Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 41
Filtrar
Más filtros

Tipo del documento
Intervalo de año de publicación
1.
Proc Natl Acad Sci U S A ; 116(4): 1347-1352, 2019 01 22.
Artículo en Inglés | MEDLINE | ID: mdl-30610177

RESUMEN

We have identified a GRAP variant (c.311A>T; p.Gln104Leu) cosegregating with autosomal recessive nonsyndromic deafness in two unrelated families. GRAP encodes a member of the highly conserved growth factor receptor-bound protein 2 (GRB2)/Sem-5/drk family of proteins, which are involved in Ras signaling; however, the function of the growth factor receptor-bound protein 2 (GRB2)-related adaptor protein (GRAP) in the auditory system is not known. Here, we show that, in mouse, Grap is expressed in the inner ear and the protein localizes to the neuronal fibers innervating cochlear and utricular auditory hair cells. Downstream of receptor kinase (drk), the Drosophila homolog of human GRAP, is expressed in Johnston's organ (JO), the fly hearing organ, and the loss of drk in JO causes scolopidium abnormalities. drk mutant flies present deficits in negative geotaxis behavior, which can be suppressed by human wild-type but not mutant GRAP. Furthermore, drk specifically colocalizes with synapsin at synapses, suggesting a potential role of such adaptor proteins in regulating actin cytoskeleton dynamics in the nervous system. Our findings establish a causative link between GRAP mutation and nonsyndromic deafness and suggest a function of GRAP/drk in hearing.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/metabolismo , Proteína Adaptadora GRB2/metabolismo , Pérdida Auditiva Sensorineural/metabolismo , Secuencia de Aminoácidos , Animales , Proteínas Portadoras/metabolismo , Sordera/microbiología , Drosophila/metabolismo , Femenino , Humanos , Masculino , Ratones , Ratones Endogámicos C57BL , Unión Proteica/fisiología , Transducción de Señal/fisiología
2.
Hum Mol Genet ; 28(8): 1286-1297, 2019 04 15.
Artículo en Inglés | MEDLINE | ID: mdl-30561639

RESUMEN

Molecular mechanisms governing the development of the human cochlea remain largely unknown. Through genome sequencing, we identified a homozygous FOXF2 variant c.325A>T (p.I109F) in a child with profound sensorineural hearing loss (SNHL) associated with incomplete partition type I anomaly of the cochlea. This variant is not found in public databases or in over 1000 ethnicity-matched control individuals. I109 is a highly conserved residue in the forkhead box (Fox) domain of FOXF2, a member of the Fox protein family of transcription factors that regulate the expression of genes involved in embryogenic development as well as adult life. Our in vitro studies show that the half-life of mutant FOXF2 is reduced compared to that of wild type. Foxf2 is expressed in the cochlea of developing and adult mice. The mouse knockout of Foxf2 shows shortened and malformed cochleae, in addition to altered shape of hair cells with innervation and planar cell polarity defects. Expressions of Eya1 and Pax3, genes essential for cochlear development, are reduced in the cochleae of Foxf2 knockout mice. We conclude that FOXF2 plays a major role in cochlear development and its dysfunction leads to SNHL and developmental anomalies of the cochlea in humans and mice.


Asunto(s)
Cóclea/embriología , Factores de Transcripción Forkhead/genética , Factores de Transcripción Forkhead/fisiología , Adulto , Animales , Niño , Cóclea/metabolismo , Cóclea/fisiología , Desarrollo Embrionario , Femenino , Células Ciliadas Auditivas/metabolismo , Humanos , Péptidos y Proteínas de Señalización Intracelular/genética , Péptidos y Proteínas de Señalización Intracelular/fisiología , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Proteínas Nucleares/genética , Proteínas Nucleares/fisiología , Organogénesis , Factor de Transcripción PAX3/genética , Factor de Transcripción PAX3/fisiología , Proteínas Tirosina Fosfatasas/genética , Proteínas Tirosina Fosfatasas/fisiología , Transducción de Señal/genética , Secuenciación Completa del Genoma
3.
Clin Genet ; 96(6): 575-578, 2019 12.
Artículo en Inglés | MEDLINE | ID: mdl-31432506

RESUMEN

Auditory reception relies on the perception of mechanical stimuli by stereocilia and its conversion to electrochemical signal. Mechanosensory stereocilia are abundant in actin, which provides them with structural conformity necessary for perception of auditory stimuli. Out of three major classes of actin-bundling proteins, plastin 1 encoded by PLS1, is highly expressed in stereocilia and is necessary for their regular maintenance. A missense PLS1 variant associated with autosomal dominant hearing loss (HL) in a small family has recently been reported. Here, we present another PLS1 missense variant, c.805G > A (p.E269K), in a Turkish family with autosomal dominant non-syndromic HL confirming the causative role of PLS1 mutations in HL. We propose that HL due to the p.E269K variant is from the loss of a stable PLS1-ACTB interaction.


Asunto(s)
Genes Dominantes , Pérdida Auditiva/genética , Glicoproteínas de Membrana/genética , Proteínas de Microfilamentos/genética , Mutación/genética , Secuencia de Aminoácidos , Secuencia de Bases , Familia , Femenino , Humanos , Masculino , Glicoproteínas de Membrana/química , Proteínas de Microfilamentos/química , Proteínas Mutantes/química , Linaje , Turquía
4.
Proc Natl Acad Sci U S A ; 113(21): 5993-8, 2016 May 24.
Artículo en Inglés | MEDLINE | ID: mdl-27162350

RESUMEN

Hair cells of the inner ear, the mechanosensory receptors, convert sound waves into neural signals that are passed to the brain via the auditory nerve. Little is known about the molecular mechanisms that govern the development of hair cell-neuronal connections. We ascertained a family with autosomal recessive deafness associated with a common cavity inner ear malformation and auditory neuropathy. Via whole-exome sequencing, we identified a variant (c.2207G>C, p.R736T) in ROR1 (receptor tyrosine kinase-like orphan receptor 1), cosegregating with deafness in the family and absent in ethnicity-matched controls. ROR1 is a tyrosine kinase-like receptor localized at the plasma membrane. At the cellular level, the mutation prevents the protein from reaching the cellular membrane. In the presence of WNT5A, a known ROR1 ligand, the mutated ROR1 fails to activate NF-κB. Ror1 is expressed in the inner ear during development at embryonic and postnatal stages. We demonstrate that Ror1 mutant mice are severely deaf, with preserved otoacoustic emissions. Anatomically, mutant mice display malformed cochleae. Axons of spiral ganglion neurons show fasciculation defects. Type I neurons show impaired synapses with inner hair cells, and type II neurons display aberrant projections through the cochlear sensory epithelium. We conclude that Ror1 is crucial for spiral ganglion neurons to innervate auditory hair cells. Impairment of ROR1 function largely affects development of the inner ear and hearing in humans and mice.


Asunto(s)
Células Ciliadas Auditivas/metabolismo , Pérdida Auditiva Sensorineural/metabolismo , Mutación , Receptores Huérfanos Similares al Receptor Tirosina Quinasa/metabolismo , Ganglio Espiral de la Cóclea/metabolismo , Animales , Axones/metabolismo , Axones/patología , Línea Celular , Células Ciliadas Auditivas/patología , Pérdida Auditiva Sensorineural/genética , Pérdida Auditiva Sensorineural/patología , Humanos , Ratones , Ratones Mutantes , Receptores Huérfanos Similares al Receptor Tirosina Quinasa/genética , Ganglio Espiral de la Cóclea/patología , Proteína Wnt-5a/genética , Proteína Wnt-5a/metabolismo
5.
Hum Genet ; 137(6-7): 479-486, 2018 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-29982980

RESUMEN

While recent studies have revealed a substantial portion of the genes underlying human hearing loss, the extensive genetic landscape has not been completely explored. Here, we report a loss-of-function variant (c.72delA) in MPZL2 in three unrelated multiplex families from Turkey and Iran with autosomal recessive nonsyndromic hearing loss. The variant co-segregates with moderate sensorineural hearing loss in all three families. We show a shared haplotype flanking the variant in our families implicating a single founder. While rare in other populations, the allele frequency of the variant is ~ 0.004 in Ashkenazi Jews, suggesting that it may be an important cause of moderate hearing loss in that population. We show that Mpzl2 is expressed in mouse inner ear, and the protein localizes in the auditory inner and outer hair cells, with an asymmetric subcellular localization. We thus present MPZL2 as a novel gene associated with sensorineural hearing loss.


Asunto(s)
Moléculas de Adhesión Celular/genética , Sordera/genética , Células Ciliadas Auditivas Internas/metabolismo , Pérdida Auditiva Sensorineural/genética , Animales , Sordera/fisiopatología , Oído Interno/crecimiento & desarrollo , Oído Interno/fisiopatología , Femenino , Frecuencia de los Genes , Genes Recesivos , Células Ciliadas Auditivas Internas/patología , Haplotipos/genética , Pérdida Auditiva Sensorineural/fisiopatología , Humanos , Irán/epidemiología , Judíos/genética , Masculino , Ratones , Mutación , Linaje , Células de Schwann/patología , Turquía
6.
Proc Natl Acad Sci U S A ; 111(27): 9864-8, 2014 Jul 08.
Artículo en Inglés | MEDLINE | ID: mdl-24958875

RESUMEN

In a large consanguineous Turkish kindred with recessive nonsyndromic, prelingual, profound hearing loss, we identified in the gene FAM65B (MIM611410) a splice site mutation (c.102-1G>A) that perfectly cosegregates with the phenotype in the family. The mutation leads to exon skipping and deletion of 52-amino acid residues of a PX membrane localization domain. FAM65B is known to be involved in myotube formation and in regulation of cell adhesion, polarization, and migration. We show that wild-type Fam65b is expressed during embryonic and postnatal development stages in murine cochlea, and that the protein localizes to the plasma membranes of the stereocilia of inner and outer hair cells of the inner ear. The wild-type protein targets the plasma membrane, whereas the mutant protein accumulates in cytoplasmic inclusion bodies and does not reach the membrane. In zebrafish, knockdown of fam65b leads to significant reduction of numbers of saccular hair cells and neuromasts and to hearing loss. We conclude that FAM65B is a plasma membrane-associated protein of hair cell stereocilia that is essential for hearing.


Asunto(s)
Audición/fisiología , Proteínas/fisiología , Estereocilios/fisiología , Animales , Moléculas de Adhesión Celular , Modelos Animales de Enfermedad , Femenino , Regulación del Desarrollo de la Expresión Génica , Técnicas de Silenciamiento del Gen , Audición/genética , Pérdida Auditiva Sensorineural/genética , Humanos , Masculino , Ratones , Linaje , Proteínas/genética , Proteínas/metabolismo , Empalme del ARN , Fracciones Subcelulares/metabolismo , Turquía , Pez Cebra
7.
Genet Med ; 18(4): 364-71, 2016 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-26226137

RESUMEN

PURPOSE: Autosomal recessive nonsyndromic deafness (ARNSD) is characterized by a high degree of genetic heterogeneity, with reported mutations in 58 different genes. This study was designed to detect deafness-causing variants in a multiethnic cohort with ARNSD by using whole-exome sequencing (WES). METHODS: After excluding mutations in the most common gene, GJB2, we performed WES in 160 multiplex families with ARNSD from Turkey, Iran, Mexico, Ecuador, and Puerto Rico to screen for mutations in all known ARNSD genes. RESULTS: We detected ARNSD-causing variants in 90 (56%) families, 54% of which had not been previously reported. Identified mutations were located in 31 known ARNSD genes. The most common genes with mutations were MYO15A (13%), MYO7A (11%), SLC26A4 (10%), TMPRSS3 (9%), TMC1 (8%), ILDR1 (6%), and CDH23 (4%). Nine mutations were detected in multiple families with shared haplotypes, suggesting founder effects. CONCLUSION: We report on a large multiethnic cohort with ARNSD in which comprehensive analysis of all known ARNSD genes identifies causative DNA variants in 56% of the families. In the remaining families, WES allows us to search for causative variants in novel genes, thus improving our ability to explain the underlying etiology in more families.Genet Med 18 4, 364-371.


Asunto(s)
Sordera/diagnóstico , Sordera/genética , Exoma , Genes Recesivos , Pérdida Auditiva Sensorineural/diagnóstico , Pérdida Auditiva Sensorineural/genética , Secuenciación de Nucleótidos de Alto Rendimiento , Alelos , Estudios de Cohortes , Etnicidad/genética , Genotipo , Humanos , Mutación
8.
Hum Genet ; 134(2): 181-90, 2015 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-25413698

RESUMEN

Mutations in ANKRD11 have recently been reported to cause KBG syndrome, an autosomal dominant condition characterized by intellectual disability (ID), behavioral problems, and macrodontia. To understand the pathogenic mechanism that relates ANKRD11 mutations with the phenotype of KBG syndrome, we studied the cellular characteristics of wild-type ANKRD11 and the effects of mutations in humans and mice. We show that the abundance of wild-type ANKRD11 is tightly regulated during the cell cycle, and that the ANKRD11 C-terminus is required for the degradation of the protein. Analysis of 11 pathogenic ANKRD11 variants in humans, including six reported in this study, and one reported in the Ankrd11 (Yod/+) mouse, shows that all mutations affect the C-terminal regions and that the mutant proteins accumulate aberrantly. In silico analysis shows the presence of D-box sequences that are signals for proteasome degradation. We suggest that ANKRD11 C-terminus plays an important role in regulating the abundance of the protein, and a disturbance of the protein abundance due to the mutations leads to KBG syndrome.


Asunto(s)
Anomalías Múltiples , Enfermedades del Desarrollo Óseo , Ciclo Celular/genética , Proteínas de Unión al ADN , Facies , Discapacidad Intelectual , Mutación , Proteolisis , Proteínas Represoras , Anomalías Dentarias , Anomalías Múltiples/genética , Anomalías Múltiples/metabolismo , Animales , Enfermedades del Desarrollo Óseo/genética , Enfermedades del Desarrollo Óseo/metabolismo , Línea Celular Tumoral , Proteínas de Unión al ADN/genética , Proteínas de Unión al ADN/metabolismo , Femenino , Humanos , Discapacidad Intelectual/genética , Discapacidad Intelectual/metabolismo , Masculino , Ratones , Ratones Mutantes , Complejo de la Endopetidasa Proteasomal/genética , Complejo de la Endopetidasa Proteasomal/metabolismo , Estabilidad Proteica , Estructura Terciaria de Proteína , Proteínas Represoras/genética , Proteínas Represoras/metabolismo , Anomalías Dentarias/genética , Anomalías Dentarias/metabolismo
9.
Am J Hum Genet ; 91(5): 872-82, 2012 11 02.
Artículo en Inglés | MEDLINE | ID: mdl-23122586

RESUMEN

Hereditary hearing loss is characterized by a high degree of genetic heterogeneity. Here we present OTOGL mutations, a homozygous one base pair deletion (c.1430 delT) causing a frameshift (p.Val477Glufs(∗)25) in a large consanguineous family and two compound heterozygous mutations, c.547C>T (p.Arg183(∗)) and c.5238+5G>A, in a nonconsanguineous family with moderate nonsyndromic sensorineural hearing loss. OTOGL maps to the DFNB84 locus at 12q21.31 and encodes otogelin-like, which has structural similarities to the epithelial-secreted mucin protein family. We demonstrate that Otogl is expressed in the inner ear of vertebrates with a transcription level that is high in embryonic, lower in neonatal, and much lower in adult stages. Otogelin-like is localized to the acellular membranes of the cochlea and the vestibular system and to a variety of inner ear cells located underneath these membranes. Knocking down of otogl with morpholinos in zebrafish leads to sensorineural hearing loss and anatomical changes in the inner ear, supporting that otogelin-like is essential for normal inner ear function. We propose that OTOGL mutations affect the production and/or function of acellular structures of the inner ear, which ultimately leads to sensorineural hearing loss.


Asunto(s)
Pérdida Auditiva Sensorineural/genética , Proteínas de la Membrana/genética , Mutación , Adolescente , Animales , Preescolar , Aberraciones Cromosómicas , Cóclea/metabolismo , Cóclea/patología , Exoma , Perfilación de la Expresión Génica , Técnicas de Silenciamiento del Gen , Pérdida Auditiva Sensorineural/diagnóstico , Humanos , Mutación INDEL , Masculino , Ratones , Polimorfismo de Nucleótido Simple , Ratas , Pez Cebra
10.
Mol Genet Genomics ; 290(4): 1327-34, 2015 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-25633957

RESUMEN

Hearing loss (HL) is a major public health issue. It is clinically and genetically heterogeneous.The identification of the causal mutation is important for early diagnosis, clinical follow-up, and genetic counseling. HL due to mutations in COL11A2, encoding collagen type XI alpha-2, can be non-syndromic autosomal-dominant or autosomal-recessive, and also syndromic as in Otospondylomegaepiphyseal Dysplasia, Stickler syndrome type III, and Weissenbacher-Zweymuller syndrome. However, thus far only one mutation co-segregating with autosomal recessive non-syndromic hearing loss (ARNSHL) in a single family has been reported. In this study, whole exome sequencing of two consanguineous families with ARNSHL from Tunisia and Turkey revealed two novel causative COL11A2 mutations, c.109G > T (p.Ala37Ser) and c.2662C > A (p.Pro888Thr). The variants identified co-segregated with deafness in both families. All homozygous individuals in those families had early onset profound hearing loss across all frequencies without syndromic findings. The variants are predicted to be damaging the protein function. The p.Pro888Thr mutation affects a -Gly-X-Y- triplet repeat motif. The novel p.Ala37Ser is the first missense mutation located in the NC4 domain of the COL11A2 protein. Structural model suggests that this mutation will likely obliterate, or at least partially compromise, the ability of NC4 domain to interact with its cognate ligands. In conclusion, we confirm that COL11A2 mutations cause ARNSHL and broaden the mutation spectrum that may shed new light on genotype-phenotype correlation for the associated phenotypes and clinical follow-up.


Asunto(s)
Colágeno Tipo XI/genética , Genes Recesivos , Predisposición Genética a la Enfermedad/genética , Pérdida Auditiva Sensorineural/genética , Mutación Missense , Secuencia de Aminoácidos , Secuencia de Bases , Colágeno Tipo XI/química , Consanguinidad , Exoma/genética , Salud de la Familia , Femenino , Frecuencia de los Genes , Genotipo , Pérdida Auditiva Sensorineural/patología , Humanos , Masculino , Modelos Moleculares , Datos de Secuencia Molecular , Linaje , Estructura Terciaria de Proteína , Análisis de Secuencia de ADN/métodos , Homología de Secuencia de Aminoácido
11.
Genet Res (Camb) ; 97: e4, 2015 Mar 31.
Artículo en Inglés | MEDLINE | ID: mdl-25825321

RESUMEN

Next-generation sequencing (NGS) technologies have played a central role in the genetic revolution. These technologies, especially whole-exome sequencing, have become the primary tool of geneticists to identify the causative DNA variants in Mendelian disorders, including hereditary deafness. Current research estimates that 1% of all human genes have a function in hearing. To date, mutations in over 80 genes have been reported to cause nonsyndromic hearing loss (NSHL). Strikingly, more than a quarter of all known genes related to NSHL were discovered in the past 5 years via NGS technologies. In this article, we review recent developments in the usage of NGS for hereditary deafness, with an emphasis on whole-exome sequencing.


Asunto(s)
Exoma/genética , Pérdida Auditiva/diagnóstico , Pérdida Auditiva/genética , Secuenciación de Nucleótidos de Alto Rendimiento/métodos , Sordera/diagnóstico , Sordera/genética , Predisposición Genética a la Enfermedad/genética , Humanos , Reproducibilidad de los Resultados , Sensibilidad y Especificidad
12.
Hum Genet ; 133(6): 737-42, 2014 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-24337657

RESUMEN

Megacystis-microcolon-intestinal hypoperistalsis syndrome (MMIHS) is characterized by prenatal-onset distended urinary bladder with functional intestinal obstruction, requiring extensive surgical intervention for survival. While it is believed to be an autosomal recessive disorder, most cases are sporadic. Through whole-exome sequencing in a child with MMIHS, we identified a de novo mutation, p.R178L, in the gene encoding the smooth muscle gamma-2 actin, ACTG2. We subsequently detected another de novo ACTG2 mutation, p.R178C, in an additional child with MMIHS. Actg2 transcripts were primarily found in murine urinary bladder and intestinal tissues. Structural analysis and functional experiments suggested that both ACTG2 mutants interfere with proper polymerization of ACTG2 into thin filaments, leading to impaired contractility of the smooth muscle. In conclusion, our study suggests a pathogenic mechanism for MMIHS by identifying causative ACTG2 mutations.


Asunto(s)
Anomalías Múltiples/genética , Actinas/genética , Colon/anomalías , Seudoobstrucción Intestinal/genética , Mutación , Vejiga Urinaria/anomalías , Anomalías Múltiples/metabolismo , Anomalías Múltiples/patología , Animales , Secuencia de Bases , Niño , Preescolar , Colon/metabolismo , Colon/patología , Análisis Mutacional de ADN , Exoma , Femenino , Genes Recesivos , Humanos , Seudoobstrucción Intestinal/metabolismo , Seudoobstrucción Intestinal/patología , Masculino , Ratones , Modelos Moleculares , Datos de Secuencia Molecular , Músculo Liso/metabolismo , Músculo Liso/patología , Linaje , Vejiga Urinaria/metabolismo , Vejiga Urinaria/patología
13.
Arthritis Rheum ; 65(8): 2183-9, 2013 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-23666765

RESUMEN

OBJECTIVE: Hypocomplementemic urticarial vasculitis syndrome (HUVS) is characterized by recurrent urticaria along with dermal vasculitis, arthritis, and glomerulonephritis. Systemic lupus erythematosus (SLE) develops in >50% of patients with HUVS, although the pathogenesis is unknown. The aim of this study was to identify the causative DNA mutations in 2 families with autosomal-recessive HUVS, in order to reveal the pathogenesis and facilitate the laboratory diagnosis. METHODS: Autozygosity mapping was combined with whole-exome sequencing. RESULTS: In a family with 3 affected children, we identified a homozygous frameshift mutation, c.289_290delAC, in DNASE1L3. We subsequently identified another homozygous DNASE1L3 mutation leading to exon skipping, c.320+4delAGTA, in an unrelated family. The detected mutations led to loss of function, via either nonsense-mediated messenger RNA decay or abolished endonuclease activity, as demonstrated by a plasmid nicking assay. CONCLUSION: These results show that HUVS is caused by mutations in DNASE1L3, encoding an endonuclease that previously has been associated with SLE.


Asunto(s)
Proteínas del Sistema Complemento/deficiencia , Endodesoxirribonucleasas/genética , Enfermedades del Sistema Inmune/genética , Mutación , Urticaria/genética , Vasculitis/genética , Edad de Inicio , Niño , Preescolar , Análisis Mutacional de ADN , Femenino , Genes Recesivos , Humanos , Enfermedades del Sistema Inmune/diagnóstico , Enfermedades del Sistema Inmune/inmunología , Masculino , Urticaria/diagnóstico , Urticaria/inmunología , Vasculitis/diagnóstico , Vasculitis/inmunología
14.
Ann Hum Biol ; 37(6): 778-88, 2010 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-20569042

RESUMEN

BACKGROUND: Incidence of type 1 diabetes varies widely around the world, probably due to ethnic differences across populations among other factors. AIMS: To determine whether there is an association between disease and ancestry proportions; and to control disease-HLA associations for possible confounding by admixture or population stratification. SUBJECTS AND METHODS: 100 cases and 129 controls participated in the study. Ancestry informative markers, which have considerable differences in frequency between European, West African and Native American populations were used. Type 1 diabetes associated HLA susceptibility/protection alleles were ascertained by PCR using specific primers. Statistical analyses were conducted using STRUCTURE 2.1, ADMIXMAP 3.7, SPSS 16.0 and STRAT 1.0 packages. RESULTS: The results of logistic regression implemented in ADMIXMAP 3.7 indicated that European ancestry was associated with type 1 diabetes mellitus with an odds ratio of 5.7 corresponding to one unit change in European admixture proportion. Association was found between HLA alleles and disease, DQA1*0501, *0301 DQB1*0201 and DRB1*0301, *0401 being susceptibility alleles and DRB1*1501, DQA1*0102/3 and DQB1*0602 being protective alleles. CONCLUSIONS: We found an association between European ancestry and type 1 diabetes in our sample, indicating the contribution of ethnicity to incidence differences. Previously reported associations of HLA DR/DQ alleles with disease are confirmed for the admixed Cuban population.


Asunto(s)
Diabetes Mellitus Tipo 1/genética , Diabetes Mellitus Tipo 1/inmunología , Genes MHC Clase II , Antígenos HLA-DQ/genética , Antígenos HLA-DR/genética , Adulto , Alelos , Estudios de Casos y Controles , Cuba , Diabetes Mellitus Tipo 1/epidemiología , Susceptibilidad a Enfermedades , Femenino , Frecuencia de los Genes , Marcadores Genéticos , Predisposición Genética a la Enfermedad , Genotipo , Antígenos HLA-DQ/inmunología , Antígenos HLA-DR/inmunología , Haplotipos , Humanos , Masculino , Reacción en Cadena de la Polimerasa , Polimorfismo de Nucleótido Simple , Medición de Riesgo
15.
J Mol Med (Berl) ; 96(11): 1227-1238, 2018 11.
Artículo en Inglés | MEDLINE | ID: mdl-30280293

RESUMEN

RIPOR2 (previously known as FAM65B) localizes to stereocilia of auditory hair cells and causes deafness when its function is disturbed by mutations. Here, we demonstrate that during the morphogenesis of the hair cell bundle, absence of Ripor2 affects the orientation of this key subcellular structure. We show that Ripor2 interacts with Myh9, a protein encoded by a known deafness gene. Absence of Ripor2 is associated with low Myh9 abundance in the mouse cochlea despite increased amount of Myh9 transcripts. While Myh9 is mainly expressed in stereocilia, a phosphorylated form of Myh9 is particularly enriched in the kinocilium. In Ripor2-deficient mice, kinocilium shows an aberrant localization which associates with a reduced content of phosphorylated Myh9. Acetylated alpha tubulin, another specific kinociliary protein which contributes to microtubule stabilization, is reduced in the absence of Ripor2 as well. We propose that Ripor2 deficiency influences abundance and/or post-translational modifications of proteins expressed in both stereocilia and kinocilia. This effect may have a negative impact on the structure and function of the auditory hair cell bundle.


Asunto(s)
Proteínas Portadoras/fisiología , Células Ciliadas Auditivas/fisiología , Proteínas de la Membrana/fisiología , Miosina Tipo IIA no Muscular/fisiología , Animales , Moléculas de Adhesión Celular , Cilios/fisiología , Oído Interno/fisiología , Epitelio/fisiología , Células HEK293 , Humanos , Masculino , Ratones Endogámicos C57BL , Ratones Noqueados , Cadenas Pesadas de Miosina , ARN Mensajero/metabolismo
16.
Autoimmunity ; 40(2): 103-7, 2007 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-17364501

RESUMEN

BACKGROUND: Celiac Disease (CD) is present in 1-16.4% of patients with type 1 diabetes mellitus. The most important serological markers of CD are anti-endomysial (EMA), anti-tissue transglutaminase (tTGA) and antigliadin antibodies (AGA). AIM/HYPOTHESIS: The objective of this work is to determine the frequency of tTGA and/or AGA in latent autoimmune diabetes of adult (LADA) and subjects with type 2 diabetes (T2DM), as well as to evaluate their relation with several clinical and biochemical characteristics. SUBJECTS AND METHODS: Forty three subjects with LADA and 99 with T2DM were studied. The presence of AGA, tTGA was determined in the sera of these patients. The variables: sex, age, duration of diabetes, treatment, body mass index (BMI) and fasting blood glucose concentration were also recorded. RESULTS: No differences were found in the frequency of celiac disease associated antibodies between LADA and T2DM subjects. The presence of celiac disease related antibodies was more frequent in patients with a normal or low BMI. CONCLUSIONS: Celiac disease does not seem to be related with pancreatic autoimmunity in type 2 diabetes. Celiac disease causes a decrease of body mass index in type 2 diabetes while pancreatic islet autoimmunity in this entity masks this effect.


Asunto(s)
Autoanticuerpos/inmunología , Enfermedad Celíaca/inmunología , Diabetes Mellitus Tipo 1/inmunología , Diabetes Mellitus Tipo 2/inmunología , Adulto , Anciano , Índice de Masa Corporal , Femenino , Gliadina/inmunología , Humanos , Masculino , Persona de Mediana Edad , Transglutaminasas/inmunología
19.
Autoimmunity ; 39(4): 333-40, 2006 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-16891222

RESUMEN

BACKGROUND: Nicotinamide has been used with success to prevent type 1 diabetes in animal models and humans. This vitamin B3 derivative has attracting effects on beta-cell protection and regeneration. AIM/HYPOTHESIS: To evaluate the effect of standard nicotinamide administration on type 1 diabetes prevention in first degree relatives of persons with type 1 diabetes as well as on the concentrations of islet-cell-related autoantibodies, insulin secretion and peripheral sensitivity. SUBJECTS AND METHODS: A randomized double-blind placebo controlled intervention trial was conducted in 40 first degree relatives of type 1 diabetic patients. Persistence of ICA ( >or= 10 JDF units) was among inclusion criteria. Participants were randomly allocated oral standard nicotinamide (1.2 g/m2) or placebo for 5 years. Groups were also stratified by age. Islet associated antibodies, fasting blood glucose, fasting plasma insulin concentrations, OGTT, IVGTT and HLA-DR genotyping were performed in all participants. The main criterion to stop treatment was type 1 diabetes development as defined by WHO. RESULTS: Type 1 diabetes development frequencies were similar between the treatment groups. ICA frequencies at the end of the study, first phase insulin release, and insulin sensitivity did not differ between groups as well. None of the participants suffered from any adverse events described for nicotinamide. CONCLUSIONS: Type 1 diabetes prevention trial using standard nicotinamide is feasible but fails to prevent or delay the disease onset at the dose we used.


Asunto(s)
Diabetes Mellitus Tipo 1/prevención & control , Niacinamida/uso terapéutico , Estado Prediabético/tratamiento farmacológico , Complejo Vitamínico B/uso terapéutico , Adolescente , Adulto , Autoanticuerpos/sangre , Autoanticuerpos/inmunología , Glucemia/metabolismo , Niño , Preescolar , Diabetes Mellitus Tipo 1/sangre , Diabetes Mellitus Tipo 1/genética , Diabetes Mellitus Tipo 1/inmunología , Método Doble Ciego , Femenino , Predisposición Genética a la Enfermedad , Prueba de Tolerancia a la Glucosa , Antígenos HLA-DR/sangre , Humanos , Insulina/sangre , Insulina/metabolismo , Secreción de Insulina , Masculino , Niacinamida/análogos & derivados , Niacinamida/orina , Estado Prediabético/sangre , Estado Prediabético/inmunología
20.
Diabetes ; 51(6): 1815-24, 2002 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-12031969

RESUMEN

Ca(2+) may trigger programmed cell death (apoptosis) and regulate death-specific enzymes. Therefore, the development of strategies to control Ca(2+) homeostasis may represent a potential approach to prevent or enhance cell apoptosis. To test this hypothesis, the plasma membrane Na/Ca exchanger (NCX1.7 isoform) was stably overexpressed in insulin-secreting tumoral cells. NCX1.7 overexpression increased apoptosis induced by endoplasmic reticulum (ER) Ca(2+)-ATPase inhibitors, but not by agents increasing intracellular calcium concentration ([Ca(2+)](i)), through the opening of plasma membrane Ca(2+)-channels. NCX1.7 overexpression reduced the rise in [Ca(2+)](i) induced by all agents, depleted ER Ca(2+) stores, sensitized the cells to Ca(2+)-independent proapoptotic signaling pathways, and reduced cell proliferation by approximately 40%. ER Ca(2+) stores depletion was accompanied by the activation of the ER-specific caspase (caspase-12), and the activation was enhanced by ER Ca(2+)-ATPase inhibitors. Hence, Na/Ca exchanger overexpression, by depleting ER Ca(2+) stores, triggers the activation of caspase-12 and increases apoptotic cell death. By increasing apoptosis and decreasing cell proliferation, overexpression of Na/Ca exchanger may represent a new potential approach in cancer gene therapy. On the other hand, our results open the way to the development of new strategies to control cellular Ca(2+) homeostasis that could, on the contrary, prevent the process of apoptosis that mediates, in part, beta-cell autoimmune destruction in type 1 diabetes.


Asunto(s)
Apoptosis , Caspasas/metabolismo , Retículo Endoplásmico/fisiología , Insulina/metabolismo , Islotes Pancreáticos/metabolismo , Intercambiador de Sodio-Calcio/genética , Animales , Western Blotting , Calcio/metabolismo , ATPasas Transportadoras de Calcio/antagonistas & inhibidores , Caspasa 12 , División Celular , Línea Celular , Fragmentación del ADN , Diabetes Mellitus Tipo 1/patología , Activación Enzimática , Inhibidores Enzimáticos/farmacología , Expresión Génica , Humanos , Secreción de Insulina , Islotes Pancreáticos/enzimología , Islotes Pancreáticos/patología , Microscopía Fluorescente , Ratas
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA