Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 11 de 11
Filter
1.
Glia ; 64(1): 175-94, 2016 Jan.
Article in English | MEDLINE | ID: mdl-26507463

ABSTRACT

In mammals, large caliber axons are ensheathed by myelin, a glial specialization supporting axon integrity and conferring accelerated and energy-efficient action potential conduction. Myelin basic protein (MBP) is required for normal myelin elaboration with maximal mbp transcription in oligodendrocytes requiring the upstream M3 enhancer. To further characterize the mechanism regulating mbp transcription, we defined M3 structure/function relationships by evaluating its evolutionary conservation, DNA footprints and the developmental programing conferred in mice by M3 derivatives. Multiple M3 regulatory element combinations were found to drive expression in oligodendrocytes and Schwann cells with a minimal 129 bp sequence conferring expression in oligodendrocytes throughout myelin elaboration, maintenance and repair. Unexpectedly, M3 derivatives conferred markedly different spatial and temporal expression programs thus illuminating striking transcriptional heterogeneity within post-mitotic oligodendrocytes. Finally, one M3 derivative engaged only during primary myelination, not during adult remyelination, demonstrating that transcriptional regulation in the two states is not equivalent.


Subject(s)
Gene Regulatory Networks , Myelin Basic Protein/genetics , Myelin Basic Protein/metabolism , Myelin Sheath/genetics , Myelin Sheath/metabolism , Animals , Avian Proteins/genetics , Avian Proteins/metabolism , Base Sequence , Brain/growth & development , Brain/metabolism , Chickens , Conserved Sequence , Immunohistochemistry , In Situ Hybridization , Male , Mice, Transgenic , Molecular Sequence Data , Mutation , Optic Nerve/growth & development , Optic Nerve/metabolism , Sciatic Nerve/growth & development , Sciatic Nerve/metabolism , Sequence Alignment , Spinal Cord/growth & development , Spinal Cord/metabolism , beta-Galactosidase/metabolism
2.
J Mol Diagn ; 14(5): 467-75, 2012 Sep.
Article in English | MEDLINE | ID: mdl-22874498

ABSTRACT

Individuals and families carrying mutations in BRCA1 and BRCA2 (BRCA1/2) have a markedly elevated risk of developing breast and ovarian cancers. The first-generation of BRCA1/2 mutation analysis targeted only the coding exons and has implicated protein-truncating mutations (indel, nonsense) in BRCA1/2 inactivation. Recently, heritable breast cancers have also been attributed to other exonic mutations (missense, silent) and mutations in introns and untranslated regions. However, analysis of these alterations has been prohibitively laborious and cost intensive, and the proportion of cases carrying mutations in unscreened regions of BRCA1/2 and other predisposition genes is unknown. We have developed and validated a next-generation sequencing (NGS) approach for BRCA1/2 mutation analysis by applying long-range PCR and deep sequencing. Genomic DNA from familial breast cancer patients (N = 12) were screened and NGS successfully identified all 19 distinct (51 total) BRCA1 and 35 distinct (63 total) BRCA2 sequence alterations detectable by the Sanger sequencing, with no false-negative or positive results. In addition, we report the robust detection of variants from introns and untranslated regions. These results illustrate that NGS can provide comprehensive genetic information more quickly, accurately, and at a lower cost than conventional approaches, and we propose NGS to be a more effective method for BRCA1/2 mutational analysis. Advances in NGS will play an important role in enabling molecular diagnostics and personalized treatment of breast and ovarian cancers.


Subject(s)
Breast Neoplasms/genetics , Genes, BRCA1 , Genes, BRCA2 , High-Throughput Nucleotide Sequencing , Polymerase Chain Reaction , Breast Neoplasms/diagnosis , Female , Humans , Mutation , Reproducibility of Results , Sensitivity and Specificity
3.
Am J Hum Genet ; 90(3): 524-32, 2012 Mar 09.
Article in English | MEDLINE | ID: mdl-22365150

ABSTRACT

We have previously shown that rheumatoid arthritis (RA) risk alleles overlap between different ethnic groups. Here, we utilize a multiethnic approach to show that we can effectively discover RA risk alleles. Thirteen putatively associated SNPs that had not yet exceeded genome-wide significance (p < 5 × 10(-8)) in our previous RA genome-wide association study (GWAS) were analyzed in independent sample sets consisting of 4,366 cases and 17,765 controls of European, African American, and East Asian ancestry. Additionally, we conducted an overall association test across all 65,833 samples (a GWAS meta-analysis plus the replication samples). Of the 13 SNPs investigated, four were significantly below the study-wide Bonferroni corrected p value threshold (p < 0.0038) in the replication samples. Two SNPs (rs3890745 at the 1p36 locus [p = 2.3 × 10(-12)] and rs2872507 at the 17q12 locus [p = 1.7 × 10(-9)]) surpassed genome-wide significance in all 16,659 RA cases and 49,174 controls combined. We used available GWAS data to fine map these two loci in Europeans and East Asians, and we found that the same allele conferred risk in both ethnic groups. A series of bioinformatic analyses identified TNFRSF14-MMEL1 at the 1p36 locus and IKZF3-ORMDL3-GSDMB at the 17q12 locus as the genes most likely associated with RA. These findings demonstrate empirically that a multiethnic approach is an effective strategy for discovering RA risk loci, and they suggest that combining GWASs across ethnic groups represents an efficient strategy for gaining statistical power.


Subject(s)
Arthritis, Rheumatoid/ethnology , Arthritis, Rheumatoid/genetics , Chromosomes, Human, Pair 17 , Chromosomes, Human, Pair 1 , Genetic Loci , Alleles , Case-Control Studies , Computational Biology/methods , Ethnicity/genetics , Genetic Predisposition to Disease , Genome-Wide Association Study/methods , Genotype , Humans , Ikaros Transcription Factor/genetics , Linkage Disequilibrium , Membrane Proteins/genetics , Neoplasm Proteins/genetics , Neprilysin/genetics , Polymorphism, Single Nucleotide , Receptors, Tumor Necrosis Factor, Member 14/genetics
4.
Gynecol Oncol ; 109(3): 384-7, 2008 Jun.
Article in English | MEDLINE | ID: mdl-18405947

ABSTRACT

OBJECTIVES: Inherited mutations in the MLH1 gene are associated with a proportion of families with the hereditary non-polyposis colon cancer syndrome (HNPCC). The cardinal features of the syndrome are a predisposition to colon, endometrial and ovarian cancers. Recently, it has been shown that a non-coding polymorphic variant in MLH1 (G>A nt-93) predisposes to colon and endometrial cancer, but with much reduced penetrance. We sought to establish whether or not this polymorphic variant also predisposes to ovarian cancer. METHODS: We genotyped 899 women with invasive ovarian cancer and 931 controls for the G>A nt-93 variant. RESULTS: The presence of the variant was associated with a modest, but highly significant risk of ovarian cancer (OR=1.5; 95% CI 1.3-1.9; p=0.00005). The association was present in cancers of all histologies except clear cell, and in all ethnic groups. CONCLUSIONS: The G>A nt-93 variant of the MLH1 gene is associated with an increased risk of invasive ovarian cancer.


Subject(s)
Adaptor Proteins, Signal Transducing/genetics , Nuclear Proteins/genetics , Ovarian Neoplasms/genetics , Case-Control Studies , Ethnicity/genetics , Family Health , Female , Genetic Predisposition to Disease , Genotype , Humans , Middle Aged , MutL Protein Homolog 1 , Ontario/epidemiology , Ovarian Neoplasms/epidemiology , Ovarian Neoplasms/pathology , Polymorphism, Single Nucleotide , Promoter Regions, Genetic
5.
Mol Diagn Ther ; 10(6): 353-66, 2006.
Article in English | MEDLINE | ID: mdl-17154652

ABSTRACT

Genetic variation in specific G-protein coupled receptors (GPCRs) is associated with a spectrum of respiratory disease predispositions and drug response phenotypes. Although certain GPCR gene variants can be disease-causing through the expression of inactive, overactive, or constitutively active receptor proteins, many more GPCR gene variants confer risk for potentially deleterious endophenotypes. Endophenotypes are traits, such as bronchiole hyperactivity, atopy, and aspirin intolerant asthma, which have a strong genetic component and are risk factors for a variety of more complex outcomes that may include disease states. GPCR genes implicated in asthma endophenotypes include variants of the cysteinyl leukotriene receptors (CYSLTR1 and CYSLTR2), and prostaglandin D2 receptors (PTGDR and CRTH2), thromboxane A2 receptor (TBXA2R), beta2-adrenergic receptor (ADRB2), chemokine receptor 5 (CCR5), and the G protein-coupled receptor associated with asthma (GPRA). This review of the contribution of variability in these genes places the contribution of the cysteinyl leukotriene system to respiratory endophenotypes in perspective. The genetic variant(s) of receptors that are associated with endophenotypes are discussed in the context of the extent to which they contribute to a disease phenotype or altered drug efficacy.


Subject(s)
Asthma/genetics , Drug Hypersensitivity/genetics , Leukotrienes/genetics , Receptors, G-Protein-Coupled/genetics , Receptors, Leukotriene/genetics , Aspirin/metabolism , Asthma/metabolism , Genetic Variation , Humans , Inactivation, Metabolic/genetics , Pharmacogenetics , Phenotype , Polymorphism, Single Nucleotide , Receptors, G-Protein-Coupled/metabolism , Signal Transduction/drug effects , Signal Transduction/genetics
6.
Can J Cardiol ; 22(13): 1133-6, 2006 Nov.
Article in English | MEDLINE | ID: mdl-17102831

ABSTRACT

BACKGROUND: It has long been debated whether patients with atrial septal defect (ASD) Eisenmenger syndrome have idiopathic pulmonary arterial hypertension with an incidental ASD or severe pulmonary hypertension on the basis of their ASD shunt magnitude alone. HYPOTHESIS: It was hypothesized that if ASD Eisenmenger patients had idiopathic pulmonary arterial hypertension with an incidental ASD, a mutation in the bone morphogenetic protein receptor-2 (BMPR2) would be found in some of these patients. PATIENTS AND METHODS: All adult patients with ASD Eisenmenger syndrome were identified from the databases of two adult congenital cardiac units, and were matched to a control group with similar types of ASDs and no pulmonary hypertension. Gene coding for BMPR2 was examined for mutation using denaturing high-performance liquid chromatography of the entire coding sequence. RESULTS: Eighteen adult patients with ASD Eisenmenger syndrome and 18 control patients were identified. ASD Eisenmenger patients had significantly larger ASDs than the control patients (3.7+/-1.2 cm versus 1.9+/-0.7 cm, P<0.01). A mutation in BMPR2 was not detected in either group. CONCLUSION: ASD Eisenmenger syndrome may occur without BMPR2 mutation. Whether shunt magnitude alone or in combination with yet another genetic mutation is responsible for the development of pulmonary hypertension in these patients remains to be determined.


Subject(s)
Bone Morphogenetic Protein Receptors, Type II/genetics , Eisenmenger Complex/genetics , Heart Septal Defects, Atrial/genetics , Mutation , Adult , Case-Control Studies , DNA Primers , Eisenmenger Complex/complications , Eisenmenger Complex/physiopathology , Exons , Female , Genetic Predisposition to Disease , Heart Septal Defects, Atrial/complications , Heart Septal Defects, Atrial/physiopathology , Humans , Hypertension, Pulmonary/etiology , Hypertension, Pulmonary/genetics , Hypertension, Pulmonary/physiopathology , Introns , Male , Middle Aged , Ontario , Pulmonary Wedge Pressure/genetics , Quebec , Sequence Analysis, DNA , Transcription, Genetic
7.
Clin Cancer Res ; 12(21): 6452-8, 2006 Nov 01.
Article in English | MEDLINE | ID: mdl-17085659

ABSTRACT

PURPOSE: Increased levels of serum human kallikrein-2 (hK2) and an hK2 gene (KLK2) variant are positively associated for prostate cancer, but the relationships between them remain unclear. We examined five variants of the KLK2 gene to further define its relevance to prostate cancer susceptibility and hK2 levels. EXPERIMENTAL DESIGN: We genotyped 645 men with biopsy-proven prostate cancer (cases) and 606 males with biopsies negative for prostate cancer (controls) for five additional single nucleotide polymorphisms (SNP) across the KLK2 gene and also tested for serum hK2 levels. These SNPs were identified from sequencing the KLK2 gene among 20 patients with aggressive prostate cancer. Odds ratios (OR) for prostate cancer detection and haplotype analysis were done. RESULTS: Among the SNPs studied, the A allele of the KLK2-SNP1 (G>A, rs2664155) and the T allele of the KLK2-SNP5 (C>T, rs198977) polymorphisms showed positive associations with prostate cancer, adjusted ORs for KLK2-SNP1 AG and AA genotypes being 1.4 [95% confidence interval (95% CI), 1.2-1.8; P=0.002] and for KLK2-SNP5 TT or CT genotypes being 1.3 (95% CI, 1.1-1.6; P=0.05). Haplotype analyses also revealed a significant association between prostate cancer and the haplotype containing both risk alleles (ACCTT), OR being 5.1 (95% CI, 1.6-6.5; P=0.005). Analysis of serum hK2 revealed hK2 levels to be significantly increased in association with KLK2-SNP1 AA and AG risk genotypes compared with the GG genotype (P=0.001) and also in association with the ACCTT risk haplotype compared with the most common non-risk haplotype (P=0.05). CONCLUSIONS: These findings suggest a role for the KLK2 gene in prostate cancer susceptibility and imply that this role may be realized at least in part by the induction of increases in hK2 production.


Subject(s)
Adenocarcinoma/blood , Biomarkers, Tumor/blood , Genetic Predisposition to Disease , Prostatic Neoplasms/blood , Tissue Kallikreins/blood , Tissue Kallikreins/genetics , Adult , Aged , Aged, 80 and over , Biopsy , DNA Primers , Genotype , Haplotypes , Humans , Male , Middle Aged , Polymorphism, Single Nucleotide
8.
Cancer Epidemiol Biomarkers Prev ; 15(9): 1636-40, 2006 Sep.
Article in English | MEDLINE | ID: mdl-16985024

ABSTRACT

Women with germ-line mutations in the mismatch repair genes (responsible for hereditary nonpolyposis colorectal cancer) face an increased risk of colonic and endometrial cancer. However, these germ-line mutations are rare and are responsible for fewer than 1% of endometrial cancers. Therefore, we examined whether or not common variants of the hereditary nonpolyposis colorectal cancer-associated genes might also be associated with an increased risk of endometrial cancer. Three single-nucleotide polymorphisms were selected in the MLH1 and MSH2 mismatch repair genes. All the various 672 women with endometrial cancer and 880 controls were genotyped. Each of these three single-nucleotide polymorphisms was associated with an increased risk of endometrial cancer. Carriers of the MLH1 nt-93 A allele were at a 1.5-fold increased risk of developing endometrial cancer compared with controls [95% confidence interval (95% CI), 1.2-2.0; P = 0.001]. The risk was higher for homozygote carriers [odds ratio (OR), 1.9; 95% CI, 1.2-3.2; P = 0.009]. For carriers of the MSH2 rs2303428 C allele, the OR was 1.4 (95% CI, 1.0-1.9; P = 0.05), and for carriers of the MSH2 rs2059520 G allele, the OR was 1.3 (95% CI, 1.0-1.7; P = 0.03). More than 9% of endometrial cancer cases carried a variant allele in both MLH1 and MSH2. For these women, the risk of endometrial cancer was particularly high (OR, 2.1; 95% CI, 1.2-3.6; P = 0.005). For patients younger than 50 years at diagnosis who carried both variants, the risk was even higher (OR, 3.4; 95% CI, 1.7-6.6; P = 0.0005). In summary, two common variant alleles of the MLH1 and MSH2 genes make a substantial contribution to endometrial cancer incidence in Ontario.


Subject(s)
Carrier Proteins/genetics , DNA Repair/genetics , Endometrial Neoplasms/genetics , MutS Homolog 2 Protein/genetics , Nuclear Proteins/genetics , Polymorphism, Single Nucleotide , Adaptor Proteins, Signal Transducing , Adolescent , Adult , Aged , Aged, 80 and over , Endometrial Neoplasms/etiology , Female , Humans , Middle Aged , MutL Protein Homolog 1
9.
Am J Gastroenterol ; 101(3): 572-80, 2006 Mar.
Article in English | MEDLINE | ID: mdl-16542294

ABSTRACT

OBJECTIVES: The complete elucidation of genetic variants that contribute to inflammatory bowel disease (IBD) will likely include variants that increase risk to both Crohn's disease and ulcerative colitis as well as variants that increase risk for particular phenotypic subsets. The purpose of this study was to assess phenotypic subsets that contribute to the major IBD susceptibility loci. METHODS: This linkage study encompassed 904 affected relative pairs, representing the largest combined phenotyped cohort to date, and allowing for meaningful subset analyses. Genetic linkage data were stratified by disease location and age at diagnosis. RESULTS: We establish that some loci, notably the IBD3 and chromosome 3q linkage regions demonstrate contributions from both small intestine and colon cohorts, whereas others, notably the IBD1 (NOD2/CARD15) and IBD2 regions increase risk for small intestine or colon inflammation, respectively. The strongest linkage evidence in this study was for the subset of extensive ulcerative colitis in the region of IBD2 (lod 3.27; p < 0.001). Evidence for linkage in the region of NOD2/CARD15 (IBD1) was stronger for the subset of Crohn's patients with ileal disease (lod 2.56; p= 0.035) compared to the overall Crohn's group, consistent with previous findings that NOD2/CARD15 variants are associated with ileal disease. CONCLUSIONS: Analyses incorporating disease location in IBD increase the power and enhance the accuracy of genomic localization. Our data provide strong evidence that extensive ulcerative colitis represents a pathophysiologic subset of IBD.


Subject(s)
Cell Cycle Proteins/genetics , Chromosome Mapping , Colitis, Ulcerative/genetics , Intracellular Signaling Peptides and Proteins/genetics , Phenotype , Saccharomyces cerevisiae Proteins/genetics , Adaptor Proteins, Signal Transducing/genetics , Cohort Studies , Colon/immunology , Crohn Disease/genetics , Genetic Predisposition to Disease/genetics , Genetic Variation , Humans , Intestinal Mucosa/immunology , Intestine, Small/immunology , Lod Score , Nod1 Signaling Adaptor Protein , Nod2 Signaling Adaptor Protein
10.
Pharmacogenetics ; 13(10): 641-9, 2003 Oct.
Article in English | MEDLINE | ID: mdl-14515063

ABSTRACT

The clinical heterogeneity of asthma suggests that the contribution of genetic variability in candidate gene loci to well-defined phenotypes, such as atopy, may be examined to identify appropriate genetic risk factors for asthma. The gene encoding the cysteinyl leukotriene 2 (CysLT2) receptor has been implicated in atopy since it is localized to a region of chromosome 13q14 that has been linked to atopy in several populations and the cysteinyl leukotrienes are known to activate eosinophils and mast cells in atopy. Accordingly, we analysed the contribution of CysLT2 receptor gene variation to atopy in the inhabitants of Tristan da Cunha, a population characterized by both a founder effect and a 47% prevalence of atopy. Single-stranded conformational polymorphism analysis revealed four variants. Among these, the M201V [corrected] variant was activated with four-fold less potency by leukotriene D4 (LTD4) in a calcium flux assay. The CysLT2 receptor partial agonist, BAY u9773, also showed four-fold lower potency on the M201V [corrected] variant. The M201V [corrected] mutation is located within the extracellular region of the fifth transmembrane spanning domain of CysLT2 receptor, a position that may alter ligand binding and effector signalling. The novel M201V [corrected] CysLT2 receptor variant was associated with atopy (21%) on Tristan da Cunha compared with those who were non-atopic (7%) (Fisher's exact test, P=0.0016) in a manner that was independent of asthma (two-way ANOVA, P=0.0015). This represents the first association of a coding mutation in the CysLT2 receptor gene, located on chromosome 13q14, with the atopic phenotype found in the Tristan da Cunha population.


Subject(s)
Genetic Variation , Hypersensitivity, Immediate/genetics , Membrane Proteins/genetics , Receptors, Leukotriene/genetics , SRS-A/analogs & derivatives , Black or African American/genetics , Asthma/blood , Asthma/ethnology , Asthma/genetics , Atlantic Islands/epidemiology , Calcium/metabolism , Case-Control Studies , Chromosomes, Human, Pair 13/genetics , DNA/blood , DNA/genetics , DNA Primers/chemistry , Founder Effect , Humans , Hypersensitivity, Immediate/epidemiology , Hypersensitivity, Immediate/ethnology , Leukotriene D4/metabolism , Membrane Proteins/agonists , Phenotype , Polymerase Chain Reaction , Polymorphism, Restriction Fragment Length , Polymorphism, Single-Stranded Conformational , Receptors, Leukotriene/agonists , SRS-A/pharmacology , White People/genetics
11.
J Biol Chem ; 278(41): 40057-66, 2003 Oct 10.
Article in English | MEDLINE | ID: mdl-12869565

ABSTRACT

Src family protein-tyrosine kinases, which play an important role in signal integration, have been implicated in tumorigenesis in multiple lineages, including breast cancer. We demonstrate, herein, that Src kinases regulate the phosphatidylinositol 3-kinase (PI3K) signaling cascade via altering the function of the PTEN tumor suppressor. Overexpression of activated Src protein-tyrosine kinases in PTEN-deficient breast cancer cells does not alter AKT phosphorylation, an indicator of signal transduction through the PI3K pathway. However, in the presence of functional PTEN, Src reverses the activity of PTEN, resulting in an increase in AKT phosphorylation. Activated Src reduces the ability of PTEN to dephosphorylate phosphatidylinositols in micelles and promotes AKT translocation to cellular plasma membranes but does not alter PTEN activity toward water-soluble phosphatidylinositols. Thus, Src may alter the capacity of the PTEN C2 domain to bind cellular membranes rather than directly interfering with PTEN enzymatic activity. Tyrosine phosphorylation of PTEN is increased in breast cancer cells treated with pervanadate, suggesting that PTEN contains sites for tyrosine phosphorylation. Src kinase inhibitors markedly decreased pervanadate-mediated tyrosine phosphorylation of PTEN. Further, expression of activated Src results in marked tyrosine phosphorylation of PTEN. SHP-1, a SH2 domain-containing protein-tyrosine phosphatase, selectively binds and dephosphorylates PTEN in Src transfected cells. Both Src inhibitors and SHP-1 overexpression reverse Src-induced loss of PTEN function. Coexpression of PTEN with activated Src reduces the stability of PTEN. Taken together, the data indicate that activated Src inhibits PTEN function leading to alterations in signaling through the PI3K/AKT pathway.


Subject(s)
Phosphatidylinositol 3-Kinases/metabolism , Phosphoric Monoester Hydrolases/metabolism , Protein Serine-Threonine Kinases , Proto-Oncogene Proteins/metabolism , Tumor Suppressor Proteins/metabolism , src-Family Kinases/metabolism , Animals , Breast Neoplasms/metabolism , COS Cells , Cell Line, Tumor , Enzyme Activation , Female , Humans , In Vitro Techniques , Intracellular Signaling Peptides and Proteins , Mutation , PTEN Phosphohydrolase , Phosphoric Monoester Hydrolases/chemistry , Phosphoric Monoester Hydrolases/genetics , Phosphorylation , Protein Tyrosine Phosphatase, Non-Receptor Type 6 , Protein Tyrosine Phosphatases/metabolism , Proto-Oncogene Proteins c-akt , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Signal Transduction , Transfection , Tumor Suppressor Proteins/chemistry , Tumor Suppressor Proteins/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...