Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 37
Filtrar
Más filtros

Banco de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Clin Genet ; 97(1): 114-124, 2020 01.
Artículo en Inglés | MEDLINE | ID: mdl-31355911

RESUMEN

Hirschsprung disease (HSCR) is a rare congenital disorder caused by an incorrect enteric nervous system development due to a failure in migration, proliferation, differentiation and/or survival of enteric neural crest cells. HSCR is a complex genetic disease, where alterations at different molecular levels are required for the manifestation of the disease. In addition, a wide spectrum of mutations affecting many different genes cause HSCR, although the occurrence and severity of HSCR from many cases still remain unexplained. This review summarizes the current knowledge about molecular genetic basis of HSCR.


Asunto(s)
Enfermedad de Hirschsprung/genética , Enfermedades del Sistema Nervioso/genética , Sistema Nervioso/patología , Proteínas Proto-Oncogénicas c-ret/genética , Enfermedad de Hirschsprung/patología , Humanos , Mutación/genética , Sistema Nervioso/metabolismo , Enfermedades del Sistema Nervioso/patología , Cresta Neural/crecimiento & desarrollo , Cresta Neural/patología , Transducción de Señal/genética
2.
Hum Mol Genet ; 25(23): 5265-5275, 2016 12 01.
Artículo en Inglés | MEDLINE | ID: mdl-27702942

RESUMEN

Hirschsprung disease (HSCR) is the most common cause of neonatal intestinal obstruction. It is characterized by the absence of ganglia in the nerve plexuses of the lower gastrointestinal tract. So far, three common disease-susceptibility variants at the RET, SEMA3 and NRG1 loci have been detected through genome-wide association studies (GWAS) in Europeans and Asians to understand its genetic etiologies. Here we present a trans-ethnic meta-analysis of 507 HSCR cases and 1191 controls, combining all published GWAS results on HSCR to fine-map these loci and narrow down the putatively causal variants to 99% credible sets. We also demonstrate that the effects of RET and NRG1 are universal across European and Asian ancestries. In contrast, we detected a European-specific association of a low-frequency variant, rs80227144, in SEMA3 [odds ratio (OR) = 5.2, P = 4.7 × 10-10]. Conditional analyses on the lead SNPs revealed a secondary association signal, corresponding to an Asian-specific, low-frequency missense variant encoding RET p.Asp489Asn (rs9282834, conditional OR = 20.3, conditional P = 4.1 × 10-14). When in trans with the RET intron 1 enhancer risk allele, rs9282834 increases the risk of HSCR from 1.1 to 26.7. Overall, our study provides further insights into the genetic architecture of HSCR and has profound implications for future study designs.


Asunto(s)
Predisposición Genética a la Enfermedad , Enfermedad de Hirschsprung/genética , Neurregulina-1/genética , Proteínas Proto-Oncogénicas c-ret/genética , Semaforina-3A/genética , Alelos , Pueblo Asiatico/genética , Etnicidad/genética , Femenino , Estudio de Asociación del Genoma Completo , Genotipo , Enfermedad de Hirschsprung/patología , Humanos , Intrones/genética , Masculino , Polimorfismo de Nucleótido Simple , Población Blanca/genética
3.
Am J Hum Genet ; 96(4): 581-96, 2015 Apr 02.
Artículo en Inglés | MEDLINE | ID: mdl-25839327

RESUMEN

Innervation of the gut is segmentally lost in Hirschsprung disease (HSCR), a consequence of cell-autonomous and non-autonomous defects in enteric neuronal cell differentiation, proliferation, migration, or survival. Rare, high-penetrance coding variants and common, low-penetrance non-coding variants in 13 genes are known to underlie HSCR risk, with the most frequent variants in the ret proto-oncogene (RET). We used a genome-wide association (220 trios) and replication (429 trios) study to reveal a second non-coding variant distal to RET and a non-coding allele on chromosome 7 within the class 3 Semaphorin gene cluster. Analysis in Ret wild-type and Ret-null mice demonstrates specific expression of Sema3a, Sema3c, and Sema3d in the enteric nervous system (ENS). In zebrafish embryos, sema3 knockdowns show reduction of migratory ENS precursors with complete ablation under conjoint ret loss of function. Seven candidate receptors of Sema3 proteins are also expressed within the mouse ENS and their expression is also lost in the ENS of Ret-null embryos. Sequencing of SEMA3A, SEMA3C, and SEMA3D in 254 HSCR-affected subjects followed by in silico protein structure modeling and functional analyses identified five disease-associated alleles with loss-of-function defects in semaphorin dimerization and binding to their cognate neuropilin and plexin receptors. Thus, semaphorin 3C/3D signaling is an evolutionarily conserved regulator of ENS development whose dys-regulation is a cause of enteric aganglionosis.


Asunto(s)
Epistasis Genética/genética , Predisposición Genética a la Enfermedad/genética , Variación Genética , Enfermedad de Hirschsprung/genética , Proteínas Proto-Oncogénicas c-ret/genética , Semaforinas/genética , Animales , Secuencia de Bases , Estudio de Asociación del Genoma Completo , Ratones , Datos de Secuencia Molecular , Semaforinas/deficiencia , Semaforinas/metabolismo , Análisis de Secuencia de ADN
4.
Am J Med Genet A ; 170A(5): 1268-73, 2016 May.
Artículo en Inglés | MEDLINE | ID: mdl-26762557

RESUMEN

Monosomy 10p is a rare chromosomal disorder with a prevalence <1/1,000,000, in which a terminal or interstitial distal region of chromosome 10 is deleted resulting in a variable phenotype depending on the size of the deletion. Two main phenotypes have been defined depending on the location of the deletion: HDR syndrome (Hypoparathyroidism, sensorineural Deafness, and Renal disease), and DGS2 (DiGeorge syndrome type 2). The vast majority of cases reported so far have resulted from de novo events. Here, we present the first familial presentation of this contiguous gene deletion syndrome, affecting two family members in different generations: a child and his maternal uncle. In both cases, the deletion was due to a malsegregation of a maternal balanced rearrangement, ins(16;10)(q22;p13p15.2). The identification and characterization of this rearrangement was possible using a combination of different genetic analyses such as karyotype, MLPA, FISH, and array CGH. We underline the importance of the present results in terms of genetic and reproductive counseling for the carriers of the balanced rearrangement within the family, and demonstrate again the utility of expanding the genetic studies to the relatives of the affected patients.


Asunto(s)
Trastornos de los Cromosomas/genética , Cromosomas Humanos Par 10/genética , Síndrome de DiGeorge/genética , Pérdida Auditiva Sensorineural/genética , Hipoparatiroidismo/genética , Nefrosis/genética , Adulto , Niño , Deleción Cromosómica , Trastornos de los Cromosomas/fisiopatología , Hibridación Genómica Comparativa , Discapacidades del Desarrollo/genética , Discapacidades del Desarrollo/fisiopatología , Síndrome de DiGeorge/fisiopatología , Eliminación de Gen , Pérdida Auditiva Sensorineural/fisiopatología , Humanos , Hipoparatiroidismo/fisiopatología , Hibridación Fluorescente in Situ , Cariotipificación , Masculino , Nefrosis/fisiopatología
5.
Dev Biol ; 382(1): 320-9, 2013 Oct 01.
Artículo en Inglés | MEDLINE | ID: mdl-23707863

RESUMEN

Finding genes for complex diseases has been the goal of many genetic studies. Most of these studies have been successful by searching for genes and mutations in rare familial cases, by screening candidate genes and by performing genome wide association studies. However, only a small fraction of the total genetic risk for these complex genetic diseases can be explained by the identified mutations and associated genetic loci. In this review we focus on Hirschsprung disease (HSCR) as an example of a complex genetic disorder. We describe the genes identified in this congenital malformation and postulate that both common 'low penetrant' variants in combination with rare or private 'high penetrant' variants determine the risk on HSCR, and likely, on other complex diseases. We also discuss how new technological advances can be used to gain further insights in the genetic background of complex diseases. Finally, we outline a few steps to develop functional assays in order to determine the involvement of these variants in disease development.


Asunto(s)
Variación Genética , Enfermedad de Hirschsprung/genética , Modelos Biológicos , Animales , Estudios de Asociación Genética , Predisposición Genética a la Enfermedad , Enfermedad de Hirschsprung/patología , Humanos
6.
Am J Med Genet A ; 164A(2): 542-7, 2014 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-24311220

RESUMEN

Shah-Waardenburg syndrome or Waardenburg syndrome type 4 (WS4) is a neurocristopathy characterized by the association of deafness, depigmentation and Hirschsprung disease. Three disease-causing genes have been identified so far for WS4: EDNRB, EDN3, and SOX10. SOX10 mutations, found in 45-55% of WS4 patients, are inherited in autosomal dominant way. In addition, mutations in SOX10 are also responsible for an extended syndrome involving peripheral and central neurological phenotypes, referred to as PCWH (peripheral demyelinating neuropathy, central dysmyelinating leucodystrophy, Waardenburg syndrome, Hirschsprung disease). Such mutations are mostly private, and a high intra- and inter-familial variability exists. In this report, we present a patient with WS4 and a second with PCWH due to SOX10 mutations supporting again the genetic and phenotypic heterogeneity of these syndromes. Interestingly, the WS4 family carries an insertion of 19 nucleotides in exon 5 of SOX10, which results in distinct phenotypes along three different generations: hypopigmentation in the maternal grandmother, hearing loss in the mother, and WS4 in the proband. Since mosaicism cannot explain the three different related-WS features observed in this family, we propose as the most plausible explanation the existence of additional molecular events, acting in an additive or multiplicative fashion, in genes or regulatory regions unidentified so far. On the other hand, the PCWH case was due to a de novo deletion in exon 5 of the gene. Efforts should be devoted to unravel the mechanisms underlying the intrafamilial phenotypic variability observed in the families affected, and to identify new genes responsible for the still unsolved WS4 cases.


Asunto(s)
Mutación , Factores de Transcripción SOXE/genética , Síndrome de Waardenburg/diagnóstico , Síndrome de Waardenburg/genética , Secuencia de Aminoácidos , Sustitución de Aminoácidos , Preescolar , Análisis Mutacional de ADN , Exones , Orden Génico , Enfermedad de Hirschsprung , Humanos , Masculino , Datos de Secuencia Molecular , Fenotipo , España
7.
Am J Hum Genet ; 87(1): 60-74, 2010 Jul 09.
Artículo en Inglés | MEDLINE | ID: mdl-20598273

RESUMEN

The major gene for Hirschsprung disease (HSCR) encodes the receptor tyrosine kinase RET. In a study of 690 European- and 192 Chinese-descent probands and their parents or controls, we demonstrate the ubiquity of a >4-fold susceptibility from a C-->T allele (rs2435357: p = 3.9 x 10(-43) in European ancestry; p = 1.1 x 10(-21) in Chinese samples) that probably arose once within the intronic RET enhancer MCS+9.7. With in vitro assays, we now show that the T variant disrupts a SOX10 binding site within MCS+9.7 that compromises RET transactivation. The T allele, with a control frequency of 20%-30%/47% and case frequency of 54%-62%/88% in European/Chinese-ancestry individuals, is involved in all forms of HSCR. It is marginally associated with proband gender (p = 0.13) and significantly so with length of aganglionosis (p = 7.6 x 10(-5)) and familiality (p = 6.2 x 10(-4)). The enhancer variant is more frequent in the common forms of male, short-segment, and simplex families whereas multiple, rare, coding mutations are the norm in the less common and more severe forms of female, long-segment, and multiplex families. The T variant also increases penetrance in patients with rare RET coding mutations. Thus, both rare and common mutations, individually and together, make contributions to the risk of HSCR. The distribution of RET variants in diverse HSCR patients suggests a "cellular-recessive" genetic model where both RET alleles' function is compromised. The RET allelic series, and its genotype-phenotype correlations, shows that success in variant identification in complex disorders may strongly depend on which patients are studied.


Asunto(s)
Enfermedad de Hirschsprung/genética , Proteínas Proto-Oncogénicas c-ret/genética , Pueblo Asiatico , Secuencia de Bases , Estudios de Casos y Controles , Elementos de Facilitación Genéticos , Femenino , Frecuencia de los Genes , Estudio de Asociación del Genoma Completo , Haplotipos , Enfermedad de Hirschsprung/etnología , Enfermedad de Hirschsprung/fisiopatología , Humanos , Masculino , Mutación , Penetrancia , Polimorfismo de Nucleótido Simple , Unión Proteica , Proteínas Proto-Oncogénicas c-ret/metabolismo , Factores de Transcripción SOXE/metabolismo , Factores Sexuales , Activación Transcripcional , Población Blanca
8.
Am J Med Genet A ; 158A(4): 816-20, 2012 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-22344793

RESUMEN

X-linked hydrocephalus (XLH) has an incidence of 1/30,000 male births and is characterized by intellectual disability, spastic paraplegia, adducted thumbs, and agenesis of corpus callosum, and/or corticospinal tract. The great proportion of cases is ascribed to loss of function mutations of L1CAM gene. Hirschsprung disease (HSCR) is characterized by the absence of ganglion cells in the submucosal and myenteric plexuses along a variable portion of the intestinal tract and has incidence of about 1/5,000. Although with several genes involved in its pathogenesis, the major HSCR gene is the RET proto-oncogene. To date only a few patients have been reported with both phenotypes and mutations in the L1CAM gene. In this report, we describe a new patient with concurrent XLH and HSCR. L1CAM mutational screening showed the presence of the G698R hemizygous mutation, which is a deleterious substitution affecting a key residue necessary for the correct folding of the protein. Moreover, the patient also carried the transcriptional enhancer RET mutation (c.73 + 9277T > C) in heterozygosis. We speculate that both the RET enhancer variant, and the L1CAM mutation may act in combination to produce the enteric phenotype, probably with the participation of other still unidentified molecular events. While it is obvious that additional studies are necessary to further delineate the association between XLH and HSCR in the presence of L1CAM mutations, the documentation of this new patient reinforces the role of this gene acting either in a direct or indirect way in the pathogenesis of Hirschsprung disease.


Asunto(s)
Enfermedades Genéticas Ligadas al Cromosoma X/genética , Enfermedad de Hirschsprung/genética , Hidrocefalia/genética , Molécula L1 de Adhesión de Célula Nerviosa/genética , Anomalías Múltiples/genética , Secuencia de Bases , Acueducto del Mesencéfalo/anomalías , Acueducto del Mesencéfalo/patología , Cuerpo Calloso/patología , Análisis Mutacional de ADN , Enfermedades Genéticas Ligadas al Cromosoma X/diagnóstico , Enfermedades Genéticas Ligadas al Cromosoma X/patología , Enfermedad de Hirschsprung/diagnóstico , Enfermedad de Hirschsprung/patología , Humanos , Hidrocefalia/diagnóstico , Hidrocefalia/patología , Lactante , Masculino , Proto-Oncogenes Mas , Análisis de Secuencia de ADN
9.
J Pediatr Endocrinol Metab ; 35(10): 1306-1308, 2022 Oct 26.
Artículo en Inglés | MEDLINE | ID: mdl-35932482

RESUMEN

OBJECTIVES: Congenital hypogonadotropic hypogonadism combined with anosmia or hyposmia is considered Kallmann syndrome (KS). It is often accompanied by bone defects. CASE PRESENTATION: Here, we report a girl and her mother with KS caused by a novel mutation in the fibroblast growth factor receptor 1 gene (FGFR1). Interestingly, the daughter presented syndactyly and oligodactyly of the feet. CONCLUSIONS: The presence of bone malformations in a KS patient should direct the geneticist towards a search for specific mutations in FGFR1. Our finding contributes to enrich the spectrum of FGFR1 mutations in patients with KS.


Asunto(s)
Hipogonadismo , Síndrome de Kallmann , Femenino , Humanos , Hipogonadismo/genética , Síndrome de Kallmann/genética , Madres , Mutación , Núcleo Familiar , Receptor Tipo 1 de Factor de Crecimiento de Fibroblastos/genética , Receptor Tipo 1 de Factor de Crecimiento de Fibroblastos/metabolismo
10.
BMC Med Genet ; 12: 138, 2011 Oct 13.
Artículo en Inglés | MEDLINE | ID: mdl-21995290

RESUMEN

BACKGROUND: RET is the major gene associated to Hirschsprung disease (HSCR) with differential contributions of its rare and common, coding and noncoding mutations to the multifactorial nature of this pathology. In the present study, we have performed a comprehensive study of our HSCR series evaluating the involvement of both RET rare variants (RVs) and common variants (CVs) in the context of the disease. METHODS: RET mutational screening was performed by dHPLC and direct sequencing for the identification of RVs. In addition Taqman technology was applied for the genotyping of 3 RET CVs previously associated to HSCR, including a variant lying in an enhancer domain within RET intron 1 (rs2435357). Statistical analyses were performed using the SPSS v.17.0 to analyze the distribution of the variants. RESULTS: Our results confirm the strongest association to HSCR for the "enhancer" variant, and demonstrate a significantly higher impact of it in male versus female patients. Integration of the RET RVs and CVs analysis showed that in 91.66% of cases with both kinds of mutational events, the enhancer allele is in trans with the allele bearing the RET RV. CONCLUSIONS: A gender effect exists on both the transmission and distribution of rare coding and common HSCR causing mutations. In addition, these RET CVs and RVs seem to act in a synergistic way leading to HSCR phenotype.


Asunto(s)
Enfermedad de Hirschsprung/genética , Mutación , Proteínas Proto-Oncogénicas c-ret/genética , Alelos , Estudios de Casos y Controles , Análisis Mutacional de ADN , Elementos de Facilitación Genéticos , Exones , Femenino , Frecuencia de los Genes , Estudios de Asociación Genética , Prueba de Complementación Genética , Variación Genética , Mutación de Línea Germinal , Humanos , Intrones , Masculino , Polimorfismo de Nucleótido Simple , Caracteres Sexuales , España
11.
BMC Med Genet ; 11: 71, 2010 May 11.
Artículo en Inglés | MEDLINE | ID: mdl-20459765

RESUMEN

BACKGROUND: Hirschsprung disease is characterized by the absence of intramural ganglion cells in the enteric plexuses, due to a fail during enteric nervous system formation. Hirschsprung has a complex genetic aetiology and mutations in several genes have been related to the disease. There is a clear predominance of missense/nonsense mutations in these genes whereas copy number variations (CNVs) have been seldom described, probably due to the limitations of conventional techniques usually employed for mutational analysis. In this study, we have looked for CNVs in some of the genes related to Hirschsprung (EDNRB, GFRA1, NRTN and PHOX2B) using the Multiple Ligation-dependent Probe Amplification (MLPA) approach. METHODS: CNVs screening was performed in 208 HSCR patients using a self-designed set of MLPA probes, covering the coding region of those genes. RESULTS: A deletion comprising the first 4 exons in GFRA1 gene was detected in 2 sporadic HSCR patients and in silico approaches have shown that the critical translation initiation signal in the mutant gene was abolished. In this study, we have been able to validate the reliability of this technique for CNVs screening in HSCR. CONCLUSIONS: The implemented MLPA based technique presented here allows CNV analysis of genes involved in HSCR that have not been not previously evaluated. Our results indicate that CNVs could be implicated in the pathogenesis of HSCR, although they seem to be an uncommon molecular cause of HSCR.


Asunto(s)
Variaciones en el Número de Copia de ADN , Enfermedad de Hirschsprung/genética , Técnicas de Amplificación de Ácido Nucleico , Sondas de ADN , Femenino , Humanos , Masculino
12.
BMC Med Genet ; 11: 137, 2010 Sep 22.
Artículo en Inglés | MEDLINE | ID: mdl-20860806

RESUMEN

BACKGROUND: Hirschsprung disease (HSCR) is a neurocristopathy characterized by the absence of parasympathetic intrinsic ganglion cells in the submucosal and myenteric plexuses along a variable portion of the intestinal tract. In approximately 18% of the cases HSCR also presents with multiple congenital anomalies including recognized syndromes. METHODS: A combination of MLPA and microarray data analysis have been undertaken to refine a duplication at the Xq28 region. RESULTS: In this study we present a new clinical association of severe neonatal encephalopathy (Lubs syndrome) and HSCR, in a male patient carrying a duplication at the Xq28 region which encompasses the MECP2 and L1CAM genes. CONCLUSIONS: While the encephalopathy has been traditionally attributed to the MECP2 gene duplication in patients with Lubs syndrome, here we propose that the enteric phenotype in our patient might be due to the dosage variation of the L1CAM protein, together with additional molecular events not identified yet. This would be in agreement with the hypothesis previously forwarded that mutations in L1CAM may be involved in HSCR development in association with a predisposing genetic background.


Asunto(s)
Encefalopatías/genética , Cromosomas Humanos X , Duplicación de Gen , Enfermedad de Hirschsprung/genética , Encefalopatías/complicaciones , Encefalopatías/congénito , Análisis Mutacional de ADN , Estudio de Asociación del Genoma Completo , Enfermedad de Hirschsprung/complicaciones , Humanos , Recién Nacido , Masculino , Linaje , Índice de Severidad de la Enfermedad , Síndrome
13.
BMC Med Genet ; 10: 119, 2009 Nov 19.
Artículo en Inglés | MEDLINE | ID: mdl-19925665

RESUMEN

BACKGROUND: Hirschsprung disease (HSCR) is a congenital malformation of the hindgut produced by a disruption in neural crest cell migration during embryonic development. HSCR has a complex genetic etiology and mutations in several genes, mainly the RET proto-oncogene, have been related to the disease. There is a clear predominance of missense/nonsense mutations in these genes whereas copy number variations (CNVs) have been seldom described, probably due to the limitations of conventional techniques usually employed for mutational analysis. METHODS: In this study we have aimed to analyze the presence of CNVs in some HSCR genes (RET, EDN3, GDNF and ZFHX1B) using the Multiple Ligation-dependent Probe Amplification (MLPA) approach. RESULTS: Two alterations in the MLPA profiles of RET and EDN3 were detected, but a detailed inspection showed that the decrease in the corresponding dosages were due to point mutations affecting the hybridization probes regions. CONCLUSION: Our results indicate that CNVs of the gene coding regions analyzed here are not a common molecular cause of Hirschsprung disease. However, further studies are required to determine the presence of CNVs affecting non-coding regulatory regions, as well as other candidate genes.


Asunto(s)
Variaciones en el Número de Copia de ADN/genética , Enfermedad de Hirschsprung/genética , Técnicas de Amplificación de Ácido Nucleico/métodos , Endotelina-3/genética , Femenino , Humanos , Masculino , Mutación , Proto-Oncogenes Mas , Proteínas Proto-Oncogénicas c-ret/genética , Secuencias Reguladoras de Ácidos Nucleicos , España
14.
Neuromuscul Disord ; 28(3): 208-215, 2018 03.
Artículo en Inglés | MEDLINE | ID: mdl-29433793

RESUMEN

Spinal muscular atrophy (SMA) is a neuromuscular disorder caused by loss or mutations in SMN1. According to age of onset, achieved motor abilities, and life span, SMA patients are classified into type I (never sit), II (never walk unaided) or III (achieve independent walking abilities). SMN2, the highly homologous copy of SMN1, is considered the most important phenotypic modifier of the disease. Determination of SMN2 copy number is essential to establish careful genotype-phenotype correlations, predict disease evolution, and to stratify patients for clinical trials. We have determined SMN2 copy numbers in 625 unrelated Spanish SMA patients with loss or mutation of both copies of SMN1 and a clear assignation of the SMA type by clinical criteria. Furthermore, we compiled data from relevant worldwide reports that link SMN2 copy number with SMA severity published from 1999 to date (2834 patients with different ethnic and geographic backgrounds). Altogether, we have assembled a database with a total of 3459 patients to delineate more universal prognostic rules regarding the influence of SMN2 copy number on SMA phenotype. This issue is crucial in the present scenario of therapeutic advances with the perspective of SMA neonatal screening and early diagnosis to initiate treatments.


Asunto(s)
Variaciones en el Número de Copia de ADN , Estudios de Asociación Genética , Atrofia Muscular Espinal/genética , Bases de Datos Genéticas , Femenino , Dosificación de Gen , Predisposición Genética a la Enfermedad , Genotipo , Humanos , Masculino , Mutación , Fenotipo , Pronóstico , España , Proteína 2 para la Supervivencia de la Neurona Motora/genética
15.
Pediatr Dev Pathol ; 20(1): 28-37, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-28276298

RESUMEN

Objectives Hirschsprung disease (HSCR) has a wide range of severity. There are nonsevere forms treated conservatively until surgery and severe forms that require an early stoma and prolonged hospitalization. Our objective was to establish a clinical scoring system to predict the severity of HSCR and to evaluate the possible existence of a clinical-genetic correlation. Methods We carried out a retrospective observational study including all HSCR cases treated in our hospital. The sample was divided into severe and nonsevere disease according to the number of surgical procedures, hospitalization time, and episodes of enterocolitis. The proposed score was applied at diagnosis, and the sensitivity, specificity, and optimal cut-point were determined. We conducted a prospective molecular study of RET, EDNRB, and EDN3 on all patients, as well as SOX10 in Waardenburg Syndrome type 4 forms. Results Among the 42 patients treated between 1983 and 2013, 15 met the severe disease criteria. This group had a higher mean score (13.15 ± 2.36) than the nonsevere group (8.15 ± 2.13; p < 0.001). A score ≥11 had a sensitivity of 87% and a specificity of 81% in detecting the severe cases. Causative mutations were identified in 12 patients, 8 of them in the severe group ( p = 0.015). Most of these mutations (75%) were located in the RET proto-oncogene. Conclusion The proposed scoring system enables the early selection of patients with severe behavior of HSCR. A value ≥11 showed good sensitivity and specificity for this purpose. Causative mutations were identified in more than 50% of patients who met the criteria for severe disease.


Asunto(s)
Estudios de Asociación Genética , Marcadores Genéticos , Enfermedad de Hirschsprung/diagnóstico , Mutación , Índice de Severidad de la Enfermedad , Adolescente , Niño , Preescolar , Femenino , Estudios de Seguimiento , Enfermedad de Hirschsprung/genética , Humanos , Lactante , Recién Nacido , Modelos Logísticos , Masculino , Proto-Oncogenes Mas , Estudios Retrospectivos , Sensibilidad y Especificidad
16.
Thyroid ; 16(4): 411-7, 2006 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-16646689

RESUMEN

CONTEXT: Little is known about the etiology of sporadic medullary thyroid carcinoma (sMTC). While germline gain-of-function mutations in the RET proto-oncogene cause hereditary MTC, the molecular mechanisms leading to the sporadic forms remain obscure. Our group had evidence about the existence of a low-penetrance susceptibility locus for sMTC in linkage disequilibrium with RET variants S836S/IVS1-126G>T, and probably in 5' with respect to both variants. In this study we sought to identify such locus. On the other hand, because an overrepresentation of G691S/S904S variants in patients with sMTC had been previously reported, we sought to determine if such association was present in our series. DESIGN: We performed a case-control study analysing a wide spectrum of RET variants in the 5' region of the gene, as well as the variants G691S/S904S. Haplotype distribution were also analyzed. A total of 58 patients with sMTC were included in the study. In addition, 100 unselected, unrelated race-, age-, and gender-matched normal controls were also evaluated. MAIN OUTCOME: Although the overrepresentation of IVS1-126G>T remains present in our current sMTC series, thus supporting our previous hypothesis, no differences were obtained among cases and controls in the distribution of the variants tested upstream this position. On the other hand, the frequency and distribution of G691S/S904S variants were similar in both groups of study, leading to exclude their role in sMTC in our series. CONCLUSIONS: These findings would suggest that the major genetic events contributing to the appearance of sMTC may reside in several different RET loci. In this way, we could hypothesize about the existence of at least two sMTC loci, linked to S836S-IVS1-126G>T, or to G691S-S904S, respectively.


Asunto(s)
Carcinoma Medular/genética , Proteínas Proto-Oncogénicas c-ret/genética , Neoplasias de la Tiroides/genética , Transferencia Resonante de Energía de Fluorescencia , Haplotipos , Humanos , Polimorfismo de Nucleótido Simple , Proto-Oncogenes Mas
17.
Clin Case Rep ; 4(9): 879-84, 2016 09.
Artículo en Inglés | MEDLINE | ID: mdl-27648268

RESUMEN

Despite co-segregation of two different genetic neurological disorders within a family is rare, clinicians should take into consideration this possibility in patients presenting with unusual complex phenotypes or with unexpected electrophysiological findings. Here, we report a Spanish 11-month-old patient with spinal muscular atrophy type 2 and Charcot-Marie-Tooth 1A.

18.
Biomed Res Int ; 2015: 965839, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-26713318

RESUMEN

Fragile X syndrome (FXS) accounts for about one-half of cases of X-linked intellectual disability and is the most common monogenic cause of mental impairment. Reproductive options for the FXS carriers include preimplantation genetic diagnosis (PGD). However, this strategy is considered by some centers as wasteful owing to the high prevalence of premature ovarian failure in FXS carriers and the difficulties in genetic diagnosis of the embryos. Here we present the results of our PGD Program applied to FXS, at the Department of Genetics, Reproduction and Fetal Medicine of the University Hospital Virgen del Rocío in Seville. A total of 11 couples have participated in our PGD Program for FXS since 2010. Overall, 15 cycles were performed, providing a total of 43 embryos. The overall percentage of transfers per cycle was 46.67% and the live birth rate per cycle was 13.33%. As expected, these percentages are considerably lower than the ones obtained in PGD for other pathologies. Our program resulted in the birth of 3 unaffected babies of FXS for 2 of the 11 couples (18.2%) supporting that, despite the important drawbacks of PGD for FXS, efforts should be devoted in offering this reproductive option to the affected families.


Asunto(s)
Síndrome del Cromosoma X Frágil/diagnóstico , Pruebas Genéticas , Diagnóstico Preimplantación , Insuficiencia Ovárica Primaria/diagnóstico , Adulto , Tasa de Natalidad , Transferencia de Embrión , Femenino , Síndrome del Cromosoma X Frágil/genética , Síndrome del Cromosoma X Frágil/patología , Heterocigoto , Hospitales Universitarios , Humanos , Embarazo , Insuficiencia Ovárica Primaria/genética , Insuficiencia Ovárica Primaria/patología , España
19.
Biomed Res Int ; 2015: 406096, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-26258137

RESUMEN

Hemophilia A and B are the most common hereditary hemorrhagic disorders, with an X-linked mode of inheritance. Reproductive options for the families affected with hemophilia, aiming at the prevention of the birth of children with severe coagulation disorders, include preimplantation genetic diagnosis (PGD). Here we present the results of our PGD Program applied to hemophilia, at the Department of Genetics, Reproduction and Fetal Medicine of the University Hospital Virgen del Rocío in Seville. A total of 34 couples have been included in our program since 2005 (30 for hemophilia A and 4 for hemophilia B). Overall, 60 cycles were performed, providing a total of 508 embryos. The overall percentage of transfers per cycle was 81.7% and the live birth rate per cycle ranged from 10.3 to 24.1% depending on the methodological approach applied. Although PGD for hemophilia can be focused on gender selection of female embryos, our results demonstrate that methodological approaches that allow the diagnosis of the hemophilia status of every embryo have notorious advantages. Our PGD Program resulted in the birth of 12 healthy babies for 10 out of the 34 couples (29.4%), constituting a relevant achievement for the Spanish Public Health System within the field of haematological disorders.


Asunto(s)
Pruebas Genéticas/métodos , Hemofilia A/diagnóstico , Hemofilia A/genética , Hemofilia B/diagnóstico , Hemofilia B/genética , Hospitales Universitarios , Diagnóstico Preimplantación/métodos , Adulto , Embrión de Mamíferos/fisiología , Femenino , Humanos , Hibridación Fluorescente in Situ , Masculino , Repeticiones de Microsatélite/genética , Reacción en Cadena de la Polimerasa , Embarazo , España
20.
BMC Med Genomics ; 8: 83, 2015 Dec 21.
Artículo en Inglés | MEDLINE | ID: mdl-26690675

RESUMEN

BACKGROUND: The molecular mechanisms leading to sporadic medullary thyroid carcinoma (sMTC) and juvenile papillary thyroid carcinoma (PTC), two rare tumours of the thyroid gland, remain poorly understood. Genetic studies on thyroid carcinomas have been conducted, although just a few loci have been systematically associated. Given the difficulties to obtain single-loci associations, this work expands its scope to the study of epistatic interactions that could help to understand the genetic architecture of complex diseases and explain new heritable components of genetic risk. METHODS: We carried out the first screening for epistasis by Multifactor-Dimensionality Reduction (MDR) in genome-wide association study (GWAS) on sMTC and juvenile PTC, to identify the potential simultaneous involvement of pairs of variants in the disease. RESULTS: We have identified two significant epistatic gene interactions in sMTC (CHFR-AC016582.2 and C8orf37-RNU1-55P) and three in juvenile PTC (RP11-648k4.2-DIO1, RP11-648k4.2-DMGDH and RP11-648k4.2-LOXL1). Interestingly, each interacting gene pair included a non-coding RNA, providing thus support to the relevance that these elements are increasingly gaining to explain carcinoma development and progression. CONCLUSIONS: Overall, this study contributes to the understanding of the genetic basis of thyroid carcinoma susceptibility in two different case scenarios such as sMTC and juvenile PTC.


Asunto(s)
Carcinoma Neuroendocrino/genética , Carcinoma/genética , Epistasis Genética , Estudio de Asociación del Genoma Completo , Neoplasias de la Tiroides/genética , Adolescente , Carcinoma Papilar , Estudios de Casos y Controles , Niño , Preescolar , Femenino , Predisposición Genética a la Enfermedad/genética , Humanos , Masculino , Polimorfismo de Nucleótido Simple , Cáncer Papilar Tiroideo , Adulto Joven
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA