Biol ; 25(14): 6140-53, 2005 Jul.
Article in English | MEDLINE | ID: mdl-15988025

ABSTRACT

Genome replication in eukaryotic cells necessitates the stringent coupling of histone biosynthesis with the onset of DNA replication at the G1/S phase transition. A fundamental question is the mechanism that links the restriction (R) point late in G1 with histone gene expression at the onset of S phase. Here we demonstrate that HiNF-P, a transcriptional regulator of replication-dependent histone H4 genes, interacts directly with p220(NPAT), a substrate of cyclin E/CDK2, to coactivate histone genes during S phase. HiNF-P and p220 are targeted to, and colocalize at, subnuclear foci (Cajal bodies) in a cell cycle-dependent manner. Genetic or biochemical disruption of the HiNF-P/p220 interaction compromises histone H4 gene activation at the G1/S phase transition and impedes cell cycle progression. Our results show that HiNF-P and p220 form a critical regulatory module that directly links histone H4 gene expression at the G1/S phase transition to the cyclin E/CDK2 signaling pathway at the R point.


Subject(s)
Cell Cycle Proteins/metabolism , Gene Expression Regulation , Histones/genetics , Nuclear Proteins/metabolism , Repressor Proteins/metabolism , S Phase/genetics , Amino Acid Motifs , Animals , CDC2-CDC28 Kinases/metabolism , Cell Cycle Proteins/analysis , Cell Cycle Proteins/genetics , Cell Nucleus/chemistry , Cell Nucleus/metabolism , Chromatin/metabolism , Cyclin E/metabolism , Cyclin-Dependent Kinase 2 , G1 Phase/genetics , Humans , Mutation , Nuclear Proteins/analysis , Nuclear Proteins/genetics , Promoter Regions, Genetic , Protein Interaction Mapping , Repressor Proteins/analysis , Transcription, Genetic , Transcriptional Activation
8.
Gene ; 342(1): 35-40, 2004 Nov 10.
Article in English | MEDLINE | ID: mdl-15527963

ABSTRACT

Histones are the major protein component of nucleosomes, and de novo histone synthesis is essential for packaging newly replicated DNA into chromatin. As a result, histone gene expression is exquisitely and functionally coupled with DNA replication. Vastly divergent organisms such as yeast, fly and human all demonstrate the phylogenetically conserved propensity to maintain clustering of histone genes at one or more genomic loci. Although specific mechanisms are unclear, clustering is presumed to be important for common stringent transcriptional control of these genes at the G1/S phase transition. In this study, we describe a genomic duplication of the human histone gene cluster located at chromosome 1q21, which effectively doubles the previously known size and gene number of that cluster. The duplication persists in all examined tissues and cell lines, and the duplicated genes are transcriptionally active. Levels of messenger RNAs for duplicated histone H4 genes are high relative to those for non-duplicated H4 genes. Our data suggest that transcriptionally robust histone H4 genes may have been preferentially duplicated during evolution.


Subject(s)
Histones/genetics , Multigene Family/genetics , Amino Acid Sequence , Base Sequence , Cell Line, Tumor , Chromosomes, Human, Pair 1/genetics , Gene Duplication , Genes, Duplicate/genetics , Genome, Human , HL-60 Cells , HeLa Cells , Humans , Molecular Sequence Data , RNA, Messenger/genetics , RNA, Messenger/metabolism , Sequence Homology, Nucleic Acid
9.
Int J Oncol ; 25(2): 503-9, 2004 Aug.
Article in English | MEDLINE | ID: mdl-15254750

ABSTRACT

A promising family of anticancer agents, the camptothecins, is noted for their ability to induce apoptosis specifically in malignant cells. However, a major obstacle for successful cancer treatment by these and other chemotherapeutic agents is the intrinsic or acquired resistance to drug treatment. Resistance to 9NC6, a camptothecin derivative, has been modeled in vitro using a human prostate cancer cell line. To elucidate the mechanism for acquired 9NC resistance, we have used a subtractive cloning approach to identify genes whose altered expression level is reflective of 9NC resistance or susceptibility. Differential gene expression was compared between wild-type human prostate cancer cell line, DU-145, and a 9NC-resistant subline, RC1. Results were confirmed by Northern and Western blot analyses. In this report, we focus on one gene, 14-3-3zeta. An expression vector of a full-length myc-epitope-tagged 14-3-3zeta cDNA was constructed and used for transfection into DU-145 cells. We consistently observed that 14-3-3zeta message and protein levels were dramatically increased in 9NC resistant cells. The expression levels of other 14-3-3 family members were unaffected. Strikingly, ectopic overexpression of 14-3-3zeta in wild-type 9NC-susceptible prostate cancer cells decreased 9NC-induced apoptosis. Our results suggest a novel direct or indirect role of 14-3-3zeta in mediating resistance of DU-145 cells to the topoisomerase I inhibitor, 9NC. We are currently exploring whether this represents a more general pathway for drug resistance as well.


Subject(s)
14-3-3 Proteins/metabolism , Antineoplastic Agents/pharmacology , Apoptosis , Camptothecin/analogs & derivatives , Camptothecin/pharmacology , Prostatic Neoplasms/metabolism , 14-3-3 Proteins/genetics , Caspase 3 , Caspases/metabolism , Cloning, Molecular , Drug Resistance, Neoplasm/genetics , Gene Expression , Humans , Male , Poly(ADP-ribose) Polymerases/metabolism , Prostatic Neoplasms/genetics , Transfection , Tumor Cells, Cultured
10.
Trends Cell Biol ; 13(11): 584-92, 2003 Nov.
Article in English | MEDLINE | ID: mdl-14573352

ABSTRACT

The organization and sorting of regulatory information for transcription, replication and repair depends on components of nuclear architecture. It is necessary, therefore, to understand cellular processes within the context of intranuclear microenvironments that mediate the focal assembly of the machinery for transcription, replication and repair and which facilitate the orchestration of these essential processes. Here, we discuss how nuclear anatomy supports the temporal and spatial coordination of regulatory protein recruitment for combinatorial control.


Subject(s)
Cell Nucleus/physiology , DNA Repair , DNA Replication , Gene Expression Regulation , Animals , Apoptosis/physiology , Cell Nucleus/genetics , Cell Nucleus/ultrastructure , DNA Methylation , Humans , Models, Genetic , Nuclear Matrix/genetics , Nuclear Matrix/physiology
11.
J Biol Chem ; 278(10): 8261-8, 2003 Mar 07.
Article in English | MEDLINE | ID: mdl-12475992

ABSTRACT

The ability of p53 to alter, at the transcriptional level, the gene expression of downstream targets is critical for its role as a tumor suppressor. Most models of p53 activation postulate the stepwise recruitment by p53 of coactivators, histone acetyltransferases, and/or chromatin remodeling factors to a promoter region to facilitate the subsequent access of the general transcriptional machinery required for transcriptional induction. We demonstrate here, however, that the promoter regions for the p53 target genes, p21, 14-3-3sigma, and KARP-1, exist in a constitutively open conformation that is readily accessible to DNase I. This conformation was not altered by DNA damage or by whether p53 was present or absent in the cell. In contrast, p53 response elements, which resided outside the immediate promoter regions, existed within DNase I-resistant chromatin domains. Thus, p53 activation of downstream target genes occurs without p53 inducing chromatin alterations detectable by DNase I accessibility at either the promoter or the response element. As such, these data support models of p53 activation that do not require extensive chromatin alterations to support cognate gene expression.


Subject(s)
Biomarkers, Tumor , DNA Helicases , Deoxyribonuclease I/metabolism , Neoplasm Proteins , Promoter Regions, Genetic , Tumor Suppressor Protein p53/physiology , 14-3-3 Proteins , Base Sequence , Carrier Proteins/genetics , Cell Line , Cyclin-Dependent Kinase Inhibitor p21 , Cyclins/genetics , DNA Primers , Exonucleases/genetics , Exoribonucleases , Infrared Rays , Ku Autoantigen , Protein Binding , Reverse Transcriptase Polymerase Chain Reaction , Tumor Suppressor Protein p53/metabolism , Up-Regulation/radiation effects
12.
Nucleic Acids Res ; 30(8): 1713-24, 2002 Apr 15.
Article in English | MEDLINE | ID: mdl-11937624

ABSTRACT

The human Ku86 gene and an isoform, KARP-1 (Ku86 autoantigen related protein-1), encode overlapping, but differentially regulated, transcripts. Ku86 is constitutively transcribed at high levels and, although it plays a seminal role in DNA double-strand break repair, its expression is not induced by DNA damage. KARP-1, in contrast, is expressed constitutively only at low levels and its expression is induced by DNA damage in a p53-dependent fashion. The regulatory elements promoting KARP-1 gene expression and p53 responsiveness, however, were unknown. Here, we report that a strong DNase I hypersensitive site (DHS) resides approximately 25 kb upstream from the Ku86 promoter. This DHS is encompassed by a hypomethylated CpG island. Reporter assays demonstrated that this region corresponded to a promoter(s), which promoted transcription of peroxisomal trans-2-enoyl CoA reductase in the centromeric direction and KARP-1 in the telomeric direction. KARP-1 primer extension products were mapped to this CpG island in the correct transcriptional orientation confirming that KARP-1 transcription initiates from this site. Moreover, a p53 response element within the first intron of the KARP-1 transcriptional unit was identified using chromatin immunoprecipitation and antibodies specific to activated forms of p53. These data expand our understanding of this important DNA repair locus.


Subject(s)
Carrier Proteins/genetics , CpG Islands , DNA Helicases , NADH, NADPH Oxidoreductases , Response Elements , Transcriptional Activation , Tumor Suppressor Protein p53/metabolism , 5' Flanking Region , Base Sequence , Carrier Proteins/biosynthesis , DNA Methylation , Deoxyribonuclease I/chemistry , Fatty Acid Desaturases/biosynthesis , Fatty Acid Desaturases/genetics , Humans , Ku Autoantigen , Molecular Sequence Data , Oxidoreductases Acting on CH-CH Group Donors , RNA, Messenger/biosynthesis , Radiation, Ionizing , Transcription Initiation Site , Tumor Cells, Cultured
SELECTION OF CITATIONS
SEARCH DETAIL
...