Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 145
Filter
Add more filters

Publication year range
1.
Nature ; 581(7809): 459-464, 2020 05.
Article in English | MEDLINE | ID: mdl-32461653

ABSTRACT

Naturally occurring human genetic variants that are predicted to inactivate protein-coding genes provide an in vivo model of human gene inactivation that complements knockout studies in cells and model organisms. Here we report three key findings regarding the assessment of candidate drug targets using human loss-of-function variants. First, even essential genes, in which loss-of-function variants are not tolerated, can be highly successful as targets of inhibitory drugs. Second, in most genes, loss-of-function variants are sufficiently rare that genotype-based ascertainment of homozygous or compound heterozygous 'knockout' humans will await sample sizes that are approximately 1,000 times those presently available, unless recruitment focuses on consanguineous individuals. Third, automated variant annotation and filtering are powerful, but manual curation remains crucial for removing artefacts, and is a prerequisite for recall-by-genotype efforts. Our results provide a roadmap for human knockout studies and should guide the interpretation of loss-of-function variants in drug development.


Subject(s)
Genes, Essential/drug effects , Genes, Essential/genetics , Loss of Function Mutation/genetics , Molecular Targeted Therapy , Artifacts , Automation , Consanguinity , Exons/genetics , Gain of Function Mutation/genetics , Gene Frequency , Gene Knockdown Techniques , Heterozygote , Homozygote , Humans , Huntingtin Protein/genetics , Leucine-Rich Repeat Serine-Threonine Protein Kinase-2/genetics , Neurodegenerative Diseases/genetics , Prion Proteins/genetics , Reproducibility of Results , Sample Size , tau Proteins/genetics
2.
Eur Respir J ; 61(2)2023 02.
Article in English | MEDLINE | ID: mdl-36302552

ABSTRACT

Pulmonary arterial hypertension (PAH) is a rare disease that can be caused by (likely) pathogenic germline genomic variants. In addition to the most prevalent disease gene, BMPR2 (bone morphogenetic protein receptor 2), several genes, some belonging to distinct functional classes, are also now known to predispose to the development of PAH. As a consequence, specialist and non-specialist clinicians and healthcare professionals are increasingly faced with a range of questions regarding the need for, approaches to and benefits/risks of genetic testing for PAH patients and/or related family members. We provide a consensus-based approach to recommendations for genetic counselling and assessment of current best practice for disease gene testing. We provide a framework and the type of information to be provided to patients and relatives through the process of genetic counselling, and describe the presently known disease causal genes to be analysed. Benefits of including molecular genetic testing within the management protocol of patients with PAH include the identification of individuals misclassified by other diagnostic approaches, the optimisation of phenotypic characterisation for aggregation of outcome data, including in clinical trials, and importantly through cascade screening, the detection of healthy causal variant carriers, to whom regular assessment should be offered.


Subject(s)
Hypertension, Pulmonary , Pulmonary Arterial Hypertension , Humans , Pulmonary Arterial Hypertension/genetics , Genetic Counseling/methods , Hypertension, Pulmonary/diagnosis , Hypertension, Pulmonary/genetics , Mutation , Familial Primary Pulmonary Hypertension/genetics , Genetic Testing , Bone Morphogenetic Protein Receptors, Type II/genetics , Genetic Predisposition to Disease
3.
PLoS Med ; 19(5): e1003981, 2022 05.
Article in English | MEDLINE | ID: mdl-35587468

ABSTRACT

BACKGROUND: Type 2 diabetes (T2D) is highly prevalent in British South Asians, yet they are underrepresented in research. Genes & Health (G&H) is a large, population study of British Pakistanis and Bangladeshis (BPB) comprising genomic and routine health data. We assessed the extent to which genetic risk for T2D is shared between BPB and European populations (EUR). We then investigated whether the integration of a polygenic risk score (PRS) for T2D with an existing risk tool (QDiabetes) could improve prediction of incident disease and the characterisation of disease subtypes. METHODS AND FINDINGS: In this observational cohort study, we assessed whether common genetic loci associated with T2D in EUR individuals were replicated in 22,490 BPB individuals in G&H. We replicated fewer loci in G&H (n = 76/338, 22%) than would be expected given power if all EUR-ascertained loci were transferable (n = 101, 30%; p = 0.001). Of the 27 transferable loci that were powered to interrogate this, only 9 showed evidence of shared causal variants. We constructed a T2D PRS and combined it with a clinical risk instrument (QDiabetes) in a novel, integrated risk tool (IRT) to assess risk of incident diabetes. To assess model performance, we compared categorical net reclassification index (NRI) versus QDiabetes alone. In 13,648 patients free from T2D followed up for 10 years, NRI was 3.2% for IRT versus QDiabetes (95% confidence interval (CI): 2.0% to 4.4%). IRT performed best in reclassification of individuals aged less than 40 years deemed low risk by QDiabetes alone (NRI 5.6%, 95% CI 3.6% to 7.6%), who tended to be free from comorbidities and slim. After adjustment for QDiabetes score, PRS was independently associated with progression to T2D after gestational diabetes (hazard ratio (HR) per SD of PRS 1.23, 95% CI 1.05 to 1.42, p = 0.028). Using cluster analysis of clinical features at diabetes diagnosis, we replicated previously reported disease subgroups, including Mild Age-Related, Mild Obesity-related, and Insulin-Resistant Diabetes, and showed that PRS distribution differs between subgroups (p = 0.002). Integrating PRS in this cluster analysis revealed a Probable Severe Insulin Deficient Diabetes (pSIDD) subgroup, despite the absence of clinical measures of insulin secretion or resistance. We also observed differences in rates of progression to micro- and macrovascular complications between subgroups after adjustment for confounders. Study limitations include the absence of an external replication cohort and the potential biases arising from missing or incorrect routine health data. CONCLUSIONS: Our analysis of the transferability of T2D loci between EUR and BPB indicates the need for larger, multiancestry studies to better characterise the genetic contribution to disease and its varied aetiology. We show that a T2D PRS optimised for this high-risk BPB population has potential clinical application in BPB, improving the identification of T2D risk (especially in the young) on top of an established clinical risk algorithm and aiding identification of subgroups at diagnosis, which may help future efforts to stratify care and treatment of the disease.


Subject(s)
Diabetes Mellitus, Type 2 , Asian People , Cohort Studies , Diabetes Mellitus, Type 2/diagnosis , Diabetes Mellitus, Type 2/epidemiology , Diabetes Mellitus, Type 2/genetics , Female , Humans , Insulin , Pakistan/epidemiology , Risk Factors
4.
Eur Respir J ; 59(3)2022 03.
Article in English | MEDLINE | ID: mdl-34588193

ABSTRACT

BACKGROUND: Inflammation and dysregulated immunity are important in the development of pulmonary arterial hypertension (PAH). Compelling preclinical data supports the therapeutic blockade of interleukin-6 (IL-6) signalling. METHODS: We conducted a phase 2 open-label study of intravenous tocilizumab (8 mg·kg-1) over 6 months in patients with group 1 PAH. Co-primary end-points were safety, defined by incidence and severity of adverse events, and change in pulmonary vascular resistance. Separately, a mendelian randomisation study was undertaken on 11 744 individuals with European ancestry including 2085 patients with idiopathic/heritable disease for the IL-6 receptor (IL6R) variant (rs7529229), known to associate with circulating IL-6R levels. RESULTS: We recruited 29 patients (male/female 10/19; mean±sd age 54.9±11.4 years). Of these, 19 had heritable/idiopathic PAH and 10 had connective tissue disease-associated PAH. Six were withdrawn prior to drug administration; 23 patients received at least one dose of tocilizumab. Tocilizumab was discontinued in four patients owing to serious adverse events. There were no deaths. Despite evidence of target engagement in plasma IL-6 and C-reactive protein levels, both intention-to-treat and modified intention-to-treat analyses demonstrated no change in pulmonary vascular resistance. Inflammatory markers did not predict treatment response. Mendelian randomisation did not support an effect of the lead IL6R variant on risk of PAH (OR 0.99, p=0.88). CONCLUSION: Adverse events were consistent with the known safety profile of tocilizumab. Tocilizumab did not show any consistent treatment effect.


Subject(s)
Biomedical Research , Pulmonary Arterial Hypertension , Adult , Aged , Familial Primary Pulmonary Hypertension , Female , Humans , Interleukin-6 , Male , Middle Aged , Treatment Outcome
6.
Am J Respir Crit Care Med ; 202(4): 586-594, 2020 08 15.
Article in English | MEDLINE | ID: mdl-32352834

ABSTRACT

Rationale: Idiopathic and heritable pulmonary arterial hypertension (PAH) are rare but comprise a genetically heterogeneous patient group. RNA sequencing linked to the underlying genetic architecture can be used to better understand the underlying pathology by identifying key signaling pathways and stratify patients more robustly according to clinical risk.Objectives: To use a three-stage design of RNA discovery, RNA validation and model construction, and model validation to define a set of PAH-associated RNAs and a single summarizing RNA model score. To define genes most likely to be involved in disease development, we performed Mendelian randomization (MR) analysis.Methods: RNA sequencing was performed on whole-blood samples from 359 patients with idiopathic, heritable, and drug-induced PAH and 72 age- and sex-matched healthy volunteers. The score was evaluated against disease severity markers including survival analysis using all-cause mortality from diagnosis. MR used known expression quantitative trait loci and summary statistics from a PAH genome-wide association study.Measurements and Main Results: We identified 507 genes with differential RNA expression in patients with PAH compared with control subjects. A model of 25 RNAs distinguished PAH with 87% accuracy (area under the curve 95% confidence interval: 0.791-0.945) in model validation. The RNA model score was associated with disease severity and long-term survival (P = 4.66 × 10-6) in PAH. MR detected an association between SMAD5 levels and PAH disease susceptibility (odds ratio, 0.317; 95% confidence interval, 0.129-0.776; P = 0.012).Conclusions: A whole-blood RNA signature of PAH, which includes RNAs relevant to disease pathogenesis, associates with disease severity and identifies patients with poor clinical outcomes. Genetic variants associated with lower SMAD5 expression may increase susceptibility to PAH.


Subject(s)
Familial Primary Pulmonary Hypertension/blood , Familial Primary Pulmonary Hypertension/genetics , RNA/blood , Adult , Cohort Studies , Female , Gene Expression Profiling , Humans , Male , Mendelian Randomization Analysis , Middle Aged
7.
Eur Respir J ; 55(2)2020 02.
Article in English | MEDLINE | ID: mdl-31744833

ABSTRACT

Pulmonary arterial hypertension (PAH) is a rare disease that leads to premature death from right heart failure. It is strongly associated with elevated red cell distribution width (RDW), a correlate of several iron status biomarkers. High RDW values can signal early-stage iron deficiency or iron deficiency anaemia. This study investigated whether elevated RDW is causally associated with PAH.A two-sample Mendelian randomisation (MR) approach was applied to investigate whether genetic predisposition to higher levels of RDW increases the odds of developing PAH. Primary and secondary MR analyses were performed using all available genome-wide significant RDW variants (n=179) and five genome-wide significant RDW variants that act via systemic iron status, respectively.We confirmed the observed association between RDW and PAH (OR 1.90, 95% CI 1.80-2.01) in a multicentre case-control study (cases n=642, disease controls n=15 889). The primary MR analysis was adequately powered to detect a causal effect (odds ratio) between 1.25 and 1.52 or greater based on estimates reported in the RDW genome-wide association study or from our own data. There was no evidence for a causal association between RDW and PAH in either the primary (ORcausal 1.07, 95% CI 0.92-1.24) or the secondary (ORcausal 1.09, 95% CI 0.77-1.54) MR analysis.The results suggest that at least some of the observed association of RDW with PAH is secondary to disease progression. Results of iron therapeutic trials in PAH should be interpreted with caution, as any improvements observed may not be mechanistically linked to the development of PAH.


Subject(s)
Hypertension, Pulmonary , Pulmonary Arterial Hypertension , Case-Control Studies , Erythrocyte Indices , Genome-Wide Association Study , Humans , Hypertension, Pulmonary/genetics
8.
Hum Mol Genet ; 26(21): 4301-4313, 2017 11 01.
Article in English | MEDLINE | ID: mdl-28973304

ABSTRACT

Psoriasis is a common inflammatory skin disorder for which multiple genetic susceptibility loci have been identified, but few resolved to specific functional variants. In this study, we sought to identify common and rare psoriasis-associated gene-centric variation. Using exome arrays we genotyped four independent cohorts, totalling 11 861 psoriasis cases and 28 610 controls, aggregating the dataset through statistical meta-analysis. Single variant analysis detected a previously unreported risk locus at TNFSF15 (rs6478108; P = 1.50 × 10-8, OR = 1.10), and association of common protein-altering variants at 11 loci previously implicated in psoriasis susceptibility. We validate previous reports of protective low-frequency protein-altering variants within IFIH1 (encoding an innate antiviral receptor) and TYK2 (encoding a Janus kinase), in each case establishing a further series of protective rare variants (minor allele frequency < 0.01) via gene-wide aggregation testing (IFIH1: pburden = 2.53 × 10-7, OR = 0.707; TYK2: pburden = 6.17 × 10-4, OR = 0.744). Both genes play significant roles in type I interferon (IFN) production and signalling. Several of the protective rare and low-frequency variants in IFIH1 and TYK2 disrupt conserved protein domains, highlighting potential mechanisms through which their effect may be exerted.


Subject(s)
Psoriasis/genetics , Tumor Necrosis Factor Ligand Superfamily Member 15/genetics , Alleles , Case-Control Studies , Cohort Studies , Exome , Female , Gene Frequency/genetics , Genetic Predisposition to Disease/genetics , Genetic Variation/genetics , Genome-Wide Association Study , Genotype , Humans , Interferon-Induced Helicase, IFIH1/genetics , Interferon-Induced Helicase, IFIH1/metabolism , Male , Polymorphism, Single Nucleotide/genetics , Psoriasis/physiopathology , Risk Factors , TYK2 Kinase/genetics , TYK2 Kinase/metabolism , Tumor Necrosis Factor Ligand Superfamily Member 15/metabolism , Exome Sequencing
9.
Am J Hum Genet ; 98(6): 1092-1100, 2016 Jun 02.
Article in English | MEDLINE | ID: mdl-27236921

ABSTRACT

Bacteremia (bacterial bloodstream infection) is a major cause of illness and death in sub-Saharan Africa but little is known about the role of human genetics in susceptibility. We conducted a genome-wide association study of bacteremia susceptibility in more than 5,000 Kenyan children as part of the Wellcome Trust Case Control Consortium 2 (WTCCC2). Both the blood-culture-proven bacteremia case subjects and healthy infants as controls were recruited from Kilifi, on the east coast of Kenya. Streptococcus pneumoniae is the most common cause of bacteremia in Kilifi and was thus the focus of this study. We identified an association between polymorphisms in a long intergenic non-coding RNA (lincRNA) gene (AC011288.2) and pneumococcal bacteremia and replicated the results in the same population (p combined = 1.69 × 10(-9); OR = 2.47, 95% CI = 1.84-3.31). The susceptibility allele is African specific, derived rather than ancestral, and occurs at low frequency (2.7% in control subjects and 6.4% in case subjects). Our further studies showed AC011288.2 expression only in neutrophils, a cell type that is known to play a major role in pneumococcal clearance. Identification of this novel association will further focus research on the role of lincRNAs in human infectious disease.


Subject(s)
Bacteremia/genetics , Pneumonia, Pneumococcal/genetics , Polymorphism, Genetic/genetics , RNA, Long Noncoding/genetics , Streptococcus pneumoniae/genetics , Adolescent , Bacteremia/microbiology , Bacteremia/pathology , Case-Control Studies , Child , Child, Preschool , Genome-Wide Association Study , Humans , Infant , Infant, Newborn , Kenya/epidemiology , Pneumonia, Pneumococcal/microbiology , Pneumonia, Pneumococcal/pathology , Risk Factors
10.
Eur Respir J ; 53(1)2019 01.
Article in English | MEDLINE | ID: mdl-30545973

ABSTRACT

Since 2000 there have been major advances in our understanding of the genetic and genomics of pulmonary arterial hypertension (PAH), although there remains much to discover. Based on existing knowledge, around 25-30% of patients diagnosed with idiopathic PAH have an underlying Mendelian genetic cause for their condition and should be classified as heritable PAH (HPAH). Here, we summarise the known genetic and genomic drivers of PAH, the insights these provide into pathobiology, and the opportunities afforded for development of novel therapeutic approaches. In addition, factors determining the incomplete penetrance observed in HPAH are discussed. The currently available approaches to genetic testing and counselling, and the impact of a genetic diagnosis on clinical management of the patient with PAH, are presented. Advances in DNA sequencing technology are rapidly expanding our ability to undertake genomic studies at scale in large cohorts. In the future, such studies will provide a more complete picture of the genetic contribution to PAH and, potentially, a molecular classification of this disease.


Subject(s)
Genetic Predisposition to Disease/epidemiology , Genomics/trends , Hypertension, Pulmonary/diagnosis , Hypertension, Pulmonary/genetics , Animals , Bone Morphogenetic Protein Receptors, Type II/genetics , Familial Primary Pulmonary Hypertension , Genome-Wide Association Study/trends , Humans , Hypertension, Pulmonary/epidemiology , Mutation
11.
Eur Respir J ; 53(3)2019 03.
Article in English | MEDLINE | ID: mdl-30655285

ABSTRACT

Chronic thromboembolic pulmonary hypertension (CTEPH) is an important consequence of pulmonary embolism that is associated with abnormalities in haemostasis. We investigated the ADAMTS13-von Willebrand factor (VWF) axis in CTEPH, including its relationship with disease severity, inflammation, ABO groups and ADAMTS13 genetic variants.ADAMTS13 and VWF plasma antigen levels were measured in patients with CTEPH (n=208), chronic thromboembolic disease without pulmonary hypertension (CTED) (n=35), resolved pulmonary embolism (n=28), idiopathic pulmonary arterial hypertension (n=30) and healthy controls (n=68). CTEPH genetic ABO associations and protein quantitative trait loci were investigated. ADAMTS13-VWF axis abnormalities were assessed in CTEPH and healthy control subsets by measuring ADAMTS13 activity, D-dimers and VWF multimeric size.Patients with CTEPH had decreased ADAMTS13 (adjusted ß -23.4%, 95% CI -30.9- -15.1%, p<0.001) and increased VWF levels (ß +75.5%, 95% CI 44.8-113%, p<0.001) compared to healthy controls. ADAMTS13 levels remained low after reversal of pulmonary hypertension by pulmonary endarterectomy surgery and were equally reduced in CTED. We identified a genetic variant near the ADAMTS13 gene associated with ADAMTS13 protein that accounted for ∼8% of the variation in levels.The ADAMTS13-VWF axis is dysregulated in CTEPH. This is unrelated to pulmonary hypertension, disease severity or markers of systemic inflammation and implicates the ADAMTS13-VWF axis in CTEPH pathobiology.


Subject(s)
ADAMTS13 Protein/genetics , Hypertension, Pulmonary/physiopathology , Pulmonary Embolism/physiopathology , von Willebrand Factor/analysis , Adult , Aged , Aged, 80 and over , Biomarkers , Case-Control Studies , Chronic Disease , Endarterectomy , Female , Humans , Hypertension, Pulmonary/genetics , Linear Models , Male , Middle Aged , Multivariate Analysis , Pulmonary Embolism/genetics , Thrombosis/genetics , Thrombosis/physiopathology
12.
Nature ; 498(7453): 232-5, 2013 Jun 13.
Article in English | MEDLINE | ID: mdl-23698362

ABSTRACT

Genome-wide association studies (GWAS) have identified common variants of modest-effect size at hundreds of loci for common autoimmune diseases; however, a substantial fraction of heritability remains unexplained, to which rare variants may contribute. To discover rare variants and test them for association with a phenotype, most studies re-sequence a small initial sample size and then genotype the discovered variants in a larger sample set. This approach fails to analyse a large fraction of the rare variants present in the entire sample set. Here we perform simultaneous amplicon-sequencing-based variant discovery and genotyping for coding exons of 25 GWAS risk genes in 41,911 UK residents of white European origin, comprising 24,892 subjects with six autoimmune disease phenotypes and 17,019 controls, and show that rare coding-region variants at known loci have a negligible role in common autoimmune disease susceptibility. These results do not support the rare-variant synthetic genome-wide-association hypothesis (in which unobserved rare causal variants lead to association detected at common tag variants). Many known autoimmune disease risk loci contain multiple, independently associated, common and low-frequency variants, and so genes at these loci are a priori stronger candidates for harbouring rare coding-region variants than other genes. Our data indicate that the missing heritability for common autoimmune diseases may not be attributable to the rare coding-region variant portion of the allelic spectrum, but perhaps, as others have proposed, may be a result of many common-variant loci of weak effect.


Subject(s)
Autoimmune Diseases/genetics , Genetic Predisposition to Disease/genetics , Genetic Variation/genetics , Open Reading Frames/genetics , Exons/genetics , Gene Frequency , Genome-Wide Association Study , Humans , Models, Genetic , Mutation/genetics , Phenotype , Sample Size , United Kingdom , White People/genetics
13.
Hum Mol Genet ; 25(9): 1836-45, 2016 05 01.
Article in English | MEDLINE | ID: mdl-26945007

ABSTRACT

Familial medullary thyroid cancer (MTC) and its precursor, C cell hyperplasia (CCH), is associated with germline RET mutations causing multiple endocrine neoplasia type 2. However, some rare families with apparent MTC/CCH predisposition do not have a detectable RET mutation. To identify novel MTC/CCH predisposition genes we undertook exome resequencing studies in a family with apparent predisposition to MTC/CCH and no identifiable RET mutation. We identified a novel ESR2 frameshift mutation, c.948delT, which segregated with histological diagnosis following thyroid surgery in family members and demonstrated loss of ESR2-encoded ERß expression in the MTC tumour. ERα and ERß form heterodimers binding DNA at specific oestrogen-responsive elements (EREs) to regulate gene transcription. ERß represses ERα-mediated activation of the ERE and the RET promoter contains three EREs. In vitro, we showed that ESR2 c.948delT results in unopposed ERα mediated increased cellular proliferation, activation of the ERE and increased RET expression. In vivo, immunostaining of CCH and MTC using an anti-RET antibody demonstrated increased RET expression. Together these findings identify germline ESR2 mutation as a novel cause of familial MTC/CCH and provide important insights into a novel mechanism causing increased RET expression in tumourigenesis.


Subject(s)
Carcinoma, Medullary/congenital , Estrogen Receptor beta/genetics , Gene Expression Regulation, Neoplastic , Germ-Line Mutation/genetics , Multiple Endocrine Neoplasia Type 2a/genetics , Multiple Endocrine Neoplasia Type 2a/metabolism , Proto-Oncogene Proteins c-ret/metabolism , Thyroid Neoplasms/genetics , Thyroid Neoplasms/metabolism , Adult , Carcinoma, Medullary/genetics , Carcinoma, Medullary/metabolism , Carcinoma, Medullary/pathology , Cell Proliferation , Disease Susceptibility , Genotype , Humans , Male , Multiple Endocrine Neoplasia Type 2a/pathology , Pedigree , Proto-Oncogene Proteins c-ret/genetics , Thyroid Neoplasms/pathology , Tumor Cells, Cultured , Up-Regulation , Young Adult
14.
Am J Med Genet B Neuropsychiatr Genet ; 177(7): 641-657, 2018 10.
Article in English | MEDLINE | ID: mdl-30325587

ABSTRACT

Individuals with psychiatric disorders have elevated rates of autoimmune comorbidity and altered immune signaling. It is unclear whether these altered immunological states have a shared genetic basis with those psychiatric disorders. The present study sought to use existing summary-level data from previous genome-wide association studies to determine if commonly varying single nucleotide polymorphisms are shared between psychiatric and immune-related phenotypes. We estimated heritability and examined pair-wise genetic correlations using the linkage disequilibrium score regression (LDSC) and heritability estimation from summary statistics methods. Using LDSC, we observed significant genetic correlations between immune-related disorders and several psychiatric disorders, including anorexia nervosa, attention deficit-hyperactivity disorder, bipolar disorder, major depression, obsessive compulsive disorder, schizophrenia, smoking behavior, and Tourette syndrome. Loci significantly mediating genetic correlations were identified for schizophrenia when analytically paired with Crohn's disease, primary biliary cirrhosis, systemic lupus erythematosus, and ulcerative colitis. We report significantly correlated loci and highlight those containing genome-wide associations and candidate genes for respective disorders. We also used the LDSC method to characterize genetic correlations among the immune-related phenotypes. We discuss our findings in the context of relevant genetic and epidemiological literature, as well as the limitations and caveats of the study.


Subject(s)
Autoimmune Diseases/genetics , Mental Disorders/genetics , Autoimmune Diseases/physiopathology , Comorbidity , Databases, Factual , Female , Genetic Predisposition to Disease , Genome-Wide Association Study/methods , Humans , Linkage Disequilibrium , Male , Mental Disorders/physiopathology , Multifactorial Inheritance , Polymorphism, Single Nucleotide , White People/genetics
15.
EMBO J ; 32(9): 1225-37, 2013 May 02.
Article in English | MEDLINE | ID: mdl-23481255

ABSTRACT

Adenosine diphosphate (ADP)-ribosylation is a post-translational protein modification implicated in the regulation of a range of cellular processes. A family of proteins that catalyse ADP-ribosylation reactions are the poly(ADP-ribose) (PAR) polymerases (PARPs). PARPs covalently attach an ADP-ribose nucleotide to target proteins and some PARP family members can subsequently add additional ADP-ribose units to generate a PAR chain. The hydrolysis of PAR chains is catalysed by PAR glycohydrolase (PARG). PARG is unable to cleave the mono(ADP-ribose) unit directly linked to the protein and although the enzymatic activity that catalyses this reaction has been detected in mammalian cell extracts, the protein(s) responsible remain unknown. Here, we report the homozygous mutation of the c6orf130 gene in patients with severe neurodegeneration, and identify C6orf130 as a PARP-interacting protein that removes mono(ADP-ribosyl)ation on glutamate amino acid residues in PARP-modified proteins. X-ray structures and biochemical analysis of C6orf130 suggest a mechanism of catalytic reversal involving a transient C6orf130 lysyl-(ADP-ribose) intermediate. Furthermore, depletion of C6orf130 protein in cells leads to proliferation and DNA repair defects. Collectively, our data suggest that C6orf130 enzymatic activity has a role in the turnover and recycling of protein ADP-ribosylation, and we have implicated the importance of this protein in supporting normal cellular function in humans.


Subject(s)
Glycoside Hydrolases/physiology , Neurodegenerative Diseases/enzymology , Poly Adenosine Diphosphate Ribose/physiology , Thiolester Hydrolases/physiology , Amino Acid Sequence , Base Sequence , Cells, Cultured , Child , Child, Preschool , Family , Female , Glycoside Hydrolases/genetics , HEK293 Cells , HeLa Cells , Humans , Male , Models, Molecular , Molecular Sequence Data , Neurodegenerative Diseases/genetics , Pedigree , Poly Adenosine Diphosphate Ribose/genetics , Protein Processing, Post-Translational/genetics , Sequence Homology, Amino Acid , Thiolester Hydrolases/genetics
16.
Am J Hum Genet ; 94(5): 790-7, 2014 May 01.
Article in English | MEDLINE | ID: mdl-24791904

ABSTRACT

Adaptor protein complex 1 (AP-1) is an evolutionary conserved heterotetramer that promotes vesicular trafficking between the trans-Golgi network and the endosomes. The knockout of most murine AP-1 complex subunits is embryonically lethal, so the identification of human disease-associated alleles has the unique potential to deliver insights into gene function. Here, we report two founder mutations (c.11T>G [p.Phe4Cys] and c.97C>T [p.Arg33Trp]) in AP1S3, the gene encoding AP-1 complex subunit σ1C, in 15 unrelated individuals with a severe autoinflammatory skin disorder known as pustular psoriasis. Because the variants are predicted to destabilize the 3D structure of the AP-1 complex, we generated AP1S3-knockdown cell lines to investigate the consequences of AP-1 deficiency in skin keratinocytes. We found that AP1S3 silencing disrupted the endosomal translocation of the innate pattern-recognition receptor TLR-3 (Toll-like receptor 3) and resulted in a marked inhibition of downstream signaling. These findings identify pustular psoriasis as an autoinflammatory phenotype caused by defects in vesicular trafficking and demonstrate a requirement of AP-1 for Toll-like receptor homeostasis.


Subject(s)
Adaptor Protein Complex 1/genetics , Psoriasis/genetics , Psoriasis/metabolism , Toll-Like Receptor 3/metabolism , Adaptor Protein Complex 1/chemistry , Amino Acid Sequence , Amino Acid Substitution , Cell Line , Female , Gene Knockdown Techniques , Humans , Male , Molecular Sequence Data , Protein Conformation , Protein Transport/genetics
17.
Hum Mutat ; 37(11): 1157-1161, 2016 11.
Article in English | MEDLINE | ID: mdl-27492651

ABSTRACT

Hereditary spastic paraplegias (HSPs) are genetically and clinically heterogeneous axonopathies primarily affecting upper motor neurons and, in complex forms, additional neurons. Here, we report two families with distinct recessive mutations in TFG, previously suggested to cause HSP based on findings in a single small family with complex HSP. The first carried a homozygous c.317G>A (p.R106H) variant and presented with pure HSP. The second carried the same homozygous c.316C>T (p.R106C) variant previously reported and displayed a similarly complex phenotype including optic atrophy. Haplotyping and bisulfate sequencing revealed evidence for a c.316C>T founder allele, as well as for a c.316_317 mutation hotspot. Expression of mutant TFG proteins in cultured neurons revealed mitochondrial fragmentation, the extent of which correlated with clinical severity. Our findings confirm the causal nature of bi-allelic TFG mutations for HSP, broaden the clinical and mutational spectra, and suggest mitochondrial impairment to represent a pathomechanistic link to other neurodegenerative conditions.


Subject(s)
Mutation, Missense , Proteins/genetics , Proteins/metabolism , Spastic Paraplegia, Hereditary/pathology , Animals , Cells, Cultured , Female , Genetic Predisposition to Disease , Humans , Magnetic Resonance Imaging/methods , Male , Mice , Mitochondria/pathology , Neurons/cytology , Neurons/metabolism , Neurons/pathology , Pedigree , Sequence Analysis, DNA , Spastic Paraplegia, Hereditary/genetics , Spastic Paraplegia, Hereditary/metabolism
18.
Hum Mutat ; 36(12): 1113-27, 2015 Dec.
Article in English | MEDLINE | ID: mdl-26387786

ABSTRACT

Pulmonary arterial hypertension (PAH) is an often fatal disorder resulting from several causes including heterogeneous genetic defects. While mutations in the bone morphogenetic protein receptor type II (BMPR2) gene are the single most common causal factor for hereditary cases, pathogenic mutations have been observed in approximately 25% of idiopathic PAH patients without a prior family history of disease. Additional defects of the transforming growth factor beta pathway have been implicated in disease pathogenesis. Specifically, studies have confirmed activin A receptor type II-like 1 (ACVRL1), endoglin (ENG), and members of the SMAD family as contributing to PAH both with and without associated clinical phenotypes. Most recently, next-generation sequencing has identified novel, rare genetic variation implicated in the PAH disease spectrum. Of importance, several identified genetic factors converge on related pathways and provide significant insight into the development, maintenance, and pathogenetic transformation of the pulmonary vascular bed. Together, these analyses represent the largest comprehensive compilation of BMPR2 and associated genetic risk factors for PAH, comprising known and novel variation. Additionally, with the inclusion of an allelic series of locus-specific variation in BMPR2, these data provide a key resource in data interpretation and development of contemporary therapeutic and diagnostic tools.


Subject(s)
Hypertension, Pulmonary/genetics , Animals , Bone Morphogenetic Protein Receptors, Type II/chemistry , Bone Morphogenetic Protein Receptors, Type II/genetics , Bone Morphogenetic Protein Receptors, Type II/metabolism , Disease Models, Animal , Genetic Association Studies , Genetic Counseling , Genetic Predisposition to Disease , Genetic Variation , High-Throughput Nucleotide Sequencing , Humans , Hypertension, Pulmonary/diagnosis , Hypertension, Pulmonary/metabolism , Multigene Family , Mutation , Signal Transduction , Transforming Growth Factor beta/genetics , Transforming Growth Factor beta/metabolism
19.
Am J Hum Genet ; 91(2): 358-64, 2012 Aug 10.
Article in English | MEDLINE | ID: mdl-22795537

ABSTRACT

Excessive growth of terminal hair around the elbows (hypertrichosis cubiti) has been reported both in isolation and in association with a variable spectrum of associated phenotypic features. We identified a cohort of six individuals with hypertrichosis cubiti associated with short stature, intellectual disability, and a distinctive facial appearance, consistent with a diagnosis of Wiedemann-Steiner syndrome (WSS). Utilizing a whole-exome sequencing approach, we identified de novo mutations in MLL in five of the six individuals. MLL encodes a histone methyltransferase that regulates chromatin-mediated transcription through the catalysis of methylation of histone H3K4. Each of the five mutations is predicted to result in premature termination of the protein product. Furthermore, we demonstrate that transcripts arising from the mutant alleles are subject to nonsense-mediated decay. These findings define the genetic basis of WSS, provide additional evidence for the role of haploinsufficency of histone-modification enzymes in multiple-congenital-anomaly syndromes, and further illustrate the importance of the regulation of histone modification in development.


Subject(s)
Abnormalities, Multiple/genetics , Growth Disorders/genetics , Hypertrichosis/congenital , Myeloid-Lymphoid Leukemia Protein/genetics , Abnormalities, Multiple/pathology , Base Sequence , Exome/genetics , Gene Components , Growth Disorders/pathology , Haploinsufficiency/genetics , Histone-Lysine N-Methyltransferase , Humans , Hypertrichosis/genetics , Hypertrichosis/pathology , Molecular Sequence Data , Mutation/genetics , Sequence Analysis, DNA
20.
Am J Hum Genet ; 90(2): 290-4, 2012 Feb 10.
Article in English | MEDLINE | ID: mdl-22265017

ABSTRACT

Genitopatellar syndrome (GPS) is a rare disorder in which patellar aplasia or hypoplasia is associated with external genital anomalies and severe intellectual disability. Using an exome-sequencing approach, we identified de novo mutations of KAT6B in five individuals with GPS; a single nonsense variant and three frameshift indels, including a 4 bp deletion observed in two cases. All identified mutations are located within the terminal exon of the gene and are predicted to generate a truncated protein product lacking evolutionarily conserved domains. KAT6B encodes a member of the MYST family of histone acetyltranferases. We demonstrate a reduced level of both histone H3 and H4 acetylation in patient-derived cells suggesting that dysregulation of histone acetylation is a direct functional consequence of GPS alleles. These findings define the genetic basis of GPS and illustrate the complex role of the regulation of histone acetylation during development.


Subject(s)
Histone Acetyltransferases/genetics , Musculoskeletal Abnormalities/genetics , Mutation , Urogenital Abnormalities/genetics , Acetylation , Alleles , Animals , Exome , Exons , Female , Histones/metabolism , Humans , Intellectual Disability/enzymology , Intellectual Disability/genetics , Male , Mice , Musculoskeletal Abnormalities/enzymology , Sequence Analysis, DNA/methods , Urogenital Abnormalities/enzymology
SELECTION OF CITATIONS
SEARCH DETAIL