Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 40
Filter
Add more filters

Country/Region as subject
Publication year range
1.
J Med Genet ; 59(8): 727-736, 2022 08.
Article in English | MEDLINE | ID: mdl-35393334

ABSTRACT

The purpose of this document is to provide pre-analytical, analytical and post-analytical considerations and recommendations to Canadian clinical laboratories developing, validating and offering next-generation sequencing (NGS)-based BRCA1 and BRCA2 (BRCA1/2) tumour testing in ovarian cancers. This document was drafted by the members of the Canadian College of Medical Geneticists (CCMG) somatic BRCA Ad Hoc Working Group, and representatives from the Canadian Association of Pathologists. The document was circulated to the CCMG members for comment. Following incorporation of feedback, this document has been approved by the CCMG board of directors. The CCMG is a Canadian organisation responsible for certifying medical geneticists and clinical laboratory geneticists, and for establishing professional and ethical standards for clinical genetics services in Canada. The current CCMG Practice Guidelines were developed as a resource for clinical laboratories in Canada; however, they are not inclusive of all information laboratories should consider in the validation and use of NGS for BRCA1/2 tumour testing in ovarian cancers.


Subject(s)
Clinical Laboratory Services , Ovarian Neoplasms , BRCA1 Protein/genetics , BRCA2 Protein/genetics , Canada , Carcinoma, Ovarian Epithelial , Female , Genetic Testing , Germ-Line Mutation , High-Throughput Nucleotide Sequencing , Humans , Ovarian Neoplasms/diagnosis , Ovarian Neoplasms/genetics
2.
Hum Genet ; 141(3-4): 431-444, 2022 Apr.
Article in English | MEDLINE | ID: mdl-35278131

ABSTRACT

Sequencing exomes/genomes have been successful for identifying recessive genes; however, discovery of dominant genes including deafness genes (DFNA) remains challenging. We report a new DFNA gene, ATP11A, in a Newfoundland family with a variable form of bilateral sensorineural hearing loss (SNHL). Genome-wide SNP genotyping linked SNHL to DFNA33 (LOD = 4.77), a locus on 13q34 previously mapped in a German family with variable SNHL. Whole-genome sequencing identified 51 unremarkable positional variants on 13q34. Continuous clinical ascertainment identified several key recombination events and reduced the disease interval to 769 kb, excluding all but one variant. ATP11A (NC_000013.11: chr13:113534963G>A) is a novel variant predicted to be a cryptic donor splice site. RNA studies verified in silico predictions, revealing the retention of 153 bp of intron in the 3' UTR of several ATP11A isoforms. Two unresolved families from Israel were subsequently identified with a similar, variable form of SNHL and a novel duplication (NM_032189.3:c.3322_3327+2dupGTCCAGGT) in exon 28 of ATP11A extended exon 28 by 8 bp, leading to a frameshift and premature stop codon (p.Asn1110Valfs43Ter). ATP11A is a type of P4-ATPase that transports (flip) phospholipids from the outer to inner leaflet of cell membranes to maintain asymmetry. Haploinsufficiency of ATP11A, the phospholipid flippase that specially transports phosphatidylserine (PS) and phosphatidylethanolamine (PE), could leave cells with PS/PE at the extracellular side vulnerable to phagocytic degradation. Given that surface PS can be pharmaceutically targeted, hearing loss due to ATP11A could potentially be treated. It is also likely that ATP11A is the gene underlying DFNA33.


Subject(s)
ATP-Binding Cassette Transporters , Deafness , Hearing Loss, Sensorineural , Hearing Loss , Humans , 3' Untranslated Regions , ATP-Binding Cassette Transporters/genetics , Deafness/genetics , Hearing Loss/genetics , Hearing Loss, Sensorineural/genetics , Mutation , Pedigree , Phospholipids/metabolism , RNA Splice Sites
3.
Hum Genet ; 141(3-4): 965-979, 2022 Apr.
Article in English | MEDLINE | ID: mdl-34633540

ABSTRACT

Otosclerosis is a bone disorder of the otic capsule and common form of late-onset hearing impairment. Considered a complex disease, little is known about its pathogenesis. Over the past 20 years, ten autosomal dominant loci (OTSC1-10) have been mapped but no genes identified. Herein, we map a new OTSC locus to a 9.96 Mb region within the FOX gene cluster on 16q24.1 and identify a 15 bp coding deletion in Forkhead Box L1 co-segregating with otosclerosis in a Caucasian family. Pre-operative phenotype ranges from moderate to severe hearing loss to profound sensorineural loss requiring a cochlear implant. Mutant FOXL1 is both transcribed and translated and correctly locates to the cell nucleus. However, the deletion of 5 residues in the C-terminus of mutant FOXL1 causes a complete loss of transcriptional activity due to loss of secondary (alpha helix) structure. FOXL1 (rs764026385) was identified in a second unrelated case on a shared background. We conclude that FOXL1 (rs764026385) is pathogenic and causes autosomal dominant otosclerosis and propose a key inhibitory role for wildtype Foxl1 in bone remodelling in the otic capsule. New insights into the molecular pathology of otosclerosis from this study provide molecular targets for non-invasive therapeutic interventions.


Subject(s)
Otosclerosis , Forkhead Transcription Factors/genetics , Humans , Otosclerosis/genetics
4.
BMC Med Genet ; 20(1): 68, 2019 05 02.
Article in English | MEDLINE | ID: mdl-31046701

ABSTRACT

BACKGROUND: Usher syndrome, the most common form of inherited deaf-blindness, is unlike many other forms of syndromic hereditary hearing loss in that the extra aural clinical manifestations are also detrimental to communication. Usher syndrome patients with early onset deafness also experience vision loss due to progressive retinitis pigmentosa that can lead to legal blindness in their third or fourth decade. METHODS: Using a multi-omic approach, we identified three novel pathogenic variants in two Usher syndrome genes (USH2A and ADGRV1) in cases initially referred for isolated vision or hearing loss. RESULTS: In a multiplex hearing loss family, two affected sisters, the product of a second cousin union, are homozygous for a novel nonsense pathogenic variant in ADGRV1 (c.17062C > T, p.Arg5688*), predicted to create a premature stop codon near the N-terminus of ADGRV1. Ophthalmological examination of the sisters confirmed typical retinitis pigmentosa and prompted a corrected Usher syndrome diagnosis. In an unrelated clinical case, a child with hearing loss tested positive for two novel USH2A splicing variants (c.5777-1G > A, p. Glu1926_Ala1952del and c.10388-2A > G, p.Asp3463Alafs*6) and RNA studies confirmed that both pathogenic variants cause splicing errors. Interestingly, these same USH2A variants are also identified in another family with vision loss where subsequent clinical follow-up confirmed pre-existing hearing loss since early childhood, eventually resulting in a reassigned diagnosis of Usher syndrome. CONCLUSION: These findings provide empirical evidence to increase Usher syndrome surveillance of at-risk children. Given that novel antisense oligonucleotide therapies have been shown to rescue retinal degeneration caused by USH2A splicing pathogenic variants, these solved USH2A patients may now be eligible to be enrolled in therapeutic trials.


Subject(s)
Deaf-Blind Disorders/genetics , Usher Syndromes/genetics , Child , Child, Preschool , Female , Genotype , Humans , Male , Pedigree , Phenotype
5.
J Rheumatol Suppl ; 95: 46-50, 2019 Jun.
Article in English | MEDLINE | ID: mdl-31154404

ABSTRACT

Psoriatic arthritis (PsA) is an inflammatory arthritis that manifests in 20-30% of patients diagnosed with psoriasis. Epidemiologic studies suggest a substantial genetic contribution to PsA. There is a strong need for genome-wide association studies on patients with PsA, including PsA-weighted or specific variants, and a need for a better understanding of the relevance of HLA alleles in disease expression. Interferon signaling and the nuclear factor-κB cascade are involved in PsA, and there are genetic differences between purely cutaneous psoriasis (PsC) and PsA. Psoriasis susceptibility genes for which putative functional coding variants in TYK2 and TRAF3IP2 are strongly associated with PsC and PsA, and neutrophil extracellular traps promote Th17 induction in an Act1 D10N-dependent fashion. Genomics and serological factors may also predict treatment response in tumor necrosis factor inhibitors (TNFi) in PsA, and genetics may play a role in treatment response to TNFi. Collaborations through the Group for Research and Assessment of Psoriasis and Psoriatic Arthritis (GRAPPA) are essential to increase study population size, which will enhance the ability to detect the genetic variants that create a predisposition to psoriatic disease and to predict response to biological therapy.


Subject(s)
Arthritis, Psoriatic/genetics , Genetic Predisposition to Disease , HLA-C Antigens/genetics , Polymorphism, Single Nucleotide , Psoriasis/genetics , Alleles , Genome-Wide Association Study , Genotype , Humans
6.
Curr Rheumatol Rep ; 20(8): 51, 2018 06 27.
Article in English | MEDLINE | ID: mdl-29951774

ABSTRACT

PURPOSE OF REVIEW: The purpose of this review is to highlight recent evidence with respect to expression and metabolomic profiling in axial spondyloarthritis (axSpA) that included ankylosing spondylitis (AS). RECENT FINDINGS: AxSpA is not only characterized by the strongest genetic contribution for any complex rheumatic disease but is also influenced by environmental and immunological factors. Large-scale association-based studies have identified over 100 genetic variants contributing to 30% of the genetic risk of ankylosing spondylitis. Recent studies in global expression and metabolomic profiling appear to highlight common themes despite differences in tissues, populations, techniques, and relative paucity of patients in many of these studies. Expression studies support a role for immunomodulation and bone remodeling in the pathogenesis and progression of axSpA/AS, while metabolomic studies implicate the importance of the intestinal microbial metabolism as well as fat and choline metabolic pathways in AS.


Subject(s)
Metabolome , MicroRNAs/metabolism , Spondylarthritis/metabolism , Disease Progression , Humans , Metabolomics
7.
Hum Genet ; 136(1): 107-118, 2017 01.
Article in English | MEDLINE | ID: mdl-27838790

ABSTRACT

Genetic isolates provide unprecedented opportunities to identify pathogenic mutations and explore the full natural history of clinically heterogeneous phenotypes such as hearing loss. We noticed a unique audioprofile, characterized by prelingual and rapid deterioration of hearing thresholds at frequencies >0.5 kHz in several adults from unrelated families from the island population of Newfoundland. Targeted serial Sanger sequencing of probands for deafness alleles (n = 23) that we previously identified in this founder population was negative. Whole exome sequencing in four members of the largest family (R2010) identified a CLDN14 (DFNB29) variant [c.488C>T; p. (Ala163Val)], likely pathogenic, sensorineural hearing loss, autosomal recessive. Although not associated with deafness or disease, CLDN14 p.(Ala163Val) has been previously reported as a variant of uncertain significance (VUS). Targeted sequencing of 169 deafness probands identified one homozygote and one heterozygous carrier. Genealogical studies, cascade sequencing and haplotype analysis across four unrelated families showed all subjects with the unique audioprofile (n = 12) were also homozygous for p.(Ala163Val) and shared a 1.4 Mb DFNB29-associated haplotype on chromosome 21. Most significantly, sequencing 175 population controls revealed 1% of the population are heterozygous for CLDN14 p.(Ala163Val), consistent with a major founder effect in Newfoundland. The youngest CLDN14 [c.488C>T; p.(Ala163Val)] homozygote passed newborn screening and had normal hearing thresholds up to 3 years of age, which then deteriorated to a precipitous loss >1 kHz during the first decade. Our study suggests that genetic testing may be necessary to identify at-risk children in time to prevent speech, language and developmental delay.


Subject(s)
Claudins/genetics , Founder Effect , Hearing Loss, Sensorineural/diagnosis , Alleles , Amino Acid Sequence , Child , Child, Preschool , Claudins/metabolism , Deafness/diagnosis , Deafness/genetics , Female , Gene Expression Regulation , Genetic Variation , Genome-Wide Association Study , Haplotypes , Hearing Loss, Sensorineural/genetics , Heterozygote , Humans , Male , Newfoundland and Labrador , Pedigree , Sequence Analysis, DNA
8.
Curr Opin Rheumatol ; 28(4): 337-45, 2016 Jul.
Article in English | MEDLINE | ID: mdl-27224740

ABSTRACT

PURPOSE OF REVIEW: This article discusses genomic investigations in ankylosing spondylitis (AS) beyond genome-wide association (GWA) studies, but prior to this, genetic variants achieving genome-wide significance will be summarized highlighting key pathways contributing to disease pathogenesis. RECENT FINDINGS: Evidence suggests that disease pathogenesis is attributed to a complex interplay of genetic, environmental and immunological factors. GWA studies have greatly enhanced our understanding of AS pathogenesis by illuminating distinct immunomodulatory pathways affecting innate and acquired immunity, most notably the interleukin-23/interleukin-17 pathway. However, despite the wealth of new information gleaned from such studies, a fraction of the heritability (24.4%) has been explained. This review will focus on investigations beyond GWA studies including copy number variants, gene expression profiling, including microRNA (miRNA), epigenetics, rare variants and gene-gene interactions. SUMMARY: To address the 'missing heritability' and advance beyond GWA studies, a concerted effort involving rethinking of study design and implementation of newer technologies will be required. The coming of age of next-generation sequencing and advancements in epigenetic and miRNA technologies, combined with familial-focused investigations using well-characterized cohorts, is likely to reveal some of the hidden genomic mysteries associated with AS.


Subject(s)
Spondylitis, Ankylosing/genetics , Adaptive Immunity/genetics , Epigenesis, Genetic , Epigenomics/methods , Gene Expression Profiling/methods , Genetic Predisposition to Disease , Genetic Variation , Genome-Wide Association Study , Genomics/methods , Humans , Interleukin-17/genetics , Interleukin-23/genetics , MicroRNAs/genetics , Spondylitis, Ankylosing/immunology
9.
Ann Rheum Dis ; 75(4): 772-9, 2016 Apr.
Article in English | MEDLINE | ID: mdl-25956157

ABSTRACT

OBJECTIVE: Axial spondyloarthritis (AxSpA) represents a group of inflammatory axial diseases that share common clinical and histopathological manifestations. Ankylosing spondylitis (AS) is the best characterised subset of AxSpA, and its genetic basis has been extensively investigated. Given that genome-wide association studies account for only 25% of AS heritability, the objective of this study was to discover rare, highly penetrant genetic variants in AxSpA pathogenesis using a well-characterised, multigenerational family. METHODS: HLA-B*27 genotyping and exome sequencing was performed on DNA collected from available family members. Variant frequency was assessed by mining publically available datasets and using fragment analysis of unrelated AxSpA cases and unaffected controls. Gene expression was performed by qPCR, and protein expression was assessed by western blot analysis and immunofluorescence microscopy using patient-derived B-cell lines. Circular dichroism spectroscopy was performed to assess the impact of discovered variants on secondary structure. RESULTS: This is the first report identifying two rare private familial variants in a multigenerational AxSpA family, an in-frame SEC16A deletion and an out-of-frame MAMDC4 deletion. Evidence suggests the causative mechanism for SEC16A appears to be a conformational change induced by deletion of three highly conserved amino acids from the intrinsically disordered Sec16A N-terminus and RNA-mediated decay for MAMDC4. CONCLUSIONS: The results suggest that it is the presence of rare syntenic SEC16A and MAMDC4 deletions that increases susceptibility to AxSpA in family members who carry the HLA-B*27 allele.


Subject(s)
B-Lymphocytes/metabolism , HLA-B27 Antigen/genetics , Proteins/genetics , Spondylarthropathies/genetics , Vesicular Transport Proteins/genetics , Adolescent , Adult , Blotting, Western , Child , Chromosome Deletion , Chromosomes, Human, Pair 10 , Circular Dichroism , Female , Genetic Linkage , Heterozygote , Humans , Male , Microscopy, Fluorescence , Mutation , Pedigree , Polymerase Chain Reaction , Proteins/metabolism , Vesicular Transport Proteins/metabolism
10.
BMC Genet ; 14: 67, 2013 Aug 08.
Article in English | MEDLINE | ID: mdl-23927372

ABSTRACT

BACKGROUND: The primary objective of this study is to identify novel copy number variations (CNVs) associated with familial ankylosing spondylitis (AS). A customized genome-wide microarray was designed to detect CNVs and applied to a multiplex AS family with six (6) affected family members. CNVs were detected using the built-in DNA analytics aberration detection method-2 (ADM-2) algorithm. Gene enrichment analysis was performed to observe the segregation. Subsequent validation was performed using real time quantitative fluorescence polymerase reaction (QF-PCR). The frequency of copy number variation for the UGT2B17 gene was then performed on two well-defined AS cohorts. Fisher exact test was performed to quantify the association. RESULTS: Our family-based analysis revealed ten gene-enriched CNVs that segregate with all six family members affected with AS. Based on the proposed function and the polymorphic nature of the UGT2B17 gene, the UGT2B17 gene CNV was selected for validation using real time QF-PCR with full concordance. The frequency of two copies of the UGT2B17 gene CNV was 0.41 in the Newfoundland AS cases and 0.35 in the Newfoundland controls (OR = 1.26(0.99-1.59); p < 0.05)), whereas the frequency of two (2) copies of the UGT2B17 gene CNV was 0.40 in the Alberta AS cases and 0.39 in the Alberta controls (OR = 1.05(95% CI: 0.83-1.33); p < 0.71)). CONCLUSIONS: A genome-wide microarray interrogation of a large multiplex AS family revealed segregation of the UGT2B17 gene CNV among all affected family members. The association of the UGT2B17 CNV with AS is particularly interesting given the recent association of this CNV with osteoporosis and the proposed function as it encodes a key enzyme that inhibits androgens. However, two copies of the UGT2B17 gene CNV were only marginally significant in a uniplex AS cohort from Newfoundland but not in a uniplex AS cohort from Alberta.


Subject(s)
DNA Copy Number Variations , Glucuronosyltransferase/genetics , Spondylitis, Ankylosing/genetics , Case-Control Studies , Cohort Studies , Female , Humans , Male , Minor Histocompatibility Antigens , Pedigree , Polymerase Chain Reaction
11.
Curr Rheumatol Rep ; 15(8): 347, 2013 Aug.
Article in English | MEDLINE | ID: mdl-23771560

ABSTRACT

Spondyloarthritis (SpA) is a group of inflammatory rheumatic diseases whose main clinical feature is inflammation of the axial spine. Articular, periarticular, and extra-articular manifestations can also occur, depending on the type of spondyloarthritis. The most common clinical subsets of SpA are ankylosing spondylitis (AS) and psoriatic arthritis (PsA). SpA is a major health challenge given the propensity to affect young adults and the potential requirement for lifelong treatment. Although the precise etiology of SpA is unknown, there is mounting evidence that these diseases are a result of complex interplay of genetic, environmental, and immunological factors. In this review on SpA, we will discuss genetic variants with genome-wide significance, highlight potential clinical application of genetic variants, and discuss challenges in further elucidating the genetic basis of SpA.


Subject(s)
Spondylarthritis/genetics , Antigen Presentation/genetics , Genetic Predisposition to Disease , Genetic Variation , Genome-Wide Association Study , Humans , Immunity, Innate/genetics , Signal Transduction/genetics , Spondylarthritis/immunology
12.
Eur J Hum Genet ; 31(7): 815-823, 2023 07.
Article in English | MEDLINE | ID: mdl-37072551

ABSTRACT

Genotype-phenotype correlations add value to the management of families with hereditary hearing loss (HL), where age-related typical audiograms (ARTAs) are generated from cross-sectional regression equations and used to predict the audiogram phenotype across the lifespan. A seven-generation kindred with autosomal dominant sensorineural HL (ADSNHL) was recruited and a novel pathogenic variant in POU4F3 (c.37del) was identified by combining linkage analysis with whole exome sequencing (WES). POU4F3 is noted for large intrafamilial variation including the age of onset of HL, audiogram configuration and presence of vestibular impairment. Sequential audiograms and longitudinal analyses reveal highly variable audiogram features among POU4F3 (c.37del) carriers, limiting the utility of ARTAs for clinical prognosis and management of HL. Furthermore, a comparison of ARTAs against three previously published families (1 Israeli Jewish, 2 Dutch) reveals significant interfamilial differences, with earlier onset and slower deterioration. This is the first published report of a North American family with ADSNHL due to POU4F3, the first report of the pathogenic c.37del variant, and the first study to conduct longitudinal analysis, extending the phenotypic spectrum of DFNA15.


Subject(s)
Deafness , Hearing Loss, Sensorineural , Hearing Loss , Humans , Cross-Sectional Studies , Homeodomain Proteins/genetics , Hearing Loss/genetics , Hearing Loss, Sensorineural/diagnosis , Hearing Loss, Sensorineural/genetics , Hearing Loss, Sensorineural/pathology , Pedigree , Transcription Factor Brn-3C/genetics
13.
Genes (Basel) ; 13(2)2022 01 24.
Article in English | MEDLINE | ID: mdl-35205252

ABSTRACT

Rare post-zygotic mutations in the brain are now known to contribute to several neurodevelopmental disorders, including autism spectrum disorder (ASD). However, due to the limited availability of brain tissue, most studies rely on estimates of mosaicism from peripheral samples. In this study, we undertook whole exome sequencing on brain tissue from 26 ASD brain donors from the Harvard Brain Tissue Resource Center (HBTRC) and ascertained the presence of post-zygotic and germline mutations categorized as pathological, including those impacting known ASD-implicated genes. Although quantification did not reveal enrichment for post-zygotic mutations compared with the controls (n = 15), a small number of pathogenic, potentially ASD-implicated mutations were identified, notably in TRAK1 and CLSTN3. Furthermore, germline mutations were identified in the same tissue samples in several key ASD genes, including PTEN, SC1A, CDH13, and CACNA1C. The establishment of tissue resources that are available to the scientific community will facilitate the discovery of new mutations for ASD and other neurodevelopmental disorders.


Subject(s)
Autism Spectrum Disorder , Autism Spectrum Disorder/genetics , Autism Spectrum Disorder/pathology , Brain/pathology , Calcium-Binding Proteins/genetics , Genetic Predisposition to Disease , Humans , Membrane Proteins/genetics , Mutation , Exome Sequencing
14.
Methods Mol Biol ; 2249: 335-367, 2021.
Article in English | MEDLINE | ID: mdl-33871853

ABSTRACT

Genetic and environmental factors are critical elements in most common complex disease. Genetics is increasingly being recognized to play a substantive role in the susceptibility, prognosis, and treatment of common diseases. Due to recent and rapid advancements in characterization of genetic variants and large-scale genotyping platforms, multiple genes and genetic variants have now been identified for common, complex diseases. The most efficient method for gene identification at present appears to be large-scale association-based studies, which integrate genetic and epidemiological principles. As the strategy for gene identification studies has shifted toward genetic association-based methods rather than traditional linkage analysis, epidemiological methods are increasingly being integrated into genetic investigations and in public health research. Consequently, the disciplines of genetics and epidemiology, which historically have functioned separately, have been integrated into a hybrid discipline referred to as genetic epidemiology. In this chapter, we review methods for establishing the genetic burden of complex genetic disease, followed by methods for gene and/or genetic variant identification. When appropriate, we will highlight the epidemiological issues and clinical applications that guide these methods.


Subject(s)
Genetic Predisposition to Disease/genetics , Genetic Variation , Genome-Wide Association Study/methods , Genetic Linkage , High-Throughput Nucleotide Sequencing , Humans , Molecular Epidemiology , Phenotype , Sequence Analysis, DNA
15.
Best Pract Res Clin Rheumatol ; 35(2): 101691, 2021 06.
Article in English | MEDLINE | ID: mdl-34020887

ABSTRACT

Psoriatic arthritis (PsA) is caused by a combination of environmental and multiple genetic factors, with clear evidence for a strong genetic basis. The remarkable accumulation of knowledge gained from genetic, pharmacogenetic, and therapeutic response of biologic agents in PsA has fundamentally changed and advanced our understanding of disease pathogenesis and has identified key signalling pathways. However, only one-quarter of the genetic contribution of PsA has been accounted for; and dissecting the genetic contributors of the cutaneous disease from those that would identify joint disease has been challenging. More importantly, the clinical utility of multiple proposed loci is unclear. In this review, we summarize the potential clinical relevance from established genetic associations and provide insight on the proposed molecular pathways that arise from these associations.


Subject(s)
Arthritis, Psoriatic , Arthritis, Psoriatic/drug therapy , Arthritis, Psoriatic/genetics , Biological Factors , Genetic Loci , Genetic Predisposition to Disease , Genome-Wide Association Study , Humans
16.
NPJ Genom Med ; 6(1): 14, 2021 Feb 16.
Article in English | MEDLINE | ID: mdl-33594065

ABSTRACT

Collectively, rare genetic diseases affect a significant number of individuals worldwide. In this study, we have conducted whole-exome sequencing (WES) and identified underlying pathogenic or likely pathogenic variants in five children with rare genetic diseases. We present evidence for disease-causing autosomal recessive variants in a range of disease-associated genes such as DHH-associated 46,XY gonadal dysgenesis (GD) or 46,XY sex reversal 7, GNPTAB-associated mucolipidosis II alpha/beta (ML II), BBS1-associated Bardet-Biedl Syndrome (BBS), SURF1-associated Leigh Syndrome (LS) and AP4B1-associated spastic paraplegia-47 (SPG47) in unrelated affected members from Bangladesh. Our analysis pipeline detected three homozygous mutations, including a novel c. 863 G > C (p.Pro288Arg) variant in DHH, and two compound heterozygous variants, including two novel variants: c.2972dupT (p.Met991Ilefs*) in GNPTAB and c.229 G > C (p.Gly77Arg) in SURF1. All mutations were validated by Sanger sequencing. Collectively, this study adds to the genetic heterogeneity of rare genetic diseases and is the first report elucidating the genetic profile of (consanguineous and nonconsanguineous) rare genetic diseases in the Bangladesh population.

17.
Curr Rheumatol Rep ; 12(4): 300-8, 2010 Aug.
Article in English | MEDLINE | ID: mdl-20480402

ABSTRACT

Psoriasis and psoriatic arthritis (PsA) are interrelated heritable diseases. Polymorphisms in the genes encoded in the major histocompatibility complex region have consistently been associated with psoriasis and PsA and account for about 30% of the genetic risk. Recently, the results of multiple well-powered genome-wide association studies have identified several additional loci outside the major histocompatibility complex region associated with psoriasis risk, including three genes involved in interleukin (IL)-23 signaling (IL-23R, IL-23A, IL-12B), two genes that regulate nuclear factor-kappaB signaling (TNIP1, TNFAIP3), and two genes involved in the modulation of T-helper type 2 immune responses (IL-4, IL-13). These initial findings are beginning to illuminate the molecular pathways implicated in the pathogenesis of psoriasis and suggest priority targets for study in PsA cohorts. Of course, the next step will be to identify genetic risk factors specifically associated with PsA, and indeed, a formal genome-wide association study on PsA is being undertaken.


Subject(s)
Arthritis, Psoriatic/genetics , Major Histocompatibility Complex/genetics , Genetic Association Studies , Genetic Predisposition to Disease , Humans , Interleukins/genetics , Polymorphism, Genetic
18.
Sci Rep ; 10(1): 21703, 2020 12 10.
Article in English | MEDLINE | ID: mdl-33303908

ABSTRACT

Biological therapies have dramatically improved the therapeutic landscape of psoriatic arthritis (PsA); however, 40-50% of patients are primary non-responders with response rates declining significantly with each successive biological therapy. Therefore, there is a pressing need to develop a coherent strategy for effective initial and subsequent selection of biologic agents. We interrogated 40 PsA patients initiating either tumour necrosis factor inhibitors (TNFi) or interleukin-17A inhibitors (17Ai) for active PsA. Patients achieving low disease activity according to the Disease Activity Index for PsA (DAPSA) at 3 months were classified as responders. Baseline and 3-month CD4+ transcript profiling were performed, and novel signaling pathways were identified using a multi-omics profiling and integrative computational analysis approach. Using transcriptomic data at initiation of therapy, we identified over 100 differentially expressed genes (DEGs) that differentiated IL-17Ai response from non-response and TNFi response from non-response. Integration of cell-type-specific DEGs with protein-protein interactions and further comprehensive pathway enrichment analysis revealed several pathways. Rho GTPase signaling pathway exhibited a strong signal specific to IL-17Ai response and the genes, RAC1 and ROCKs, are supported by results from prior research. Our detailed network and pathway analyses have identified the rewiring of Rho GTPase pathways as potential markers of response to IL17Ai but not TNFi. These results need further verification.


Subject(s)
Antibodies, Monoclonal, Humanized/therapeutic use , Antirheumatic Agents/therapeutic use , Arthritis, Psoriatic/drug therapy , Arthritis, Psoriatic/genetics , Biological Therapy/methods , Interleukin-17/antagonists & inhibitors , Signal Transduction/drug effects , Signal Transduction/genetics , Tumor Necrosis Factor-alpha/antagonists & inhibitors , rho GTP-Binding Proteins/metabolism , Adalimumab , Antibodies, Monoclonal , Antibodies, Monoclonal, Humanized/pharmacology , Antirheumatic Agents/pharmacology , Arthritis, Psoriatic/diagnosis , Signal Transduction/physiology , Treatment Outcome , rac1 GTP-Binding Protein
19.
Eur J Hum Genet ; 28(7): 925-937, 2020 07.
Article in English | MEDLINE | ID: mdl-32467599

ABSTRACT

Stargardt disease (STGD1) is a form of inherited retinal dystrophy attributed to variants affecting function of the large ABCA4 gene and is arguably the most complex monogenic disease. Therapeutic trials in patients depend on identifying causal ABCA4 variants in trans, which is complicated by extreme allelic and clinical heterogeneity. We report the genetic architecture of STGD1 in the young genetically isolated population of Newfoundland, Canada. Population-based clinical recruitment over several decades yielded 29 STGD1 and STGD1-like families (15 multiplex, 14 singleton). Family interviews and public archival records reveal the vast majority of pedigree founders to be of English extraction. Full gene sequencing and haplotype analysis yielded a high solve rate (38/41 cases; 92.7%) for STGD1 and identified 16 causative STGD1 alleles, including a novel deletion (NM_000350.3: ABCA4 c.67-1delG). Several STGD1 alleles of European origin (including NM_000350.3: ABCA4 c.5714 + 5G>A and NM_000350.3: ABCA4 c.5461-10T>C) have drifted to a relatively high population frequency due to founder effect. We report on retinal disease progression in homozygous patients, providing valuable allele-specific insights. The least involved retinal disease is seen in patients homozygous for c.5714 + 5G>A variant, a so-called "mild" variant which is sufficient to precipitate a STGD1 phenotype in the absence of other pathogenic variants in the coding region and intron/exon boundaries of ABCA4. The most severe retinal disease is observed in cases with ABCA4 c.[5461-10T>C;5603A>T] complex allele. We discuss the advantages of determining genetic architecture in genetic isolates in order to begin to meet the grand challenge of human genetics.


Subject(s)
ATP-Binding Cassette Transporters/genetics , Gene Frequency , Stargardt Disease/genetics , Female , Founder Effect , Homozygote , Humans , Male , Mutation , Pedigree
20.
Mol Genet Genomic Med ; 8(2): e1070, 2020 02.
Article in English | MEDLINE | ID: mdl-31782267

ABSTRACT

BACKGROUND: RAD51C is important in DNA repair and individuals with pathogenic RAD51C variants have increased risk of hereditary breast and ovarian cancer syndrome (HBOC), an autosomal dominant genetic predisposition to early onset breast and/or ovarian cancer. METHODS: Five female HBOC probands sequenced negative for moderate- and high-risk genes but shared a recurrent variant of uncertain significance in RAD51C (NM_058216.3: c.571 + 4A > G). Participant recruitment was followed by haplotype and case/control analyses, RNA splicing analysis, gene and protein expression assays, and Sanger sequencing of tumors. RESULTS: The RAD51C c.571 + 4A > G variant segregates with HBOC, with heterozygotes sharing a 5.07 Mbp haplotype. RAD51C c.571 + 4A > G is increased ~52-fold in the Newfoundland population compared with the general Caucasian population and positive population controls share disease-associated alleles, providing evidence of a founder effect. Splicing analysis confirmed in silico predictions that RAD51C c.571 + 4A > G causes exon 3 skipping, creating an immediate premature termination codon. Gene and protein expression were significantly reduced in a RAD51C c.571 + 4G > A heterozygote compared with a wild-type relative. Sanger sequencing of tumors from two probands indicates loss-of-heterozygosity, suggesting loss of function. CONCLUSION: The RAD51C c.571 + 4A > G variant affects mRNA splicing and should be re-classified as pathogenic according to American College of Medical Genetics and Genomics guidelines.


Subject(s)
DNA-Binding Proteins/genetics , Founder Effect , Hereditary Breast and Ovarian Cancer Syndrome/genetics , Population/genetics , Aged , Aged, 80 and over , Female , Genes, Dominant , Haplotypes , Humans , Male , Middle Aged , Mutation , Newfoundland and Labrador , RNA Splicing
SELECTION OF CITATIONS
SEARCH DETAIL