Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 19 de 19
Filter
1.
Genes Chromosomes Cancer ; 62(10): 589-596, 2023 10.
Article in English | MEDLINE | ID: mdl-37222498

ABSTRACT

BACKGROUND: Approximately half of ovarian tumors have defects within the homologous recombination repair pathway. Tumors carrying pathogenic variants (PVs) in BRCA1/BRCA2 are more likely to respond to poly-ADP ribose polymerase (PARP) inhibitor treatment. Large rearrangements (LRs) are a challenging class of variants to identify and characterize in tumor specimens and may therefore be underreported. This study describes the prevalence of pathogenic BRCA1/BRCA2 LRs in ovarian tumors and discusses the importance of their identification using a comprehensive testing strategy. METHODS: Sequencing and LR analyses of BRCA1/BRCA2 were conducted in 20 692 ovarian tumors received between March 18, 2016 and February 14, 2023 for MyChoice CDx testing. MyChoice CDx uses NGS dosage analysis to detect LRs in BRCA1/BRCA2 genes using dense tiling throughout the coding regions and limited flanking regions. RESULTS: Of the 2217 PVs detected, 6.3% (N = 140) were LRs. Overall, 0.67% of tumors analyzed carried a pathogenic LR. The majority of detected LRs were deletions (89.3%), followed by complex LRs (5.7%), duplications (4.3%), and retroelement insertions (0.7%). Notably, 25% of detected LRs encompassed a single or partial single exon. This study identified 84 unique LRs, 2 samples each carried 2 unique LRs in the same gene. We identified 17 LRs that occurred in multiple samples, some of which were specific to certain ancestries. Several cases presented here illustrate the intricacies involved in characterizing LRs, particularly when multiple events occur within the same gene. CONCLUSIONS: Over 6% of PVs detected in the ovarian tumors analyzed were LRs. It is imperative for laboratories to utilize testing methodologies that will accurately detect LRs at a single exon resolution to optimize the identification of patients who may benefit from PARP inhibitor treatment.


Subject(s)
Breast Neoplasms , Ovarian Neoplasms , Female , Humans , BRCA1 Protein/genetics , Ovarian Neoplasms/genetics , Ovarian Neoplasms/pathology , BRCA2 Protein/genetics , Genes, BRCA2 , Gene Rearrangement , DNA Repair , High-Throughput Nucleotide Sequencing , Breast Neoplasms/genetics , Germ-Line Mutation
2.
Front Oncol ; 13: 1069467, 2023.
Article in English | MEDLINE | ID: mdl-36793599

ABSTRACT

Background and Aims: Tumor immunohistochemical staining (IHC) of DNA mismatch repair (MMR) proteins is often used to guide germline genetic testing and variant classification for patients with suspected Lynch syndrome. This analysis examined the spectrum of germline findings in a cohort of individuals showing abnormal tumor IHC. Methods: We assessed individuals with reported abnormal IHC findings and referred for testing with a six-gene syndrome-specific panel (n=703). Pathogenic variants (PVs) and variants of uncertain significance (VUS) in MMR genes were designated expected/unexpected relative to IHC results. Results: The PV positive rate was 23.2% (163/703; 95% confidence interval [CI], 20.1%-26.5%); 8.0% (13/163; 95% CI, 4.3%-13.3%) of PV carriers had a PV in an unexpected MMR gene. Overall, 121 individuals carried VUS in MMR genes expected to be mutated based on IHC results. Based on independent evidence, in 47.1% (57/121; 95% CI, 38.0%-56.4%) of these individuals the VUSs were later reclassified as benign and in 14.0% (17/121; 95% CI, 8.4%-21.5%) of these individuals the VUSs were reclassified as pathogenic. Conclusions: Among patients with abnormal IHC findings, IHC-guided single-gene genetic testing may miss 8% of individuals with Lynch syndrome. In addition, in patients with VUS identified in MMR genes predicted to be mutated by IHC, extreme caution must be taken when the IHC results are considered in variant classification.

3.
Hum Mutat ; 41(1): 203-211, 2020 01.
Article in English | MEDLINE | ID: mdl-31490007

ABSTRACT

Previous analysis of next-generation sequencing (NGS) hereditary pan-cancer panel testing demonstrated that approximately 40% of TP53 pathogenic and likely pathogenic variants (PVs) detected have NGS allele frequencies between 10% and 30%, indicating that they likely are acquired somatically. These are seen more frequently in older adults, suggesting that most result from normal aging-related clonal hematopoiesis. For this analysis, apparent heterozygous germline TP53 PV carriers (NGS allele frequency 30-70%) were offered follow-up testing to confirm variant origin. Ninety-eight probands had samples submitted for follow-up family member testing, fibroblast testing, or both. The apparent heterozygous germline TP53 PV was not detected in 32.6% (15/46) of submitted fibroblast samples, indicating that it was acquired somatically, either through clonal hematopoiesis or via constitutional mosaicism. Notably, no individuals with confirmed germline or likely germline TP53 PVs met classic Li-Fraumeni syndrome (LFS) criteria, only 41% met Chompret LFS criteria, and 59% met neither criteria, based upon provider-reported personal and family cancer history. Comprehensive reporting of TP53 PVs detected using NGS, combined with follow-up analysis to confirm variant origin, is advised for clinical testing laboratories. These findings underscore the investment required to provide individuals and family members with clinically accurate genetic test results pertaining to their LFS risk.


Subject(s)
Genetic Association Studies , Genetic Predisposition to Disease , Heterozygote , Mutation , Neoplasms/diagnosis , Neoplasms/genetics , Tumor Suppressor Protein p53/genetics , Adolescent , Adult , Aged , Aged, 80 and over , Alleles , Female , Genetic Association Studies/methods , Genetic Testing , Germ-Line Mutation , High-Throughput Nucleotide Sequencing , Humans , Male , Middle Aged , Young Adult
4.
BMC Med Genomics ; 12(1): 138, 2019 10 17.
Article in English | MEDLINE | ID: mdl-31623605

ABSTRACT

BACKGROUND: Healthcare providers increasingly use information about pathogenic variants in cancer predisposition genes, including sequence variants and large rearrangements (LRs), in medical management decisions. While sequence variant detection is typically robust, LRs can be difficult to detect and characterize and may be underreported as a cause for hereditary cancer risk. This report describes the outcomes of hereditary cancer genetic testing using a comprehensive strategy that employs next-generation sequencing (NGS) for LR detection, coupled with LR confirmation using repeat hybrid capture NGS, microarray comparative genomic hybridization (microarray-CGH), and/or multiplex ligation-dependent probe amplification (MLPA). METHODS: Sequencing and LR analysis were conducted in a consecutive series of 376,159 individuals who received clinical testing with a hereditary pan-cancer gene panel from September 2013 through May 2017. NGS dosage analysis was used to evaluate potential deletions or duplications, with controls in place to exclude pseudogene reads. Samples positive for a putative LR based on NGS were confirmed using a comprehensive approach that included targeted microarray-CGH and/or MLPA analysis, with further examination as needed to ascertain the nature of the LR. RESULTS: A total of 3461 LRs were identified and classified as a deleterious mutation (DM), suspected deleterious mutation (SDM) or variant of uncertain significance. Pathogenic LRs (DM/SDM) accounted for the majority of LRs (67.7%), the largest proportion of which were deletions (86.1%), followed by duplications (11.3%), insertions (1.8%), triplications (0.5%), and inversions (0.3%). Several cases presented illustrate that the laboratory approach employed here can ensure consistent identification and accurate characterization of LRs. In the absence of this comprehensive testing strategy, 9% of LRs identified in this testing population might have been missed, potentially leading to inappropriate medical management in as many as 210 individuals referred for hereditary cancer testing. CONCLUSIONS: These data show that copy number analysis using NGS coupled with confirmatory testing reliably detects and characterizes LRs. Further, LRs comprise a substantial proportion (7.2%) of pathogenic variants identified by the test. A robust and accurate LR identification strategy is an essential component of a high-quality genetic testing program, enabling clinicians to optimize patient medical management decisions.


Subject(s)
Gene Rearrangement , High-Throughput Nucleotide Sequencing/methods , Neoplasms/genetics , Case-Control Studies , Comparative Genomic Hybridization , DNA Copy Number Variations , Gene Duplication , Humans , Mutagenesis, Insertional , Neoplasms/diagnosis , Sequence Analysis, DNA , Sequence Deletion
5.
Cancer Genet ; 235-236: 31-38, 2019 06.
Article in English | MEDLINE | ID: mdl-31056428

ABSTRACT

Next-generation sequencing (NGS) hereditary pan-cancer panel testing can identify somatic variants, which exhibit lower allele frequencies than do germline variants and may confound hereditary cancer predisposition testing. This analysis examined the prevalence and characteristics of likely-somatic variants among 348,543 individuals tested using a clinical NGS hereditary pan-cancer panel. Variants showing allele frequencies between 10% and 30% were interpreted as likely somatic and identified in 753 (0.22%) individuals. They were most frequent in TP53, CHEK2 and ATM, commonly as C-to-T transitions. Among individuals who carried a likely-somatic variant and reported no personal cancer history, 54.2% (78/144) carried a variant in TP53, CHEK2 or ATM. With a reported cancer history, this percentage increased to 81.1% (494/609), predominantly in CHEK2 and TP53. Their presence was associated with age (OR=3.1, 95% CI 2.5, 3.7; p<0.001) and personal history of cancer (OR=3.3, 95% CI 2.7, 4.0; p<0.001), particularly ovarian cancer. Germline ATM pathogenic variant carriers showed significant enrichment of likely-somatic variants (OR=2.8, 95% CI 1.6, 4.9; p = 0.005), regardless of cancer status. The appearance of likely-somatic variants is consistent with clonal hematopoiesis, possibly influenced by cancer treatment. These findings highlight the precision required of diagnostic laboratories to deliver accurate germline testing results.


Subject(s)
Genetic Predisposition to Disease/genetics , Germ-Line Mutation/genetics , Neoplastic Syndromes, Hereditary/genetics , Adult , Aged , Ataxia Telangiectasia Mutated Proteins/genetics , Base Sequence , Checkpoint Kinase 2/genetics , Gene Frequency/genetics , High-Throughput Nucleotide Sequencing , Humans , Mass Screening , Middle Aged , Tumor Suppressor Protein p53/genetics
6.
Future Oncol ; 15(1): 65-79, 2019 Jan.
Article in English | MEDLINE | ID: mdl-30113232

ABSTRACT

Expanded genetic test utilization to guide cancer management has driven the development of larger gene panels and greater diversity in the patient population pursuing testing, resulting in increased identification of atypical or technically challenging genetic findings. To ensure appropriate patient care, it is critical that genetic tests adequately identify and characterize these findings. We describe genetic testing challenges frequently encountered by our laboratory and the methodologies we employ to improve test accuracy for the identification and characterization of atypical genetic findings. While these findings may be individually rare, 15,745 (9%) individuals tested by our laboratory for hereditary cancer risk had an atypical genetic finding, highlighting the importance of employing highly accurate and comprehensive methods in clinical genetic testing.


Subject(s)
Genetic Testing/methods , High-Throughput Nucleotide Sequencing/methods , Neoplastic Syndromes, Hereditary/genetics , Gene Rearrangement , Genetic Predisposition to Disease , Genetic Testing/standards , High-Throughput Nucleotide Sequencing/standards , Humans , Mismatch Repair Endonuclease PMS2/genetics , Mosaicism , Pseudogenes , Quality Control , Reproducibility of Results
7.
Cancer Genet ; 216-217: 159-169, 2017 Oct.
Article in English | MEDLINE | ID: mdl-29025590

ABSTRACT

Cancer risks have been previously reported for some retrotransposon element (RE) insertions; however, detection of these insertions is technically challenging and very few oncogenic RE insertions have been reported. Here we evaluate RE insertions identified during hereditary cancer genetic testing using a comprehensive testing strategy. Individuals who had single-syndrome or pan-cancer hereditary cancer genetic testing from February 2004 to March 2017 were included. RE insertions were identified using Sanger sequencing, Next Generation Sequencing, or multiplex quantitative PCR, and further characterized using targeted PCR and sequencing analysis. Personal cancer history, ancestry, and haplotype were evaluated. A total of 37 unique RE insertions were identified in 10 genes, affecting 211 individuals. BRCA2 accounted for 45.9% (17/37) of all unique RE insertions. Several RE insertions were detected with high frequency in populations of conserved ancestry wherein up to 100% of carriers shared a high degree of haplotype conservation, suggesting founder effects. Our comprehensive testing strategy resulted in a substantial increase in the number of reported oncogenic RE insertions, several of which may have possible founder effects. Collectively, these data show that the detection of RE insertions is an important component of hereditary cancer genetic testing and may be more prevalent than previously reported.


Subject(s)
Genes, Neoplasm , Genetic Predisposition to Disease , Mutagenesis, Insertional/genetics , Neoplasms/genetics , Retroelements/genetics , Alu Elements/genetics , Base Sequence , Founder Effect , Haplotypes/genetics , Humans , Mutation/genetics , Risk Factors
8.
Cancer Genet ; 211: 5-8, 2017 02.
Article in English | MEDLINE | ID: mdl-28279308

ABSTRACT

Next Generation Sequencing (NGS) multigene panels, which are routinely used to assess hereditary cancer risk, can detect both inherited germline variants and somatic variants in cancer-risk genes. We evaluated the frequency and distribution of likely somatic Pathogenic and Likely Pathogenic variants (PVs) detected in >220,000 individuals who underwent clinical testing with a 25-gene panel between September 2013 and March 2016. Likely somatic PVs are defined as variants with NGS read frequencies from 10% to 30%. Overall, 137 (0.06%) individuals were identified as carrying likely somatic PVs, most commonly in TP53 (73), CHEK2 (27), and ATM (20). Among this group, a second PV with a NGS read frequency consistent with a germline variant within the same gene or a different gene on the panel was detected in 21 individuals (15.3%), which is similar to the detection rate in our general testing population. Likely somatic PVs accounted for 38.8% of all PVs in TP53. In comparison, likely somatic PVs accounted for <1% of PVs in most other genes. Likely somatic PVs were more frequently identified in older individuals (p < 0.001). Additional studies are ongoing to further investigate the incidence and clinical implications of somatic variants, enabling the appropriate medical management for these patients.


Subject(s)
High-Throughput Nucleotide Sequencing/methods , Lymphocytes/physiology , Neoplasms/blood , Neoplasms/genetics , Genetic Predisposition to Disease , Humans , Lymphocytes/chemistry , Lymphocytes/metabolism
10.
J Exp Clin Cancer Res ; 33: 74, 2014 Sep 11.
Article in English | MEDLINE | ID: mdl-25204323

ABSTRACT

BACKGROUND: Conventional Sanger sequencing reliably detects the majority of genetic mutations associated with hereditary cancers, such as single-base changes and small insertions or deletions. However, detection of genomic rearrangements, such as large deletions and duplications, requires special technologies. Microarray analysis has been successfully used to detect large rearrangements (LRs) in genetic disorders. METHODS: We designed and validated a high-density oligonucleotide microarray for the detection of gene-level genomic rearrangements associated with hereditary breast and ovarian cancer (HBOC), Lynch, and polyposis syndromes. The microarray consisted of probes corresponding to the exons and flanking introns of BRCA1 and BRCA2 (≈1,700) and Lynch syndrome/polyposis genes MLH1, MSH2, MSH6, APC, MUTYH, and EPCAM (≈2,200). We validated the microarray with 990 samples previously tested for LR status in BRCA1, BRCA2, MLH1, MSH2, MSH6, APC, MUTYH, or EPCAM. Microarray results were 100% concordant with previous results in the validation studies. Subsequently, clinical microarray analysis was performed on samples from patients with a high likelihood of HBOC mutations (13,124), Lynch syndrome mutations (18,498), and polyposis syndrome mutations (2,739) to determine the proportion of LRs. RESULTS: Our results demonstrate that LRs constitute a substantial proportion of genetic mutations found in patients referred for hereditary cancer genetic testing. CONCLUSION: The use of microarray comparative genomic hybridization (CGH) for the detection of LRs is well-suited as an adjunct technology for both single syndrome (by Sanger sequencing analysis) and extended gene panel testing by next generation sequencing analysis. Genetic testing strategies using microarray analysis will help identify additional patients carrying LRs, who are predisposed to various hereditary cancers.


Subject(s)
Genomics , Neoplastic Syndromes, Hereditary/genetics , Oligonucleotide Array Sequence Analysis , Translocation, Genetic , Exons , Genomics/methods , Humans , MutS Homolog 2 Protein/genetics , Mutation , Neoplastic Syndromes, Hereditary/diagnosis , Oligonucleotide Array Sequence Analysis/methods , Reproducibility of Results
11.
Genet Med ; 12(5): 268-78, 2010 May.
Article in English | MEDLINE | ID: mdl-20474083

ABSTRACT

PURPOSE: Genetic tests for the most commonly mutated genes in dilated cardiomyopathy (DCM) can confirm a clinical diagnosis in the proband and inform family management. Presymptomatic family members can be identified, allowing for targeted clinical monitoring to minimize adverse outcomes. However, the marked locus and allelic heterogeneity associated with DCM have made clinical genetic testing challenging. Novel sequencing platforms have now opened up avenues for more comprehensive diagnostic testing while simultaneously decreasing test cost and turn around time. METHODS: By using a custom design based on triplicate resequencing and separate genotyping of known disease-causing variants, we developed the DCM CardioChip for efficient analysis of 19 genes previously implicated in causing DCM. RESULTS: The chip's analytical sensitivity for known and novel substitution variants is 100% and 98%, respectively. In screening 73 previously tested DCM patients who did not carry clinically significant variants in 10 genes, 7 variants of likely clinical significance were identified in the remaining 9 genes included on the chip. Compared with traditional Sanger-based sequencing, test cost and turn around time were reduced by approximately 50%. CONCLUSIONS: The DCM CardioChip is a highly efficient screening test with a projected clinical sensitivity of 26-29%.


Subject(s)
Cardiomyopathy, Dilated/diagnosis , Cardiomyopathy, Dilated/genetics , Family , Female , Genetic Testing , Humans , Mutation , Pregnancy
12.
Circ Res ; 106(9): 1549-52, 2010 May 14.
Article in English | MEDLINE | ID: mdl-20378854

ABSTRACT

RATIONALE: The myosin-binding protein C isoform 3 (MYBPC3) variant Arg502Trp has been identified in multiple hypertrophic cardiomyopathy (HCM) cases, but compelling evidence to support or refute the pathogenicity of this variant is lacking. OBJECTIVE: To determine the prevalence, origin and clinical significance of the MYBPC3 Arg502Trp variant. METHODS AND RESULTS: The prevalence of MYBPC3 Arg502Trp was ascertained in 1414 sequential HCM patients of primarily European descent. MYBPC3 Arg502Trp was identified in 34 of these 1414 unrelated HCM patients. Segregation of MYBPC3 Arg502Trp with clinical status was assessed in family members. Disease haplotypes were examined in 17 families using two loci flanking MYBPC3. Family studies identified an additional 43 variant carriers, many with manifest disease, yielding a calculated odds ratio of 11 000:1 for segregation of MYBPC3 Arg502Trp with HCM. Analyses in 17 families showed at least 4 independent haplotypes flanked MYBPC3 Arg502Trp. Eight individuals (4 probands and 4 family members) also had another sarcomere protein gene mutation. Major adverse clinical events occurred in approximately 30% of MYBPC3 Arg502Trp carriers by age 50; these were significantly more likely (P<0.0001) when another sarcomere mutation was present. CONCLUSIONS: MYBPC3 Arg502Trp is the most common and recurrent pathogenic mutation in a diverse primarily European descent HCM cohort, occurring in 2.4% of patients. MYBPC3 Arg502Trp conveys a 340-fold increased risk for HCM by 45 years of age, when more than 50% of carriers have overt disease. HCM prognosis worsens when MYBPC3 Arg502Trp occurs in the setting of another sarcomere protein gene mutation.


Subject(s)
Cardiomyopathy, Hypertrophic/genetics , Carrier Proteins/genetics , Point Mutation , Adolescent , Adult , Aged , Aged, 80 and over , Cardiomyopathy, Hypertrophic/diagnosis , Child , Child, Preschool , Humans , Infant , Middle Aged , Young Adult
13.
Circ Cardiovasc Genet ; 3(2): 129-37, 2010 Apr.
Article in English | MEDLINE | ID: mdl-20173215

ABSTRACT

BACKGROUND: Danon disease is an X-linked dominant disorder characterized by the clinical triad of hypertrophic cardiomyopathy, skeletal myopathy, and variable mental retardation. Pathologically, autophagic vacuoles are noted in both skeletal and cardiac muscle. It exhibits an X-linked dominant mode of inheritance, and male carriers are severely affected, whereas female carriers develop milder and later-onset cardiac symptoms. Danon disease has been associated with mutations in the lysosome-associated membrane glycoprotein 2 (LAMP2) gene located at Xq24, typically resulting in splicing defects or protein truncation affecting the LAMP2. Because of its rarity, the full spectrum of genetic mutation resulting in Danon disease has not been elucidated. METHODS AND RESULTS: We analyzed 3 male cases with clinical and pathological findings consistent with Danon disease. Comprehensive mutational analysis failed to yield detectable products for selected LAMP2 exons, and genomic DNA deletion was suspected. Genomic junction fragment polymerase chain reaction analysis in case 1 identified a novel Alu-mediated 34-kb microdeletion encompassing the entire 5'-untranslated region and exon 1 of LAMP2. In case 2 and 3, junctional polymerase chain reaction and Southern blot analyses mapped the breakpoint to an MIRb and (TA)(n) simple repeats present in intron 3, which determined a 64-kb and a 58-kb deletion, respectively, thereby ablating exons 4 to 10. Western blot analysis confirmed the absence of LAMP2 in protein extract from lymphocytes of index case 2. CONCLUSIONS: This article is the first report of Danon disease caused by microdeletions at Xq24, which functionally ablate LAMP2. The microdeletion mechanism appears to involve 1 Alu-mediated unequal recombination and 2 chromosomal breakage points involving TA-rich repeat sequences.


Subject(s)
Glycogen Storage Disease Type IIb/genetics , Lysosomal Membrane Proteins/genetics , Sequence Deletion , Adolescent , Adult , Chromosomes, Human, X , Electrocardiography , Exons , Humans , Lysosomal-Associated Membrane Protein 2 , Male
14.
Per Med ; 6(4): 449-457, 2009 Jul.
Article in English | MEDLINE | ID: mdl-29783542

ABSTRACT

AIMS: Warfarin is a commonly prescribed drug with a narrow therapeutic index. Adverse drug reactions owing to over- or under-dosing are common. It is now established that genetic differences between individuals play a major role in warfarin metabolism. In particular, common variants in CYP2C9 (*2 and *3) and VKORC1 (-1639G>A) have been associated with a reduced drug-dosage requirement. MATERIALS & METHODS: We have evaluated the performance of five platforms that can be used to genotype individuals for these variants. These include Third Wave Technologies Invader®, Applied Biosystems TaqMan®, AutoGenomics INFINITI™ 2C9-VKORC1 assay, Osmetech eSensor® XT-8 warfarin sensitivity test and the Idaho Technologies LightScanner®. RESULTS & CONCLUSIONS: Excluding failures, all of these technologies had 100% concordance rates with either Sanger sequencing or another validated technology. All of these platforms had high sensitivity and specificity and are therefore appropriate for clinical molecular diagnostics. Therefore, platform choice is likely to be driven by clinical laboratories interested in performing this service taking other factors into account, including turnaround time, capacity, cost and ease of use.

15.
Mol Cell ; 32(2): 232-46, 2008 Oct 24.
Article in English | MEDLINE | ID: mdl-18951091

ABSTRACT

Recent investigations have implicated long antisense noncoding RNAs in the epigenetic regulation of chromosomal domains. Here we show that Kcnq1ot1 is an RNA polymerase II-encoded, 91 kb-long, moderately stable nuclear transcript and that its stability is important for bidirectional silencing of genes in the Kcnq1 domain. Kcnq1ot1 interacts with chromatin and with the H3K9- and H3K27-specific histone methyltransferases G9a and the PRC2 complex in a lineage-specific manner. This interaction correlates with the presence of extended regions of chromatin enriched with H3K9me3 and H3K27me3 in the Kcnq1 domain in placenta, whereas fetal liver lacks both chromatin interactions and heterochromatin structures. In addition, the Kcnq1 domain is more often found in contact with the nucleolar compartment in placenta than in liver. Taken together, our data describe a mechanism whereby Kcnq1ot1 establishes lineage-specific transcriptional silencing patterns through recruitment of chromatin remodeling complexes and maintenance of these patterns through subsequent cell divisions occurs via targeting the associated regions to the perinucleolar compartment.


Subject(s)
Chromatin/metabolism , Gene Silencing/physiology , RNA, Antisense/physiology , RNA, Messenger/metabolism , RNA, Untranslated/physiology , Animals , Cell Nucleolus/genetics , Cell Nucleolus/metabolism , Chromatin Immunoprecipitation , Histocompatibility Antigens/metabolism , Histone-Lysine N-Methyltransferase/metabolism , Histones/metabolism , Humans , Mice , Potassium Channels, Voltage-Gated/genetics , Potassium Channels, Voltage-Gated/metabolism , Potassium Channels, Voltage-Gated/physiology , RNA Polymerase II/metabolism , RNA Stability , RNA, Antisense/metabolism , RNA, Untranslated/metabolism
16.
Curr Protoc Hum Genet ; Chapter 7: Unit 7.15, 2008 Jan.
Article in English | MEDLINE | ID: mdl-18428425

ABSTRACT

New mutation detection technologies must keep pace by becoming more cost-effective while offering improved technical sensitivity and higher throughput capacity. In recent years, the number of mutation detection platforms available to the clinical researcher has grown to a point where it is difficult to keep track of all available options as well as their benefits and pitfalls. This unit provides an entry point for a variety of researchers who wish to analyze samples for known or novel mutations and need to determine which platform is most suited for their particular needs. A practical guide is provided in this unit, including a brief overview, information on assay parameters, design and cost considerations, as well as platform flexibility and scalability of the assay. Although the focus here is on applications involving human disease, many of these platforms can be easily adapted to the study of other organisms.


Subject(s)
DNA Mutational Analysis/methods , Biomedical Research/methods , DNA Mutational Analysis/economics , Genetic Testing/methods , Humans
17.
Genes Dev ; 20(10): 1268-82, 2006 May 15.
Article in English | MEDLINE | ID: mdl-16702402

ABSTRACT

The imprinted gene cluster at the telomeric end of mouse chromosome 7 contains a differentially methylated CpG island, KvDMR, that is required for the imprinting of multiple genes, including the genes encoding the maternally expressed placental-specific transcription factor ASCL2, the cyclin-dependent kinase CDKN1C, and the potassium channel KCNQ1. The KvDMR, which maps within intron 10 of Kcnq1, contains the promoter for a paternally expressed, noncoding, antisense transcript, Kcnq1ot1. A 244-base-pair deletion of the promoter on the paternal allele leads to the derepression of all silent genes tested. To distinguish between the loss of silencing as the consequence of the absence of transcription or the transcript itself, we prematurely truncated the Kcnq1ot1 transcript by inserting a transcriptional stop signal downstream of the promoter. We show that the lack of a full-length Kcnq1ot1 transcript on the paternal chromosome leads to the expression of genes that are normally paternally repressed. Finally, we demonstrate that five highly conserved repeats residing at the 5' end of the Kcnq1ot1 transcript are not required for imprinting at this locus.


Subject(s)
CpG Islands , DNA Methylation , Genomic Imprinting/genetics , RNA, Antisense/genetics , RNA, Messenger, Stored/genetics , RNA, Untranslated/genetics , Terminator Regions, Genetic , Animals , Cyclin-Dependent Kinase Inhibitor p57/genetics , Methyltransferases/genetics , Mice , Promoter Regions, Genetic/genetics , RNA, Messenger, Stored/metabolism , Sequence Deletion , Transcription, Genetic
18.
Int J Cancer ; 111(5): 669-78, 2004 Sep 20.
Article in English | MEDLINE | ID: mdl-15252835

ABSTRACT

Changes in DNA methylation patterns are frequently observed in human cancers and are associated with a decrease in tumor suppressor gene expression. Hypermethylation of the BRCA1 promoter has been reported in a portion of sporadic breast tumours that correspond to a reduction in BRCA1 transcription and expression. Questions remain concerning the maintenance of methylation free zones in promoter regions of tumor suppressor genes in normal tissues. Sodium bisulfite based analysis of the BRCA1 promoter defines a methylation free zone in normal breast tissue that starts 650 bp upstream of the transcription start site and extends for 1.4 kb through most of the BRCA1 CpG island. We provide data implicating 2 proteins, Sp1 and CTCF, in the maintenance of this methylation-free zone. Both of these proteins have been shown to function as methylation boundaries in other genes. Four Sp1 sites have been identified in the BRCA1 promoter by gel shift assays. In vivo binding of Sp1 has been confirmed at 2 of these sites in the BRCA1 promoter and at 2 CTCF sites that flank the unmethylated region. Our data suggest that these DNA binding sites for Sp1 and CTCF may act as boundary elements that are important in maintaining genomic integrity by delineating the normal methylation free BRCA1 promoter. Inactivation or disruption of these boundaries may facilitate an epigenetic "hit", in this case DNA methylation, leading to BRCA1 downregulation and contributing to tumorigenesis, particularly the genesis of sporadic breast tumours.


Subject(s)
BRCA1 Protein/biosynthesis , Breast Neoplasms/genetics , DNA Methylation , DNA, Neoplasm/genetics , DNA-Binding Proteins/pharmacology , Genes, BRCA1 , Repressor Proteins/pharmacology , Sp1 Transcription Factor/pharmacology , Base Sequence , CCCTC-Binding Factor , Cell Transformation, Neoplastic , Down-Regulation , Female , Humans , Molecular Sequence Data , Polymerase Chain Reaction , Promoter Regions, Genetic , Zinc Fingers
19.
Hum Mol Genet ; 12(3): 283-94, 2003 Feb 01.
Article in English | MEDLINE | ID: mdl-12554682

ABSTRACT

The imprinted gene cluster on mouse distal chromosome 7 contains a differentially methylated CpG island that maps within the Kcnq1 gene that has been shown to be required for the imprinting of multiple genes. To evaluate models for how this imprinting control region (ICR) regulates imprinting, we have characterized it structurally and functionally. We show that the region contains a promoter for a paternally expressed anti-sense transcript, Kcnq1ot1, and we define the extent of the minimal promoter. We describe three paternal-specific nuclease hypersensitive sites immediately upstream from the start site and show that they are required for full promoter activity. The expression of Kcnq1ot1 during pre- and postnatal development is compared to that of other imprinted genes in its vicinity, Cdnkn1c and Kcnq1. The lack of coordination in their expression tends to rule out an enhancer competition model for the action of the ICR in imprinting control. Using a stable transfection assay we show that the region contains a position-independent and orientation-independent silencer. We propose, on the basis of these findings, that the Kcnq1 ICR functions as a silencer on the paternal chromosome to effect the repression of neighboring genes.


Subject(s)
Gene Silencing , Genomic Imprinting , Potassium Channels, Voltage-Gated , Potassium Channels/genetics , Promoter Regions, Genetic , Animals , CpG Islands , Enhancer Elements, Genetic , Gene Expression Regulation , Humans , KCNQ Potassium Channels , KCNQ1 Potassium Channel , Mice , Molecular Sequence Data , Transcription Initiation Site
SELECTION OF CITATIONS
SEARCH DETAIL
...