Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 44
Filtrar
1.
Nat Commun ; 14(1): 3855, 2023 06 29.
Artículo en Inglés | MEDLINE | ID: mdl-37386006

RESUMEN

Microsatellites are polymorphic tracts of short tandem repeats with one to six base-pair (bp) motifs and are some of the most polymorphic variants in the genome. Using 6084 Icelandic parent-offspring trios we estimate 63.7 (95% CI: 61.9-65.4) microsatellite de novo mutations (mDNMs) per offspring per generation, excluding one bp repeats motifs (homopolymers) the estimate is 48.2 mDNMs (95% CI: 46.7-49.6). Paternal mDNMs occur at longer repeats than maternal ones, which are in turn larger with a mean size of 3.4 bp vs 3.1 bp for paternal ones. mDNMs increase by 0.97 (95% CI: 0.90-1.04) and 0.31 (95% CI: 0.25-0.37) per year of father's and mother's age at conception, respectively. Here, we find two independent coding variants that associate with the number of mDNMs transmitted to offspring; The minor allele of a missense variant (allele frequency (AF) = 1.9%) in MSH2, a mismatch repair gene, increases transmitted mDNMs from both parents (effect: 13.1 paternal and 7.8 maternal mDNMs). A synonymous variant (AF = 20.3%) in NEIL2, a DNA damage repair gene, increases paternally transmitted mDNMs (effect: 4.4 mDNMs). Thus, the microsatellite mutation rate in humans is in part under genetic control.


Asunto(s)
Reparación de la Incompatibilidad de ADN , Mutación de Línea Germinal , Humanos , Alelos , Mutación de Línea Germinal/genética , Repeticiones de Microsatélite/genética , Células Germinativas
2.
Nat Commun ; 13(1): 5701, 2022 09 28.
Artículo en Inglés | MEDLINE | ID: mdl-36171188

RESUMEN

By the end of July 2021, the majority of the Icelandic population had received vaccination against COVID-19. In mid-July a wave of SARS-CoV-2 infections, dominated by the Delta variant, spread through the population, followed by an Omicron wave in December. A booster vaccination campaign was initiated to curb the spread of the virus. We estimate the risk of infection for different vaccine combinations using vaccination data from 276,028 persons and 963,557 qPCR tests for 277,687 persons. We measure anti-Spike-RBD antibody levels and ACE2-Spike binding inhibitory activity in 371 persons who received one of four recommended vaccination schedules with or without an mRNA vaccine booster. Overall, we find different antibody levels and inhibitory activity in recommended vaccination schedules, reflected in the observed risk of SARS-CoV-2 infections. We observe an increased protection following mRNA boosters, against both Omicron and Delta variant infections, although BNT162b2 boosters provide greater protection against Omicron than mRNA-1273 boosters.


Asunto(s)
COVID-19 , Vacunas Virales , Enzima Convertidora de Angiotensina 2 , Anticuerpos Antivirales/metabolismo , Vacuna BNT162 , COVID-19/epidemiología , COVID-19/prevención & control , Humanos , Islandia/epidemiología , ARN Mensajero , SARS-CoV-2/genética , Vacunación , Vacunas Sintéticas , Vacunas de ARNm
3.
Nature ; 607(7920): 732-740, 2022 07.
Artículo en Inglés | MEDLINE | ID: mdl-35859178

RESUMEN

Detailed knowledge of how diversity in the sequence of the human genome affects phenotypic diversity depends on a comprehensive and reliable characterization of both sequences and phenotypic variation. Over the past decade, insights into this relationship have been obtained from whole-exome sequencing or whole-genome sequencing of large cohorts with rich phenotypic data1,2. Here we describe the analysis of whole-genome sequencing of 150,119 individuals from the UK Biobank3. This constitutes a set of high-quality variants, including 585,040,410 single-nucleotide polymorphisms, representing 7.0% of all possible human single-nucleotide polymorphisms, and 58,707,036 indels. This large set of variants allows us to characterize selection based on sequence variation within a population through a depletion rank score of windows along the genome. Depletion rank analysis shows that coding exons represent a small fraction of regions in the genome subject to strong sequence conservation. We define three cohorts within the UK Biobank: a large British Irish cohort, a smaller African cohort and a South Asian cohort. A haplotype reference panel is provided that allows reliable imputation of most variants carried by three or more sequenced individuals. We identified 895,055 structural variants and 2,536,688 microsatellites, groups of variants typically excluded from large-scale whole-genome sequencing studies. Using this formidable new resource, we provide several examples of trait associations for rare variants with large effects not found previously through studies based on whole-exome sequencing and/or imputation.


Asunto(s)
Bancos de Muestras Biológicas , Bases de Datos Genéticas , Variación Genética , Genoma Humano , Genómica , Secuenciación Completa del Genoma , África/etnología , Asia/etnología , Estudios de Cohortes , Secuencia Conservada , Exones/genética , Genoma Humano/genética , Haplotipos/genética , Humanos , Mutación INDEL , Irlanda/etnología , Repeticiones de Microsatélite , Polimorfismo de Nucleótido Simple/genética , Reino Unido
4.
Clin Microbiol Infect ; 28(6): 852-858, 2022 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-35182757

RESUMEN

OBJECTIVES: The spread of SARS-CoV-2 is dependent on several factors, both biological and behavioural. The effectiveness of nonpharmaceutical interventions can be attributed largely to changes in human behaviour, but quantifying this effect remains challenging. Reconstructing the transmission tree of the third wave of SARS-CoV-2 infections in Iceland using contact tracing and viral sequence data from 2522 cases enables us to directly compare the infectiousness of distinct groups of persons. METHODS: The transmission tree enables us to model the effect that a given population prevalence of vaccination would have had on the third wave had one of three different vaccination strategies been implemented before that time. This allows us to compare the effectiveness of the strategies in terms of minimizing the number of cases, deaths, critical cases, and severe cases. RESULTS: We found that people diagnosed outside of quarantine (Rˆ=1.31) were 89% more infectious than those diagnosed while in quarantine (Rˆ=0.70) and that infectiousness decreased as a function of time spent in quarantine before diagnosis, with people diagnosed outside of quarantine being 144% more infectious than those diagnosed after ≥3 days in quarantine (Rˆ=0.54). People of working age, 16 to 66 years (Rˆ=1.08), were 46% more infectious than those outside of that age range (Rˆ=0.74). DISCUSSION: We found that vaccinating the population in order of ascending age or uniformly at random would have prevented more infections per vaccination than vaccinating in order of descending age, without significantly affecting the expected number of deaths, critical cases, or severe cases.


Asunto(s)
COVID-19 , Adolescente , Adulto , Anciano , COVID-19/epidemiología , COVID-19/prevención & control , Brotes de Enfermedades/prevención & control , Humanos , Islandia/epidemiología , Persona de Mediana Edad , Modelos Teóricos , SARS-CoV-2 , Vacunación , Adulto Joven
5.
Nat Commun ; 12(1): 3633, 2021 06 15.
Artículo en Inglés | MEDLINE | ID: mdl-34131116

RESUMEN

A pressing concern in the SARS-CoV-2 epidemic and other viral outbreaks, is the extent to which the containment measures are halting the viral spread. A straightforward way to assess this is to tally the active cases and the recovered ones throughout the epidemic. Here, we show how epidemic control can be assessed with molecular information during a well characterized epidemic in Iceland. We demonstrate how the viral concentration decreased in those newly diagnosed as the epidemic transitioned from exponential growth phase to containment phase. The viral concentration in the cases identified in population screening decreased faster than in those symptomatic and considered at high risk and that were targeted by the healthcare system. The viral concentration persists in recovering individuals as we found that half of the cases are still positive after two weeks. We demonstrate that accumulation of mutations in SARS-CoV-2 genome can be exploited to track the rate of new viral generations throughout the different phases of the epidemic, where the accumulation of mutations decreases as the transmission rate decreases in the containment phase. Overall, the molecular signatures of SARS-CoV-2 infections contain valuable epidemiological information that can be used to assess the effectiveness of containment measures.


Asunto(s)
Benchmarking/métodos , COVID-19/epidemiología , Epidemias , SARS-CoV-2/genética , Animales , COVID-19/virología , Humanos , Islandia/epidemiología , Epidemiología Molecular , Mutación , ARN Viral
6.
Commun Biol ; 4(1): 706, 2021 06 09.
Artículo en Inglés | MEDLINE | ID: mdl-34108613

RESUMEN

Age-related hearing impairment (ARHI) is the most common sensory disorder in older adults. We conducted a genome-wide association meta-analysis of 121,934 ARHI cases and 591,699 controls from Iceland and the UK. We identified 21 novel sequence variants, of which 13 are rare, under either additive or recessive models. Of special interest are a missense variant in LOXHD1 (MAF = 1.96%) and a tandem duplication in FBF1 covering 4 exons (MAF = 0.22%) associating with ARHI (OR = 3.7 for homozygotes, P = 1.7 × 10-22 and OR = 4.2 for heterozygotes, P = 5.7 × 10-27, respectively). We constructed an ARHI genetic risk score (GRS) using common variants and showed that a common variant GRS can identify individuals at risk comparable to carriers of rare high penetrance variants. Furthermore, we found that ARHI and tinnitus share genetic causes. This study sheds a new light on the genetic architecture of ARHI, through several rare variants in both Mendelian deafness genes and genes not previously linked to hearing.


Asunto(s)
Pérdida Auditiva/genética , Adulto , Anciano , Anciano de 80 o más Años , Envejecimiento/genética , Femenino , Genes/genética , Predisposición Genética a la Enfermedad , Variación Genética/genética , Estudio de Asociación del Genoma Completo , Humanos , Masculino , Persona de Mediana Edad , Factores de Riesgo
7.
Cancer Res ; 81(8): 1954-1964, 2021 04 15.
Artículo en Inglés | MEDLINE | ID: mdl-33602785

RESUMEN

The success of genome-wide association studies (GWAS) in identifying common, low-penetrance variant-cancer associations for the past decade is undisputed. However, discovering additional high-penetrance cancer mutations in unknown cancer predisposing genes requires detection of variant-cancer association of ultra-rare coding variants. Consequently, large-scale next-generation sequence data with associated phenotype information are needed. Here, we used genotype data on 166,281 Icelanders, of which, 49,708 were whole-genome sequenced and 408,595 individuals from the UK Biobank, of which, 41,147 were whole-exome sequenced, to test for association between loss-of-function burden in autosomal genes and basal cell carcinoma (BCC), the most common cancer in Caucasians. A total of 25,205 BCC cases and 683,058 controls were tested. Rare germline loss-of-function variants in PTPN14 conferred substantial risks of BCC (OR, 8.0; P = 1.9 × 10-12), with a quarter of carriers getting BCC before age 70 and over half in their lifetime. Furthermore, common variants at the PTPN14 locus were associated with BCC, suggesting PTPN14 as a new, high-impact BCC predisposition gene. A follow-up investigation of 24 cancers and three benign tumor types showed that PTPN14 loss-of-function variants are associated with high risk of cervical cancer (OR, 12.7, P = 1.6 × 10-4) and low age at diagnosis. Our findings, using power-increasing methods with high-quality rare variant genotypes, highlight future prospects for new discoveries on carcinogenesis. SIGNIFICANCE: This study identifies the tumor-suppressor gene PTPN14 as a high-impact BCC predisposition gene and indicates that inactivation of PTPN14 by germline sequence variants may also lead to increased risk of cervical cancer.


Asunto(s)
Carcinoma Basocelular/genética , Mutación con Pérdida de Función , Penetrancia , Proteínas Tirosina Fosfatasas no Receptoras/genética , Neoplasias Cutáneas/genética , Neoplasias del Cuello Uterino/genética , Factores de Edad , Carcinoma Basocelular/epidemiología , Estudios de Casos y Controles , Femenino , Frecuencia de los Genes , Genes Supresores de Tumor , Predisposición Genética a la Enfermedad , Pruebas Genéticas , Estudio de Asociación del Genoma Completo , Genotipo , Mutación de Línea Germinal , Humanos , Islandia/epidemiología , Masculino , Oportunidad Relativa , Neoplasias Cutáneas/epidemiología , Bancos de Tejidos/estadística & datos numéricos , Reino Unido/epidemiología , Neoplasias del Cuello Uterino/epidemiología , Secuenciación del Exoma/estadística & datos numéricos , Secuenciación Completa del Genoma/estadística & datos numéricos
8.
Nat Genet ; 53(1): 27-34, 2021 01.
Artículo en Inglés | MEDLINE | ID: mdl-33414551

RESUMEN

Despite the important role that monozygotic twins have played in genetics research, little is known about their genomic differences. Here we show that monozygotic twins differ on average by 5.2 early developmental mutations and that approximately 15% of monozygotic twins have a substantial number of these early developmental mutations specific to one of them. Using the parents and offspring of twins, we identified pre-twinning mutations. We observed instances where a twin was formed from a single cell lineage in the pre-twinning cell mass and instances where a twin was formed from several cell lineages. CpG>TpG mutations increased in frequency with embryonic development, coinciding with an increase in DNA methylation. Our results indicate that allocations of cells during development shapes genomic differences between monozygotic twins.


Asunto(s)
Genoma Humano , Células Germinativas/metabolismo , Gemelos Monocigóticos/genética , Desarrollo Embrionario/genética , Femenino , Frecuencia de los Genes/genética , Humanos , Masculino , Mosaicismo , Mutación/genética , Cigoto/metabolismo
9.
BMJ Open ; 11(12): e049709, 2021 12 30.
Artículo en Inglés | MEDLINE | ID: mdl-36070241

RESUMEN

PURPOSE: The aim of Copenhagen Hospital Biobank-Cardiovascular Disease Cohort (CHB-CVDC) is to establish a cohort that can accelerate our understanding of CVD initiation and progression by jointly studying genetics, diagnoses, treatments and risk factors. PARTICIPANTS: The CHB-CVDC is a large genomic cohort of patients with CVD. CHB-CVDC currently includes 96 308 patients. The cohort is part of CHB initiated in 2009 in the Capital Region of Denmark. CHB is continuously growing with ~40 000 samples/year. Patients in CHB were included in CHB-CVDC if they were above 18 years of age and assigned at least one cardiovascular diagnosis. Additionally, up-to 110 000 blood donors can be analysed jointly with CHB-CVDC. Linkage with the Danish National Health Registries, Electronic Patient Records, and Clinical Quality Databases allow up-to 41 years of medical history. All individuals are genotyped using the Infinium Global Screening Array from Illumina and imputed using a reference panel consisting of whole-genome sequence data from 8429 Danes along with 7146 samples from North-Western Europe. Currently, 39 539 of the patients are deceased. FINDINGS TO DATE: Here, we demonstrate the utility of the cohort by showing concordant effects between known variants and selected CVDs, that is, >93% concordance for coronary artery disease, atrial fibrillation, heart failure and cholesterol measurements and 85% concordance for hypertension. Furthermore, we evaluated multiple study designs and the validity of using Danish blood donors as part of CHB-CVDC. Lastly, CHB-CVDC has already made major contributions to studies of sick sinus syndrome and the role of phytosterols in development of atherosclerosis. FUTURE PLANS: In addition to genetics, electronic patient records, national socioeconomic and health registries extensively characterise each patient in CHB-CVDC and provides a promising framework for improved understanding of risk and protective variants. We aim to include other measurable biomarkers for example, proteins in CHB-CVDC making it a platform for multiomics cardiovascular studies.


Asunto(s)
Enfermedades Cardiovasculares , Cardiopatías , Bancos de Muestras Biológicas , Enfermedades Cardiovasculares/epidemiología , Enfermedades Cardiovasculares/genética , Estudios de Cohortes , Hospitales , Humanos
10.
N Engl J Med ; 383(18): 1724-1734, 2020 10 29.
Artículo en Inglés | MEDLINE | ID: mdl-32871063

RESUMEN

BACKGROUND: Little is known about the nature and durability of the humoral immune response to infection with severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2). METHODS: We measured antibodies in serum samples from 30,576 persons in Iceland, using six assays (including two pan-immunoglobulin [pan-Ig] assays), and we determined that the appropriate measure of seropositivity was a positive result with both pan-Ig assays. We tested 2102 samples collected from 1237 persons up to 4 months after diagnosis by a quantitative polymerase-chain-reaction (qPCR) assay. We measured antibodies in 4222 quarantined persons who had been exposed to SARS-CoV-2 and in 23,452 persons not known to have been exposed. RESULTS: Of the 1797 persons who had recovered from SARS-CoV-2 infection, 1107 of the 1215 who were tested (91.1%) were seropositive; antiviral antibody titers assayed by two pan-Ig assays increased during 2 months after diagnosis by qPCR and remained on a plateau for the remainder of the study. Of quarantined persons, 2.3% were seropositive; of those with unknown exposure, 0.3% were positive. We estimate that 0.9% of Icelanders were infected with SARS-CoV-2 and that the infection was fatal in 0.3%. We also estimate that 56% of all SARS-CoV-2 infections in Iceland had been diagnosed with qPCR, 14% had occurred in quarantined persons who had not been tested with qPCR (or who had not received a positive result, if tested), and 30% had occurred in persons outside quarantine and not tested with qPCR. CONCLUSIONS: Our results indicate that antiviral antibodies against SARS-CoV-2 did not decline within 4 months after diagnosis. We estimate that the risk of death from infection was 0.3% and that 44% of persons infected with SARS-CoV-2 in Iceland were not diagnosed by qPCR.


Asunto(s)
Infecciones por Coronavirus/inmunología , Inmunidad Humoral , Neumonía Viral/inmunología , Estudios Seroepidemiológicos , Adulto , Anciano , Anticuerpos Antivirales/sangre , Betacoronavirus , COVID-19 , Infecciones por Coronavirus/mortalidad , Femenino , Humanos , Islandia/epidemiología , Masculino , Persona de Mediana Edad , Pandemias , Neumonía Viral/mortalidad , Reacción en Cadena de la Polimerasa , Cuarentena , SARS-CoV-2
11.
N Engl J Med ; 382(24): 2302-2315, 2020 06 11.
Artículo en Inglés | MEDLINE | ID: mdl-32289214

RESUMEN

BACKGROUND: During the current worldwide pandemic, coronavirus disease 2019 (Covid-19) was first diagnosed in Iceland at the end of February. However, data are limited on how SARS-CoV-2, the virus that causes Covid-19, enters and spreads in a population. METHODS: We targeted testing to persons living in Iceland who were at high risk for infection (mainly those who were symptomatic, had recently traveled to high-risk countries, or had contact with infected persons). We also carried out population screening using two strategies: issuing an open invitation to 10,797 persons and sending random invitations to 2283 persons. We sequenced SARS-CoV-2 from 643 samples. RESULTS: As of April 4, a total of 1221 of 9199 persons (13.3%) who were recruited for targeted testing had positive results for infection with SARS-CoV-2. Of those tested in the general population, 87 (0.8%) in the open-invitation screening and 13 (0.6%) in the random-population screening tested positive for the virus. In total, 6% of the population was screened. Most persons in the targeted-testing group who received positive tests early in the study had recently traveled internationally, in contrast to those who tested positive later in the study. Children under 10 years of age were less likely to receive a positive result than were persons 10 years of age or older, with percentages of 6.7% and 13.7%, respectively, for targeted testing; in the population screening, no child under 10 years of age had a positive result, as compared with 0.8% of those 10 years of age or older. Fewer females than males received positive results both in targeted testing (11.0% vs. 16.7%) and in population screening (0.6% vs. 0.9%). The haplotypes of the sequenced SARS-CoV-2 viruses were diverse and changed over time. The percentage of infected participants that was determined through population screening remained stable for the 20-day duration of screening. CONCLUSIONS: In a population-based study in Iceland, children under 10 years of age and females had a lower incidence of SARS-CoV-2 infection than adolescents or adults and males. The proportion of infected persons identified through population screening did not change substantially during the screening period, which was consistent with a beneficial effect of containment efforts. (Funded by deCODE Genetics-Amgen.).


Asunto(s)
Infecciones por Coronavirus/epidemiología , Monitoreo Epidemiológico , Neumonía Viral/epidemiología , Adolescente , Adulto , Anciano , Anciano de 80 o más Años , Betacoronavirus/genética , COVID-19 , Niño , Preescolar , Trazado de Contacto , Femenino , Haplotipos , Humanos , Islandia/epidemiología , Lactante , Masculino , Tamizaje Masivo , Persona de Mediana Edad , Pandemias , SARS-CoV-2 , Viaje , Adulto Joven
12.
Nat Commun ; 11(1): 393, 2020 01 20.
Artículo en Inglés | MEDLINE | ID: mdl-31959851

RESUMEN

Asthma is one of the most common chronic diseases affecting both children and adults. We report a genome-wide association meta-analysis of 69,189 cases and 702,199 controls from Iceland and UK biobank. We find 88 asthma risk variants at 56 loci, 19 previously unreported, and evaluate their effect on other asthma and allergic phenotypes. Of special interest are two low frequency variants associated with protection against asthma; a missense variant in TNFRSF8 and 3' UTR variant in TGFBR1. Functional studies show that the TNFRSF8 variant reduces TNFRSF8 expression both on cell surface and in soluble form, acting as loss of function. eQTL analysis suggests that the TGFBR1 variant acts through gain of function and together with an intronic variant in a downstream gene, SMAD3, points to defective TGFßR1 signaling as one of the biological perturbations increasing asthma risk. Our results increase the number of asthma variants and implicate genes with known role in T cell regulation, inflammation and airway remodeling in asthma pathogenesis.


Asunto(s)
Remodelación de las Vías Aéreas (Respiratorias)/genética , Asma/genética , Antígeno Ki-1/genética , Receptor Tipo I de Factor de Crecimiento Transformador beta/genética , Linfocitos T/inmunología , Regiones no Traducidas 3'/genética , Remodelación de las Vías Aéreas (Respiratorias)/inmunología , Asma/inmunología , Eosinófilos , Predisposición Genética a la Enfermedad , Estudio de Asociación del Genoma Completo , Humanos , Islandia , Antígeno Ki-1/inmunología , Antígeno Ki-1/metabolismo , Recuento de Leucocitos , MicroARNs/metabolismo , Polimorfismo de Nucleótido Simple/inmunología , Sitios de Carácter Cuantitativo/inmunología , Receptor Tipo I de Factor de Crecimiento Transformador beta/inmunología , Receptor Tipo I de Factor de Crecimiento Transformador beta/metabolismo , Reino Unido
13.
J Am Coll Cardiol ; 74(24): 2982-2994, 2019 12 17.
Artículo en Inglés | MEDLINE | ID: mdl-31865966

RESUMEN

BACKGROUND: Lipoprotein(a) [Lp(a)] is a causal risk factor for cardiovascular diseases that has no established therapy. The attribute of Lp(a) that affects cardiovascular risk is not established. Low levels of Lp(a) have been associated with type 2 diabetes (T2D). OBJECTIVES: This study investigated whether cardiovascular risk is conferred by Lp(a) molar concentration or apolipoprotein(a) [apo(a)] size, and whether the relationship between Lp(a) and T2D risk is causal. METHODS: This was a case-control study of 143,087 Icelanders with genetic information, including 17,715 with coronary artery disease (CAD) and 8,734 with T2D. This study used measured and genetically imputed Lp(a) molar concentration, kringle IV type 2 (KIV-2) repeats (which determine apo(a) size), and a splice variant in LPA associated with small apo(a) but low Lp(a) molar concentration to disentangle the relationship between Lp(a) and cardiovascular risk. Loss-of-function homozygotes and other subjects genetically predicted to have low Lp(a) levels were evaluated to assess the relationship between Lp(a) and T2D. RESULTS: Lp(a) molar concentration was associated dose-dependently with CAD risk, peripheral artery disease, aortic valve stenosis, heart failure, and lifespan. Lp(a) molar concentration fully explained the Lp(a) association with CAD, and there was no residual association with apo(a) size. Homozygous carriers of loss-of-function mutations had little or no Lp(a) and increased the risk of T2D. CONCLUSIONS: Molar concentration is the attribute of Lp(a) that affects risk of cardiovascular diseases. Low Lp(a) concentration (bottom 10%) increases T2D risk. Pharmacologic reduction of Lp(a) concentration in the 20% of individuals with the greatest concentration down to the population median is predicted to decrease CAD risk without increasing T2D risk.


Asunto(s)
Enfermedad de la Arteria Coronaria/sangre , Variaciones en el Número de Copia de ADN , Diabetes Mellitus Tipo 2/sangre , Lipoproteína(a)/sangre , Estudios de Casos y Controles , Enfermedad de la Arteria Coronaria/genética , Diabetes Mellitus Tipo 2/genética , Humanos , Islandia , Kringles , Lipoproteína(a)/genética , Análisis de la Aleatorización Mendeliana , Peso Molecular , Isoformas de Proteínas/sangre , Factores de Riesgo
14.
Nat Commun ; 10(1): 2358, 2019 May 24.
Artículo en Inglés | MEDLINE | ID: mdl-31127096

RESUMEN

The original HTML version of this Article was updated shortly after publication to add links to the Peer Review file.In addition, affiliations 16 and 17 incorrectly read 'School of Medicine Sydney, University of Notre Dame Australia, Sydney, WA, 6160, Australia' and 'St Vincent's Clinical School, University of New South Wales Medicine, University of New South Wales, Sydney, NSW, 2052, Australia.' This has now been corrected in both the PDF and HTML versions of the Article.

15.
Nat Commun ; 10(1): 2054, 2019 05 03.
Artículo en Inglés | MEDLINE | ID: mdl-31053729

RESUMEN

Bone area is one measure of bone size that is easily derived from dual-energy X-ray absorptiometry (DXA) scans. In a GWA study of DXA bone area of the hip and lumbar spine (N ≥ 28,954), we find thirteen independent association signals at twelve loci that replicate in samples of European and East Asian descent (N = 13,608 - 21,277). Eight DXA area loci associate with osteoarthritis, including rs143384 in GDF5 and a missense variant in COL11A1 (rs3753841). The strongest DXA area association is with rs11614913[T] in the microRNA MIR196A2 gene that associates with lumbar spine area (P = 2.3 × 10-42, ß = -0.090) and confers risk of hip fracture (P = 1.0 × 10-8, OR = 1.11). We demonstrate that the risk allele is less efficient in repressing miR-196a-5p target genes. We also show that the DXA area measure contributes to the risk of hip fracture independent of bone density.


Asunto(s)
Densidad Ósea/genética , Fracturas de Cadera/genética , MicroARNs/genética , Osteoartritis/genética , Absorciometría de Fotón , Adulto , Anciano , Anciano de 80 o más Años , Alelos , Estatura/genética , Huesos/diagnóstico por imagen , Huesos/fisiología , Estudios de Casos y Controles , Colágeno Tipo XI/genética , Femenino , Estudios de Seguimiento , Sitios Genéticos , Predisposición Genética a la Enfermedad , Estudio de Asociación del Genoma Completo , Factor 5 de Diferenciación de Crecimiento/genética , Fracturas de Cadera/epidemiología , Humanos , Masculino , Persona de Mediana Edad , Osteoartritis/epidemiología , Factores de Riesgo
16.
Nat Genet ; 50(12): 1674-1680, 2018 12.
Artículo en Inglés | MEDLINE | ID: mdl-30397338

RESUMEN

De novo mutations (DNMs) cause a large proportion of severe rare diseases of childhood. DNMs that occur early may result in mosaicism of both somatic and germ cells. Such early mutations can cause recurrence of disease. We scanned 1,007 sibling pairs from 251 families and identified 878 DNMs shared by siblings (ssDNMs) at 448 genomic sites. We estimated DNM recurrence probability based on parental mosaicism, sharing of DNMs among siblings, parent-of-origin, mutation type and genomic position. We detected 57.2% of ssDNMs in the parental blood. The recurrence probability of a DNM decreases by 2.27% per year for paternal DNMs and 1.78% per year for maternal DNMs. Maternal ssDNMs are more likely to be T>C mutations than paternal ssDNMs, and less likely to be C>T mutations. Depending on the properties of the DNM, the recurrence probability ranges from 0.011% to 28.5%. We have launched an online calculator to allow estimation of DNM recurrence probability for research purposes.


Asunto(s)
Familia , Patrón de Herencia , Mutación , Relaciones Padres-Hijo , Adulto , Niño , Células Germinales Embrionarias/metabolismo , Composición Familiar , Femenino , Mutación de Línea Germinal , Humanos , Patrón de Herencia/genética , Masculino , Mosaicismo , Linaje
17.
Nat Genet ; 50(11): 1616, 2018 11.
Artículo en Inglés | MEDLINE | ID: mdl-30237445

RESUMEN

In the version of this article published, statements about the impact of insertions and deletions on gene conversions were incorrect. We reported a bias toward deletions, whereas in fact the bias was toward insertions. We are deeply indebted to Laurent Duret and Brice Letcher for noticing this mistake in our manuscript. The following statements are incorrect in the published manuscript.

18.
Sci Data ; 4: 170115, 2017 09 21.
Artículo en Inglés | MEDLINE | ID: mdl-28933420

RESUMEN

Understanding of sequence diversity is the cornerstone of analysis of genetic disorders, population genetics, and evolutionary biology. Here, we present an update of our sequencing set to 15,220 Icelanders who we sequenced to an average genome-wide coverage of 34X. We identified 39,020,168 autosomal variants passing GATK filters: 31,079,378 SNPs and 7,940,790 indels. Calling de novo mutations (DNMs) is a formidable challenge given the high false positive rate in sequencing datasets relative to the mutation rate. Here we addressed this issue by using segregation of alleles in three-generation families. Using this transmission assay, we controlled the false positive rate and identified 108,778 high quality DNMs. Furthermore, we used our extended family structure and read pair tracing of DNMs to a panel of phased SNPs, to determine the parent of origin of 42,961 DNMs.


Asunto(s)
Genoma Humano , Humanos , Mutación INDEL , Islandia , Polimorfismo de Nucleótido Simple
19.
Nature ; 549(7673): 519-522, 2017 09 28.
Artículo en Inglés | MEDLINE | ID: mdl-28959963

RESUMEN

The characterization of mutational processes that generate sequence diversity in the human genome is of paramount importance both to medical genetics and to evolutionary studies. To understand how the age and sex of transmitting parents affect de novo mutations, here we sequence 1,548 Icelanders, their parents, and, for a subset of 225, at least one child, to 35× genome-wide coverage. We find 108,778 de novo mutations, both single nucleotide polymorphisms and indels, and determine the parent of origin of 42,961. The number of de novo mutations from mothers increases by 0.37 per year of age (95% CI 0.32-0.43), a quarter of the 1.51 per year from fathers (95% CI 1.45-1.57). The number of clustered mutations increases faster with the mother's age than with the father's, and the genomic span of maternal de novo mutation clusters is greater than that of paternal ones. The types of de novo mutation from mothers change substantially with age, with a 0.26% (95% CI 0.19-0.33%) decrease in cytosine-phosphate-guanine to thymine-phosphate-guanine (CpG>TpG) de novo mutations and a 0.33% (95% CI 0.28-0.38%) increase in C>G de novo mutations per year, respectively. Remarkably, these age-related changes are not distributed uniformly across the genome. A striking example is a 20 megabase region on chromosome 8p, with a maternal C>G mutation rate that is up to 50-fold greater than the rest of the genome. The age-related accumulation of maternal non-crossover gene conversions also mostly occurs within these regions. Increased sequence diversity and linkage disequilibrium of C>G variants within regions affected by excess maternal mutations indicate that the underlying mutational process has persisted in humans for thousands of years. Moreover, the regional excess of C>G variation in humans is largely shared by chimpanzees, less by gorillas, and is almost absent from orangutans. This demonstrates that sequence diversity in humans results from evolving interactions between age, sex, mutation type, and genomic location.


Asunto(s)
Envejecimiento/genética , Mutación de Línea Germinal/genética , Edad Materna , Mutagénesis , Padres , Edad Paterna , Adolescente , Adulto , Anciano , Animales , Niño , Cromosomas Humanos Par 8/genética , Evolución Molecular , Femenino , Secuencia Rica en GC , Genoma Humano/genética , Gorilla gorilla/genética , Humanos , Mutación INDEL , Islandia , Desequilibrio de Ligamiento/genética , Masculino , Persona de Mediana Edad , Tasa de Mutación , Pan troglodytes/genética , Polimorfismo de Nucleótido Simple , Pongo/genética , Adulto Joven
20.
Nat Genet ; 49(8): 1182-1191, 2017 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-28628107

RESUMEN

Immunoglobulins are the effector molecules of the adaptive humoral immune system. In a genome-wide association study of 19,219 individuals, we found 38 new variants and replicated 5 known variants associating with IgA, IgG or IgM levels or with composite immunoglobulin traits, accounted for by 32 loci. Variants at these loci also affect the risk of autoimmune diseases and blood malignancies and influence blood cell development. Notable associations include a rare variant at RUNX3 decreasing IgA levels by shifting isoform proportions (rs188468174[C>T]: P = 8.3 × 10-55, ß = -0.90 s.d.), a rare in-frame deletion in FCGR2B abolishing IgG binding to the encoded receptor (p.Asn106del: P = 4.2 × 10-8, ß = 1.03 s.d.), four IGH locus variants influencing class switching, and ten new associations with the HLA region. Our results provide new insight into the regulation of humoral immunity.


Asunto(s)
Variación Genética , Inmunoglobulinas/genética , Estudios de Cohortes , Femenino , Predisposición Genética a la Enfermedad , Estudio de Asociación del Genoma Completo , Hematopoyesis/genética , Humanos , Islandia , Inmunidad Humoral/genética , Cambio de Clase de Inmunoglobulina/genética , Isotipos de Inmunoglobulinas/genética , Masculino , Polimorfismo de Nucleótido Simple , Suecia
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...