Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 80
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
Clin Genet ; 98(3): 293-298, 2020 09.
Artículo en Inglés | MEDLINE | ID: mdl-32613604

RESUMEN

Primary ovarian insufficiency (POI) implies the cessation of menstruation for several months in women before the age of 40 years and is a major cause of infertility. The study of the contribution of genetic factors to POI has been fueled by the use of whole exome sequencing (WES). Here, to uncover novel causative pathogenic variants and risk alleles, WES has been performed in 12 patients with familial POI (eight unrelated index cases and two pairs of sisters) and six women with early menopause and family history of POI (four index cases and one pair of sisters). Likely causative variants in NR5A1 and MCM9 genes were identified as well as a variant in INHA that requires further investigation. Moreover, we have identified more than one candidate variant in 3 out of 15 familial cases. Taken together, our results highlight the genetic heterogeneity of POI and early menopause and support the hypothesis of an oligogenic inheritance of such conditions, in addition to monogenic inheritance.


Asunto(s)
Inhibinas/genética , Proteínas de Mantenimiento de Minicromosoma/genética , Insuficiencia Ovárica Primaria/genética , Factor Esteroidogénico 1/genética , Adolescente , Adulto , Alelos , Exoma/genética , Femenino , Predisposición Genética a la Enfermedad , Humanos , Insuficiencia Ovárica Primaria/patología , Factores de Riesgo , Secuenciación del Exoma , Adulto Joven
2.
Clin Genet ; 98(4): 379-383, 2020 10.
Artículo en Inglés | MEDLINE | ID: mdl-32632923

RESUMEN

Chromosomal microarray analysis (CMA) has now replaced karyotyping in the analysis of prenatal cases with a fetal structural anomaly, whereas in those pregnancies undergoing invasive prenatal diagnosis with a normal fetal ultrasound, conventional karyotyping is still performed. The aims of this study were to establish the diagnostic yield of CMA in prenatal diagnosis, and to provide new data that might contribute to reconsider current practices. We reviewed 2905 prenatal samples with a normal rapid aneuploidy detection test referred for evaluation by CMA testing. Our study revealed pathogenic and reported susceptibility copy number variants associated with syndromic disorders in 4.8% (n = 138/2905) of cases, being 2.8% (n = 81/2905) the estimated added diagnostic value of CMA over karyotyping. Clinically significant CMA abnormality was detected in 5.4% (107/1975) of the fetuses with ultrasound anomalies and in 1.4% (5/345) of those considered as low-risk pregnancies. Our series shows that in prenatal samples, CMA increases 2-fold the diagnostic yield achieved by conventional karyotyping.


Asunto(s)
Cromosomas/genética , Enfermedades Genéticas Congénitas/genética , Pruebas Genéticas , Diagnóstico Prenatal , Aneuploidia , Variaciones en el Número de Copia de ADN/genética , Femenino , Enfermedades Genéticas Congénitas/diagnóstico , Enfermedades Genéticas Congénitas/patología , Humanos , Análisis por Micromatrices/tendencias , Embarazo , Síndrome
3.
J Med Genet ; 56(7): 481-490, 2019 07.
Artículo en Inglés | MEDLINE | ID: mdl-30894412

RESUMEN

BACKGROUND: Mapping the genetic component of molecular mechanisms responsible for the reduced penetrance (RP) of rare disorders constitutes one of the most challenging problems in human genetics. Heritable pulmonary arterial hypertension (PAH) is one such disorder characterised by rare mutations mostly occurring in the bone morphogenetic protein receptor type 2 (BMPR2) gene and a wide heterogeneity of penetrance modifier mechanisms. Here, we analyse 32 genotyped individuals from a large Iberian family of 65 members, including 22 carriers of the pathogenic BMPR2 mutation c.1472G>A (p.Arg491Gln), 8 of them diagnosed with PAH by right-heart catheterisation, leading to an RP rate of 36.4%. METHODS: We performed a linkage analysis on the genotyping data to search for genetic modifiers of penetrance. Using functional genomics data, we characterised the candidate region identified by linkage analysis. We also predicted the haplotype segregation within the family. RESULTS: We identified a candidate chromosome region in 2q24.3, 38 Mb upstream from BMPR2, with significant linkage (LOD=4.09) under a PAH susceptibility model. This region contains common variants associated with vascular aetiology and shows functional evidence that the putative genetic modifier is located in the upstream distal promoter of the fidgetin (FIGN) gene. CONCLUSION: Our results suggest that the genetic modifier acts through FIGN transcriptional regulation, whose expression variability would contribute to modulating heritable PAH. This finding may help to advance our understanding of RP in PAH across families sharing the p.Arg491Gln pathogenic mutation in BMPR2.


Asunto(s)
ATPasas Asociadas con Actividades Celulares Diversas/genética , Hipertensión Pulmonar Primaria Familiar/diagnóstico , Hipertensión Pulmonar Primaria Familiar/genética , Ligamiento Genético , Predisposición Genética a la Enfermedad , Proteínas Asociadas a Microtúbulos/genética , Penetrancia , Alelos , Sustitución de Aminoácidos , Presión Sanguínea , Cromosomas Humanos Par 2 , Familia , Estudios de Asociación Genética , Estudio de Asociación del Genoma Completo , Genotipo , Hemodinámica , Heterocigoto , Humanos , Desequilibrio de Ligamiento , Mutación , Linaje , Fenotipo , Polimorfismo de Nucleótido Simple
4.
Int J Cancer ; 144(3): 513-524, 2019 02 01.
Artículo en Inglés | MEDLINE | ID: mdl-30350313

RESUMEN

Somatically acquired uniparental disomies (aUPDs) are frequent events in solid tumors and have been associated with cancer-related genes. Studies assessing their functional consequences across several cancer types are therefore necessary. Here, we aimed at integrating aUPD profiles with the mutational status of cancer-related genes in a tumor-type specific manner. Using TCGA datasets for 1,032 gastrointestinal cancers, including colon (COAD), rectum (READ), stomach (STAD), esophageal adenocarcinoma (EAC) and esophageal squamous cell carcinoma (ESCC), we show a non-random distribution of aUPD, suggesting the existence of a cancer-specific landscape of aUPD events. Our analysis indicates that aUPD acts as a "second hit" in Knudson's model in order to achieve biallelic inactivation of tumor suppressor genes. In particular, APC, ARID1A and NOTCH1 were recurrently inactivated by the presence of homozygous mutation as a consequence of aUPD in COAD and READ, STAD and ESCC, respectively. Furthermore, while TP53 showed inactivation caused by aUPD at chromosome arm 17p across all tumor types, copy number losses at this genomic position were also frequent. By experimental and computationally inferring genome ploidy, we demonstrate that an increased number of aUPD events, both affecting the whole chromosome or segments of it, were present in highly aneuploid genomes compared to near-diploid tumors. Finally, the presence of mosaic UPD was detected at a higher frequency in DNA extracted from peripheral blood lymphocytes of patients with colorectal cancer compared to healthy individuals. In summary, our study defines specific profiles of aUPD in gastrointestinal cancers and provides unequivocal evidence of their relevance in cancer.


Asunto(s)
Neoplasias Gastrointestinales/genética , Disomía Uniparental/genética , Aneuploidia , Estudios de Casos y Controles , Análisis Mutacional de ADN , Neoplasias Gastrointestinales/patología , Perfil Genético , Estudio de Asociación del Genoma Completo , Humanos , Polimorfismo de Nucleótido Simple , Análisis de Matrices Tisulares , Disomía Uniparental/patología
5.
Am J Med Genet A ; 179(7): 1148-1156, 2019 07.
Artículo en Inglés | MEDLINE | ID: mdl-31050164

RESUMEN

Instability of the FMR1 repeat, commonly observed in transmissions of premutation alleles (55-200 repeats), is influenced by the size of the repeat, its internal structure and the sex of the transmitting parent. We assessed these three factors in unstable transmissions of 14/3,335 normal (~5 to 44 repeats), 54/293 intermediate (45-54 repeats), and 1561/1,880 premutation alleles. While most unstable transmissions led to expansions, contractions to smaller repeats were observed in all size classes. For normal alleles, instability was more frequent in paternal transmissions and in alleles with long 3' uninterrupted repeat lengths. For premutation alleles, contractions also occurred more often in paternal than maternal transmissions and the frequency of paternal contractions increased linearly with repeat size. All paternal premutation allele contractions were transmitted as premutation alleles, but maternal premutation allele contractions were transmitted as premutation, intermediate, or normal alleles. The eight losses of AGG interruptions in the FMR1 repeat occurred exclusively in contractions of maternal premutation alleles. We propose a refined model of FMR1 repeat progression from normal to premutation size and suggest that most normal alleles without AGG interruptions are derived from contractions of maternal premutation alleles.


Asunto(s)
Alelos , Proteína de la Discapacidad Intelectual del Síndrome del Cromosoma X Frágil/genética , Síndrome del Cromosoma X Frágil/genética , Patrón de Herencia , Expansión de Repetición de Trinucleótido , Femenino , Síndrome del Cromosoma X Frágil/diagnóstico , Síndrome del Cromosoma X Frágil/patología , Expresión Génica , Frecuencia de los Genes , Humanos , Masculino , Linaje
6.
Am J Med Genet A ; 173(10): 2795-2797, 2017 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-28815939

RESUMEN

Fragile X syndrome (FXS) is the most common form of inherited intellectual disability (ID) and autism. In most of cases, the molecular basis of this syndrome is a CGG repeat expansion in the 5' untranslated region of the FMR1 gene. It is inherited as an X linked dominant trait, with a reduced penetrance (80% for males and 30% for females). Full mutation (FM) expansion from premutated alleles (PM) is only acquired via maternal meiosis, while paternal transmission always remains in the PM range. We present a 16-year-old girl with a mild fragile X syndrome phenotype. FMR1 gene study showed that the patient inherited a mosaic premutation-full mutation with an unmethylated uninterrupted allele (175, >200 CGG) from her father. The father showed an 88 CGG uninterrupted unmethylated allele in blood and sperm cells. To our knowledge, this is the first case of a FMR1 mosaic premutation-full mutation allele inherited from a PM father. In our opinion, the most likely explanation could be a postzygotic somatic expansion. We can conclude that in rare cases of a child with a full mutation whose mother does not carry a premutation, the possibility of paternal transmission should be considered.


Asunto(s)
Proteína de la Discapacidad Intelectual del Síndrome del Cromosoma X Frágil/genética , Síndrome del Cromosoma X Frágil/genética , Mutación , Herencia Paterna , Adolescente , Adulto , Femenino , Síndrome del Cromosoma X Frágil/patología , Humanos , Masculino , Adulto Joven
7.
Cytogenet Genome Res ; 148(2-3): 156-64, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-27216161

RESUMEN

Complex chromosome rearrangements (CCRs) are unusual structural chromosome alterations found in humans, and to date only a few have been characterized molecularly. New mechanisms, such as chromothripsis, have been proposed to explain the presence of the CCRs in cancer cells and in patients with congenital disorders and/or mental retardation. The aim of the present study was the molecular characterization of a constitutional CCR in a girl with multiple congenital disorders and intellectual disability in order to determine the genotype-phenotype relation and to clarify whether the CCR could have been caused by chromosomal catastrophic events. The present CCR was characterized by G-banding, high-resolution CGH, multiplex ligation-dependent probe amplification and subtelomeric 2q-FISH analyses. Preliminary results indicate that the de novo CCR is unbalanced showing a 2q37.3 deletion and 2q34q37.2 partial trisomy. Our patient shows some of the typical traits and intellectual disability described in patients with 2q37 deletion and also in carriers of 2q34q37.2 partial trisomy; thus, the clinical disorders could be explained by additional effects of both chromosome alterations (deletions and duplications). A posterior, sequential FISH study using BAC probes revealed the unexpected presence of at least 17 different reorganizations affecting 2q34q37.2, suggesting the existence of chromosome instability in this region. The present CCR is the first case described in the literature of heterogeneity of unbalanced CCRs affecting a small region of 2q, indicating that the mechanisms involved in constitutional chromosome rearrangement may be more complex than previously thought.


Asunto(s)
Trisomía/genética , Anomalías Múltiples/genética , Niño , Preescolar , Bandeo Cromosómico , Deleción Cromosómica , Cromosomas Humanos Par 2/genética , Femenino , Humanos , Hibridación Fluorescente in Situ , Lactante , Discapacidad Intelectual/genética , Reacción en Cadena de la Polimerasa Multiplex , Translocación Genética/genética
8.
Genet Med ; 18(4): 378-85, 2016 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-26181491

RESUMEN

PURPOSE: Autism spectrum disorders are associated with defects in social response and communication that often occur in the context of intellectual disability. Rett syndrome is one example in which epilepsy, motor impairment, and motor disturbance may co-occur. Mutations in histone demethylases are known to occur in several of these syndromes. Herein, we aimed to identify whether mutations in the candidate histone demethylase JMJD1C (jumonji domain containing 1C) are implicated in these disorders. METHODS: We performed the mutational and functional analysis of JMJD1C in 215 cases of autism spectrum disorders, intellectual disability, and Rett syndrome without a known genetic defect. RESULTS: We found seven JMJD1C variants that were not present in any control sample (~ 6,000) and caused an amino acid change involving a different functional group. From these, two de novo JMJD1C germline mutations were identified in a case of Rett syndrome and in a patient with intellectual disability. The functional study of the JMJD1C mutant Rett syndrome patient demonstrated that the altered protein had abnormal subcellular localization, diminished activity to demethylate the DNA damage-response protein MDC1, and reduced binding to MECP2. We confirmed that JMJD1C protein is widely expressed in brain regions and that its depletion compromises dendritic activity. CONCLUSIONS: Our findings indicate that mutations in JMJD1C contribute to the development of Rett syndrome and intellectual disability.Genet Med 18 1, 378-385.


Asunto(s)
Discapacidad Intelectual/genética , Histona Demetilasas con Dominio de Jumonji/genética , Mutación , Oxidorreductasas N-Desmetilantes/genética , Síndrome de Rett/genética , Adulto , Secuencias de Aminoácidos , Secuencia de Aminoácidos , Trastorno del Espectro Autista/diagnóstico , Trastorno del Espectro Autista/genética , Encéfalo/metabolismo , Encéfalo/patología , Secuencia Conservada , Análisis Mutacional de ADN , Femenino , Expresión Génica , Orden Génico , Estudios de Asociación Genética , Sitios Genéticos , Humanos , Discapacidad Intelectual/diagnóstico , Histona Demetilasas con Dominio de Jumonji/química , Histona Demetilasas con Dominio de Jumonji/metabolismo , Masculino , Persona de Mediana Edad , Modelos Moleculares , Neuronas/metabolismo , Oxidorreductasas N-Desmetilantes/química , Oxidorreductasas N-Desmetilantes/metabolismo , Posición Específica de Matrices de Puntuación , Conformación Proteica , Transporte de Proteínas , Síndrome de Rett/diagnóstico
9.
Ann Neurol ; 77(5): 889-94, 2015 May.
Artículo en Inglés | MEDLINE | ID: mdl-25631192

RESUMEN

Epidemiological studies have reported the co-occurrence of Parkinson disease (PD) and melanoma. Common genetic variants in the MC1R (melanocortin 1 receptor) gene, which determines skin and hair color, are associated with melanoma. Here we investigated whether genetic variants in MC1R modulate the risk of PD by sequencing the entire gene in 870 PD patients and 736 controls ascertained from Spain. We found that the MC1R variant p.R160W (rs1805008) is marginally associated with PD (odds ratio = 2.10, gender- and age-adjusted p = 0.009, Bonferroni-corrected p = 0.063). Our results suggest that MC1R genetic variants modulate the risk of PD disease in the Spanish population.


Asunto(s)
Predisposición Genética a la Enfermedad/genética , Variación Genética/genética , Melanoma/genética , Enfermedad de Parkinson/genética , Receptor de Melanocortina Tipo 1/genética , Adulto , Anciano , Secuencia de Aminoácidos , Estudios de Casos y Controles , Femenino , Estudios de Asociación Genética/métodos , Predisposición Genética a la Enfermedad/epidemiología , Humanos , Masculino , Melanoma/diagnóstico , Melanoma/epidemiología , Persona de Mediana Edad , Datos de Secuencia Molecular , Enfermedad de Parkinson/diagnóstico , Enfermedad de Parkinson/epidemiología , España/epidemiología
10.
Cerebellum ; 15(5): 543-5, 2016 10.
Artículo en Inglés | MEDLINE | ID: mdl-27338822

RESUMEN

The wide spectrum of clinical phenotypes associated with the FMR1 premutation affect more than two million people worldwide. The clinical implications have only been recognized recently despite this disorder constitutes a relevant health problem. The present issue of The Cerebellum is focused on the "2(nd) International Conference on the FMR1 Premutation: Basic Mechanisms and Clinical Involvement" held in Sitges, Barcelona (Spain), from September 30th to October 2nd, 2015. The conference was attended by professionals from different countries in Europe, the USA, Chile, Israel, Australia, and Indonesia and covered the latest clinical and molecular findings resulting from FMR1 premutation studies. Although the pathologies associated with the FMR1 premutation are considered as rare diseases, seventy abstracts were presented. This reflects the relevance of this topic in the medical community and the growing interest among professionals from other disciplines. The major topics discussed included why and how the mRNA toxicity due to a gain of function and non-canonical RAN are responsible for disorders associated with the premutation. Several presentations addressed the impact of these mechanisms in FXTAS and FXPOI, two clinical presentations caused by the FMR1 premutation. Interestingly, a deterioration of the DNA repair machinery was first proposed as the pathogenicity cause of premutation alleles. Communications related to FXTAS and FXPOI animal models were also presented. These models facilitate studies aimed to understand disease progression and early treatment interventions. Finally, there were presentations related to psychiatric, psychological, neurological, and radiological aspects. Interesting discussion on intermediate alleles and their involvement in clinical and reproductive aspects was generated. In this regards, genetic counselling is improved by taking into account the AGG interruptions and including information about the FMR1 premutation associated pathologies although there are still some uncertainties linked to the spectrum of these pathologies. Overall, the meeting covered all aspects of the different pathologies associated with the premutation of the FMR1 gene.


Asunto(s)
Proteína de la Discapacidad Intelectual del Síndrome del Cromosoma X Frágil/genética , Proteína de la Discapacidad Intelectual del Síndrome del Cromosoma X Frágil/metabolismo , Animales , Congresos como Asunto , Humanos , España
11.
Cerebellum ; 15(5): 570-7, 2016 10.
Artículo en Inglés | MEDLINE | ID: mdl-27315125

RESUMEN

Fragile X-associated tremor/ataxia syndrome (FXTAS) is a late-onset neurodegenerative disorder that occurs in FMR1 premutation carriers. The prevalence of FMR1 premutation carriers in the general population is relatively high, and although rare, a premutation in both X chromosomes may occur in females inheriting a premutation allele from each of both parent carriers. Here, we report the first female with an autozygous (homozygous by descendent) FMR1 premutation allele, who fulfills neurological and radiological FXTAS findings/criteria. Molecular characterization included CGG repeat length, AGG interruption pattern, FMR1 messenger RNA (mRNA), fragile X mental retardation protein (FMRP) level quantification, and single-nucleotide polymorphism (SNP) microarray. Neuroradiological assessment of 3-T magnetic resonance imaging and neurological and cognitive/neuropsychological evaluations were performed. Neurological and neuroradiological examination of the female with the same FMR1 allele in the premutation range (77 CGGs) demonstrated FXTAS features. Further familial evaluation showed a similar neuropsychiatric profile, with impairments in cognitive flexibility and visuospatial function, mainly. A unique family with an autozygous FMR1 premutation female is presented. Neurological/cognitive and neuroradiological examinations revealed FXTAS-specific findings in the female with the autozygous FMR1 premutation allele. The consistent molecular and cognitive/psychiatric phenotype in family members suggests that carrying one or two FMR1 premutation alleles has no effect on illness severity.


Asunto(s)
Ataxia/genética , Proteína de la Discapacidad Intelectual del Síndrome del Cromosoma X Frágil/genética , Síndrome del Cromosoma X Frágil/genética , Dosificación de Gen , Temblor/genética , Adulto , Anciano , Anciano de 80 o más Años , Alelos , Ataxia/diagnóstico por imagen , Encéfalo/diagnóstico por imagen , Familia , Femenino , Síndrome del Cromosoma X Frágil/diagnóstico por imagen , Heterocigoto , Humanos , Masculino , Persona de Mediana Edad , Índice de Severidad de la Enfermedad , Temblor/diagnóstico por imagen
12.
Cerebellum ; 15(5): 578-86, 2016 10.
Artículo en Inglés | MEDLINE | ID: mdl-27287737

RESUMEN

Fragile X-associated tremor/ataxia syndrome (FXTAS) is a progressive neurodegenerative disorder caused by a repeat expansion in the fragile X mental retardation 1 (FMR1) gene. The disorder is characterized by kinetic tremor and cerebellar ataxia, shows age-dependent penetrance, and occurs more frequently in men. This paper summarizes the key emerging issues in FXTAS as presented at the Second International Conference on the FMR1 Premutation: Basic Mechanisms & Clinical Involvement in 2015. The topics discussed include phenotype-genotype relationships, neurobehavioral function, and updates on FXTAS genetics and imaging.


Asunto(s)
Ataxia/diagnóstico por imagen , Ataxia/fisiopatología , Proteína de la Discapacidad Intelectual del Síndrome del Cromosoma X Frágil/genética , Síndrome del Cromosoma X Frágil/diagnóstico por imagen , Síndrome del Cromosoma X Frágil/fisiopatología , Heterocigoto , Temblor/diagnóstico por imagen , Temblor/fisiopatología , Animales , Ataxia/genética , Ataxia/terapia , Congresos como Asunto , Síndrome del Cromosoma X Frágil/genética , Síndrome del Cromosoma X Frágil/terapia , Humanos , Fenotipo , Temblor/genética , Temblor/terapia
13.
Neurodegener Dis ; 16(3-4): 290-2, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-26609701

RESUMEN

BACKGROUND: Fragile-X-associated tremor/ataxia syndrome (FXTAS) is a late-onset multisystem neurological disorder characterized by intention tremor and cerebellar ataxia. OBJECTIVE: We hypothesized that in FMR1 premutation females with FXTAS, a normal X chromosome might more frequently be inactivated; therefore, the aim of this study was to determine the relationship between skewed X chromosome inactivation (XCI) and FXTAS. METHODS: We studied the XCI patterns of cases of FMR1 premutation in 10 women with FXTAS and 21 without FXTAS. RESULTS: The distribution of XCI patterns in the FXTAS and no-FXTAS groups showed differences regarding the allele presenting severe skewed XCI. In the FXTAS group, all cases preferentially inactivated the non-expanded X chromosome, whereas in the no-FXTAS group, all inactivated the expanded X chromosome. Nevertheless, no significant differences were found on comparing XCI frequencies among FMR1 premutation carriers with and without FXTAS. As expected, we found statistically significant differences in the skewed XCI on comparing FMR1 premutation women and controls. CONCLUSION: Although the reduced sample size and blood XCI patterns are two limitations of this study, our results suggest that the skewed XCI of the normal FMR1 allele may be a risk factor for the development of FXTAS. Furthermore, our findings also support the protective effect of the expression of a normal FMR1 allele.


Asunto(s)
Ataxia/sangre , Ataxia/genética , Proteína de la Discapacidad Intelectual del Síndrome del Cromosoma X Frágil/genética , Síndrome del Cromosoma X Frágil/sangre , Síndrome del Cromosoma X Frágil/genética , Temblor/sangre , Temblor/genética , Inactivación del Cromosoma X , Análisis Químico de la Sangre , Metilación de ADN , Análisis Mutacional de ADN , Femenino , Heterocigoto , Humanos , Persona de Mediana Edad
14.
Neurobiol Dis ; 65: 43-54, 2014 May.
Artículo en Inglés | MEDLINE | ID: mdl-24418349

RESUMEN

Male premutation carriers presenting between 55 and 200 CGG repeats in the Fragile-X-associated (FMR1) gene are at risk of developing Fragile X Tremor/Ataxia Syndrome (FXTAS), and females undergo Premature Ovarian Failure (POF1). Here, we have evaluated gene expression profiles from blood in male FMR1 premutation carriers and detected a strong deregulation of genes enriched in FXTAS relevant biological pathways, including inflammation, neuronal homeostasis and viability. Gene expression profiling distinguished between control individuals, carriers with FXTAS and carriers without FXTAS, with levels of expanded FMR1 mRNA being increased in FXTAS patients. In vitro studies in a neuronal cell model indicate that expression levels of expanded FMR1 5'-UTR are relevant in modulating the transcriptome. Thus, perturbations of the transcriptome may be an interplay between the CGG expansion size and FMR1 expression levels. Several deregulated genes (DFFA, BCL2L11, BCL2L1, APP, SOD1, RNF10, HDAC5, KCNC3, ATXN7, ATXN3 and EAP1) were validated in brain samples of a FXTAS mouse model. Downregulation of EAP1, a gene involved in the female reproductive system physiology, was confirmed in female carriers. Decreased levels were detected in female carriers with POF1 compared to those without POF1, suggesting that EAP1 levels contribute to ovarian insufficiency. In summary, gene expression profiling in blood has uncovered mechanisms that may underlie different pathological aspects of the premutation. A better understanding of the transcriptome dynamics in relation with expanded FMR1 mRNA expression levels and CGG expansion size may provide mechanistic insights into the disease process and a more accurate FXTAS diagnosis to the myriad of phenotypes associated with the premutation.


Asunto(s)
Proteína de la Discapacidad Intelectual del Síndrome del Cromosoma X Frágil/genética , Regulación de la Expresión Génica/genética , Infertilidad/genética , Enfermedades Neurodegenerativas/sangre , Expansión de Repetición de Trinucleótido/genética , Adulto , Anciano , Animales , Modelos Animales de Enfermedad , Femenino , Humanos , Masculino , Ratones , Análisis por Micromatrices , Proteínas de Microfilamentos , Persona de Mediana Edad , Neuroblastoma/patología , Enfermedades Neurodegenerativas/genética , Fenotipo , Proteínas/genética , Proteínas/metabolismo , Securina/genética , Securina/metabolismo
15.
Am J Med Genet B Neuropsychiatr Genet ; 165B(6): 472-8, 2014 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-24980697

RESUMEN

The phenotype overlap between autism spectrum disorders (ASD) & intellectual disabilities (ID) is mirrored at the genetic level, with common genes being reported mutated in variety of developmental disabilities. However despite widespread genetic screening for mutations, in approximately 40-60% of childhood developmental disorders the genetic cause remains unknown. Several genome-wide linkage screens in ASD have identified a locus mapping to distal 8q. We have recently identified a novel brain-specific imprinted cluster at this location, which contains the reciprocally expressed maternal KCNK9 and paternally expressed non-coding PEG13 transcripts, the latter located within an intron of TRAPPC9. Interestingly, mutations of KCNK9 and TRAPPC9 have been reported in Birk-Barel mental retardation and non-syndromic familial forms of ID, respectively. Here, we report a genetic screen for KCNK9 coding mutations and potential epigenetic aberrations that could result in deregulated imprinting in a cohort of 120 ID, 86 ASD and 86 Tourette syndrome patients. Fifteen of the ID patients had clinical characteristics overlapping with Birk-Barel syndrome. Sequencing of the two coding exons of KCNK9 failed to identify pathologic mutations, with only one variant, rs2615374, being present with allele frequencies similar to those described in dbSNP database. DNA methylation profiling of the KCNK9 and TRAPPC9 promoters, the maternally methylated PEG13 DMR and a long-range enhancer region were normal in all patients. Our findings suggest that mutations of KCNK9 or epigenetic disturbances within the PEG13 imprinted cluster do not significantly contribute to the cause of the developmental disabilities tested in this study.


Asunto(s)
Trastorno Autístico/genética , Cromosomas Humanos Par 8/genética , Metilación de ADN/genética , Pruebas Genéticas , Impresión Genómica/genética , Discapacidad Intelectual/genética , Canales de Potasio de Dominio Poro en Tándem/genética , Síndrome de Tourette/genética , Epigénesis Genética , Frecuencia de los Genes/genética , Predisposición Genética a la Enfermedad , Humanos , Mutación , Polimorfismo de Nucleótido Simple/genética
16.
Genet Med ; 15(8): 639-42, 2013 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-23492875

RESUMEN

PURPOSE: Fragile X-associated tremor/ataxia syndrome is a late-onset neurodegenerative disorder that occurs in FMR1 premutation carriers. It is well known that the apolipoprotein E ε4 allele is a risk factor for neurodegenerative disease. The main goal of this work was to evaluate the apolipoprotein E genotypes and allelic distribution among patients with fragile X-associated tremor/ataxia syndrome. METHODS: A total of 44 unrelated FMR1 premutation carriers (22 presenting with fragile X-associated tremor/ataxia syndrome and 22 without fragile X-associated tremor/ataxia syndrome) were genotyped. RESULTS: All the apolipoprotein E ε4/4 genotype carriers detected (100%), and six of the seven apolipoprotein E ε4/3 genotype carriers (85.7%) are patients presenting with fragile X-associated tremor/ataxia syndrome symptoms, whereas only 40% of the apolipoprotein E ε3/3 genotype carriers belong to the fragile X-associated tremor/ataxia syndrome group. The results showed that the presence of the apolipoprotein E ε4 allele increases the risk of developing fragile X-associated tremor/ataxia syndrome (odds ratio = 12.041; P = 0.034). CONCLUSION: On the basis of these results, we conclude that the presence of at least one apolipoprotein E ε4 allele might act as a genetic factor predisposing individuals to develop fragile X-associated tremor/ataxia syndrome.


Asunto(s)
Apolipoproteína E3/genética , Apolipoproteína E4/genética , Ataxia/genética , Síndrome del Cromosoma X Frágil/genética , Temblor/genética , Anciano , Anciano de 80 o más Años , Femenino , Proteína de la Discapacidad Intelectual del Síndrome del Cromosoma X Frágil/genética , Frecuencia de los Genes , Predisposición Genética a la Enfermedad , Genotipo , Heterocigoto , Humanos , Masculino , Persona de Mediana Edad , Factores de Riesgo , España , Población Blanca/genética
17.
Tumour Biol ; 34(3): 1337-47, 2013 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-23504524

RESUMEN

Although widely used, the value of prostate-specific antigen (PSA) in the detection of prostate cancer is controversial. The percentage of free PSA increases the specificity of PSA, but results are not enough. Prostate cancer gene 3 (PCA3) has been proposed as an option that may complement these markers in the detection and management of early prostate cancer. Our aim has been to review the value of PCA3 as tumor marker. The available results suggest that PCA3 is particularly useful to select in which patients the biopsy should be repeated when the first biopsy was negative. However, some points should be specified with further studies, including the most appropriate PCA3 cutoff level and the significance of a high PCA3 score in patients with negative biopsy. False-negative results are also a conflictive point in the use of PCA3, because prostate cancer, including aggressive tumors, may be present in patients with a low PCA3 score. Probably, a proper interpretation of this test requires its management together with other tests, through multivariate models for the detection of prostate cancer. On the other hand, several studies showed the relation between PCA3 score and Gleason score, and also the utility of PCA3 to select patients for active surveillance. To summarize, the available studies show the utility of PCA3 in the detection and management of early prostate cancer, although some aspects referred to its use need to be retested after further studies to confirm the actual value and the limitations of this test.


Asunto(s)
Antígenos de Neoplasias/metabolismo , Biomarcadores de Tumor/metabolismo , Neoplasias de la Próstata/diagnóstico , Neoplasias de la Próstata/terapia , Manejo de la Enfermedad , Humanos , Masculino , Neoplasias de la Próstata/metabolismo
19.
J Cell Mol Med ; 16(9): 2017-21, 2012 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-22129439

RESUMEN

In idiopathic portal hypertension (IPH) typical vascular lesions are present in the branches of the portal vein or in the perisinusoidal area of the liver. Similar histological alterations have been reported in the pulmonary vasculature of patients with idiopathic pulmonary artery hypertension (IPAH). As IPAH is associated with mutations of the bone morphogenetic protein receptor 2 (BMPR2) gene, the aim of this study was to investigate whether this association might also be found in patients with IPH. Twenty-three samples belonging to 21 unrelated caucasian patients with IPH followed in the hepatic haemodynamic laboratory of the Hospital Clinic in Barcelona were included in the study. All patients were studied for the entire open reading frame and splice site of the BMPR2 gene by direct sequencing and multiple ligation probe amplification (MLPA) in order to detect large deletions/duplications. None of the 23 patients had pulmonary artery hypertension. Four patients presented one single nucleotide polymorphism (SNP) in intron 5, four patients had a SNP in exon 12 and a SNP in exon 1 was found in two cases. Two patients had both intron 5 and exon 12 polymorphisms. All SNPs were previously described. Except for these three SNPs, neither mutations nor rearrangements have been identified in the BMPR2 gene in this population. We did not detect mutations or rearrangements in the coding region of the BMPR2 gene in our patients with IPH. These findings suggest that, in contrast to IPAH, mutations in BMPR2 are not involved in the pathogenesis of IPH.


Asunto(s)
Receptores de Proteínas Morfogenéticas Óseas de Tipo II/genética , Hipertensión Portal/genética , Cirrosis Hepática/genética , Pancitopenia/genética , Esplenomegalia/genética , Adolescente , Adulto , Anciano , Anciano de 80 o más Años , Receptores de Proteínas Morfogenéticas Óseas de Tipo II/metabolismo , Niño , Exones , Femenino , Reordenamiento Génico , Humanos , Hipertensión Portal/fisiopatología , Intrones , Cirrosis Hepática/fisiopatología , Masculino , Persona de Mediana Edad , Reacción en Cadena de la Polimerasa Multiplex/métodos , Mutación , Pancitopenia/fisiopatología , Polimorfismo de Nucleótido Simple , ARN/genética , ARN/aislamiento & purificación , Esplenomegalia/fisiopatología , Adulto Joven , Hipertensión Portal Idiopática no Cirrótica
20.
Am J Hum Genet ; 85(6): 809-22, 2009 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-20004760

RESUMEN

We report on the identification of a 0.3 Mb inherited recurrent but variable copy-number gain at Xq28 in affected males of four unrelated families with X-linked mental retardation (MR). All aberrations segregate with the disease in the families, and the carrier mothers show nonrandom X chromosome inactivation. Tiling Xq28-region-specific oligo array revealed that all aberrations start at the beginning of the low copy repeat LCR-K1, at position 153.20 Mb, and end just distal to LCR-L2, at 153.54 Mb. The copy-number gain always includes 18 annotated genes, of which RPL10, ATP6AP1 and GDI1 are highly expressed in brain. From these, GDI1 is the most likely candidate gene. Its copy number correlates with the severity of clinical features, because it is duplicated in one family with nonsyndromic moderate MR, is triplicated in males from two families with mild MR and additional features, and is present in five copies in a fourth family with a severe syndromic form of MR. Moreover, expression analysis revealed copy-number-dependent increased mRNA levels in affected patients compared to control individuals. Interestingly, analysis of the breakpoint regions suggests a recombination mechanism that involves two adjacent but different sets of low copy repeats. Taken together, our data strongly suggest that an increased expression of GDI1 results in impaired cognition in a dosage-dependent manner. Moreover, these data also imply that a copy-number gain of an individual gene present in the larger genomic aberration that leads to the severe MECP2 duplication syndrome can of itself result in a clinical phenotype as well.


Asunto(s)
Cromosomas Humanos X , Dosificación de Gen , Discapacidad Intelectual/genética , Recombinación Genética , Adulto , Encéfalo/metabolismo , Niño , Preescolar , Aberraciones Cromosómicas , Mapeo Cromosómico , Femenino , Humanos , Masculino , Modelos Genéticos , Hibridación de Ácido Nucleico , Linaje , Fenotipo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA