Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 89
Filtrar
Más filtros

Banco de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
J Hum Genet ; 61(9): 811-21, 2016 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-27225849

RESUMEN

Autosomal recessive polycystic kidney disease (ARPKD) is a rare severe genetic disorder arising in the perinatal period, although a late-onset presentation of the disease has been described. Pulmonary hypoplasia is the major cause of morbidity and mortality in the newborn period. ARPKD is caused by mutations in the PKHD1 (polycystic kidney and hepatic disease 1) gene that is among the largest human genes. To achieve a molecular diagnosis of the disease, a large series of Italian affected subjects were recruited. Exhaustive mutation analysis of PKHD1 gene was carried out by Sanger sequencing and multiple ligation probe amplification (MLPA) technique in 110 individuals. A total of 173 mutations resulting in a detection rate of 78.6% were identified. Additional 20 unrelated patients, in whom it was not possible to analyze the whole coding sequence, have been included in this study. Taking into account the total number (n=130) of this cohort of patients, 107 different types of mutations have been detected in 193 mutated alleles. Out of 107 mutations, 62 were novel: 11 nonsense, 6 frameshift, 7 splice site mutations, 2 in-frame deletions and 2 multiexon deletion detected by MLPA. Thirty-four were missense variants. In conclusion, our report expands the spectrum of PKHD1 mutations and confirms the heterogeneity of this disorder. The population under study represents the largest Italian ARPKD cohort reported to date. The estimated costs and the time invested for molecular screening of genes with large size and allelic heterogeneity such as PKHD1 demand the use of next-generation sequencing (NGS) technologies for a faster and cheaper screening of the affected subjects.


Asunto(s)
Mutación , Riñón Poliquístico Autosómico Recesivo/genética , Receptores de Superficie Celular/genética , Adolescente , Adulto , Alelos , Sustitución de Aminoácidos , Niño , Preescolar , Análisis Mutacional de ADN , Femenino , Genotipo , Humanos , Lactante , Masculino , Persona de Mediana Edad , Reacción en Cadena de la Polimerasa Multiplex , Riñón Poliquístico Autosómico Recesivo/diagnóstico , Análisis de Secuencia de ADN , Adulto Joven
2.
BMC Med Genet ; 16: 69, 2015 Aug 23.
Artículo en Inglés | MEDLINE | ID: mdl-26297663

RESUMEN

BACKGROUND: Deletions on the distal portion of the long arm of chromosome 6 are relatively uncommon, and only a small number occurs in the paternal copy, causing growth abnormalities. As a result, extensive clinical descriptions are lacking. CASE PRESENTATION: We describe a male of Italian descent born at 35 weeks by elective caesarean delivery presenting hypoplastic left colon, bilateral inguinal hernia, dysplastic tricuspid and pulmonary valves, premature ventricular contractions, recurrent otitis media, poor feeding, gastro-oesophageal reflux, bilateral pseudopapilledema, and astigmatism. He also showed particular facial dysmorphisms and postnatal growth failure. Early psychomotor development was mildly delayed. At 3.75 years, he was evaluated for severe short stature (-2.98 SD) and delayed bone age. He showed an insulin-like growth factor 1 concentration (IGF-1) in the low-normal range. Growth hormone stimulation tests showed a low response to clonidine and insulin. Magnetic resonance imaging showed hypophyseal hypoplasia. Genetic evaluation by Single Nucleotide Polymorphism arrays showed a de novo 6q24.2-q25.2 deletion on paternal chromosome 6. CONCLUSION: We confirm that this is a new congenital malformation syndrome associated with a deletion of 6q24.2-q25.2 on paternal chromosome 6. We suggest evaluating the growth hormone axis in children with 6q24.2-q25.2 deletions and growth failure.


Asunto(s)
Anomalías Múltiples/genética , Deleción Cromosómica , Cromosomas Humanos Par 6/genética , Trastornos del Crecimiento/genética , Hormona del Crecimiento/deficiencia , Anomalías Múltiples/tratamiento farmacológico , Anomalías Múltiples/patología , Hibridación Genómica Comparativa , Ecocardiografía , Estudios de Seguimiento , Trastornos del Crecimiento/tratamiento farmacológico , Trastornos del Crecimiento/patología , Hormona del Crecimiento/administración & dosificación , Humanos , Italia , Cariotipificación , Imagen por Resonancia Magnética , Masculino
3.
Am J Med Genet A ; 167A(2): 438-44, 2015 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-25428890

RESUMEN

We provide a detailed clinical and molecular characterization of an 11-year-old female patient presenting with neurodevelopmental delay (NDD), intellectual disability (ID), seizures, stereotypies and dysmorphic features. Chromosomal microarrays analysis (CMA) detected a small, rare de novo deletion on chromosome 12q24.31 encompassing 31 protein-coding RefSeq genes and a microRNA. Phenotypic comparison with molecularly well-defined cases previously reported in the literature harboring an overlapping 12q24.31 microdeletion indicate that these patients shared common clinical features including neurodevelopmental delay, intellectual disability and behavioral problems. Also, seizures and dysmorphic features are frequent and a consistent pattern was recognized. Since there are remarkable resemblance between the patient described here and at least another one previously reported, our report is provides supportive evidence for the existence of an emerging syndrome caused by a microdeletion in 12q24.31. We propose a minimal region shared among patients contributing to the etiology of the common clinical features observed suggesting as candidate, for the first time, the gene SETD1B which is a component of a histone methyltransferase complex. In addition, we speculate on the possible contributive role of the MIR4304 to some clinical features observed in our patient. Evaluation of more patients with well-characterized deletions within 12q24.31, as well as careful clinical assessment of them, is needed to corroborate our hypothesis, to perform a more detailed genotype-phenotype correlation and, finally, to fully delineate this emerging microdeletion syndrome.


Asunto(s)
Deleción Cromosómica , Cromosomas Humanos Par 12 , Huesos Faciales/anomalías , Discapacidad Intelectual/genética , Convulsiones/genética , Conducta Estereotipada , Preescolar , Mapeo Cromosómico , Hibridación Genómica Comparativa , Variaciones en el Número de Copia de ADN , Facies , Femenino , Humanos , Discapacidad Intelectual/diagnóstico , Fenotipo , Convulsiones/diagnóstico
4.
Hum Mutat ; 35(4): 447-51, 2014 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-24515783

RESUMEN

TBC1D7 forms a complex with TSC1 and TSC2 that inhibits mTORC1 signaling and limits cell growth. Mutations in TBC1D7 were reported in a family with intellectual disability (ID) and macrocrania. Using exome sequencing, we identified two sisters homozygote for the novel c.17_20delAGAG, p.R7TfsX21 TBC1D7 truncating mutation. In addition to the already described macrocephaly and mild ID, they share osteoarticular defects, patella dislocation, behavioral abnormalities, psychosis, learning difficulties, celiac disease, prognathism, myopia, and astigmatism. Consistent with a loss-of-function of TBC1D7, the patient's cell lines show an increase in the phosphorylation of 4EBP1, a direct downstream target of mTORC1 and a delay in the initiation of the autophagy process. This second family allows enlarging the phenotypic spectrum associated with TBC1D7 mutations and defining a TBC1D7 syndrome. Our work reinforces the involvement of TBC1D7 in the regulation of mTORC1 pathways and suggests an altered control of autophagy as possible cause of this disease.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/metabolismo , Proteínas Portadoras/genética , Enfermedad Celíaca/genética , Discapacidad Intelectual/genética , Megalencefalia/genética , Luxación de la Rótula/genética , Fosfoproteínas/metabolismo , Autofagia , Proteínas Portadoras/metabolismo , Enfermedad Celíaca/patología , Proteínas de Ciclo Celular , Línea Celular , Exoma , Femenino , Predisposición Genética a la Enfermedad , Variación Genética , Secuenciación de Nucleótidos de Alto Rendimiento , Homocigoto , Humanos , Discapacidad Intelectual/patología , Péptidos y Proteínas de Señalización Intracelular , Megalencefalia/patología , Mutación , Luxación de la Rótula/patología , Linaje
5.
Hum Mutat ; 35(7): 841-50, 2014 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-24633898

RESUMEN

Kabuki syndrome (KS) is a multiple congenital anomalies syndrome characterized by characteristic facial features and varying degrees of mental retardation, caused by mutations in KMT2D/MLL2 and KDM6A/UTX genes. In this study, we performed a mutational screening on 303 Kabuki patients by direct sequencing, MLPA, and quantitative PCR identifying 133 KMT2D, 62 never described before, and four KDM6A mutations, three of them are novel. We found that a number of KMT2D truncating mutations result in mRNA degradation through the nonsense-mediated mRNA decay, contributing to protein haploinsufficiency. Furthermore, we demonstrated that the reduction of KMT2D protein level in patients' lymphoblastoid and skin fibroblast cell lines carrying KMT2D-truncating mutations affects the expression levels of known KMT2D target genes. Finally, we hypothesized that the KS patients may benefit from a readthrough therapy to restore physiological levels of KMT2D and KDM6A proteins. To assess this, we performed a proof-of-principle study on 14 KMT2D and two KDM6A nonsense mutations using specific compounds that mediate translational readthrough and thereby stimulate the re-expression of full-length functional proteins. Our experimental data showed that both KMT2D and KDM6A nonsense mutations displayed high levels of readthrough in response to gentamicin treatment, paving the way to further studies aimed at eventually treating some Kabuki patients with readthrough inducers.


Asunto(s)
Anomalías Múltiples/genética , Cara/anomalías , Enfermedades Hematológicas/genética , Enfermedades Vestibulares/genética , Anomalías Múltiples/tratamiento farmacológico , Línea Celular , Codón sin Sentido/efectos de los fármacos , Estudios de Cohortes , Análisis Mutacional de ADN , Proteínas de Unión al ADN/genética , Proteínas de Unión al ADN/metabolismo , Expresión Génica , Regulación de la Expresión Génica/efectos de los fármacos , Estudios de Asociación Genética , Gentamicinas/farmacología , Gentamicinas/uso terapéutico , Haploinsuficiencia , Enfermedades Hematológicas/tratamiento farmacológico , Histona Demetilasas/genética , Proteínas de Homeodominio/genética , Humanos , Mutación , Proteínas de Neoplasias/genética , Proteínas de Neoplasias/metabolismo , Degradación de ARNm Mediada por Codón sin Sentido , Proteínas Nucleares/genética , Sitios de Empalme de ARN , Análisis de Secuencia de ADN , Transcripción Genética , Enfermedades Vestibulares/tratamiento farmacológico
6.
BMC Med Genet ; 15: 15, 2014 Jan 28.
Artículo en Inglés | MEDLINE | ID: mdl-24472332

RESUMEN

BACKGROUND: Kabuki syndrome (KS) is a rare, multiple congenital anomalies/intellectual disability syndrome caused by mutations of MLL2 gene, which codifies for a histone methyltrasferase that regulates the embryogenesis and the tissue development. Left-bronchial isomerism is a rare congenital abnormality that can be defined as the absence of the normal lateralizing features which distinguish right and left-sides in the lungs. To date, this is the first report of left-bronchial isomerism in association with KS. CASE PRESENTATION: A one-month-old Caucasian male patient underwent our attention for microcephaly, dysmorphic features (long palpebral fissures, eyebrows with sparse lateral third, everted lower eyelids, blue sclerae, large dysplastic ears, lower lip pits), persistent fetal fingertip pads, short stature, heart defects (interventricular defect and aortic coarctation), unilateral cryptorchidism, hypotonia and delay in gross motor skills. These features suggested a diagnosis of KS and a molecular analysis confirmed a novel frame-shift mutation in the exon 11 of MLL2 gene. Subsequently, given recurrent respiratory infections with a normal immunological status, he underwent a chest CT scan that showed a left bronchial isomerism. CONCLUSION: We report a patient affected by KS, with a novel MLL2 mutation and an atypical phenotype characterized by left-side bronchial isomerism. Interestingly, genes involved in the heterotaxia/isomerism such as ROCK2 and SHROOM3 are known to interact with MLL2 gene. In order to achieve a correct diagnosis and an appropriate therapy, the presence of pulmonary anatomical variations should be investigated in KS patients with respiratory signs not associated to immunological deficiency. Finally, our findings support the hypothesis that the mutations leading to a complete loss of function of MLL2 gene is often associated with complex visceral malformations.


Asunto(s)
Anomalías Múltiples/genética , Proteínas de Unión al ADN/genética , Cara/anomalías , Enfermedades Hematológicas/genética , Pulmón/anomalías , Mutación , Proteínas de Neoplasias/genética , Enfermedades Vestibulares/genética , Análisis Citogenético , Exones/genética , Femenino , Enfermedades Hematológicas/complicaciones , Humanos , Hipoglucemia/complicaciones , Lactante , Recién Nacido , Masculino , Embarazo , Eliminación de Secuencia , Enfermedades Vestibulares/complicaciones
7.
Am J Med Genet A ; 164A(4): 966-74, 2014 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-24616287

RESUMEN

Jacobsen syndrome (11q-) is a rare chromosomal disorder caused by deletions in distal11q. Many of the most common and severe congenital heart defects that occur in the general population occur in 11q-. Previous studies have demonstrated that gene-targeted deletion in mice of ETS-1, a cardiac transcription factor in distal 11q, causes ventricular septal defects with 100% penetrance. It is unclear whether deletion of other genes in distal 11q contributes to the full spectrum of congenital heart defects that occur in 11q-. Three patients with congenital heart defects have been identified that carry a translocation or paracentric inversion with a breakpoint in distal 11q disrupting one of two functionally related genes, OPCML and Neurotrimin. OPCML and Neurotrimin are two members of the IgLON subfamily of cell adhesion molecules. In this study, we report the generation and cardiac phenotype of single and double heterozygous gene-targeted OPCML and Neurotrimin knockout mice. No cardiac phenotype was detected, consistent with a single gene model as the cause of the congenital heart defects in 11q-.


Asunto(s)
Moléculas de Adhesión Celular/genética , Cardiopatías Congénitas/genética , Moléculas de Adhesión de Célula Nerviosa/genética , Animales , Secuencia de Bases , Proteínas Ligadas a GPI/genética , Eliminación de Gen , Humanos , Síndrome de Deleción Distal 11q de Jacobsen/genética , Ratones , Ratones Noqueados , Datos de Secuencia Molecular , Fenotipo , Translocación Genética
8.
Am J Med Genet A ; 161A(11): 2894-901, 2013 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-24124115

RESUMEN

Gorlin syndrome or nevoid basal cell carcinoma syndrome (NBCCS) is an autosomal dominant condition mainly characterized by the development of mandibular keratocysts which often have their onset during the second decade of life and/or multiple basal cell carcinoma (BCC) normally arising during the third decade. Cardiac and ovarian fibromas can be found. Patients with NBCCS develop the childhood brain malignancy medulloblastoma (now often called primitive neuro-ectodermal tumor [PNET]) in 5% of cases. The risk of other malignant neoplasms is not clearly increased, although lymphoma and meningioma can occur in this condition. Wilms tumor has been mentioned in the literature four times. We describe a patient with a 10.9 Mb 9q22.3 deletion spanning 9q22.2 through 9q31.1 that includes the entire codifying sequence of the gene PTCH1, with Wilms tumor, multiple neoplasms (lung, liver, mesenteric, gastric and renal leiomyomas, lung typical carcinoid tumor, adenomatoid tumor of the pleura) and a severe clinical presentation. We propose including leiomyomas among minor criteria of the NBCCS.


Asunto(s)
Síndrome del Nevo Basocelular/complicaciones , Síndrome del Nevo Basocelular/genética , Deleción Cromosómica , Cromosomas Humanos Par 9 , Proteína del Grupo de Complementación C de la Anemia de Fanconi/genética , Leiomioma/etiología , Receptores de Superficie Celular/genética , Tumor de Wilms/etiología , Anomalías Múltiples/diagnóstico , Anomalías Múltiples/genética , Adolescente , Adulto , Síndrome del Nevo Basocelular/diagnóstico , Causas de Muerte , Niño , Preescolar , Análisis Mutacional de ADN , Facies , Resultado Fatal , Femenino , Humanos , Lactante , Leiomioma/diagnóstico , Hígado/patología , Mutación , Receptores Patched , Receptor Patched-1 , Fenotipo , Tumor de Wilms/diagnóstico , Adulto Joven
9.
Mol Genet Metab ; 107(3): 627-9, 2012 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-22840376

RESUMEN

Kabuki syndrome is a rare, multiple congenital anomaly/mental retardation syndrome caused by MLL2 point mutations and KDM6A microdeletions. We screened a large cohort of MLL2 mutation-negative patients for MLL2 and KDM6A exon(s) microdeletion and microduplication. Our assays failed to detect such rearrangements in MLL2 as well as in KDM6A gene. These results show that these genomic events are extremely rare in the Kabuki syndrome, substantiating its genetic heterogeneity and the search for additional causative gene(s).


Asunto(s)
Anomalías Múltiples/genética , Proteínas de Unión al ADN/genética , Heterogeneidad Genética , Enfermedades Hematológicas/genética , Histona Demetilasas/genética , Proteínas de Neoplasias/genética , Proteínas Nucleares/genética , Enfermedades Vestibulares/genética , Anomalías Múltiples/diagnóstico , Adolescente , Niño , Preescolar , Exones , Cara/anomalías , Femenino , Eliminación de Gen , Duplicación de Gen , Enfermedades Hematológicas/diagnóstico , Humanos , Lactante , Masculino , Fenotipo , Análisis de Secuencia de ADN , Enfermedades Vestibulares/diagnóstico , Adulto Joven
10.
Mol Genet Metab ; 107(3): 548-52, 2012 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-22789683

RESUMEN

BACKGROUND: Autosomal dominant hypocalcemia (ADH) is an endocrine disorder caused by activating mutations of the calcium-sensing receptor (CASR) gene which plays a major role in maintaining calcium homeostasis. Biochemical features of ADH are hypocalcemia and hypercalciuria with inappropriately low levels of parathyroid hormone (PTH). We report on two four-generation families affected by ADH. AIM: To identify mutations of CASR gene in subjects affected by familial idiopathic hypoparathyroidism. To perform functional assays of identified CASR variants by transient transfection on HEK293 cells. RESULTS: We identified two CASR variants (Q681R and P221L): the Q681R variant was novel while the P221L had been previously published. Functional assays on the Q681R variant showed that it did not alter the whole expression nor the correct plasmamembrane localization, but enhanced the signaling function, increasing the sensitivity of the receptor as compared to the WT. CONCLUSIONS: We report two activating CASR mutations in two families affected by ADH and the functional assays performed on the novel variant Q681R. Our work enlarged the spectrum of mutations of the CASR and contributed to a better elucidation of the protein function.


Asunto(s)
Calcio/metabolismo , Hipercalciuria/genética , Hipocalcemia/genética , Hipoparatiroidismo/congénito , Hipoparatiroidismo/genética , Hormona Paratiroidea/deficiencia , Receptores Sensibles al Calcio/genética , Adulto , Anciano , Señalización del Calcio , Análisis Mutacional de ADN , Femenino , Células HEK293 , Homeostasis , Humanos , Hipercalciuria/metabolismo , Hipocalcemia/metabolismo , Hipoparatiroidismo/metabolismo , Mutación , Linaje , Receptores Sensibles al Calcio/metabolismo , Transfección
11.
Am J Med Genet A ; 158A(12): 3182-9, 2012 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-23166063

RESUMEN

Interstitial deletions of chromosome 15q25.2 are rare. To date, only nine patients with microdeletions within this chromosomal region have been described. Here, we report on a girl with severe speech and psychomotor delay, behavioral problems and mild dysmorphic features with a 1.6 Mb deletion in 15q25.2 region. In order to study the parental origin of the rearrangement, we analyzed selected SNPs in the deleted area in the patient and her parents, showing Mendelian incompatibilities suggesting a de novo deletion on the chromosome of maternal origin. By comparing the clinical and molecular features of our patient with five previously reported cases of an overlapping deletion, we suggest that 15q25.2 deletion is an emerging syndrome characterized by a distinct although variable spectrum of clinical manifestations, including mild dysmorphic features, neurodevelopmental delay, and a recognizable pattern of congenital malformation. Furthermore, our patient is the second one in which a behavioral phenotype characterized by hyperactivity, anxiety, and autistic features was reported, indicating that these features might be part of this new syndromic condition. Breakpoints of the deletion in the patient reported here are useful to better define the smallest region of overlap (SRO) among all the patients. Selected genes that are present in the hemizygous state and which might be important for the phenotype of these patients, are discussed in context of the clinical features. In conclusion, our patient increases the knowledge about the molecular and phenotypic consequences of interstitial 15q25.2 deletions, highlighting that deletions of this region may be responsible for a new microdeletion syndrome.


Asunto(s)
Deleción Cromosómica , Cromosomas Humanos Par 15 , Discapacidades del Desarrollo/genética , Eliminación de Secuencia , Anomalías Múltiples/genética , Niño , Femenino , Humanos , Fenotipo
12.
Mol Vis ; 17: 2482-94, 2011.
Artículo en Inglés | MEDLINE | ID: mdl-21976959

RESUMEN

PURPOSE: To evaluate the involvement of Visual System Homeobox 1 (VSX1), Secreted Protein Acidic and Rich in Cysteine (SPARC), Superoxide Dismutase 1 (SOD1), Lysyl Oxidase (LOX), and Tissue Inhibitor of Metalloproteinase 3 (TIMP3) in sporadic and familial keratoconus. METHODS: Mutational analysis of the five genes was performed by sequencing and fragment analysis in a large cohort of 302 Italian patients, with a diagnosis of keratoconus based on clinical examination and corneal topography. The variants identified in VSX1 and SPARC were also assessed in the available relatives of the probands. RESULTS: A novel mutation p.G239R and previously reported mutations were found in VSX1. Novel and already reported variants were identified in SPARC and SOD1, whose pathogenic significance has not been established. No pathogenic variants have been identified in LOX and TIMP3. CONCLUSIONS: Molecular analysis of the five genes in a cohort of 225 sporadic and 77 familial keratoconus cases confirms the possible pathogenic role of VSX1 though in a small number of patients; a possible involvement of LOX and TIMP3 could be excluded; and the role played by SOD1 and SPARC in determining the disease as not been definitively clarified. Further studies are required to identify other important genetic factors involved in the pathogenesis and progression of the disease that in the authors' opinion, and according with several authors, should be considered as a complex disease.


Asunto(s)
Córnea/metabolismo , Proteínas del Ojo/genética , Proteínas de Homeodominio/genética , Queratocono/genética , Osteonectina/genética , Proteína-Lisina 6-Oxidasa/genética , Superóxido Dismutasa/genética , Inhibidor Tisular de Metaloproteinasa-3/genética , Adolescente , Adulto , Secuencia de Bases , Niño , Estudios de Cohortes , Córnea/patología , Topografía de la Córnea , Análisis Mutacional de ADN , Pruebas Genéticas , Humanos , Italia , Queratocono/epidemiología , Persona de Mediana Edad , Datos de Secuencia Molecular , Mutación , Linaje , Polimorfismo de Nucleótido Simple , Superóxido Dismutasa-1
13.
Eur J Orthod ; 33(5): 498-502, 2011 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-21131390

RESUMEN

Cleidocranial dysplasia (CCD) is a rare, well-defined skeletal disorder with autosomal dominant inheritance and complete penetrance. Although it involves the whole skeletal system, the main clinical manifestations of CCD are malformations of the skull and clavicles, which lead to a typical appearance of the face and shoulders. Dental aspects are particularly evident and often eruption difficulties are the first indication for the patient, who does not present any other problem. It has been established that insufficiency of the RUNX-related transcription factor, the core-binding factor alpha 1 (CBFA1) protein, causes CCD. This protein is essential in skeletal development by regulating osteoblast differentiation and chondrocyte maturation. CBFA1 protein is encoded by the RUNX2 gene located on chromosome 6p21. The molecular characterization of the novel RUNX2 gene mutation c.580 + 1G > A in an Italian family (a 27-year-old female, her 54-year-old mother and 24-year-old sister) affected by the typical CCD phenotype, which was proven to alter splicing of the RUNX2 messenger RNA, underscoring the contribution of novel altered splicing mechanism to the aetiology of this disease is presented.


Asunto(s)
Displasia Cleidocraneal/genética , Subunidad alfa 1 del Factor de Unión al Sitio Principal/genética , ARN Mensajero/genética , Adulto , Empalme Alternativo , Femenino , Humanos , Persona de Mediana Edad , Linaje , Adulto Joven
14.
Biochim Biophys Acta ; 1792(1): 27-32, 2009 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-19027848

RESUMEN

The role of myosins in the pathogenesis of hearing loss is well established: five genes encoding unconventional myosins and two genes encoding nonmuscle conventional myosins have so far been described to be essential for normal auditory function and mutations in these genes associated with hearing impairment. To better understand the role of this gene family we performed a mutational screening on two candidate genes, MYO1C and MYO1F, analyzing hundreds of patients, affected by bilateral sensorineural hearing loss and coming from different European countries. This research activity led to the identification of 6 heterozygous missense mutations in MYO1C and additional 5 heterozygous missense mutations in MYO1F. Homology modelling suggests that some of these mutations could have a potential influence on the structure of the ATP binding site and could probably affect the ATPase activity or the actin binding process of both myosins. This study suggests a role of the above mentioned myosin genes in the pathogenesis of hearing loss.


Asunto(s)
Pérdida Auditiva Sensorineural/genética , Miosina Tipo I/genética , Secuencia de Bases , Análisis Mutacional de ADN , Cartilla de ADN/genética , Variación Genética , Heterocigoto , Humanos , Modelos Moleculares , Mutación Missense , Miosina Tipo I/química , Estructura Terciaria de Proteína
15.
Mol Genet Metab ; 99(1): 42-52, 2010 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-19782624

RESUMEN

Cystinuria is a rare inherited disorder characterized by defective renal reabsorption of cystine and the dibasic amino acids. SLC3A1 and SLC7A9 have been identified as responsible genes. The large majority of the more than 200 mutations so far identified in the two genes are point mutations, while only few alleles carrying gross genomic alterations have been reported. We screened 39 cystinuric patients for large rearrangements, by two home-made multiplex ligation-dependent probe amplification (MLPA) assays. MLPA analysis led to the identification of 6 different alleles in SLC3A1 and 2 in SLC7A9 accounting for a total of 25 copy number changes, 11 in SLC3A1 and 14 in SLC7A9. Three large rearrangements in SLC3A1, deletion of exons 2-4 (E2_E4del), deletion of exons 5-6 (E5_E6del) and duplication of exons 8-9 (E8_E9dup) are novel. A complete SLC7A9 gene deletion was found in three patients. In addition, we report the identification of three novel point mutations in SLC7A9 (p.G105E, p.R250K, c.1416_1417insAC), the frequency and the occurrence of cystinuria mutations in a cohort of 172 Italian patients. In conclusion, we developed a reliable and robust MLPA analytic method for SLC3A1 and SLC7A9 genes that represents an optimal complement to DNA sequence analysis in patients with cystinuria, enabling the screening for deletions and duplications.


Asunto(s)
Sistemas de Transporte de Aminoácidos Básicos/genética , Sistemas de Transporte de Aminoácidos Neutros/genética , Cistinuria/genética , Mutación Puntual , Estudios de Cohortes , Cistinuria/diagnóstico , Análisis Mutacional de ADN , Salud de la Familia , Femenino , Frecuencia de los Genes , Reordenamiento Génico , Pruebas Genéticas/métodos , Humanos , Italia , Masculino , Técnicas de Amplificación de Ácido Nucleico/métodos , Polimorfismo de Nucleótido Simple , Reproducibilidad de los Resultados , Sensibilidad y Especificidad
17.
Am J Med Genet A ; 152A(7): 1756-63, 2010 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-20583153

RESUMEN

We report on a boy with three cell lines: 46,XY, r(11)(p15.5,q25)[90]/45,XY,-11 [8]/47,XY, r(11)(p15.5,q25)x2[2], with minor anomalies and mental retardation who developed asynchronous bilateral Wilms tumors (WTs). Array comparative genomic hybridization (CGH) performed on peripheral blood leukocytes of the patient led to the identification of a constitutional duplication of 4.8 Mb at 11p15.5-11p15.4. This duplication was found to involve the chromosome of paternal origin, and occurred in tandem on the ring chromosome 11. Despite the constitutive duplication of the paternal 11p15 chromosome region, the patient showed no sign of Beckwith-Wiedemann syndrome. However, the molecular characterization of the two neoplasias was consistent with their independent origin and showed that they arose from the two distinct cellular clones with the ring chromosome, indicating that this anomaly is likely to have caused the patient's susceptibility to WT development.


Asunto(s)
Cromosomas Humanos Par 11/genética , Análisis Citogenético , Mosaicismo , Cromosomas en Anillo , Tumor de Wilms/genética , Tumor de Wilms/patología , Adulto , Preescolar , Hibridación Genómica Comparativa , Variaciones en el Número de Copia de ADN/genética , Metilación de ADN/genética , Femenino , Genotipo , Humanos , Hibridación Fluorescente in Situ , Lactante , Recién Nacido , Cariotipificación , Leucocitos Mononucleares/metabolismo , Masculino , Embarazo , Regiones Promotoras Genéticas/genética , Adulto Joven
18.
Artículo en Inglés | MEDLINE | ID: mdl-20798775

RESUMEN

Cerebral cavernous malformations (CCMs) represent a common autosomal dominant disorder that predisposes patients to haemorrhagic strokes and focal neurological signs. About 56% of the hereditary forms of CCMs have been so far associated with mutations in the KRIT1 (Krev Interaction Trapped 1) gene, located at 7q21.2 (CCM1 locus). We described the complete loss of 7q21.2 locus encompassing the KRIT1 gene and 4 flanking genes in a CCM family by using a dense set of 12 microsatellite markers. The complete loss of the maternal copy of KRIT1 gene region was confirmed by Real-Time Quantitative Polymerase Chain Reaction (RT-QPCR) and the same approach was used for expression analysis. Additional RT-QPCR analysis showed the extension of the deletion, for a total of 700 kb, to the adjacent downstream and upstream-located genes, MTERF, AKAP9, CYP51A1, as well as a partial loss of the ANKIB1 gene. Here we report the molecular characterization of an interstitial small genomic deletion of the 7q21.2 region in a CCMs affected family, encompassing the KRIT1 gene. Our findings confirm the loss of function mechanism for the already known CCM1 locus, without any evident involvement of the other deleted genes. Moreover, our investigations highlight the usefulness of the RT-QPCR to the molecular characterization of the breakpoints genomic deletions and to the identification of internal deleted genes involved in the human genetic diseases.


Asunto(s)
Cromosomas Humanos Par 7/genética , Hemangioma Cavernoso/genética , Malformaciones Arteriovenosas Intracraneales/genética , Proteínas Asociadas a Microtúbulos/genética , Polimorfismo Genético/genética , Proteínas Proto-Oncogénicas/genética , Eliminación de Secuencia , Adulto , Neoplasias Encefálicas/genética , Niño , Femenino , Humanos , Proteína KRIT1 , Masculino , Proteínas Asociadas a Microtúbulos/deficiencia , Persona de Mediana Edad , Linaje , Proteínas Proto-Oncogénicas/deficiencia , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa
19.
J Biomed Biotechnol ; 2009: 860761, 2009.
Artículo en Inglés | MEDLINE | ID: mdl-20145706

RESUMEN

There are many well-studied examples of human phenotypes resulting from nonsense or frameshift mutations that are modulated by Nonsense-Mediated mRNA Decay (NMD), a process that typically degrades transcripts containing premature termination codons (PTCs) in order to prevent translation of unnecessary or aberrant transcripts. Different types of germline mutations in the VHL gene cause the von Hippel-Lindau disease, a dominantly inherited familial cancer syndrome with a marked phenotypic variability and age-dependent penetrance. By generating the Drosophila UAS:Upf1(D45B) line we showed the possible involvement of NMD mechanism in the modulation of the c.172delG frameshift mutation located in the exon 1 of Vhl gene. Further, by Quantitative Real-time PCR (QPCR) we demonstrated that the corresponding c.163delG human mutation is targeted by NMD in human HEK 293 cells. The UAS:Upf1(D45B) line represents a useful system to identify novel substrates of NMD pathway in Drosophila melanogaster. Finally, we suggest the possible role of NMD on the regulation of VHL mutations.


Asunto(s)
Drosophila melanogaster/genética , Mutación del Sistema de Lectura , Estabilidad del ARN/fisiología , Proteína Supresora de Tumores del Síndrome de Von Hippel-Lindau/genética , Alcohol Deshidrogenasa/genética , Alcohol Deshidrogenasa/metabolismo , Animales , Animales Modificados Genéticamente , Línea Celular , Proteínas de Drosophila/genética , Proteínas de Drosophila/metabolismo , Femenino , Humanos , Proteínas de Insectos/genética , Proteínas de Insectos/metabolismo , Masculino , Ovario/química , Ovario/metabolismo , Reacción en Cadena de la Polimerasa , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Testículo/química , Testículo/metabolismo , Factores de Transcripción/genética , Factores de Transcripción/metabolismo , Proteína Supresora de Tumores del Síndrome de Von Hippel-Lindau/metabolismo
20.
J Nephrol ; 22(1): 152-9, 2009.
Artículo en Inglés | MEDLINE | ID: mdl-19229831

RESUMEN

BACKGROUND: IgA nephropathy (IgAN) is a common form of primary glomerulonephritis characterized by diffuse glomerular mesangial IgA1 deposition that leads to mesangial proliferation and chronic glomerular inflammation. Analyses of serum IgA1 from IgAN patients revealed an abnormal galactosylation of the O-linked carbohydrate moieties of IgA that may be a result of altered activity of core 1 beta1,3-galactosyltransferase (C1GalT1). To evaluate the association between C1GalT1 single nucleotide polymorphisms (SNPs) and IgAN, we performed a case control study on cohorts from the Italian population. METHODS: We sequenced C1GalT1 coding and promoter regions in 284 IgAN patients and 210 healthy controls. The functional role of 3' untranslated region (3'UTR) SNPs was studied using electrophoretic mobility shift assays and real-time quantitative PCR. RESULTS: We analyzed 8 SNPs in the C1GalT1 gene: 5 SNPs were in the promoter region and 3 SNPs in the 3'UTR. The allele 1365G in the 3'UTR was significantly more frequent in IgAN patients than in healthy controls. CONCLUSION: The 1365G allele and 1365G/G genotype seem to confer susceptibility to IgAN.


Asunto(s)
Galactosiltransferasas/genética , Predisposición Genética a la Enfermedad/genética , Glomerulonefritis por IGA/genética , Polimorfismo de Nucleótido Simple/genética , Adulto , Alelos , Estudios de Casos y Controles , Femenino , Galactosiltransferasas/sangre , Genotipo , Glomerulonefritis por IGA/etnología , Humanos , Inmunoglobulina A/sangre , Italia , Masculino , Persona de Mediana Edad , Regiones Promotoras Genéticas/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA