Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 51
Filtrar
1.
Eur J Hum Genet ; 32(1): 44-51, 2024 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-37684520

RESUMEN

Marfan syndrome (MFS) is an autosomal dominant condition characterized by aortic aneurysm, skeletal abnormalities, and lens dislocation, and is caused by variants in the FBN1 gene. To explore causes of MFS and the prevalence of the disease in Iceland we collected information from all living individuals with a clinical diagnosis of MFS in Iceland (n = 32) and performed whole-genome sequencing of those who did not have a confirmed genetic diagnosis (27/32). Moreover, to assess a potential underdiagnosis of MFS in Iceland we attempted a genotype-based approach to identify individuals with MFS. We interrogated deCODE genetics' database of 35,712 whole-genome sequenced individuals to search for rare sequence variants in FBN1. Overall, we identified 15 pathogenic or likely pathogenic variants in FBN1 in 44 individuals, only 22 of whom were previously diagnosed with MFS. The most common of these variants, NM_000138.4:c.8038 C > T p.(Arg2680Cys), is present in a multi-generational pedigree, and was found to stem from a single forefather born around 1840. The p.(Arg2680Cys) variant associates with a form of MFS that seems to have an enrichment of abdominal aortic aneurysm, suggesting that this may be a particularly common feature of p.(Arg2680Cys)-associated MFS. Based on these combined genetic and clinical data, we show that MFS prevalence in Iceland could be as high as 1/6,600 in Iceland, compared to 1/10,000 based on clinical diagnosis alone, which indicates underdiagnosis of this actionable genetic disorder.


Asunto(s)
Síndrome de Marfan , Humanos , Síndrome de Marfan/diagnóstico , Síndrome de Marfan/epidemiología , Síndrome de Marfan/genética , Islandia/epidemiología , Fibrilina-1/genética , Genotipo , Linaje , Mutación , Adipoquinas/genética
2.
N Engl J Med ; 389(19): 1741-1752, 2023 Nov 09.
Artículo en Inglés | MEDLINE | ID: mdl-37937776

RESUMEN

BACKGROUND: In 2021, the American College of Medical Genetics and Genomics (ACMG) recommended reporting actionable genotypes in 73 genes associated with diseases for which preventive or therapeutic measures are available. Evaluations of the association of actionable genotypes in these genes with life span are currently lacking. METHODS: We assessed the prevalence of coding and splice variants in genes on the ACMG Secondary Findings, version 3.0 (ACMG SF v3.0), list in the genomes of 57,933 Icelanders. We assigned pathogenicity to all reviewed variants using reported evidence in the ClinVar database, the frequency of variants, and their associations with disease to create a manually curated set of actionable genotypes (variants). We assessed the relationship between these genotypes and life span and further examined the specific causes of death among carriers. RESULTS: Through manual curation of 4405 sequence variants in the ACMG SF v3.0 genes, we identified 235 actionable genotypes in 53 genes. Of the 57,933 participants, 2306 (4.0%) carried at least one actionable genotype. We found shorter median survival among persons carrying actionable genotypes than among noncarriers. Specifically, we found that carrying an actionable genotype in a cancer gene was associated with survival that was 3 years shorter than that among noncarriers, with causes of death among carriers attributed primarily to cancer-related conditions. Furthermore, we found evidence of association between carrying an actionable genotype in certain genes in the cardiovascular disease group and a reduced life span. CONCLUSIONS: On the basis of the ACMG SF v3.0 guidelines, we found that approximately 1 in 25 Icelanders carried an actionable genotype and that carrying such a genotype was associated with a reduced life span. (Funded by deCODE Genetics-Amgen.).


Asunto(s)
Enfermedad , Genómica , Longevidad , Humanos , Alelos , Pruebas Genéticas , Variación Genética , Genotipo , Islandia/epidemiología , Longevidad/genética , Enfermedad/genética , Enfermedades Cardiovasculares/genética , Neoplasias/genética
3.
Nat Genet ; 55(11): 1843-1853, 2023 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-37884687

RESUMEN

Migraine is a complex neurovascular disease with a range of severity and symptoms, yet mostly studied as one phenotype in genome-wide association studies (GWAS). Here we combine large GWAS datasets from six European populations to study the main migraine subtypes, migraine with aura (MA) and migraine without aura (MO). We identified four new MA-associated variants (in PRRT2, PALMD, ABO and LRRK2) and classified 13 MO-associated variants. Rare variants with large effects highlight three genes. A rare frameshift variant in brain-expressed PRRT2 confers large risk of MA and epilepsy, but not MO. A burden test of rare loss-of-function variants in SCN11A, encoding a neuron-expressed sodium channel with a key role in pain sensation, shows strong protection against migraine. Finally, a rare variant with cis-regulatory effects on KCNK5 confers large protection against migraine and brain aneurysms. Our findings offer new insights with therapeutic potential into the complex biology of migraine and its subtypes.


Asunto(s)
Epilepsia , Trastornos Migrañosos , Migraña con Aura , Humanos , Estudio de Asociación del Genoma Completo , Trastornos Migrañosos/genética , Migraña con Aura/genética , Fenotipo
4.
Commun Biol ; 5(1): 525, 2022 06 01.
Artículo en Inglés | MEDLINE | ID: mdl-35650273

RESUMEN

The characteristic lobulated nuclear morphology of granulocytes is partially determined by composition of nuclear envelope proteins. Abnormal nuclear morphology is primarily observed as an increased number of hypolobulated immature neutrophils, called band cells, during infection or in rare envelopathies like Pelger-Huët anomaly. To search for sequence variants affecting nuclear morphology of granulocytes, we performed a genome-wide association study using band neutrophil fraction from 88,101 Icelanders. We describe 13 sequence variants affecting band neutrophil fraction at nine loci. Five of the variants are at the Lamin B receptor (LBR) locus, encoding an inner nuclear membrane protein. Mutations in LBR are linked to Pelger-Huët anomaly. In addition, we identify cosegregation of a rare stop-gain sequence variant in LBR and Pelger Huët anomaly in an Icelandic eight generation pedigree, initially reported in 1963. Two of the other loci include genes which, like LBR, play a role in the nuclear membrane function and integrity. These GWAS results highlight the role proteins of the inner nuclear membrane have as important for neutrophil nuclear morphology.


Asunto(s)
Anomalía de Pelger-Huët , Estudio de Asociación del Genoma Completo , Granulocitos/metabolismo , Humanos , Islandia , Neutrófilos/metabolismo , Anomalía de Pelger-Huët/genética
5.
Nat Commun ; 13(1): 705, 2022 02 04.
Artículo en Inglés | MEDLINE | ID: mdl-35121750

RESUMEN

Predicting the pathogenicity of biallelic missense variants can be challenging. Here, we use a deficit of observed homozygous carriers of missense variants, versus an expected number in a set of 153,054 chip-genotyped Icelanders, to identify potentially pathogenic genotypes. We follow three missense variants with a complete deficit of homozygosity and find that their pathogenic effect in homozygous state ranges from severe childhood disease to early embryonic lethality. One of these variants is in CPSF3, a gene not previously linked to disease. From a set of clinically sequenced Icelanders, and by sequencing archival samples targeted through the Icelandic genealogy, we find four homozygous carriers. Additionally, we find two homozygous carriers of Mexican descent of another missense variant in CPSF3. All six homozygous carriers of missense variants in CPSF3 show severe intellectual disability, seizures, microcephaly, and abnormal muscle tone. Here, we show how the absence of certain homozygous genotypes from a large population set can elucidate causes of previously unexplained recessive diseases and early miscarriage.


Asunto(s)
Factor de Especificidad de Desdoblamiento y Poliadenilación/genética , Predisposición Genética a la Enfermedad/genética , Homocigoto , Discapacidad Intelectual/genética , Mutación Missense , Adolescente , Alelos , Niño , Preescolar , Femenino , Frecuencia de los Genes , Genética de Población/métodos , Genotipo , Humanos , Islandia , Lactante , Discapacidad Intelectual/patología , Masculino , Linaje , Fenotipo , Síndrome , Secuenciación Completa del Genoma/métodos
6.
Arterioscler Thromb Vasc Biol ; 41(10): 2616-2628, 2021 10.
Artículo en Inglés | MEDLINE | ID: mdl-34407635

RESUMEN

Objective: Familial hypercholesterolemia (FH) is traditionally defined as a monogenic disease characterized by severely elevated LDL-C (low-density lipoprotein cholesterol) levels. In practice, FH is commonly a clinical diagnosis without confirmation of a causative mutation. In this study, we sought to characterize and compare monogenic and clinically defined FH in a large sample of Icelanders. Approach and Results: We whole-genome sequenced 49 962 Icelanders and imputed the identified variants into an overall sample of 166 281 chip-genotyped Icelanders. We identified 20 FH mutations in LDLR, APOB, and PCSK9 with combined prevalence of 1 in 836. Monogenic FH was associated with severely elevated LDL-C levels and increased risk of premature coronary disease, aortic valve stenosis, and high burden of coronary atherosclerosis. We used a modified version of the Dutch Lipid Clinic Network criteria to screen for the clinical FH phenotype among living adult participants (N=79 058). Clinical FH was found in 2.2% of participants, of whom only 5.2% had monogenic FH. Mutation-negative clinical FH has a strong polygenic basis. Both individuals with monogenic FH and individuals with mutation-negative clinical FH were markedly undertreated with cholesterol-lowering medications and only a minority attained an LDL-C target of <2.6 mmol/L (<100 mg/dL; 11.0% and 24.9%, respectively) or <1.8 mmol/L (<70 mg/dL; 0.0% and 5.2%, respectively), as recommended for primary prevention by European Society of Cardiology/European Atherosclerosis Society cholesterol guidelines. Conclusions: Clinically defined FH is a relatively common phenotype that is explained by monogenic FH in only a minority of cases. Both monogenic and clinical FH confer high cardiovascular risk but are markedly undertreated.


Asunto(s)
Apolipoproteína B-100/genética , Enfermedades Cardiovasculares/genética , Hiperlipoproteinemia Tipo II/genética , Lípidos/sangre , Mutación , Proproteína Convertasa 9/genética , Receptores de LDL/genética , Adulto , Anciano , Anciano de 80 o más Años , Biomarcadores/sangre , Enfermedades Cardiovasculares/diagnóstico , Enfermedades Cardiovasculares/etnología , Enfermedades Cardiovasculares/terapia , Femenino , Estudios de Asociación Genética , Predisposición Genética a la Enfermedad , Humanos , Inhibidores de Hidroximetilglutaril-CoA Reductasas/uso terapéutico , Hiperlipoproteinemia Tipo II/diagnóstico , Hiperlipoproteinemia Tipo II/tratamiento farmacológico , Hiperlipoproteinemia Tipo II/etnología , Islandia/epidemiología , Masculino , Persona de Mediana Edad , Fenotipo , Prevalencia , Pronóstico , Medición de Riesgo , Factores de Riesgo , Adulto Joven
7.
Eur J Hum Genet ; 29(12): 1819-1824, 2021 12.
Artículo en Inglés | MEDLINE | ID: mdl-34462577

RESUMEN

Malignant hyperthermia (MH) susceptibility is a rare life-threatening disorder that occurs upon exposure to a triggering agent. MH is commonly due to protein-altering variants in RYR1 and CACNA1S. The American College of Medical Genetics and Genomics recommends that when pathogenic and likely pathogenic variants in RYR1 and CACNA1S are incidentally found, they should be reported to the carriers. The detection of actionable variants allows the avoidance of exposure to triggering agents during anesthesia. First, we report a 10-year-old Icelandic proband with a suspected MH event, harboring a heterozygous missense variant NM_000540.2:c.6710G>A r.(6710g>a) p.(Cys2237Tyr) in the RYR1 gene that is likely pathogenic. The variant is private to four individuals within a three-generation family and absent from 62,240 whole-genome sequenced (WGS) Icelanders. Haplotype sharing and WGS revealed that the variant occurred as a somatic mosaicism also present in germline of the proband's paternal grandmother. Second, using a set of 62,240 Icelanders with WGS, we assessed the carrier frequency of actionable pathogenic and likely pathogenic variants in RYR1 and CACNA1S. We observed 13 actionable variants in RYR1, based on ClinVar classifications, carried by 43 Icelanders, and no actionable variant in CACNA1S. One in 1450 Icelanders carries an actionable variant for MH. Extensive sequencing allows for better classification and precise dating of variants, and WGS of a large fraction of the population has led to incidental findings of actionable MH genotypes.


Asunto(s)
Frecuencia de los Genes , Hipertermia Maligna/genética , Mutación Missense , Población/genética , Adulto , Canales de Calcio Tipo L/genética , Niño , Femenino , Haplotipos , Heterocigoto , Humanos , Islandia , Masculino , Hipertermia Maligna/patología , Linaje , Canal Liberador de Calcio Receptor de Rianodina/genética
8.
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
9.
Nat Genet ; 53(6): 779-786, 2021 06.
Artículo en Inglés | MEDLINE | ID: mdl-33972781

RESUMEN

Long-read sequencing (LRS) promises to improve the characterization of structural variants (SVs). We generated LRS data from 3,622 Icelanders and identified a median of 22,636 SVs per individual (a median of 13,353 insertions and 9,474 deletions). We discovered a set of 133,886 reliably genotyped SV alleles and imputed them into 166,281 individuals to explore their effects on diseases and other traits. We discovered an association of a rare deletion in PCSK9 with lower low-density lipoprotein (LDL) cholesterol levels, compared to the population average. We also discovered an association of a multiallelic SV in ACAN with height; we found 11 alleles that differed in the number of a 57-bp-motif repeat and observed a linear relationship between the number of repeats carried and height. These results show that SVs can be accurately characterized at the population scale using LRS data in a genome-wide non-targeted approach and demonstrate how SVs impact phenotypes.


Asunto(s)
Enfermedad/genética , Variación Estructural del Genoma , Secuenciación de Nucleótidos de Alto Rendimiento , Carácter Cuantitativo Heredable , Alelos , LDL-Colesterol/metabolismo , Cromosomas Humanos/genética , Femenino , Frecuencia de los Genes/genética , Humanos , Islandia , Modelos Lineales , Masculino , Proproteína Convertasa 9/genética , Recombinación Genética/genética , Eliminación de Secuencia/genética
10.
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
11.
Commun Biol ; 3(1): 189, 2020 04 23.
Artículo en Inglés | MEDLINE | ID: mdl-32327693

RESUMEN

Hemoglobin is the essential oxygen-carrying molecule in humans and is regulated by cellular iron and oxygen sensing mechanisms. To search for novel variants associated with hemoglobin concentration, we performed genome-wide association studies of hemoglobin concentration using a combined set of 684,122 individuals from Iceland and the UK. Notably, we found seven novel variants, six rare coding and one common, at the ACO1 locus associating with either decreased or increased hemoglobin concentration. Of these variants, the missense Cys506Ser and the stop-gained Lys334Ter mutations are specific to eight and ten generation pedigrees, respectively, and have the two largest effects in the study (EffectCys506Ser = -1.61 SD, CI95 = [-1.98, -1.35]; EffectLys334Ter = 0.63 SD, CI95 = [0.36, 0.91]). We also find Cys506Ser to associate with increased risk of persistent anemia (OR = 17.1, P = 2 × 10-14). The strong bidirectional effects seen in this study implicate ACO1, a known iron sensing molecule, as a major homeostatic regulator of hemoglobin concentration.


Asunto(s)
Eritropoyesis/genética , Mutación con Ganancia de Función , Hemoglobinas/metabolismo , Proteína 1 Reguladora de Hierro/genética , Mutación con Pérdida de Función , Biomarcadores/sangre , Bases de Datos Genéticas , Estudio de Asociación del Genoma Completo , Humanos , Islandia , Proteína 1 Reguladora de Hierro/metabolismo , Reino Unido
12.
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
13.
Nat Commun ; 10(1): 1777, 2019 04 16.
Artículo en Inglés | MEDLINE | ID: mdl-30992453

RESUMEN

Nerve conduction (NC) studies generate measures of peripheral nerve function that can reveal underlying pathology due to axonal loss, demyelination or both. We perform a genome-wide association study of sural NC amplitude and velocity in 7045 Icelanders and find a low-frequency splice-donor variant in PRPH (c.996+1G>A; MAF = 1.32%) associating with decreased NC amplitude but not velocity. PRPH encodes peripherin, an intermediate filament (IF) protein involved in cytoskeletal development and maintenance of neurons. Through RNA and protein studies, we show that the variant leads to loss-of-function (LoF), as when over-expressed in a cell line devoid of other IFs, it does not allow formation of the normal filamentous structure of peripherin, yielding instead punctate protein inclusions. Recall of carriers for neurological assessment confirms that from an early age, homozygotes have significantly lower sural NC amplitude than non-carriers and are at risk of a mild, early-onset, sensory-negative, axonal polyneuropathy.


Asunto(s)
Conducción Nerviosa/genética , Periferinas/genética , Polineuropatías/genética , Sitios de Empalme de ARN/genética , Nervio Sural/fisiopatología , Adulto , Edad de Inicio , Anciano , Axones/patología , Estudios de Casos y Controles , Línea Celular , Femenino , Estudios de Seguimiento , Estudio de Asociación del Genoma Completo , Homocigoto , Humanos , Islandia/epidemiología , Mutación con Pérdida de Función , Masculino , Persona de Mediana Edad , Polineuropatías/epidemiología , Polineuropatías/fisiopatología , Prevalencia , Empalme del ARN/fisiología
14.
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
15.
Nat Commun ; 9(1): 4447, 2018 10 25.
Artículo en Inglés | MEDLINE | ID: mdl-30361506

RESUMEN

Mutations in genes encoding subunits of the phagocyte NADPH oxidase complex are recognized to cause chronic granulomatous disease (CGD), a severe primary immunodeficiency. Here we describe how deficiency of CYBC1, a previously uncharacterized protein in humans (C17orf62), leads to reduced expression of NADPH oxidase's main subunit (gp91phox) and results in CGD. Analyzing two brothers diagnosed with CGD we identify a homozygous loss-of-function mutation, p.Tyr2Ter, in CYBC1. Imputation of p.Tyr2Ter into 155K chip-genotyped Icelanders reveals six additional homozygotes, all with signs of CGD, manifesting as colitis, rare infections, or a severely impaired PMA-induced neutrophil oxidative burst. Homozygosity for p.Tyr2Ter consequently associates with inflammatory bowel disease (IBD) in Iceland (P = 8.3 × 10-8; OR = 67.6), as well as reduced height (P = 3.3 × 10-4; -8.5 cm). Overall, we find that CYBC1 deficiency results in CGD characterized by colitis and a distinct profile of infections indicative of macrophage dysfunction.


Asunto(s)
Enfermedad Granulomatosa Crónica/genética , Mutación con Pérdida de Función/genética , Niño , Colitis/genética , Colitis/patología , Citocromos b/metabolismo , Femenino , Homocigoto , Humanos , Masculino , Linaje , Estallido Respiratorio
16.
Circ Genom Precis Med ; 11(8): e002151, 2018 08.
Artículo en Inglés | MEDLINE | ID: mdl-30354339

RESUMEN

BACKGROUND: Dilated cardiomyopathy (DCM) is an important cause of heart failure. Variants in >50 genes have been reported to cause DCM, but causative variants have been found in less than half of familial cases. Variants causing DCM in Iceland have not been reported before. METHODS: We performed a genome-wide association study on DCM based on whole genome sequencing. We tested the association of 32.5 million sequence variants in 424 cases and 337 689 population controls in Iceland. RESULTS: We identified 2 DCM variants in established cardiomyopathy genes, a missense variant p.Phe145Leu in NKX2-5 carried by 1 in 7100 Icelanders ( P=7.0×10-12) and a frameshift variant p.Phe1626Serfs*40 in FLNC carried by 1 in 3600 Icelanders ( P=2.1×10-10). Both variants associate with heart failure and sudden cardiac death. Additionally, p.Phe145Leu in NKX2-5 associates with high degree atrioventricular block and atrial septal defect ( P<1.4×10-4). The penetrance of serious heart disease among carriers of the NKX2-5 variant is high and higher than that of the FLNC variant. CONCLUSIONS: Two rare variants in NKX2-5 and FLNC, carried by 1 in 2400 Icelanders, cause familial DCM in Iceland. These genes have recently been associated with DCM. Given the serious consequences of these variants, we suggest screening for them in individuals with DCM and their family members, with subsequent monitoring of carriers, offering early intervention.


Asunto(s)
Cardiomiopatía Dilatada/complicaciones , Cardiomiopatía Dilatada/genética , Muerte Súbita Cardíaca/etiología , Filaminas/genética , Proteína Homeótica Nkx-2.5/genética , Adulto , Anciano , Anciano de 80 o más Años , Cardiomiopatía Dilatada/epidemiología , Estudios de Casos y Controles , Muerte Súbita Cardíaca/epidemiología , Femenino , Mutación del Sistema de Lectura , Estudio de Asociación del Genoma Completo , Humanos , Islandia/epidemiología , Masculino , Persona de Mediana Edad , Mutación Missense , Penetrancia , Adulto Joven
17.
Commun Biol ; 1: 14, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-30271901

RESUMEN

Searching for novel sequence variants associated with cholesterol levels is of particular interest due to the causative role of non-HDL cholesterol levels in cardiovascular disease. Through whole-genome sequencing of 15,220 Icelanders and imputation of the variants identified, we discovered a rare missense variant in NR1H4 (R436H) associating with lower levels of total cholesterol (effect = -0.47 standard deviations or -0.55 mmol L-1, p = 4.21 × 10-10, N = 150,211). Importantly, NR1H4 R436H also associates with lower levels of non-HDL cholesterol and, consistent with this, protects against coronary artery disease. NR1H4 encodes FXR that regulates bile acid homeostasis, however, we do not detect a significant association between R436H and biological markers of liver function. Transcriptional profiling of hepatocytes carrying R436H shows that it is not a loss-of-function variant. Rather, we observe changes in gene expression compatible with effects on lipids. These findings highlight the role of FXR in regulation of cholesterol levels in humans.

18.
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.

19.
Nat Commun ; 9(1): 3456, 2018 08 27.
Artículo en Inglés | MEDLINE | ID: mdl-30150678

RESUMEN

Discovery of coding variants in genes that confer risk of neurodevelopmental disorders is an important step towards understanding the pathophysiology of these disorders. Whole-genome sequencing of 31,463 Icelanders uncovers a frameshift variant (E712KfsTer10) in microtubule-associated protein 1B (MAP1B) that associates with ID/low IQ in a large pedigree (genome-wide corrected P = 0.022). Additional stop-gain variants in MAP1B (E1032Ter and R1664Ter) validate the association with ID and IQ. Carriers have 24% less white matter (WM) volume (ß = -2.1SD, P = 5.1 × 10-8), 47% less corpus callosum (CC) volume (ß = -2.4SD, P = 5.5 × 10-10) and lower brain-wide fractional anisotropy (P = 6.7 × 10-4). In summary, we show that loss of MAP1B function affects general cognitive ability through a profound, brain-wide WM deficit with likely disordered or compromised axons.


Asunto(s)
Proteína de la Discapacidad Intelectual del Síndrome del Cromosoma X Frágil/metabolismo , Proteínas Asociadas a Microtúbulos/metabolismo , Sustancia Blanca/metabolismo , Sustancia Blanca/patología , Animales , Encéfalo/metabolismo , Encéfalo/patología , Cognición/fisiología , Cuerpo Calloso/metabolismo , Cuerpo Calloso/patología , Femenino , Proteína de la Discapacidad Intelectual del Síndrome del Cromosoma X Frágil/genética , Discapacidad Intelectual/genética , Discapacidad Intelectual/fisiopatología , Masculino , Ratones , Ratones Noqueados , Proteínas Asociadas a Microtúbulos/genética , Mutación/genética , Linaje
20.
BMC Med Genet ; 18(1): 129, 2017 11 14.
Artículo en Inglés | MEDLINE | ID: mdl-29137621

RESUMEN

BACKGROUND: Rare missense mutations in the gene encoding coatomer subunit alpha (COPA) have recently been shown to cause autoimmune interstitial lung, joint and kidney disease, also known as COPA syndrome, under a dominant mode of inheritance. CASE PRESENTATION: Here we describe an Icelandic family with three affected individuals over two generations with a rare clinical presentation of lung and joint disease and a histological diagnosis of follicular bronchiolitis. We performed whole-genome sequencing (WGS) of the three affected as well as three unaffected members of the family, and searched for rare genotypes associated with disease using 30,067 sequenced Icelanders as a reference population. We assessed all coding and splicing variants, prioritizing variants in genes known to cause interstitial lung disease. We detected a heterozygous missense mutation, p.Glu241Lys, in the COPA gene, private to the affected family members. The mutation occurred de novo in the paternal germline of the index case and was absent from 30,067 Icelandic genomes and 141,353 individuals from the genome Aggregation Database (gnomAD). The mutation occurs within the conserved and functionally important WD40 domain of the COPA protein. CONCLUSIONS: This is the second report of the p.Glu241Lys mutation in COPA, indicating the recurrent nature of the mutation. The mutation was reported to co-segregate with COPA syndrome in a large family from the USA with five affected members, and classified as pathogenic. The two separate occurrences of the p.Glu241Lys mutation in cases and its absence from a large number of sequenced genomes confirms its role in the pathogenesis of the COPA syndrome.


Asunto(s)
Proteína Coatómero/genética , Síndromes de Inmunodeficiencia/diagnóstico , Síndromes de Inmunodeficiencia/genética , Mutación Missense , Artritis/diagnóstico , Artritis/genética , Niño , Preescolar , Femenino , Estudio de Asociación del Genoma Completo , Humanos , Islandia , Lactante , Enfermedades Pulmonares/diagnóstico , Enfermedades Pulmonares/genética , Masculino , Linaje
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...