Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 21
Filtrar
Mais filtros

Base de dados
País/Região como assunto
Tipo de documento
Intervalo de ano de publicação
1.
Hum Mol Genet ; 22(3): 544-57, 2013 Feb 01.
Artigo em Inglês | MEDLINE | ID: mdl-23118352

RESUMO

At chromosome 11p15.5, the imprinting centre 1 (IC1) controls the parent of origin-specific expression of the IGF2 and H19 genes. The 5 kb IC1 region contains multiple target sites (CTS) for the zinc-finger protein CTCF, whose binding on the maternal chromosome prevents the activation of IGF2 and allows that of H19 by common enhancers. CTCF binding helps maintaining the maternal IC1 methylation-free, whereas on the paternal chromosome gamete-inherited DNA methylation inhibits CTCF interaction and enhancer-blocking activity resulting in IGF2 activation and H19 silencing. Maternally inherited 1.4-2.2 kb deletions are associated with methylation of the residual CTSs and Beckwith-Wiedemann syndrome, although with different penetrance and expressivity. We explored the relationship between IC1 microdeletions and phenotype by analysing a number of previously described and novel mutant alleles. We used a highly quantitative assay based on next generation sequencing to measure DNA methylation in affected families and analysed enhancer-blocking activity and CTCF binding in cultured cells. We demonstrate that the microdeletions mostly affect IC1 function and CTCF binding by changing CTS spacing. Thus, the extent of IC1 inactivation and the clinical phenotype are influenced by the arrangement of the residual CTSs. A CTS spacing similar to the wild-type allele results in moderate IC1 inactivation and is associated with stochastic DNA methylation of the maternal IC1 and incomplete penetrance. Microdeletions with different CTS spacing display severe IC1 inactivation and are associated with IC1 hypermethylation and complete penetrance. Careful characterization of the IC1 microdeletions is therefore needed to predict recurrence risks and phenotypical outcomes.


Assuntos
Deleção de Genes , Impressão Genômica , Fator de Crescimento Insulin-Like II/genética , Fenótipo , RNA Longo não Codificante/genética , Proteínas Repressoras/genética , Alelos , Sítios de Ligação/genética , Fator de Ligação a CCCTC , Células Cultivadas , Imunoprecipitação da Cromatina , Cromossomos Humanos Par 11/genética , Metilação de DNA , Regulação da Expressão Gênica , Inativação Gênica , Loci Gênicos , Humanos , Fator de Crescimento Insulin-Like II/metabolismo , Linhagem , RNA Longo não Codificante/metabolismo , Proteínas Repressoras/metabolismo , Análise de Sequência de DNA
2.
BMC Med Genet ; 16: 69, 2015 Aug 23.
Artigo em Inglês | MEDLINE | ID: mdl-26297663

RESUMO

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.


Assuntos
Anormalidades Múltiplas/genética , Deleção Cromossômica , Cromossomos Humanos Par 6/genética , Transtornos do Crescimento/genética , Hormônio do Crescimento/deficiência , Anormalidades Múltiplas/tratamento farmacológico , Anormalidades Múltiplas/patologia , Hibridização Genômica Comparativa , Ecocardiografia , Seguimentos , Transtornos do Crescimento/tratamento farmacológico , Transtornos do Crescimento/patologia , Hormônio do Crescimento/administração & dosagem , Humanos , Itália , Cariotipagem , Imageamento por Ressonância Magnética , Masculino
3.
J Hum Genet ; 60(6): 287-93, 2015 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-25809938

RESUMO

Silver-Russell syndrome (SRS) is a heterogeneous disorder characterized by intrauterine and post-natal growth retardation, dysmorphic facial features and body asymmetry. About 50% of the patients carry (epi)genetic alterations involving chromosomes 7 or 11.The high proportion of patients with unidentified molecular etiology suggests the involvement of other genes. Interestingly, SRS patients share clinical features with the 12q14 microdeletion syndrome, characterized by several deletions with a 2.6 Mb region of overlap. Among the genes present in this interval, high mobility AT-hook 2 (HMGA2) appears to be the most likely cause of the growth deficiency, due to its described growth control function. To define the role of HMGA2 in SRS, we looked for 12q14 chromosome imbalances and HMGA2 mutations in a cohort of 45 patients with growth retardation and SRS-like phenotype but no 11p15 (epi)mutations or maternal uniparental disomy of chromosome 7 (matUPD7). We identified a novel 7 bp intronic deletion in HMGA2 present in heterozygosity in the proband and her mother both displaying the typical features of SRS. We demonstrated that the deletion affected normal splicing, indicating that it is a likely cause of HMGA2 deficiency. This study provides the first evidence that a loss-of-function mutation of HMGA2 can be associated with a familial form of SRS. We suggest that HMGA2 mutations leading to haploinsufficiency should be investigated in the SRS patients negative for the typical 11p15 (epi)mutations and matUPD7.


Assuntos
Proteína HMGA2/genética , Síndrome de Silver-Russell/genética , Sequência de Bases , Estudos de Casos e Controles , Pré-Escolar , Análise Mutacional de DNA , Feminino , Estudos de Associação Genética , Humanos , Linhagem , Fenótipo , Sítios de Splice de RNA , Deleção de Sequência
4.
Hum Mol Genet ; 21(1): 10-25, 2012 Jan 01.
Artigo em Inglês | MEDLINE | ID: mdl-21920939

RESUMO

A cluster of imprinted genes at chromosome 11p15.5 is associated with the growth disorders, Silver-Russell syndrome (SRS) and Beckwith-Wiedemann syndrome (BWS). The cluster is divided into two domains with independent imprinting control regions (ICRs). We describe two maternal 11p15.5 microduplications with contrasting phenotypes. The first is an inverted and in cis duplication of the entire 11p15.5 cluster associated with the maintenance of genomic imprinting and with the SRS phenotype. The second is a 160 kb duplication also inverted and in cis, but resulting in the imprinting alteration of the centromeric domain. It includes the centromeric ICR (ICR2) and the most 5' 20 kb of the non-coding KCNQ1OT1 gene. Its maternal transmission is associated with ICR2 hypomethylation and the BWS phenotype. By excluding epigenetic mosaicism, cell clones analysis indicated that the two closely located ICR2 sequences resulting from the 160 kb duplication carried discordant DNA methylation on the maternal chromosome and supported the hypothesis that the ICR2 sequence is not sufficient for establishing imprinted methylation and some other property, possibly orientation-dependent, is needed. Furthermore, the 1.2 Mb duplication demonstrated that all features are present for correct imprinting at ICR2 when this is duplicated and inverted within the entire cluster. In the individuals maternally inheriting the 160 kb duplication, ICR2 hypomethylation led to the expression of a truncated KCNQ1OT1 transcript and to down-regulation of CDKN1C. We demonstrated by chromatin RNA immunopurification that the KCNQ1OT1 RNA interacts with chromatin through its most 5' 20 kb sequence, providing a mechanism likely mediating the silencing activity of this long non-coding RNA.


Assuntos
Síndrome de Beckwith-Wiedemann/genética , Impressão Genômica , RNA não Traduzido/genética , Síndrome de Silver-Russell/genética , Adulto , Síndrome de Beckwith-Wiedemann/metabolismo , Pré-Escolar , Cromatina/genética , Cromatina/metabolismo , Cromossomos Humanos Par 11/genética , Cromossomos Humanos Par 11/metabolismo , Metilação de DNA , Feminino , Duplicação Gênica , Inativação Gênica , Humanos , Lactente , Masculino , Linhagem , Canais de Potássio de Abertura Dependente da Tensão da Membrana/genética , Ligação Proteica , RNA não Traduzido/metabolismo , Síndrome de Silver-Russell/metabolismo
5.
Am J Med Genet A ; 164A(1): 182-5, 2014 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-24214456

RESUMO

Beckwith-Wiedemann syndrome (BWS) is an overgrowth disorder with increased risk of embryonal tumors, such as Wilms tumor, hepatoblastoma, neuroblastoma, and rhabdomyosarcoma. We report on a patient with BWS that developed a giant fibroadenoma of the breast that was surgically removed. The tumor relapsed 8 months after the surgery and the patient underwent partial mastectomy. Although the patient presented several clinical features of BWS, a molecular diagnosis was not achieved despite extensive molecular investigations on both blood and tumor tissue. A SNP array revealed a de novo 7p22.1 loss in both blood and breast tumor involving the mismatch repair gene PMS2 gene that may be potentially associated with the breast tumor. In conclusion, it remains unclear whether BWS patients have an increased risk of breast lesions or a yet unknown molecular defect is responsible for the rare occurrence of this tumor in BWS.


Assuntos
Síndrome de Beckwith-Wiedemann/complicações , Neoplasias da Mama/complicações , Adolescente , Síndrome de Beckwith-Wiedemann/diagnóstico , Neoplasias da Mama/diagnóstico , Neoplasias da Mama/cirurgia , Aberrações Cromossômicas , Hibridização Genômica Comparativa , Metilação de DNA , Feminino , Fibrose , Humanos , Hiperplasia
6.
J Med Genet ; 50(2): 99-103, 2013 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-23243085

RESUMO

BACKGROUND: Heterogeneous molecular defects affecting the 11p15.5 imprinted gene cluster are associated with the opposite growth disorders Beckwith-Wiedemann Syndrome (BWS) and Silver Russell syndrome (SRS). Maternal deletions of the centromeric domain usually result in BWS, but paternal deletions have been so far associated with normal phenotype. Here we describe a case of recurrent severe Intra-Uterine Growth Restriction (IUGR) with paternal transmission of an 11p15.5 60 kb deletion. METHODS AND RESULTS: Chromosome microarray (CMA), PCR and DNA sequencing analyses showed that two fetuses conceived by a normal couple inherited from their father a 60 kb deletion encompassing the Imprinting Control Region of the 11p15.5 centromeric domain. The two fetuses died in utero with severe growth restriction. PCR amplification of parental DNAs indicated that the father carried the mutation in the mosaic state. DNA methylation and gene expression analyses showed that the deletion led to an imprinting alteration restricted to the centromeric domain and resulting in silencing of KCNQ1OT1 and activation of CDKN1C and PHLDA2. CONCLUSIONS: Our data demonstrate that the phenotype associated with 11p15.5 deletions is strongly influenced by the size of the region involved and indicate imprinting defects leading to CDKN1C and PHLDA2 activation as cause of severe IUGR.


Assuntos
Cromossomos Humanos Par 11 , Retardo do Crescimento Fetal/genética , Impressão Genômica , Deleção de Sequência , Inibidor de Quinase Dependente de Ciclina p57/genética , Metilação de DNA , Feminino , Morte Fetal/genética , Humanos , Masculino , Proteínas Nucleares/genética , Análise de Sequência com Séries de Oligonucleotídeos , Linhagem , Fenótipo , Canais de Potássio de Abertura Dependente da Tensão da Membrana/genética , Gravidez
7.
Am J Med Genet A ; 161A(10): 2652-5, 2013 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-24038823

RESUMO

Silver-Russell syndrome (SRS) is a clinically and genetically heterogeneous syndrome characterized by severe intrauterine and postnatal growth retardation, facial dysmorphism and body asymmetry. One of the main molecular mechanisms leading to the syndrome involves methylation abnormalities of chromosome 11p15. In the last decades, an increase of imprinting disorders have been reported in children born from assisted reproductive technology (ART); however there is currently little evidence linking SRS and ART. Only few infants with SRS born using ART, supported by molecular analysis, have been described. We report on a twin-girl conceived using intracytoplasmic sperm injection (ICSI) diagnosed with SRS. Molecular studies revealed a hypomethylation of the paternal H19/IGF2 Imprinting Control Region. Her twin sister had a normal prenatal and postnatal growth and a normal methylation pattern of the chromosome 11p15. This is the second reported case of a twin infant with SRS conceived using ART with hypomethylation of H19/IGF2; it provides additional evidence of a possible relationship between ART procedures and methylation defects observed in SRS. Given the clinical heterogeneity of SRS, and the increased risk of multiple and preterm births in the ART-conceived children, it is possible that a number of cases of SRS remains undiagnosed in this population. Future studies should investigate the possible link between ART and SRS, in order to better understand the causes of epimutations in ART pregnancies, and to help clinicians to adequately counsel parents who approach to ART and to assess the opportunity of a long-term follow-up of children conceived using ART.


Assuntos
Metilação de DNA , Fator de Crescimento Insulin-Like II/genética , RNA Longo não Codificante/genética , Síndrome de Silver-Russell/genética , Gêmeos , Aberrações Cromossômicas , Cromossomos Humanos Par 11 , Feminino , Fertilização in vitro , Impressão Genômica , Humanos , Recém-Nascido , Síndrome de Silver-Russell/diagnóstico
8.
Am J Med Genet A ; 161A(10): 2481-6, 2013 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-23918458

RESUMO

Although Beckwith-Wiedemann syndrome (BWS, OMIM #130650) is the most common genetic overgrowth disorder, data on its epidemiology are scanty and the estimates of its occurrence show wide variability. The aim of this study is to assess its prevalence in Piedmont Region (Italy). We included in the study all patients diagnosed with BWS born in Piedmont from 1997 to 2009 through a search in the Italian Registry for Rare Diseases. This source was further validated with data from the network of Regional Clinical Genetics services and surveys in extra-regional Clinical Genetics centres, laboratories and the Italian BWS patients association. All cases were further ascertained through physical exam, medical history and specific molecular tests. The search identified 46 clear-cut cases of BWS born across the 13-year period, providing a prevalence of 1:10 340 live births (95% confidence interval 1:7,752-13,698 live births). Among the 41 patients who underwent molecular tests, 70.7% were positive, showing hypomethylation of the IC2 imprinting center (29.3%), paternal chromosome 11 uniparental disomy (pUPD11, 24.4%), IC1 hypermethylation (14.6%), CDKN1c mutation (2.4%), whereas 29.3% had negative molecular tests. The study provides an approximate BWS prevalence of 1:10,000 live birth, the highest reported to date.


Assuntos
Síndrome de Beckwith-Wiedemann/epidemiologia , Síndrome de Beckwith-Wiedemann/diagnóstico , Feminino , Humanos , Itália/epidemiologia , Masculino , Vigilância da População , Prevalência
9.
Pediatr Nephrol ; 27(3): 397-406, 2012 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-22015620

RESUMO

Beckwith-Wiedemann syndrome (BWS), an overgrowth disorder with several congenital abnormalities, encompasses nephrourological anomalies. The objective of the report is to analyze the latter and related genotype-phenotype correlations. The study was a retrospective review of nephrourological investigations and genotype in 67 BWS patients. Imaging and laboratory studies have been correlated with the molecular anomalies typical of BWS. Thirty-eight (56.7%) patients had a total of 61 nonmalignant nephrourological findings, including nephromegaly (n = 24), collecting system abnormalities (n = 14), cryptorchidism (n = 11), nephrolithiasis (n = 5), cysts (n = 5), and dysplasia (n = 1). Four patients had Wilms' tumor, all associated with renal hyperplasia. Renal findings were almost consistent in the BWS(IC1) group, with nephromegaly in all patients and collecting system abnormalities in half of them. BWS(UPD) and negative patients also had frequent anomalies (63.6% and 61.9% respectively), whereas only 36.0% of BWS(IC2) had renal findings (p = 0.003). Cryptorchidism was associated with abdominal wall defects (p < 0.001) appearing more frequently in BWS(IC2) (p = 0.028). Urinary tract infections were observed in 17.9% of patients, with two resulting in life-threatening sepsis. Hypercalciuria was present in 10% of cases. 55.5% of BWS patients have renal findings. Although variegate, these anomalies disclose a genotype-phenotype correlation.


Assuntos
Síndrome de Beckwith-Wiedemann/patologia , Rim/anormalidades , Adolescente , Síndrome de Beckwith-Wiedemann/complicações , Síndrome de Beckwith-Wiedemann/genética , Criança , Pré-Escolar , Feminino , Genótipo , Humanos , Hipercalciúria/etiologia , Lactente , Rim/patologia , Masculino , Fenótipo , Estudos Retrospectivos , Infecções Urinárias/etiologia
10.
Eur J Pediatr ; 170(11): 1407-11, 2011 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-21448630

RESUMO

Beckwith-Wiedemann syndrome is an overgrowth disorder characterized by neonatal macrosomia, abdominal wall defects, macroglossia, renal anomalies, organomegaly, hypoglycemia, and cancer predisposition. Hepatoblastoma is the second most frequent tumor and periodic serum alpha-fetoprotein (αFP) dosage is the cornerstone of the tumor surveillance for its early detection. In this report, we describe the outstanding case of a Beckwith-Wiedemann syndrome (BWS) newborn with severe phenotype and paternal chromosome 11 uniparental disomy (UPD11) associated with a high tumor risk. Based on the clinical picture and previous reports, a close monitoring of αFP was commenced. The marker was normal immediately after birth, but rapidly raised in 20 days, leading to the diagnosis of an extremely aggressive hepatoblastoma. The latter was successfully treated with pre-surgical reductive chemotherapy, gross total mass resection, and subsequent chemotherapy. Based on this observation, the tumor surveillance routinely suggested every 3 months should be more intense and with closer time intervals in newborns with severe BWS phenotype. We suggest monitoring neonatal αFP every 20 days in such cases.


Assuntos
Síndrome de Beckwith-Wiedemann , Hepatoblastoma/diagnóstico , Neoplasias Hepáticas/diagnóstico , alfa-Fetoproteínas/metabolismo , Síndrome de Beckwith-Wiedemann/genética , Inibidor de Quinase Dependente de Ciclina p57/metabolismo , Predisposição Genética para Doença , Hepatoblastoma/genética , Humanos , Recém-Nascido , Fator de Crescimento Insulin-Like II/metabolismo , Neoplasias Hepáticas/genética , Masculino
11.
J Med Genet ; 47(6): 429-32, 2010 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-19843502

RESUMO

BACKGROUND Beckwith-Wiedemann syndrome (BWS) is a clinically variable and genetically heterogeneous disorder, providing evidence that imprinted genes play key roles in the control of fetal growth. Clinically, diagnostic criteria include macrosomia, macroglossia, abdominal wall defects, neonatal hypoglycaemia, visceromegalies and hemihyperplasia. Component clinical manifestations also include renal abnormalities, adrenocortical cytomegaly and a characteristic facial appearance, with midface hypoplasia and ear anomalies. Genetically, BWS is associated with disturbances within two different domains on 11p15 that are controlled by distinct imprinting control regions (ICR), ICR1 and ICR2. The majority of patients have abnormalities within ICR2. In particular, loss of maternal methylation accounts for 50-60% of cases, and is associated with reduction in the expression of the CDKN1C gene, a member of the cyclin dependent kinase inhibitor family acting as negative regulator of cell proliferation. Mutations in CDKN1C are detected in another 5-10% of subjects with sporadic BWS. Chromosome deletions affecting ICR2 are uncommon. METHODS AND FINDINGS We report on a patient with BWS in which a de novo 11p15 deletion was detected by array comparative genomic hybridisation. Clinically, the patient presented with mild mental retardation and minor physical anomalies. The deletion, that was demonstrated to be maternal in origin by SNP array, encompassed ICR2 and several flanking genes, including CDKN1C. A normal methylation pattern of ICR1 was observed. CONCLUSIONS This observation provides evidence that, among the genetic defects associated with BWS, a 11p15 microdeletion encompassing ICR2 identifies a peculiar clinical phenotype, with high recurrence risk in offspring of female carriers. It also supports the model of two independent domains within the BWS locus.


Assuntos
Síndrome de Beckwith-Wiedemann/genética , Centrômero/genética , Deleção Cromossômica , Cromossomos Humanos Par 11/genética , Adolescente , Síndrome de Beckwith-Wiedemann/patologia , Hibridização Genômica Comparativa , Feminino , Impressão Genômica/genética , Humanos
12.
Hum Mol Genet ; 17(10): 1427-35, 2008 May 15.
Artigo em Inglês | MEDLINE | ID: mdl-18245780

RESUMO

The parent of origin-dependent expression of the IGF2 and H19 genes is controlled by the imprinting centre 1 (IC1) consisting in a methylation-sensitive chromatin insulator. Deletions removing part of IC1 have been found in patients affected by the overgrowth- and tumour-associated Beckwith-Wiedemann syndrome (BWS). These mutations result in the hypermethylation of the remaining IC1 region, loss of IGF2/H19 imprinting and fully penetrant BWS phenotype when maternally transmitted. We now report that 12 additional cases with IC1 hypermethylation have a similar clinical phenotype but showed neither a detectable deletion nor other mutation in the local vicinity. Likewise, no IC1 deletion was detected in 40 sporadic non-syndromic Wilms' tumours. A detailed analysis of the BWS patients showed that the hypermethylation variably affected the IC1 region and was generally mosaic. We observed that all these cases were sporadic and in at least two families affected and unaffected members shared the same maternal IC1 allele but not the abnormal maternal chromosome epigenotype. Furthermore, the chromosome with the imprinting defect derived from either the maternal grandfather or maternal grandmother. Overall, these results indicate that methylation-imprinting defects at the IGF2-H19 locus can result from inherited mutations of the IC and have high recurrence risk or arise independently from the sequence context and generally not transmitted to the progeny. Despite these differences, the epigenetic abnormalities are usually present in the patients in the mosaic form and probably acquired by post-zygotic de novo methylation. Distinguishing between these two groups of cases is important for genetic counselling.


Assuntos
Síndrome de Beckwith-Wiedemann/genética , Impressão Genômica , Fator de Crescimento Insulin-Like II/genética , RNA não Traduzido/genética , Tumor de Wilms/genética , Alelos , Síndrome de Beckwith-Wiedemann/diagnóstico , Fator de Ligação a CCCTC , Segregação de Cromossomos , Cromossomos Humanos Par 11 , Metilação de DNA , Proteínas de Ligação a DNA/genética , Feminino , Deleção de Genes , Haplótipos , Humanos , Itália , Masculino , Mutação , Linhagem , RNA Longo não Codificante , Proteínas Repressoras/genética
13.
Endocr Dev ; 14: 1-9, 2009.
Artigo em Inglês | MEDLINE | ID: mdl-19293570

RESUMO

The parent-of-origin-dependent expression of IGF2 and H19 is controlled by the imprinting center 1 (IC1) consisting of a methylation-sensitive chromatin insulator. IC1 is normally methylated on the paternal chromosome and nonmethylated on the maternal chromosome. We found that 22 cases in a large cohort of patients affected by Beckwith-Wiedemann syndrome (BWS) had IC1 methylated on both parental chromosomes, resulting in biallelic activation of IGF2 and biallelic silencing of H19. These individuals had marked macrosomia and high incidence of Wilms' tumor. A subset of these patients had 1.4- to 1.8-kb deletions with hypermethylation of the remaining IC1 region and fully penetrant BWS phenotype when transmitted maternally. Another subset of individuals with IC1 hypermethylation had a similar clinical phenotype but no mutation in the local vicinity. All these cases were sporadic and in at least two families affected and unaffected members shared the same maternal IC1 allele but not the abnormal maternal epigenotype. Similarly, no IC1 deletion was detected in 10 nonsyndromic Wilms' tumors with IC1 hypermethylation. In conclusion, methylation defects at the IGF2-H19 locus can result from inherited mutations of the imprinting center and have high recurrence risk or arise independently from the sequence context and not transmitted to the progeny.


Assuntos
Síndrome de Beckwith-Wiedemann/genética , Epigênese Genética , Fator de Crescimento Insulin-Like II/genética , Neoplasias Renais/genética , Tumor de Wilms/genética , Metilação de DNA , Humanos , Mutação
14.
Eur J Hum Genet ; 24(2): 183-90, 2016 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-25898929

RESUMO

Beckwith-Wiedemann syndrome (BWS) is characterized by cancer predisposition, overgrowth and highly variable association of macroglossia, abdominal wall defects, nephrourological anomalies, nevus flammeus, ear malformations, hypoglycemia, hemihyperplasia, and organomegaly. BWS molecular defects, causing alteration of expression or activity of the genes regulated by two imprinting centres (IC) in the 11p15 chromosomal region, are also heterogeneous. In this paper we define (epi)genotype-phenotype correlations in molecularly confirmed BWS patients. The characteristics of 318 BWS patients with proven molecular defect were compared among the main four molecular subclasses: IC2 loss of methylation (IC2-LoM, n=190), IC1 gain of methylation (IC1-GoM, n=31), chromosome 11p15 paternal uniparental disomy (UPD, n=87), and cyclin-dependent kinase inhibitor 1C gene (CDKN1C) variants (n=10). A characteristic growth pattern was found in each group; neonatal macrosomia was almost constant in IC1-GoM, postnatal overgrowth in IC2-LoM, and hemihyperplasia more common in UPD (P<0.001). Exomphalos was more common in IC2/CDKN1C patients (P<0.001). Renal defects were typical of UPD/IC1 patients, uretheral malformations of IC1-GoM cases (P<0.001). Ear anomalies and nevus flammeus were associated with IC2/CDKN1C genotype (P<0.001). Macroglossia was less common among UPD patients (P<0.001). Wilms' tumor was associated with IC1-GoM or UPD and never observed in IC2-LoM patients (P<0.001). Hepatoblastoma occurred only in UPD cases. Cancer risk was lower in IC2/CDKN1C, intermediate in UPD, and very high in IC1 cases (P=0.009). In conclusion, (epi)genotype-phenotype correlations define four different phenotypic BWS profiles with some degree of clinical overlap. These observations impact clinical care allowing to move toward (epi) genotype-based follow-up and cancer screening.


Assuntos
Síndrome de Beckwith-Wiedemann/genética , Estudos de Associação Genética , Impressão Genômica , Neoplasias/genética , Síndrome de Beckwith-Wiedemann/complicações , Síndrome de Beckwith-Wiedemann/patologia , Cromossomos Humanos Par 11/genética , Inibidor de Quinase Dependente de Ciclina p57/genética , Metilação de DNA/genética , Feminino , Genótipo , Humanos , Masculino , Neoplasias/etiologia , Neoplasias/patologia , Fenótipo
15.
Orphanet J Rare Dis ; 6: 66, 2011 Oct 17.
Artigo em Inglês | MEDLINE | ID: mdl-22004918

RESUMO

BACKGROUND: Fabry disease is a rare disorder caused by a large variety of mutations in the gene encoding lysosomal alpha-galactosidase. Many of these mutations are unique to individual families. Fabry disease can be treated with enzyme replacement therapy, but a promising novel strategy relies on small molecules, so called "pharmacological chaperones", which can be administered orally. Unfortunately only 42% of genotypes respond to pharmacological chaperones. RESULTS: A procedure to predict which genotypes responsive to pharmacological chaperones in Fabry disease has been recently proposed. The method uses a position-specific substitution matrix to score the mutations. Using this method, we have screened public databases for predictable responsive cases and selected nine representative mutations as yet untested with pharmacological chaperones. Mutant lysosomal alpha galactosidases were produced by site directed mutagenesis and expressed in mammalian cells. Seven out of nine mutations responded to pharmacological chaperones. Nineteen other mutations that were tested with pharmacological chaperones, but were not included in the training of the predictive method, were gathered from literature and analyzed in silico. In this set all five mutations predicted to be positive were responsive to pharmacological chaperones, bringing the percentage of responsive mutations among those predicted to be positive and not used to train the classifier to 86% (12/14). This figure differs significantly from the percentage of responsive cases observed among all the Fabry mutants tested so far. CONCLUSIONS: In this paper we provide experimental support to an "in silico" method designed to predict missense mutations in the gene encoding lysosomal alpha galactosidase responsive to pharmacological chaperones. We demonstrated that responsive mutations can be predicted with a low percentage of false positive cases. Most of the mutations tested to validate the method were described in the literature as associated to classic or mild classic phenotype. The analysis can provide a guideline for the therapy with pharmacological chaperones supported by experimental results obtained in vitro. We are aware that our results were obtained in vitro and cannot be translated straightforwardly into benefit for patients, but need to be validated by clinical trials.


Assuntos
1-Desoxinojirimicina/farmacologia , Doença de Fabry/tratamento farmacológico , Chaperonas Moleculares/farmacologia , Mutação de Sentido Incorreto , alfa-Galactosidase/efeitos dos fármacos , alfa-Galactosidase/genética , 1-Desoxinojirimicina/uso terapêutico , Animais , Sequência de Bases , Células COS/metabolismo , Domínio Catalítico , Chlorocebus aethiops , Doença de Fabry/enzimologia , Doença de Fabry/genética , Feminino , Humanos , Masculino , Modelos Moleculares , Chaperonas Moleculares/uso terapêutico , Dados de Sequência Molecular , Muramidase , Mutagênese Sítio-Dirigida , Valor Preditivo dos Testes , alfa-Galactosidase/metabolismo
16.
Eur J Med Genet ; 54(4): e451-4, 2011.
Artigo em Inglês | MEDLINE | ID: mdl-21571108

RESUMO

The overgrowth disorder Beckwith-Wiedemann syndrome (BWS) is associated with dysregulation of imprinted genes at chromosome 11p15.5. The molecular defects are heterogeneous but most of the cases are associated with defective DNA methylation at either one of two Imprinting Control Regions (IC1 and IC2) or Uniparental paternal Disomy (UPD) at 11p15.5. In rare cases, the BWS phenotype has been found associated with maternal transmission of IC1 microdeletions. We describe a family with a novel 1.8 kb deletion that is associated with hypermethylation at IC1. The mutation results from recombination between highly homologous sequences containing target sites for the zinc-finger protein CTCF (CTSs). This finding supports the hypothesis that the function of IC1 and the penetrance of the clinical phenotype depend on the spacing of the CTSs resulting from recombination in the mutant allele.


Assuntos
Síndrome de Beckwith-Wiedemann/genética , Impressão Genômica/genética , Fator de Crescimento Insulin-Like II/genética , Mutação/genética , RNA não Traduzido/genética , Adulto , Deleção Cromossômica , Cromossomos Humanos Par 11/genética , Metilação de DNA/genética , Feminino , Ordem dos Genes , Genótipo , Humanos , Recém-Nascido , Masculino , Linhagem , Fenótipo , RNA Longo não Codificante , Dissomia Uniparental/genética
17.
Pediatr Dev Pathol ; 13(4): 326-30, 2010.
Artigo em Inglês | MEDLINE | ID: mdl-20028213

RESUMO

Genomic imprinting is an epigenetic phenomenon resulting in differential expression of maternal and paternal alleles of a subset of genes. In the mouse, mutation of imprinted genes often results in contrasting phenotypes, depending on parental origin. The overgrowth-associated Beckwith-Wiedemann syndrome (BWS) and the growth restriction-associated Silver-Russell syndrome (SRS) have been linked with a variety of epigenetic and genetic defects affecting a cluster of imprinted genes at chromosome 11p15.5. Paternally derived and maternally derived 11p15.5 duplications represent infrequent findings in BWS and SRS, respectively. Here, we report a case in which a 6.5 Mb duplication of 11p15.4-pter resulted in SRS and BWS phenotypes in a child and her mother, respectively. Molecular analyses demonstrated that the duplication involved the maternal chromosome 11p15 in the child and the paternal chromosome 11p15 in the mother. This observation provides a direct demonstration that SRS and BWS represent specular images, both at the clinical and molecular levels.


Assuntos
Síndrome de Beckwith-Wiedemann/genética , Cromossomos Humanos Par 11/genética , Duplicação Gênica , Impressão Genômica , Mães , Síndrome de Silver-Russell/genética , Adulto , Síndrome de Beckwith-Wiedemann/tratamento farmacológico , Síndrome de Beckwith-Wiedemann/patologia , Pré-Escolar , Hibridização Genômica Comparativa , Metilação de DNA , Feminino , Hormônio do Crescimento Humano/uso terapêutico , Humanos , Fenótipo , Síndrome de Silver-Russell/tratamento farmacológico , Síndrome de Silver-Russell/patologia , Dissomia Uniparental
19.
Eur J Hum Genet ; 17(5): 611-9, 2009 May.
Artigo em Inglês | MEDLINE | ID: mdl-19092779

RESUMO

Genomic imprinting is an epigenetic phenomenon restricting gene expression in a manner dependent on parent of origin. Imprinted gene products are critical regulators of growth and development, and imprinting disorders are associated with both genetic and epigenetic mutations, including disruption of DNA methylation within the imprinting control regions (ICRs) of these genes. It was recently reported that some patients with imprinting disorders have a more generalised imprinting defect, with hypomethylation at a range of maternally methylated ICRs. We report a cohort of 149 patients with a clinical diagnosis of Beckwith-Wiedemann syndrome (BWS), including 81 with maternal hypomethylation of the KCNQ1OT1 ICR. Methylation analysis of 11 ICRs in these patients showed that hypomethylation affecting multiple imprinted loci was restricted to 17 patients with hypomethylation of the KCNQ1OT1 ICR, and involved only maternally methylated loci. Both partial and complete hypomethylation was demonstrated in these cases, suggesting a possible postzygotic origin of a mosaic imprinting error. Some ICRs, including the PLAGL1 and GNAS/NESPAS ICRs implicated in the aetiology of transient neonatal diabetes and pseudohypoparathyroidism type 1b, respectively, were more frequently affected than others. Although we did not find any evidence for mutation of the candidate gene DNMT3L, these results support the hypotheses that trans-acting factors affect the somatic maintenance of imprinting at multiple maternally methylated loci and that the clinical presentation of these complex cases may reflect the loci and tissues affected with the epigenetic abnormalities.


Assuntos
Síndrome de Beckwith-Wiedemann/genética , Proteínas de Ciclo Celular/genética , Metilação de DNA , Subunidades alfa Gs de Proteínas de Ligação ao GTP/genética , Impressão Genômica , Fatores de Transcrição/genética , Proteínas Supressoras de Tumor/genética , Síndrome de Beckwith-Wiedemann/patologia , Cromograninas , DNA (Citosina-5-)-Metiltransferases/genética , Análise Mutacional de DNA , Feminino , Humanos , Masculino , Reação em Cadeia da Polimerase , Polimorfismo Genético
20.
PLoS One ; 3(3): e1849, 2008 Mar 26.
Artigo em Inglês | MEDLINE | ID: mdl-18365005

RESUMO

BACKGROUND: Differentially methylated regions (DMRs) are associated with many imprinted genes. In mice methylation at a DMR upstream of the H19 gene known as the Imprint Control region (IC1) is acquired in the male germline and influences the methylation status of DMRs 100 kb away in the adjacent Insulin-like growth factor 2 (Igf2) gene through long-range interactions. In humans, germline-derived or post-zygotically acquired imprinting defects at IC1 are associated with aberrant activation or repression of IGF2, resulting in the congenital growth disorders Beckwith-Wiedemann (BWS) and Silver-Russell (SRS) syndromes, respectively. In Wilms tumour and colorectal cancer, biallelic expression of IGF2 has been observed in association with loss of methylation at a DMR in IGF2. This DMR, known as DMR0, has been shown to be methylated on the silent maternal IGF2 allele presumably with a role in repression. The effect of IGF2 DMR0 methylation changes in the aetiology of BWS or SRS is unknown. METHODOLOGY/PRINCIPAL FINDINGS: We analysed the methylation status of the DMR0 in BWS, SRS and Wilms tumour patients by conventional bisulphite sequencing and pyrosequencing. We show here that, contrary to previous reports, the IGF2 DMR0 is actually methylated on the active paternal allele in peripheral blood and kidney. This is similar to the IC1 methylation status and is inconsistent with the proposed silencing function of the maternal IGF2 allele. Beckwith-Wiedemann and Silver-Russell patients with IC1 methylation defects have similar methylation defects at the IGF2 DMR0, consistent with IC1 regulating methylation at IGF2 in cis. In Wilms tumour, however, methylation profiles of IC1 and IGF2 DMR0 are indicative of methylation changes occurring on both parental alleles rather than in cis. CONCLUSIONS/SIGNIFICANCE: These results support a model in which DMR0 and IC1 have opposite susceptibilities to global hyper and hypomethylation during tumorigenesis independent of the parent of origin imprint. In contrast, during embryogenesis DMR0 is methylated or demethylated according to the germline methylation imprint at the IC1, indicating different mechanisms of imprinting loss in neoplastic and non-neoplastic cells.


Assuntos
Metilação de DNA , Transtornos do Crescimento/genética , Fator de Crescimento Insulin-Like II/genética , Neoplasias/genética , RNA não Traduzido/genética , Transtornos do Crescimento/congênito , Humanos , RNA Longo não Codificante
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA