Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 75
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
PLoS Genet ; 16(6): e1008725, 2020 06.
Artículo en Inglés | MEDLINE | ID: mdl-32603359

RESUMEN

Risk factors that contribute to inter-individual differences in the age-of-onset of allergic diseases are poorly understood. The aim of this study was to identify genetic risk variants associated with the age at which symptoms of allergic disease first develop, considering information from asthma, hay fever and eczema. Self-reported age-of-onset information was available for 117,130 genotyped individuals of European ancestry from the UK Biobank study. For each individual, we identified the earliest age at which asthma, hay fever and/or eczema was first diagnosed and performed a genome-wide association study (GWAS) of this combined age-of-onset phenotype. We identified 50 variants with a significant independent association (P<3x10-8) with age-of-onset. Forty-five variants had comparable effects on the onset of the three individual diseases and 38 were also associated with allergic disease case-control status in an independent study (n = 222,484). We observed a strong negative genetic correlation between age-of-onset and case-control status of allergic disease (rg = -0.63, P = 4.5x10-61), indicating that cases with early disease onset have a greater burden of allergy risk alleles than those with late disease onset. Subsequently, a multivariate GWAS of age-of-onset and case-control status identified a further 26 associations that were missed by the univariate analyses of age-of-onset or case-control status only. Collectively, of the 76 variants identified, 18 represent novel associations for allergic disease. We identified 81 likely target genes of the 76 associated variants based on information from expression quantitative trait loci (eQTL) and non-synonymous variants, of which we highlight ADAM15, FOSL2, TRIM8, BMPR2, CD200R1, PRKCQ, NOD2, SMAD4, ABCA7 and UBE2L3. Our results support the notion that early and late onset allergic disease have partly distinct genetic architectures, potentially explaining known differences in pathophysiology between individuals.


Asunto(s)
Asma/genética , Eccema/genética , Polimorfismo de Nucleótido Simple , Rinitis Alérgica Estacional/genética , Adolescente , Adulto , Edad de Inicio , Anciano , Asma/patología , Niño , Eccema/patología , Femenino , Sitios Genéticos , Estudio de Asociación del Genoma Completo/métodos , Humanos , Masculino , Persona de Mediana Edad , Rinitis Alérgica Estacional/patología
2.
Hum Mutat ; 43(10): 1472-1489, 2022 10.
Artículo en Inglés | MEDLINE | ID: mdl-35815345

RESUMEN

Aminoacyl-tRNA synthetases (ARSs) are essential enzymes for faithful assignment of amino acids to their cognate tRNA. Variants in ARS genes are frequently associated with clinically heterogeneous phenotypes in humans and follow both autosomal dominant or recessive inheritance patterns in many instances. Variants in tryptophanyl-tRNA synthetase 1 (WARS1) cause autosomal dominantly inherited distal hereditary motor neuropathy and Charcot-Marie-Tooth disease. Presently, only one family with biallelic WARS1 variants has been described. We present three affected individuals from two families with biallelic variants (p.Met1? and p.(Asp419Asn)) in WARS1, showing varying severities of developmental delay and intellectual disability. Hearing impairment and microcephaly, as well as abnormalities of the brain, skeletal system, movement/gait, and behavior were variable features. Phenotyping of knocked down wars-1 in a Caenorhabditis elegans model showed depletion is associated with defects in germ cell development. A wars1 knockout vertebrate model recapitulates the human clinical phenotypes, confirms variant pathogenicity, and uncovers evidence implicating the p.Met1? variant as potentially impacting an exon critical for normal hearing. Together, our findings provide consolidating evidence for biallelic disruption of WARS1 as causal for an autosomal recessive neurodevelopmental syndrome and present a vertebrate model that recapitulates key phenotypes observed in patients.


Asunto(s)
Aminoacil-ARNt Sintetasas , Enfermedad de Charcot-Marie-Tooth , Triptófano-ARNt Ligasa , Aminoacil-ARNt Sintetasas/genética , Enfermedad de Charcot-Marie-Tooth/genética , Exones , Humanos , Mutación , Linaje , ARN de Transferencia/genética , Síndrome , Triptófano-ARNt Ligasa/genética
3.
Hum Genet ; 140(6): 915-931, 2021 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-33496845

RESUMEN

Deafness, the most frequent sensory deficit in humans, is extremely heterogeneous with hundreds of genes involved. Clinical and genetic analyses of an extended consanguineous family with pre-lingual, moderate-to-profound autosomal recessive sensorineural hearing loss, allowed us to identify CLRN2, encoding a tetraspan protein, as a new deafness gene. Homozygosity mapping followed by exome sequencing identified a 14.96 Mb locus on chromosome 4p15.32p15.1 containing a likely pathogenic missense variant in CLRN2 (c.494C > A, NM_001079827.2) segregating with the disease. Using in vitro RNA splicing analysis, we show that the CLRN2 c.494C > A variant leads to two events: (1) the substitution of a highly conserved threonine (uncharged amino acid) to lysine (charged amino acid) at position 165, p.(Thr165Lys), and (2) aberrant splicing, with the retention of intron 2 resulting in a stop codon after 26 additional amino acids, p.(Gly146Lysfs*26). Expression studies and phenotyping of newly produced zebrafish and mouse models deficient for clarin 2 further confirm that clarin 2, expressed in the inner ear hair cells, is essential for normal organization and maintenance of the auditory hair bundles, and for hearing function. Together, our findings identify CLRN2 as a new deafness gene, which will impact future diagnosis and treatment for deaf patients.


Asunto(s)
Sustitución de Aminoácidos , Cromosomas Humanos Par 4/química , Células Ciliadas Auditivas Internas/metabolismo , Pérdida Auditiva Sensorineural/genética , Proteínas de la Membrana/genética , Mutación Puntual , Tetraspaninas/genética , Adulto , Alelos , Animales , Secuencia de Bases , Mapeo Cromosómico , Consanguinidad , Femenino , Expresión Génica , Genes Recesivos , Células Ciliadas Auditivas Internas/patología , Pérdida Auditiva Sensorineural/metabolismo , Pérdida Auditiva Sensorineural/patología , Humanos , Masculino , Proteínas de la Membrana/deficiencia , Ratones , Linaje , Tetraspaninas/deficiencia , Secuenciación del Exoma , Pez Cebra
4.
J Allergy Clin Immunol ; 143(2): 691-699, 2019 02.
Artículo en Inglés | MEDLINE | ID: mdl-29679657

RESUMEN

BACKGROUND: A recent genome-wide association study (GWAS) identified 99 loci that contain genetic risk variants shared between asthma, hay fever, and eczema. Many more risk loci shared between these common allergic diseases remain to be discovered, which could point to new therapeutic opportunities. OBJECTIVE: We sought to identify novel risk loci shared between asthma, hay fever, and eczema by applying a gene-based test of association to results from a published GWAS that included data from 360,838 subjects. METHODS: We used approximate conditional analysis to adjust the results from the published GWAS for the effects of the top risk variants identified in that study. We then analyzed the adjusted GWAS results with the EUGENE gene-based approach, which combines evidence for association with disease risk across regulatory variants identified in different tissues. Novel gene-based associations were followed up in an independent sample of 233,898 subjects from the UK Biobank study. RESULTS: Of the 19,432 genes tested, 30 had a significant gene-based association at a Bonferroni-corrected P value of 2.5 × 10-6. Of these, 20 were also significantly associated (P < .05/30 = .0016) with disease risk in the replication sample, including 19 that were located in 11 loci not reported to contain allergy risk variants in previous GWASs. Among these were 9 genes with a known function that is directly relevant to allergic disease: FOSL2, VPRBP, IPCEF1, PRR5L, NCF4, APOBR, IL27, ATXN2L, and LAT. For 4 genes (eg, ATXN2L), a genetically determined decrease in gene expression was associated with decreased allergy risk, and therefore drugs that inhibit gene expression or function are predicted to ameliorate disease symptoms. The opposite directional effect was observed for 14 genes, including IL27, a cytokine known to suppress TH2 responses. CONCLUSION: Using a gene-based approach, we identified 11 risk loci for allergic disease that were not reported in previous GWASs. Functional studies that investigate the contribution of the 19 associated genes to the pathophysiology of allergic disease and assess their therapeutic potential are warranted.


Asunto(s)
Asma/genética , Eccema/genética , Genotipo , Hipersensibilidad/genética , Rinitis Alérgica Estacional/genética , Antígeno 2 Relacionado con Fos/genética , Frecuencia de los Genes , Estudios de Asociación Genética , Sitios Genéticos/genética , Predisposición Genética a la Enfermedad , Estudio de Asociación del Genoma Completo , Humanos , Interleucina-27/genética , Polimorfismo de Nucleótido Simple , Riesgo , Balance Th1 - Th2/genética
5.
Int J Mol Sci ; 21(1)2020 Jan 02.
Artículo en Inglés | MEDLINE | ID: mdl-31906439

RESUMEN

CDC14A encodes the Cell Division Cycle 14A protein and has been associated with autosomal recessive non-syndromic hearing loss (DFNB32), as well as hearing impairment and infertile male syndrome (HIIMS) since 2016. To date, only nine variants have been associated in patients whose initial symptoms included moderate-to-profound hearing impairment. Exome analysis of Iranian and Pakistani probands who both showed bilateral, sensorineural hearing loss revealed a novel splice site variant (c.1421+2T>C, p.?) that disrupts the splice donor site and a novel frameshift variant (c.1041dup, p.Ser348Glnfs*2) in the gene CDC14A, respectively. To evaluate the pathogenicity of both loss-of-function variants, we analyzed the effects of both variants on the RNA-level. The splice variant was characterized using a minigene assay. Altered expression levels due to the c.1041dup variant were assessed using RT-qPCR. In summary, cDNA analysis confirmed that the c.1421+2T>C variant activates a cryptic splice site, resulting in a truncated transcript (c.1414_1421del, p.Val472Leufs*20) and the c.1041dup variant results in a defective transcript that is likely degraded by nonsense-mediated mRNA decay. The present study functionally characterizes two variants and provides further confirmatory evidence that CDC14A is associated with a rare form of hereditary hearing loss.


Asunto(s)
Pérdida Auditiva Sensorineural/genética , Proteínas Tirosina Fosfatasas/genética , Adulto , Exoma/genética , Femenino , Estudios de Asociación Genética , Humanos , Irán , Mutación con Pérdida de Función , Masculino , Pakistán , Linaje , Proteínas Tirosina Fosfatasas/metabolismo , Sitios de Empalme de ARN/genética , Empalme del ARN
6.
Hum Genet ; 134(7): 691-704, 2015 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-25893793

RESUMEN

We describe a large family with disproportionate short stature and bone dysplasia from Nias in which we observed differences in severity when comparing the phenotypes of affected individuals from two remote branches. We conducted a linkage scan in the more severely affected family branch and determined a critical interval of 4.7 cM on chromosome 11. Sequencing of the primary candidate gene TBX10 did not reveal a disease-causing variant. When performing whole exome sequencing we noticed a homozygous missense variant in B3GAT3, c.419C>T [p.(Pro140Leu)]. B3GAT3 encodes ß-1,3-glucuronyltransferase-I (GlcAT-I). GlcAT-I catalyzes an initial step of proteoglycan synthesis and the mutation p. (Pro140Leu) lies within the donor substrate-binding subdomain of the catalytic domain. In contrast to the previously published mutation in B3GAT3, c.830G>A [p.(Arg277Gln)], no heart phenotype could be detected in our family. Functional studies revealed a markedly reduced GlcAT-I activity in lymphoblastoid cells from patients when compared to matched controls. Moreover, relative numbers of glycosaminoglycan (GAG) side chains were decreased in patient cells. We found that Pro140Leu-mutant GlcAT-I cannot efficiently transfer GlcA to the linker region trisaccharide. This failure results in a partial deficiency of both chondroitin sulfate and heparan sulfate chains. Since the phenotype of the Nias patients differs from the Larsen-like syndrome described for patients with mutation p.(Arg277Gln), we suggest mutation B3GAT3:p.(Pro140Leu) to cause a different type of GAG linkeropathy showing no involvement of the heart.


Asunto(s)
Enfermedades del Desarrollo Óseo/genética , Enfermedades Genéticas Congénitas/genética , Mutación Missense , Adolescente , Adulto , Sustitución de Aminoácidos , Enfermedades del Desarrollo Óseo/enzimología , Enfermedades del Desarrollo Óseo/patología , Niño , Preescolar , Femenino , Enfermedades Genéticas Congénitas/enzimología , Enfermedades Genéticas Congénitas/patología , Glucuronosiltransferasa/metabolismo , Humanos , Lactante , Masculino , Linaje , Estructura Terciaria de Proteína
7.
Am J Hum Genet ; 90(2): 201-16, 2012 Feb 10.
Artículo en Inglés | MEDLINE | ID: mdl-22265013

RESUMEN

We report on an autosomal-recessive variant of Ehlers-Danlos syndrome (EDS) characterized by severe muscle hypotonia at birth, progressive scoliosis, joint hypermobility, hyperelastic skin, myopathy, sensorineural hearing impairment, and normal pyridinoline excretion in urine. Clinically, the disorder shares many features with the kyphoscoliotic type of EDS (EDS VIA) and Ullrich congenital muscular dystrophy. Linkage analysis in a large Tyrolean kindred identified a homozygous frameshift mutation in FKBP14 in two affected individuals. Based on the cardinal clinical characteristics of the disorder, four additional individuals originating from different European countries were identified who carried either homozygous or compound heterozygous mutations in FKBP14. FKBP14 belongs to the family of FK506-binding peptidyl-prolyl cis-trans isomerases (PPIases). ER-resident FKBPs have been suggested to act as folding catalysts by accelerating cis-trans isomerization of peptidyl-prolyl bonds and to act occasionally also as chaperones. We demonstrate that FKBP14 is localized in the endoplasmic reticulum (ER) and that deficiency of FKBP14 leads to enlarged ER cisterns in dermal fibroblasts in vivo. Furthermore, indirect immunofluorescence of FKBP14-deficient fibroblasts indicated an altered assembly of the extracellular matrix in vitro. These findings suggest that a disturbance of protein folding in the ER affecting one or more components of the extracellular matrix might cause the generalized connective tissue involvement in this disorder. FKBP14 mutation analysis should be considered in all individuals with apparent kyphoscoliotic type of EDS and normal urinary pyridinoline excretion, in particular in conjunction with sensorineural hearing impairment.


Asunto(s)
Anomalías Múltiples/genética , Síndrome de Ehlers-Danlos/genética , Mutación del Sistema de Lectura , Pérdida Auditiva/genética , Isomerasa de Peptidilprolil/genética , Adolescente , Aminoácidos/orina , Niño , Preescolar , Síndrome de Ehlers-Danlos/orina , Retículo Endoplásmico/genética , Matriz Extracelular/genética , Femenino , Fibroblastos/metabolismo , Variación Genética , Pérdida Auditiva/orina , Heterocigoto , Homocigoto , Humanos , Masculino , Persona de Mediana Edad , Fenotipo , Pliegue de Proteína , cis-trans-Isomerasas/genética
8.
PLoS Biol ; 10(5): e1001318, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-22563300

RESUMEN

In all vertebrates hearing and touch represent two distinct sensory systems that both rely on the transformation of mechanical force into electrical signals. There is an extensive literature describing single gene mutations in humans that cause hearing impairment, but there are essentially none for touch. Here we first asked if touch sensitivity is a heritable trait and second whether there are common genes that influence different mechanosensory senses like hearing and touch in humans. Using a classical twin study design we demonstrate that touch sensitivity and touch acuity are highly heritable traits. Quantitative phenotypic measures of different mechanosensory systems revealed significant correlations between touch and hearing acuity in a healthy human population. Thus mutations in genes causing deafness genes could conceivably negatively influence touch sensitivity. In agreement with this hypothesis we found that a proportion of a cohort of congenitally deaf young adults display significantly impaired measures of touch sensitivity compared to controls. In contrast, blind individuals showed enhanced, not diminished touch acuity. Finally, by examining a cohort of patients with Usher syndrome, a genetically well-characterized deaf-blindness syndrome, we could show that recessive pathogenic mutations in the USH2A gene influence touch acuity. Control Usher syndrome cohorts lacking demonstrable pathogenic USH2A mutations showed no impairment in touch acuity. Our study thus provides comprehensive evidence that there are common genetic elements that contribute to touch and hearing and has identified one of these genes as USH2A.


Asunto(s)
Pérdida Auditiva/genética , Mecanotransducción Celular , Tacto/genética , Síndromes de Usher/genética , Adolescente , Adulto , Factores de Edad , Barorreflejo , Estudios de Cohortes , Proteínas de la Matriz Extracelular/genética , Femenino , Pruebas Genéticas , Genotipo , Pérdida Auditiva/congénito , Humanos , Patrón de Herencia , Masculino , Mutación , Fenotipo , Temperatura , Adulto Joven
9.
Nat Genet ; 38(11): 1245-7, 2006 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-17041604

RESUMEN

Anonychia and hyponychia congenita (OMIM 206800) are rare autosomal recessive conditions in which the only presenting phenotype is the absence or severe hypoplasia of all fingernails and toenails. After determining linkage to chromosome 20p13, we identified homozygous or compound heterozygous mutations in the gene encoding R-spondin 4 (RSPO4), a secreted protein implicated in Wnt signaling, in eight affected families. Rspo4 expression was specifically localized to developing mouse nail mesenchyme at embryonic day 15.5, suggesting a crucial role in nail morphogenesis.


Asunto(s)
Uñas Malformadas/genética , Trombospondinas/genética , Proteínas Wnt/metabolismo , Secuencia de Aminoácidos , Animales , Humanos , Ratones , Ratones Transgénicos , Datos de Secuencia Molecular , Mutación , Homología de Secuencia de Aminoácido , Transducción de Señal , Trombospondinas/metabolismo
10.
Gut ; 63(1): 96-104, 2014 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-23268370

RESUMEN

OBJECTIVE: Cryptogenic multifocal ulcerating stenosing enteritis (CMUSE) is an extremely rare, but devastating, disease of unknown aetiology. We investigated the genetic basis of this autosomal recessive condition in a pair of affected siblings who have 40-year histories of catastrophic gastrointestinal and extraintestinal disease. DESIGN: Genome-wide single-nucleotide polymorphism homozygosity mapping in the two affected family members combined with whole-exome sequencing of one affected sibling. This was followed by confirmatory Sanger sequencing of the likely disease-causing sequence variant and functional studies in affected and unaffected family members. RESULTS: Insertion/deletion variation analysis revealed the presence of a homozygous 4 bp deletion (g.155574_77delGTAA) in the PLA2G4A gene, located in the splice donor site directly after exon 17 (the penultimate exon) of the gene in both affected siblings. This introduces a frameshift of 10 amino acids before a premature stop codon (p.V707fsX10), which is predicted to result in the loss of 43 amino acids (residues 707-749) at the C-terminus of cytosolic phospholipase A2-α (cPLA(2)α). cPLA(2)α protein expression was undetectable in the gut of both siblings, with platelet aggregation and thromboxane A(2) production, as functional assays for cPLA(2)α activity, grossly impaired. CONCLUSIONS: We have identified mutations in PLA2G4A as a cause of CMUSE in two affected siblings. Further studies are needed to determine if mutations in this gene are also responsible for disease of a similar phenotype in other cases.


Asunto(s)
Enteritis/genética , Fosfolipasas A2 Grupo IV/genética , Homocigoto , Úlcera Péptica/genética , Eliminación de Secuencia , Adulto , Biomarcadores/metabolismo , Western Blotting , Estudios de Casos y Controles , Codón sin Sentido , Enteritis/complicaciones , Enteritis/enzimología , Femenino , Técnica del Anticuerpo Fluorescente , Mutación del Sistema de Lectura , Marcadores Genéticos , Fosfolipasas A2 Grupo IV/metabolismo , Humanos , Obstrucción Intestinal/etiología , Masculino , Úlcera Péptica/complicaciones , Úlcera Péptica/enzimología , Polimorfismo de Nucleótido Simple , Análisis de Secuencia de ADN , Hermanos
11.
N Engl J Med ; 365(16): 1502-8, 2011 Oct 20.
Artículo en Inglés | MEDLINE | ID: mdl-22010916

RESUMEN

We performed genetic and immunohistochemical studies in a sister and brother with autosomal recessive neonatal inflammatory skin and bowel lesions. The girl died suddenly at 12 years of age from parvovirus B19-associated myocarditis; her brother had mild cardiomyopathy. We identified a loss-of-function mutation in ADAM17, which encodes a disintegrin and metalloproteinase 17 (also called tumor necrosis factor α [TNF-α]-converting enzyme, or TACE), as the probable cause of this syndrome. Peripheral-blood mononuclear cells (PBMCs) obtained from the brother at 17 years of age showed high levels of lipopolysaccharide-induced production of interleukin-1ß and interleukin-6 but impaired release of TNF-α. Despite repeated skin infections, this young man has led a relatively normal life. (Funded by Barts and the London Charity and the European Commission Seventh Framework Programme.).


Asunto(s)
Proteínas ADAM/genética , Enfermedades Inflamatorias del Intestino/genética , Eliminación de Secuencia , Enfermedades de la Piel/genética , Proteína ADAM17 , Adolescente , Niño , Resultado Fatal , Femenino , Humanos , Masculino , Miocarditis/genética , Miocarditis/virología , Linaje
13.
J Allergy Clin Immunol ; 132(2): 371-7, 2013 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-23582566

RESUMEN

BACKGROUND: Atopic dermatitis (AD) is a common inflammatory skin disease. Previous studies have revealed shared genetic determinants among different inflammatory disorders, suggesting that markers associated with immune-related traits might also play a role in AD. OBJECTIVE: We sought to identify novel genetic risk factors for AD. METHODS: We examined the results of all genome-wide association studies from a public repository and selected 318 genetic markers that were significantly associated with any inflammatory trait. These markers were considered candidates and tested for association with AD in a 3-step approach including 7 study populations with 7130 patients with AD and 9253 control subjects. RESULTS: A functional amino acid change in the IL-6 receptor (IL-6R Asp358Ala; rs2228145) was significantly associated with AD (odds ratio [OR], 1.15; P = 5 × 10(-9)). Interestingly, investigation of 2 independent population-based birth cohorts showed that IL-6R 358Ala specifically predisposes to the persistent form of AD (ORpersistent AD = 1.22, P = .0008; ORtransient AD = 1.04, P = .54). This variant determines the balance between the classical membrane-bound versus soluble IL-6R signaling pathways. Carriers of 358Ala had increased serum levels of soluble IL-6R (P = 4 × 10(-14)), with homozygote carriers showing a 2-fold increase. Moreover, we demonstrate that soluble IL-6R levels were higher in patients with AD than in control subjects (46.0 vs 37.8 ng/mL, P = .001). Additional AD risk variants were identified in RAD50, RUNX3, and ERBB3. CONCLUSION: Our study supports the importance of genetic variants influencing inflammation in the etiology of AD. Moreover, we identified a functional genetic variant in IL6R influencing disease prognosis and specifically predisposing to persistent AD.


Asunto(s)
Dermatitis Atópica/genética , Predisposición Genética a la Enfermedad , Polimorfismo de Nucleótido Simple , Receptores de Interleucina-6/genética , Adolescente , Alelos , Niño , Preescolar , Estudios de Cohortes , Femenino , Estudio de Asociación del Genoma Completo , Genotipo , Humanos , Lactante , Recién Nacido , Interleucina-6/genética , Interleucina-6/metabolismo , Masculino , Receptores de Interleucina-6/sangre , Factores de Riesgo
15.
Nat Genet ; 37(2): 166-70, 2005 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-15654338

RESUMEN

Familial glucocorticoid deficiency (FGD), or hereditary unresponsiveness to adrenocorticotropin (ACTH; OMIM 202200), is an autosomal recessive disorder resulting from resistance to the action of ACTH on the adrenal cortex, which stimulates glucocorticoid production. Affected individuals are deficient in cortisol and, if untreated, are likely to succumb to hypoglycemia or overwhelming infection in infancy or childhood. Mutations of the ACTH receptor (melanocortin 2 receptor, MC2R) account for approximately 25% of cases of FGD. FGD without mutations of MC2R is called FGD type 2. Using SNP array genotyping, we mapped a locus involved in FGD type 2 to chromosome 21q22.1. We identified mutations in a gene encoding a 19-kDa single-transmembrane domain protein, now known as melanocortin 2 receptor accessory protein (MRAP). We show that MRAP interacts with MC2R and may have a role in the trafficking of MC2R from the endoplasmic reticulum to the cell surface.


Asunto(s)
Hormona Adrenocorticotrópica/deficiencia , Proteínas de la Membrana/genética , Receptor de Melanocortina Tipo 2/genética , Animales , Células CHO , Mapeo Cromosómico , Cromosomas Humanos Par 21 , Cricetinae , Cricetulus , Femenino , Humanos , Masculino , Datos de Secuencia Molecular , Mutación , Linaje , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa
16.
Nat Genet ; 36(8): 850-4, 2004 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-15258582

RESUMEN

We identified three consanguineous Austrian kindreds with 15 members affected by autosomal recessive childhood-onset severe retinal dystrophy, a genetically heterogeneous group of disorders characterized by degeneration of the photoreceptor cells. A whole-genome scan by microarray analysis of single-nucleotide polymorphisms (ref. 2) identified a founder haplotype and defined a critical interval of 1.53 cM on chromosome 14q23.3-q24.1 that contains the gene associated with this form of retinal dystrophy. RDH12 maps in this region and encodes a retinol dehydrogenase proposed to function in the visual cycle. A homozygous 677A-->G transition (resulting in Y226C) in RDH12 was present in all affected family members studied, as well as in two Austrian individuals with sporadic retinal dystrophy. We identified additional mutations in RDH12 in 3 of 89 non-Austrian individuals with retinal dystrophy: a 5-nucleotide deletion (806delCCCTG) and the transition 565C-->T (resulting in Q189X), each in the homozygous state, and 146C-->T (resulting in T49M) and 184C-->T (resulting in R62X) in compound heterozygosity. When expressed in COS-7 cells, Cys226 and Met49 variants had diminished and aberrant activity, respectively, in interconverting isomers of retinol and retinal. The severe visual impairment of individuals with mutations in RDH12 is in marked contrast to the mild visual deficiency in individuals with fundus albipunctatus caused by mutations in RDH5, encoding another retinal dehydrogenase. Our studies show that RDH12 is associated with retinal dystrophy and encodes an enzyme with a unique, nonredundant role in the photoreceptor cells.


Asunto(s)
Oxidorreductasas de Alcohol/genética , Células Fotorreceptoras/enzimología , Degeneración Retiniana/genética , Adulto , Preescolar , Cromosomas Humanos Par 14 , Femenino , Efecto Fundador , Genes Recesivos , Humanos , Masculino , Mutación , Linaje , Polimorfismo de Nucleótido Simple
17.
Am J Hum Genet ; 84(2): 188-96, 2009 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-19185281

RESUMEN

Autosomal-recessive congenital sodium diarrhea (CSD) is characterized by perinatal onset of a persistent watery diarrhea with nonproportionally high fecal sodium excretion. Defective jejunal brush-border Na(+)/H(+) exchange has been reported in three sporadic patients, but the molecular basis of the disease has not been elucidated. We reviewed data from a large cohort of CSD patients (n = 24) and distinguished CSD associated with choanal or anal atresia, hypertelorism, and corneal erosions--i.e., a syndromic form of CSD--occurring in ten families from an isolated form--i.e., classic CSD--presenting in seven families. Patients from both groups have a high risk of mortality due to immediate electrolyte imbalances and complications from long-term parenteral nutrition in the first years of life, but survivors can eventually adapt to partial or complete enteral nutrition. A genome-wide SNP scan was applied and identified a homozygous c.593-1G-->A splicing mutation in SPINT2, encoding a Kunitz-type serine-protease inhibitor, in one extended kindred with syndromic CSD. The same mutation and four distinct, homozygous or compound heterozygous mutations (p.Y163C, c.1A-->T, c.337+2T-->C, c.553+2T-->A) were identified in all syndromic patients. No SPINT2 mutations were found in classic-CSD patients. SPINT2 mutations were associated with loss of protein synthesis or failure to inhibit the serine protease trypsin in vitro. We delineate syndromic CSD as a distinct disease entity caused by SPINT2 loss-of-function mutations. SPINT2 mutations might lead to an excess of yet unknown serine protease activity in affected tissues.


Asunto(s)
Diarrea/genética , Síndromes de Malabsorción/genética , Glicoproteínas de Membrana/genética , Mutación , Sodio/metabolismo , Secuencia de Aminoácidos , Ano Imperforado/genética , Ano Imperforado/mortalidad , Ano Imperforado/patología , Secuencia de Bases , Mapeo Cromosómico , Estudios de Cohortes , Análisis Mutacional de ADN , Diarrea/mortalidad , Diarrea/patología , Heces/química , Femenino , Genes Recesivos , Humanos , Lactante , Recién Nacido , Síndromes de Malabsorción/mortalidad , Síndromes de Malabsorción/patología , Masculino , Datos de Secuencia Molecular , Linaje , ARN Mensajero/genética , Análisis de Supervivencia
18.
PLoS Genet ; 5(1): e1000353, 2009 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-19165332

RESUMEN

The identification of recessive disease-causing genes by homozygosity mapping is often restricted by lack of suitable consanguineous families. To overcome these limitations, we apply homozygosity mapping to single affected individuals from outbred populations. In 72 individuals of 54 kindred ascertained worldwide with known homozygous mutations in 13 different recessive disease genes, we performed total genome homozygosity mapping using 250,000 SNP arrays. Likelihood ratio Z-scores (ZLR) were plotted across the genome to detect ZLR peaks that reflect segments of homozygosity by descent, which may harbor the mutated gene. In 93% of cases, the causative gene was positioned within a consistent ZLR peak of homozygosity. The number of peaks reflected the degree of inbreeding. We demonstrate that disease-causing homozygous mutations can be detected in single cases from outbred populations within a single ZLR peak of homozygosity as short as 2 Mb, containing an average of only 16 candidate genes. As many specialty clinics have access to cohorts of individuals from outbred populations, and as our approach will result in smaller genetic candidate regions, the new strategy of homozygosity mapping in single outbred individuals will strongly accelerate the discovery of novel recessive disease genes.


Asunto(s)
Genes Recesivos , Análisis Mutacional de ADN , Reacciones Falso Positivas , Salud de la Familia , Femenino , Marcadores Genéticos , Genética de Población , Homocigoto , Humanos , Enfermedades Renales Quísticas/genética , Masculino , Modelos Genéticos , Síndrome Nefrótico/genética , Linaje , Esteroides/farmacología
19.
Am J Hum Genet ; 83(6): 781-6, 2008 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-19061984

RESUMEN

Tooth eruption is a complex developmental process requiring coordinated navigation through alveolar bone and oral epithelium. Primary failure of tooth eruption (PFE) is associated with several syndromes primarily affecting skeletal development, but it is also known as a nonsyndromic autosomal-dominant condition. Teeth in the posterior quadrants of the upper and lower jaw are preferentially affected and usually result in an open bite extending from anterior to posterior. In this study, we show that familial, nonsyndromic PFE is caused by heterozygous mutations in the gene encoding the G protein-coupled receptor for parathyroid hormone and parathyroid hormone-like hormone (PTHR1). Three distinct mutations, namely c.1050-3C > G, c.543+1G > A, and c.463G > T, were identified in 15 affected individuals from four multiplex pedigrees. All mutations truncate the mature protein and therefore should lead to a functionless receptor, strongly suggesting that haplo-insufficiency of PTHR1 is the underlying cause of nonsyndromic PFE. Although complete inactivation of PTHR1 is known to underlie the autosomal-recessive Blomstrand osteochondrodysplasia (BOCD), a lethal form of short-limbed dwarfism, our data now imply that dominantly acting PTHR1 mutations that lead to haplo-insufficiency of the receptor result in a nonsyndromic phenotype affecting tooth development with high penetrance and variable expressivity.


Asunto(s)
Mutación , Receptor de Hormona Paratiroídea Tipo 1/genética , Erupción Dental/genética , Cromosomas Humanos Par 3 , Femenino , Genes Recesivos , Ligamiento Genético , Marcadores Genéticos , Heterocigoto , Humanos , Masculino , Linaje , Polimorfismo de Nucleótido Simple
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA