Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 16 de 16
Filter
1.
Am J Hum Genet ; 103(1): 3-18, 2018 07 05.
Article in English | MEDLINE | ID: mdl-29909963

ABSTRACT

Multiple primary tumors (MPTs) affect a substantial proportion of cancer survivors and can result from various causes, including inherited predisposition. Currently, germline genetic testing of MPT-affected individuals for variants in cancer-predisposition genes (CPGs) is mostly targeted by tumor type. We ascertained pre-assessed MPT individuals (with at least two primary tumors by age 60 years or at least three by 70 years) from genetics centers and performed whole-genome sequencing (WGS) on 460 individuals from 440 families. Despite previous negative genetic assessment and molecular investigations, pathogenic variants in moderate- and high-risk CPGs were detected in 67/440 (15.2%) probands. WGS detected variants that would not be (or were not) detected by targeted resequencing strategies, including low-frequency structural variants (6/440 [1.4%] probands). In most individuals with a germline variant assessed as pathogenic or likely pathogenic (P/LP), at least one of their tumor types was characteristic of variants in the relevant CPG. However, in 29 probands (42.2% of those with a P/LP variant), the tumor phenotype appeared discordant. The frequency of individuals with truncating or splice-site CPG variants and at least one discordant tumor type was significantly higher than in a control population (χ2 = 43.642; p ≤ 0.0001). 2/67 (3%) probands with P/LP variants had evidence of multiple inherited neoplasia allele syndrome (MINAS) with deleterious variants in two CPGs. Together with variant detection rates from a previous series of similarly ascertained MPT-affected individuals, the present results suggest that first-line comprehensive CPG analysis in an MPT cohort referred to clinical genetics services would detect a deleterious variant in about a third of individuals.


Subject(s)
Genetic Predisposition to Disease/genetics , Genetic Variation/genetics , Neoplasms, Multiple Primary/genetics , Adult , Aged , Biomarkers, Tumor/genetics , Female , Genetic Testing/methods , Germ-Line Mutation/genetics , Humans , Male , Middle Aged , Phenotype
2.
Am J Hum Genet ; 100(1): 75-90, 2017 01 05.
Article in English | MEDLINE | ID: mdl-28041643

ABSTRACT

Inherited retinal disease is a common cause of visual impairment and represents a highly heterogeneous group of conditions. Here, we present findings from a cohort of 722 individuals with inherited retinal disease, who have had whole-genome sequencing (n = 605), whole-exome sequencing (n = 72), or both (n = 45) performed, as part of the NIHR-BioResource Rare Diseases research study. We identified pathogenic variants (single-nucleotide variants, indels, or structural variants) for 404/722 (56%) individuals. Whole-genome sequencing gives unprecedented power to detect three categories of pathogenic variants in particular: structural variants, variants in GC-rich regions, which have significantly improved coverage compared to whole-exome sequencing, and variants in non-coding regulatory regions. In addition to previously reported pathogenic regulatory variants, we have identified a previously unreported pathogenic intronic variant in CHM in two males with choroideremia. We have also identified 19 genes not previously known to be associated with inherited retinal disease, which harbor biallelic predicted protein-truncating variants in unsolved cases. Whole-genome sequencing is an increasingly important comprehensive method with which to investigate the genetic causes of inherited retinal disease.


Subject(s)
DNA Mutational Analysis , Genetic Variation/genetics , Genome, Human/genetics , Retinal Diseases/genetics , Adaptor Proteins, Signal Transducing/genetics , Alleles , Base Sequence , Choroideremia/genetics , Ethnicity/genetics , Exome/genetics , Female , Genes, Recessive/genetics , Humans , Introns/genetics , Male , Mutation , Rare Diseases/genetics
3.
Nature ; 493(7432): 406-10, 2013 Jan 17.
Article in English | MEDLINE | ID: mdl-23242139

ABSTRACT

Improved sequencing technologies offer unprecedented opportunities for investigating the role of rare genetic variation in common disease. However, there are considerable challenges with respect to study design, data analysis and replication. Using pooled next-generation sequencing of 507 genes implicated in the repair of DNA in 1,150 samples, an analytical strategy focused on protein-truncating variants (PTVs) and a large-scale sequencing case-control replication experiment in 13,642 individuals, here we show that rare PTVs in the p53-inducible protein phosphatase PPM1D are associated with predisposition to breast cancer and ovarian cancer. PPM1D PTV mutations were present in 25 out of 7,781 cases versus 1 out of 5,861 controls (P = 1.12 × 10(-5)), including 18 mutations in 6,912 individuals with breast cancer (P = 2.42 × 10(-4)) and 12 mutations in 1,121 individuals with ovarian cancer (P = 3.10 × 10(-9)). Notably, all of the identified PPM1D PTVs were mosaic in lymphocyte DNA and clustered within a 370-base-pair region in the final exon of the gene, carboxy-terminal to the phosphatase catalytic domain. Functional studies demonstrate that the mutations result in enhanced suppression of p53 in response to ionizing radiation exposure, suggesting that the mutant alleles encode hyperactive PPM1D isoforms. Thus, although the mutations cause premature protein truncation, they do not result in the simple loss-of-function effect typically associated with this class of variant, but instead probably have a gain-of-function effect. Our results have implications for the detection and management of breast and ovarian cancer risk. More generally, these data provide new insights into the role of rare and of mosaic genetic variants in common conditions, and the use of sequencing in their identification.


Subject(s)
Breast Neoplasms/genetics , Genetic Predisposition to Disease/genetics , Mosaicism , Mutation , Ovarian Neoplasms/genetics , Phosphoprotein Phosphatases/genetics , Alleles , Cluster Analysis , Exons , Female , Humans , Isoenzymes/genetics , Lymphocytes/metabolism , Protein Phosphatase 2C , Sequence Analysis, DNA , Tumor Suppressor Protein p53/metabolism
4.
BMJ ; 345: e5660, 2012 Sep 06.
Article in English | MEDLINE | ID: mdl-22956590

ABSTRACT

OBJECTIVE: To estimate the risk of breast cancer associated with diagnostic radiation in carriers of BRCA1/2 mutations. DESIGN: Retrospective cohort study (GENE-RAD-RISK). SETTING: Three nationwide studies (GENEPSO, EMBRACE, HEBON) in France, United Kingdom, and the Netherlands, PARTICIPANTS: 1993 female carriers of BRCA1/2 mutations recruited in 2006-09. MAIN OUTCOME MEASURE: Risk of breast cancer estimated with a weighted Cox proportional hazards model with a time dependent individually estimated cumulative breast dose, based on nominal estimates of organ dose and frequency of self reported diagnostic procedures. To correct for potential survival bias, the analysis excluded carriers who were diagnosed more than five years before completion of the study questionnaire. RESULTS: In carriers of BRCA1/2 mutations any exposure to diagnostic radiation before the age of 30 was associated with an increased risk of breast cancer (hazard ratio 1.90, 95% confidence interval 1.20 to 3.00), with a dose-response pattern. The risks by quarter of estimated cumulative dose <0.0020 Gy, ≥ 0.0020-0.0065 Gy, ≥ 0.0066-0.0173 Gy, and ≥ 0.0174 Gy were 1.63 (0.96 to 2.77), 1.78 (0.88 to 3.58), 1.75 (0.72 to 4.25), and 3.84 (1.67 to 8.79), respectively. Analyses on the different types of diagnostic procedures showed a pattern of increasing risk with increasing number of radiographs before age 20 and before age 30 compared with no exposure. A history of mammography before age 30 was also associated with an increased risk of breast cancer (hazard ratio 1.43, 0.85 to 2.40). Sensitivity analysis showed that this finding was not caused by confounding by indication of family history. CONCLUSION: In this large European study among carriers of BRCA1/2 mutations, exposure to diagnostic radiation before age 30 was associated with an increased risk of breast cancer at dose levels considerably lower than those at which increases have been found in other cohorts exposed to radiation. The results of this study support the use of non-ionising radiation imaging techniques (such as magnetic resonance imaging) as the main tool for surveillance in young women with BRCA1/2 mutations.


Subject(s)
Breast Neoplasms/etiology , Genes, BRCA1 , Genes, BRCA2 , Heterozygote , Mammography/adverse effects , Adult , Age Factors , Breast/radiation effects , Breast Neoplasms/genetics , Dose-Response Relationship, Radiation , Female , Humans , Middle Aged , Mutation/radiation effects , Proportional Hazards Models , Retrospective Studies , Risk Factors , Young Adult
5.
Hum Mol Genet ; 21(4): 958-62, 2012 Feb 15.
Article in English | MEDLINE | ID: mdl-22072393

ABSTRACT

There have been few definitive examples of gene-gene interactions in humans. Through mutational analyses in 7325 individuals, we report four interactions (defined as departures from a multiplicative model) between mutations in the breast cancer susceptibility genes ATM and CHEK2 with BRCA1 and BRCA2 (case-only interaction between ATM and BRCA1/BRCA2 combined, P = 5.9 × 10(-4); ATM and BRCA1, P= 0.01; ATM and BRCA2, P= 0.02; CHEK2 and BRCA1/BRCA2 combined, P = 2.1 × 10(-4); CHEK2 and BRCA1, P= 0.01; CHEK2 and BRCA2, P= 0.01). The interactions are such that the resultant risk of breast cancer is lower than the multiplicative product of the constituent risks, and plausibly reflect the functional relationships of the encoded proteins in DNA repair. These findings have important implications for models of disease predisposition and clinical translation.


Subject(s)
Breast Neoplasms/genetics , Cell Cycle Proteins/genetics , DNA-Binding Proteins/genetics , Genes, BRCA1 , Genes, BRCA2 , Genetic Predisposition to Disease/genetics , Protein Serine-Threonine Kinases/genetics , Tumor Suppressor Proteins/genetics , Ataxia Telangiectasia Mutated Proteins , Checkpoint Kinase 2 , DNA Mutational Analysis , Family Health , Female , Humans , Models, Genetic , Pedigree , United Kingdom
6.
Hum Mol Genet ; 20(23): 4732-47, 2011 Dec 01.
Article in English | MEDLINE | ID: mdl-21890493

ABSTRACT

Mutations in the BRCA1 gene substantially increase a woman's lifetime risk of breast cancer. However, there is great variation in this increase in risk with several genetic and non-genetic modifiers identified. The BRCA1 protein plays a central role in DNA repair, a mechanism that is particularly instrumental in safeguarding cells against tumorigenesis. We hypothesized that polymorphisms that alter the expression and/or function of BRCA1 carried on the wild-type (non-mutated) copy of the BRCA1 gene would modify the risk of breast cancer in carriers of BRCA1 mutations. A total of 9874 BRCA1 mutation carriers were available in the Consortium of Investigators of Modifiers of BRCA1/2 (CIMBA) for haplotype analyses of BRCA1. Women carrying the rare allele of single nucleotide polymorphism rs16942 on the wild-type copy of BRCA1 were at decreased risk of breast cancer (hazard ratio 0.86, 95% confidence interval 0.77-0.95, P = 0.003). Promoter in vitro assays of the major BRCA1 haplotypes showed that common polymorphisms in the regulatory region alter its activity and that this effect may be attributed to the differential binding affinity of nuclear proteins. In conclusion, variants on the wild-type copy of BRCA1 modify risk of breast cancer among carriers of BRCA1 mutations, possibly by altering the efficiency of BRCA1 transcription.


Subject(s)
Alleles , BRCA1 Protein/genetics , Breast Neoplasms/genetics , Genetic Predisposition to Disease , Heterozygote , Mutation/genetics , Polymorphism, Single Nucleotide/genetics , Electrophoretic Mobility Shift Assay , Female , Genes, Reporter/genetics , Genetic Association Studies , Haplotypes/genetics , HeLa Cells , Humans , Linkage Disequilibrium/genetics , Luciferases/metabolism , Risk Factors
7.
Nat Genet ; 43(9): 879-882, 2011 Aug 07.
Article in English | MEDLINE | ID: mdl-21822267

ABSTRACT

Recently, RAD51C mutations were identified in families with breast and ovarian cancer. This observation prompted us to investigate the role of RAD51D in cancer susceptibility. We identified eight inactivating RAD51D mutations in unrelated individuals from 911 breast-ovarian cancer families compared with one inactivating mutation identified in 1,060 controls (P = 0.01). The association found here was principally with ovarian cancer, with three mutations identified in the 59 pedigrees with three or more individuals with ovarian cancer (P = 0.0005). The relative risk of ovarian cancer for RAD51D mutation carriers was estimated to be 6.30 (95% CI 2.86-13.85, P = 4.8 × 10(-6)). By contrast, we estimated the relative risk of breast cancer to be 1.32 (95% CI 0.59-2.96, P = 0.50). These data indicate that RAD51D mutation testing may have clinical utility in individuals with ovarian cancer and their families. Moreover, we show that cells deficient in RAD51D are sensitive to treatment with a PARP inhibitor, suggesting a possible therapeutic approach for cancers arising in RAD51D mutation carriers.


Subject(s)
DNA-Binding Proteins/genetics , Genetic Predisposition to Disease , Germ-Line Mutation , Ovarian Neoplasms/genetics , Age Factors , Animals , CHO Cells , Cricetinae , Cricetulus , Drug Resistance, Neoplasm/genetics , Female , Heterozygote , Humans , Pedigree , Poly(ADP-ribose) Polymerase Inhibitors
8.
Cancer Res ; 71(17): 5792-805, 2011 Sep 01.
Article in English | MEDLINE | ID: mdl-21799032

ABSTRACT

Inherited BRCA1 mutations confer elevated cancer risk. Recent studies have identified genes that encode proteins that interact with BRCA1 as modifiers of BRCA1-associated breast cancer. We evaluated a comprehensive set of genes that encode most known BRCA1 interactors to evaluate the role of these genes as modifiers of cancer risk. A cohort of 2,825 BRCA1 mutation carriers was used to evaluate the association of haplotypes at ATM, BRCC36, BRCC45 (BRE), BRIP1 (BACH1/FANCJ), CTIP, ABRA1 (FAM175A), MERIT40, MRE11A, NBS1, PALB2 (FANCN), RAD50, RAD51, RAP80, and TOPBP1, and was associated with time to breast and ovarian cancer diagnosis. Statistically significant false discovery rate (FDR) adjusted P values for overall association of haplotypes (P(FDR)) with breast cancer were identified at ATM (P(FDR) = 0.029), BRCC45 (P(FDR) = 0.019), BRIP1 (P(FDR) = 0.008), CTIP (P(FDR) = 0.017), MERIT40 (P(FDR) = 0.019), NBS1 (P(FDR) = 0.003), RAD50 (P(FDR) = 0.014), and TOPBP1 (P(FDR) = 0.011). Haplotypes at ABRA1 (P(FDR) = 0.007), BRCC45 (P(FDR) = 0.016 and P(FDR) = 0.005 in two haplotype blocks), and RAP80 (P(FDR) < 0.001) were associated with ovarian cancer risk. Overall, the data suggest that genomic variation at multiple loci that encode proteins that interact biologically with BRCA1 are associated with modified breast cancer and ovarian cancer risk in women who carry BRCA1 mutations.


Subject(s)
BRCA1 Protein/genetics , Breast Neoplasms/epidemiology , Breast Neoplasms/genetics , Ovarian Neoplasms/epidemiology , Ovarian Neoplasms/genetics , Adult , Aged , Aged, 80 and over , Cohort Studies , Female , Genetic Loci , Genetic Variation , Humans , Middle Aged , Mutation , Risk , Young Adult
9.
J Med Genet ; 48(4): 261-5, 2011 Apr.
Article in English | MEDLINE | ID: mdl-21278391

ABSTRACT

BACKGROUND: Meningiomas have been reported to occur in approximately 50% of neurofibromatosis type 2 (NF2) patients. The NF2 gene is commonly biallelically inactivated in both schwannomas and meningiomas. The spectrum of NF2 mutations consists mainly of truncating (nonsense and frameshift) mutations. A smaller number of patients have missense mutations, which are associated with a milder disease phenotype. METHODS: This study analysed the cumulative incidence and gender effects as well as the genotype-phenotype correlation between the position of the NF2 mutation and the occurrence of cranial meningiomas in a cohort of 411 NF2 patients with proven NF2 mutations. RESULTS AND CONCLUSION: Patients with mutations in exon 14 or 15 were least likely to develop meningiomas. Cumulative risk of cranial meningioma to age 50 years was 70% for exons 1-3, 81% for exons 4-6, 49% for exons 7-9, 56% for exons 10-13, and 28% for exons 14-15. In the cohort of 411 patients, no overall gender bias was found for occurrence of meningioma in NF2 disease. Cumulative incidence of meningioma was close to 80% by 70 years of age for both males and females, but incidence by age 20 years was slightly increased in males (male 25%, female 18%; p=0.023). Conversely, an increased risk of meningiomas in women with mosaic NF2 disease was also found.


Subject(s)
Genes, Neurofibromatosis 2 , Genetic Association Studies , Meningeal Neoplasms/genetics , Meningioma/genetics , Neurofibromatosis 2/genetics , Cohort Studies , Exons , Female , Humans , Male , Meningeal Neoplasms/complications , Meningeal Neoplasms/pathology , Meningioma/complications , Meningioma/pathology , Mosaicism , Mutation , Neurofibromatosis 2/complications , Risk Assessment , Risk Factors , Sex Factors
10.
Cancer Epidemiol Biomarkers Prev ; 19(11): 2859-68, 2010 Nov.
Article in English | MEDLINE | ID: mdl-20978178

ABSTRACT

BACKGROUND: The genes caspase-8 (CASP8) and caspase-10 (CASP10) functionally cooperate and play a key role in the initiation of apoptosis. Suppression of apoptosis is one of the major mechanisms underlying the origin and progression of cancer. Previous case-control studies have indicated that the polymorphisms CASP8 D302H and CASP10 V410I are associated with a reduced risk of breast cancer in the general population. METHODS: To evaluate whether the CASP8 D302H (CASP10 V410I) polymorphisms modify breast or ovarian cancer risk in BRCA1 and BRCA2 mutation carriers, we analyzed 7,353 (7,227) subjects of white European origin provided by 19 (18) study groups that participate in the Consortium of Investigators of Modifiers of BRCA1/2 (CIMBA). A weighted cohort approach was used to estimate hazard ratios (HR) and 95% confidence intervals (95% CI). RESULTS: The minor allele of CASP8 D302H was significantly associated with a reduced risk of breast cancer (per-allele HR, 0.85; 95% CI, 0.76-0.97; P(trend) = 0.011) and ovarian cancer (per-allele HR, 0.69; 95% CI, 0.53-0.89; P(trend) = 0.004) for BRCA1 but not for BRCA2 mutation carriers. The CASP10 V410I polymorphism was not associated with breast or ovarian cancer risk for BRCA1 or BRCA2 mutation carriers. CONCLUSIONS: CASP8 D302H decreases breast and ovarian cancer risk for BRCA1 mutation carriers but not for BRCA2 mutation carriers. IMPACT: The combined application of these and other recently identified genetic risk modifiers could in the future allow better individual risk calculation and could aid in the individualized counseling and decision making with respect to preventive options in BRCA1 mutation carriers.


Subject(s)
Breast Neoplasms/genetics , Caspase 10/genetics , Caspase 8/genetics , Genetic Predisposition to Disease/genetics , Mutation , Ovarian Neoplasms/genetics , Breast Neoplasms/enzymology , Female , Genes, BRCA1 , Genes, BRCA2 , Genotype , Humans , Ovarian Neoplasms/enzymology , Polymerase Chain Reaction , Polymorphism, Single Nucleotide , Risk Factors
11.
Nat Genet ; 42(10): 885-92, 2010 Oct.
Article in English | MEDLINE | ID: mdl-20852631

ABSTRACT

Germline BRCA1 mutations predispose to breast cancer. To identify genetic modifiers of this risk, we performed a genome-wide association study in 1,193 individuals with BRCA1 mutations who were diagnosed with invasive breast cancer under age 40 and 1,190 BRCA1 carriers without breast cancer diagnosis over age 35. We took forward 96 SNPs for replication in another 5,986 BRCA1 carriers (2,974 individuals with breast cancer and 3,012 unaffected individuals). Five SNPs on 19p13 were associated with breast cancer risk (P(trend) = 2.3 × 10⁻9 to P(trend) = 3.9 × 10⁻7), two of which showed independent associations (rs8170, hazard ratio (HR) = 1.26, 95% CI 1.17-1.35; rs2363956 HR = 0.84, 95% CI 0.80-0.89). Genotyping these SNPs in 6,800 population-based breast cancer cases and 6,613 controls identified a similar association with estrogen receptor-negative breast cancer (rs2363956 per-allele odds ratio (OR) = 0.83, 95% CI 0.75-0.92, P(trend) = 0.0003) and an association with estrogen receptor-positive disease in the opposite direction (OR = 1.07, 95% CI 1.01-1.14, P(trend) = 0.016). The five SNPs were also associated with triple-negative breast cancer in a separate study of 2,301 triple-negative cases and 3,949 controls (P(trend) = 1 × 10⁻7) to P(trend) = 8 × 10⁻5; rs2363956 per-allele OR = 0.80, 95% CI 0.74-0.87, P(trend) = 1.1 × 10⁻7


Subject(s)
BRCA1 Protein/genetics , Breast Neoplasms/genetics , Chromosomes, Human, Pair 19/genetics , Genetic Predisposition to Disease , Mutation/genetics , Receptor, ErbB-2/genetics , Receptors, Estrogen/genetics , Receptors, Progesterone/genetics , Adult , Breast Neoplasms/pathology , Case-Control Studies , Female , Genotype , Humans , Polymorphism, Single Nucleotide/genetics
12.
Fam Cancer ; 9(2): 239-43, 2010 Jun.
Article in English | MEDLINE | ID: mdl-19967458

ABSTRACT

Hereditary leiomyomatosis and renal cell cancer is a hereditary cancer syndrome in which affected individuals are at risk for cutaneous and uterine leiomyomas, and renal cancer. Previous reports have stressed the aggressiveness of the renal tumours, often with early metastasis, despite small primary tumour size. Almost all the previously reported patients were adults, and different studies showed variability in penetrance for the renal tumours. We report a patient in whom renal cancer was detected at the age of 11 years at his first routine screening imaging after he was found to carry a fumarate hydratase gene mutation (c.1189G > A) transmitted from his mother. This report serves to emphasize the need to improve guidelines for screening of at risk individuals, including the necessity for predictive genetic testing and early institution of tumour surveillance in childhood.


Subject(s)
Carcinoma, Renal Cell/complications , Early Diagnosis , Kidney Neoplasms/diagnosis , Leiomyoma/etiology , Leiomyomatosis/complications , Neoplastic Syndromes, Hereditary/physiopathology , Carcinoma, Papillary/etiology , Carcinoma, Renal Cell/genetics , Child , Female , Genetic Counseling/psychology , Genetic Predisposition to Disease/epidemiology , Humans , Kidney Neoplasms/genetics , Kidney Neoplasms/physiopathology , Leiomyomatosis/genetics , Male , Mutation , Neoplastic Syndromes, Hereditary/genetics , Pedigree , Uterine Neoplasms/genetics , Uterine Neoplasms/physiopathology
13.
Am J Hum Genet ; 84(6): 780-91, 2009 Jun.
Article in English | MEDLINE | ID: mdl-19500772

ABSTRACT

Alveolar capillary dysplasia with misalignment of pulmonary veins (ACD/MPV) is a rare, neonatally lethal developmental disorder of the lung with defining histologic abnormalities typically associated with multiple congenital anomalies (MCA). Using array CGH analysis, we have identified six overlapping microdeletions encompassing the FOX transcription factor gene cluster in chromosome 16q24.1q24.2 in patients with ACD/MPV and MCA. Subsequently, we have identified four different heterozygous mutations (frameshift, nonsense, and no-stop) in the candidate FOXF1 gene in unrelated patients with sporadic ACD/MPV and MCA. Custom-designed, high-resolution microarray analysis of additional ACD/MPV samples revealed one microdeletion harboring FOXF1 and two distinct microdeletions upstream of FOXF1, implicating a position effect. DNA sequence analysis revealed that in six of nine deletions, both breakpoints occurred in the portions of Alu elements showing eight to 43 base pairs of perfect microhomology, suggesting replication error Microhomology-Mediated Break-Induced Replication (MMBIR)/Fork Stalling and Template Switching (FoSTeS) as a mechanism of their formation. In contrast to the association of point mutations in FOXF1 with bowel malrotation, microdeletions of FOXF1 were associated with hypoplastic left heart syndrome and gastrointestinal atresias, probably due to haploinsufficiency for the neighboring FOXC2 and FOXL1 genes. These differences reveal the phenotypic consequences of gene alterations in cis.


Subject(s)
Bronchopulmonary Dysplasia/genetics , Chromosomes, Human, Pair 16/genetics , Forkhead Transcription Factors/genetics , Gene Deletion , Gene Silencing , Mutation/genetics , Pulmonary Alveoli/pathology , Abnormalities, Multiple/genetics , Capillaries/abnormalities , Child, Preschool , Chromosome Mapping , Doxorubicin/analogs & derivatives , Female , Humans , In Situ Hybridization, Fluorescence , Infant , Infant, Newborn , Male , Pulmonary Alveoli/blood supply , Pulmonary Veins/abnormalities
14.
Breast Cancer Res Treat ; 117(2): 371-9, 2009 Sep.
Article in English | MEDLINE | ID: mdl-19082709

ABSTRACT

GATA-binding protein 3 (GATA3) is a transcription factor that is crucial to mammary gland morphogenesis and differentiation of progenitor cells, and has been suggested to have a tumor suppressor function. The rs570613 single nucleotide polymorphism (SNP) in intron 4 of GATA3 was previously found to be associated with a reduction in breast cancer risk in the Cancer Genetic Markers of Susceptibility project and in pooled analysis of two case-control studies from Norway and Poland (P (trend) = 0.004), with some evidence for a stronger association with estrogen receptor (ER) negative tumours [Garcia-Closas M et al. (2007) Cancer Epidemiol Biomarkers Prev 16:2269-2275]. We genotyped GATA3 rs570613 in 6,388 cases and 4,995 controls from the Breast Cancer Association Consortium (BCAC) and 5,617 BRCA1 and BRCA2 carriers from the Consortium of Investigators of Modifiers of BRCA1/2 (CIMBA). We found no association between this SNP and breast cancer risk in BCAC cases overall (OR(per-allele) = 1.00, 95% CI 0.94-1.05), in ER negative BCAC cases (OR(per-allele) = 1.02, 95% CI 0.91-1.13), in BRCA1 mutation carriers RR(per-allele) = 0.99, 95% CI 0.90-1.09) or BRCA2 mutation carriers (RR(per-allele) = 0.93, 95% CI 0.80-1.07). We conclude that there is no evidence that either GATA3 rs570613, or any variant in strong linkage disequilibrium with it, is associated with breast cancer risk in women.


Subject(s)
Breast Neoplasms/genetics , GATA3 Transcription Factor/genetics , Genetic Predisposition to Disease , Polymorphism, Single Nucleotide , Female , Genes, BRCA1 , Genes, BRCA2 , Genotype , Humans , Linkage Disequilibrium , Mutation , Risk Factors
15.
Clin Dysmorphol ; 15(2): 111-3, 2006 Apr.
Article in English | MEDLINE | ID: mdl-16531739

ABSTRACT

Marshall-Smith syndrome is characterized by overgrowth, advanced bone age, failure to thrive, respiratory problems, dysmorphic facial features and variable mental retardation. Respiratory problems are a major cause of early morbidity and mortality. Ocular features have been mentioned in previous reports, but details are limited. This report describes the clinical features of a child with typical features of Marshall-Smith syndrome with emphasis on visual function. She had megalocornea, hypoplastic optic discs and was partially sighted. Aggressive management of the early respiratory and feeding problems improved survival in this child.


Subject(s)
Abnormalities, Multiple/pathology , Eye Diseases/complications , Bone and Bones/diagnostic imaging , Child, Preschool , Female , Humans , Infant , Radiography , Survival Analysis , Syndrome
16.
Cancer Res ; 66(3): 1866-72, 2006 Feb 01.
Article in English | MEDLINE | ID: mdl-16452249

ABSTRACT

High breast density as measured on mammograms is a strong risk factor for breast cancer in the general population, but its effect in carriers of germline BRCA1 and BRCA2 mutations is unclear. We obtained mammograms from 206 female carriers of BRCA1 or BRCA2 mutations, 96 of whom were subsequently diagnosed with breast cancer and 136 relatives of carriers who were themselves noncarriers. We compared the mammographic densities of affected carriers (cases) and unaffected carriers (controls), and of mutation carriers and noncarriers, using a computer-assisted method of measurement and visual assessment by two observers. Analyses were adjusted for age, parity, body mass index, menopausal status, and hormone replacement therapy use. There was no difference in the mean percent density between noncarriers and carriers. Among carriers, increasing mammographic density was associated with an increased risk of breast cancer (P(trend) = 0.024). The odds ratio (OR; 95% confidence interval) for breast cancer associated with a density of > or =50% was 2.29 (1.23-4.26; P = 0.009). The OR did not differ between BRCA1 and BRCA2 carriers or between premenopausal and postmenopausal carriers. The results suggest that the distribution of breast density in BRCA1 and BRCA2 carriers is similar to that in non-carriers. High breast density in carriers is associated with an increased risk of breast cancer, with the relative risk being similar to that observed in the general population. Use of mammographic density could improve individual risk prediction in carriers.


Subject(s)
Breast Neoplasms/genetics , Breast/anatomy & histology , Genes, BRCA1 , Genes, BRCA2 , Germ-Line Mutation , Adult , Breast Neoplasms/diagnostic imaging , Female , Genetic Predisposition to Disease , Humans , Mammography , Middle Aged
SELECTION OF CITATIONS
SEARCH DETAIL
...