Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 27
Filtrar
1.
J Med Genet ; 57(3): 160-168, 2020 03.
Artigo em Inglês | MEDLINE | ID: mdl-31586944

RESUMO

BACKGROUND: The type 1 insulin-like growth factor receptor (IGF1R) is a keystone of fetal growth regulation by mediating the effects of IGF-I and IGF-II. Recently, a cohort of patients carrying an IGF1R defect was described, from which a clinical score was established for diagnosis. We assessed this score in a large cohort of patients with identified IGF1R defects, as no external validation was available. Furthermore, we aimed to develop a functional test to allow the classification of variants of unknown significance (VUS) in vitro. METHODS: DNA was tested for either deletions or single nucleotide variant (SNV) and the phosphorylation of downstream pathways studied after stimulation with IGF-I by western blot analysis of fibroblast of nine patients. RESULTS: We detected 21 IGF1R defects in 35 patients, including 8 deletions and 10 heterozygous, 1 homozygous and 1 compound-heterozygous SNVs. The main clinical characteristics of these patients were being born small for gestational age (90.9%), short stature (88.2%) and microcephaly (74.1%). Feeding difficulties and varying degrees of developmental delay were highly prevalent (54.5%). There were no differences in phenotypes between patients with deletions and SNVs of IGF1R. Functional studies showed that the SNVs tested were associated with decreased AKT phosphorylation. CONCLUSION: We report eight new pathogenic variants of IGF1R and an original case with a homozygous SNV. We found the recently proposed clinical score to be accurate for the diagnosis of IGF1R defects with a sensitivity of 95.2%. We developed an efficient functional test to assess the pathogenicity of SNVs, which is useful, especially for VUS.


Assuntos
Anormalidades Múltiplas/genética , Desenvolvimento Fetal/genética , Retardo do Crescimento Fetal/genética , Transtornos do Crescimento/genética , Receptor IGF Tipo 1/genética , Anormalidades Múltiplas/epidemiologia , Anormalidades Múltiplas/fisiopatologia , Adolescente , Criança , Nanismo/genética , Nanismo/fisiopatologia , Feminino , Retardo do Crescimento Fetal/epidemiologia , Retardo do Crescimento Fetal/fisiopatologia , Transtornos do Crescimento/epidemiologia , Transtornos do Crescimento/fisiopatologia , Heterozigoto , Homozigoto , Humanos , Recém-Nascido Pequeno para a Idade Gestacional/crescimento & desenvolvimento , Fator de Crescimento Insulin-Like I/genética , Fator de Crescimento Insulin-Like II/genética , Masculino , Microcefalia/genética , Microcefalia/fisiopatologia , Mutação de Sentido Incorreto/genética , Linhagem , Polimorfismo de Nucleotídeo Único/genética , Receptores de Somatomedina/genética
2.
J Med Genet ; 55(3): 205-213, 2018 03.
Artigo em Inglês | MEDLINE | ID: mdl-29223973

RESUMO

BACKGROUND: The 11p15 region contains two clusters of imprinted genes. Opposite genetic and epigenetic anomalies of this region result in two distinct growth disturbance syndromes: Beckwith-Wiedemann (BWS) and Silver-Russell syndromes (SRS). Cytogenetic rearrangements within this region represent less than 3% of SRS and BWS cases. Among these, 11p15 duplications were infrequently reported and interpretation of their pathogenic effects is complex. OBJECTIVES: To report cytogenetic and methylation analyses in a cohort of patients with SRS/BWS carrying 11p15 duplications and establish genotype/phenotype correlations. METHODS: From a cohort of patients with SRS/BWS with an abnormal methylation profile (using ASMM-RTQ-PCR), we used SNP-arrays to identify and map the 11p15 duplications. We report 19 new patients with SRS (n=9) and BWS (n=10) carrying de novo or familial 11p15 duplications, which completely or partially span either both telomeric and centromeric domains or only one domain. RESULTS: Large duplications involving one complete domain or both domains are associated with either SRS or BWS, depending on the parental origin of the duplication. Genotype-phenotype correlation studies of partial duplications within the telomeric domain demonstrate the prominent role of IGF2, rather than H19, in the control of growth. Furthermore, it highlights the role of CDKN1C within the centromeric domain and suggests that the expected overexpression of KCNQ1OT1 from the paternal allele (in partial paternal duplications, excluding CDKN1C) does not affect the expression of CDKN1C. CONCLUSIONS: The phenotype associated with 11p15 duplications depends on the size, genetic content, parental inheritance and imprinting status. Identification of these rare duplications is crucial for genetic counselling.


Assuntos
Síndrome de Beckwith-Wiedemann/genética , Duplicação Gênica/genética , Impressão Molecular , Síndrome de Silver-Russell/genética , Adulto , Síndrome de Beckwith-Wiedemann/patologia , Centrômero/genética , Aberrações Cromossômicas , Cromossomos Humanos Par 11/genética , Inibidor de Quinase Dependente de Ciclina p57/genética , Análise Citogenética , Feminino , Humanos , Fator de Crescimento Insulin-Like II/genética , Masculino , Mutação , Fenótipo , Síndrome de Silver-Russell/patologia , Telômero/genética
3.
Genet Med ; 20(2): 250-258, 2018 02.
Artigo em Inglês | MEDLINE | ID: mdl-28796236

RESUMO

PurposeFetal growth is a complex process involving maternal, placental and fetal factors. The etiology of fetal growth retardation remains unknown in many cases. The aim of this study is to identify novel human mutations and genes related to Silver-Russell syndrome (SRS), a syndromic form of fetal growth retardation, usually caused by epigenetic downregulation of the potent fetal growth factor IGF2.MethodsWhole-exome sequencing was carried out on members of an SRS familial case. The candidate gene from the familial case and two other genes were screened by targeted high-throughput sequencing in a large cohort of suspected SRS patients. Functional experiments were then used to link these genes into a regulatory pathway.ResultsWe report the first mutations of the PLAG1 gene in humans, as well as new mutations in HMGA2 and IGF2 in six sporadic and/or familial cases of SRS. We demonstrate that HMGA2 regulates IGF2 expression through PLAG1 and in a PLAG1-independent manner.ConclusionGenetic defects of the HMGA2-PLAG1-IGF2 pathway can lead to fetal and postnatal growth restriction, highlighting the role of this oncogenic pathway in the fine regulation of physiological fetal/postnatal growth. This work defines new genetic causes of SRS, important for genetic counseling.


Assuntos
Proteínas de Ligação a DNA/genética , Retardo do Crescimento Fetal/genética , Retardo do Crescimento Fetal/metabolismo , Predisposição Genética para Doença , Variação Genética , Proteína HMGA2/genética , Fator de Crescimento Insulin-Like II/genética , Linhagem Celular , Proteínas de Ligação a DNA/metabolismo , Epigênese Genética , Fácies , Feminino , Retardo do Crescimento Fetal/diagnóstico , Regulação da Expressão Gênica no Desenvolvimento , Estudos de Associação Genética , Genótipo , Gráficos de Crescimento , Proteína HMGA2/metabolismo , Humanos , Fator de Crescimento Insulin-Like II/metabolismo , Modelos Biológicos , Mutação , Linhagem , Transdução de Sinais , Síndrome de Silver-Russell/diagnóstico , Síndrome de Silver-Russell/genética , Síndrome de Silver-Russell/metabolismo , Sequenciamento Completo do Genoma
4.
Hum Mutat ; 38(1): 105-111, 2017 01.
Artigo em Inglês | MEDLINE | ID: mdl-27701793

RESUMO

The 11p15 region harbors the IGF2/H19 imprinted domain, implicated in fetal and postnatal growth. Silver-Russell syndrome (SRS) is characterized by fetal and postnatal growth failure, and is caused principally by hypomethylation of the 11p15 imprinting control region 1 (ICR1). However, the mechanisms leading to ICR1 hypomethylation remain unknown. Maternally inherited genetic defects affecting the ICR1 domain have been associated with ICR1 hypermethylation and Beckwith-Wiedemann syndrome (an overgrowth syndrome, the clinical and molecular mirror of SRS), and paternal deletions of IGF2 enhancers have been detected in four SRS patients. However, no paternal deletions of ICR1 have ever been associated with hypomethylation of the IGF2/H19 domain in SRS. We screened for new genetic defects within the ICR1 in a cohort of 234 SRS patients with hypomethylated IGF2/H19 domain. We report deletions close to the boundaries of ICR1 on the paternal allele in one familial and two sporadic cases of SRS with ICR1 hypomethylation. These deletions are associated with hypomethylation of the remaining CBS, and decreased IGF2 expression. These results suggest that these regions are most likely required to maintain methylation after fertilization. We estimate these anomalies to occur in about 1% of SRS cases with ICR1 hypomethylation.


Assuntos
Cromossomos Humanos Par 11 , Metilação de DNA , Impressão Genômica , Fator de Crescimento Insulin-Like II/genética , RNA Longo não Codificante/genética , Deleção de Sequência , Síndrome de Silver-Russell/genética , Pré-Escolar , Feminino , Fibroblastos , Expressão Gênica , Estudos de Associação Genética , Predisposição Genética para Doença , Humanos , Masculino , Linhagem
5.
Hum Mol Genet ; 23(21): 5763-73, 2014 Nov 01.
Artigo em Inglês | MEDLINE | ID: mdl-24916376

RESUMO

Isolated gain of methylation (GOM) at the IGF2/H19 imprinting control region 1 (ICR1) accounts for about 10% of patients with BWS. A subset of these patients have genetic defects within ICR1, but the frequency of these defects has not yet been established in a large cohort of BWS patients with isolated ICR1 GOM. Here, we carried out a genetic analysis in a large cohort of 57 BWS patients with isolated ICR1 GOM and analyzed the methylation status of the entire domain. We found a new point mutation in two unrelated families and a 21 bp deletion in another unrelated child, both of which were maternally inherited and affected the OCT4/SOX2 binding site in the A2 repeat of ICR1. Based on data from this and previous studies, we estimate that cis genetic defects account for about 20% of BWS patients with isolated ICR1 GOM. Methylation analysis at eight loci of the IGF2/H19 domain revealed that sites surrounding OCT4/SOX2 binding site mutations were fully methylated and methylation indexes declined as a function of distance from these sites. This was not the case in BWS patients without genetic defects identified. Thus, GOM does not spread uniformly across the IGF2/H19 domain, suggesting that OCT4/SOX2 protects against methylation at local sites. These findings add new insights to the mechanism of the regulation of the ICR1 domain. Our data show that mutations and deletions within ICR1 are relatively common. Systematic identification is therefore necessary to establish appropriate genetic counseling for BWS patients with isolated ICR1 GOM.


Assuntos
Síndrome de Beckwith-Wiedemann/genética , Síndrome de Beckwith-Wiedemann/metabolismo , Metilação de DNA , Impressão Genômica , Fator de Crescimento Insulin-Like II/genética , Fator 3 de Transcrição de Octâmero/metabolismo , RNA Longo não Codificante/genética , Fatores de Transcrição SOXB1/metabolismo , Sequência de Bases , Síndrome de Beckwith-Wiedemann/diagnóstico , Sítios de Ligação , Estudos de Casos e Controles , Cromossomos Humanos Par 11 , Feminino , Frequência do Gene , Heterozigoto , Humanos , Masculino , Mutação , Motivos de Nucleotídeos , Linhagem , Fenótipo , Deleção de Sequência
6.
J Med Genet ; 52(7): 446-53, 2015 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-25951829

RESUMO

BACKGROUND: Multiple clinical scoring systems have been proposed for Silver-Russell syndrome (SRS). Here we aimed to test a clinical scoring system for SRS and to analyse the correlation between (epi)genotype and phenotype. SUBJECTS AND METHODS: Sixty-nine patients were examined by two physicians. Clinical scores were generated for all patients, with a new, six-item scoring system: (1) small for gestational age, birth length and/or weight ≤-2SDS, (2) postnatal growth retardation (height ≤-2SDS), (3) relative macrocephaly at birth, (4) body asymmetry, (5) feeding difficulties and/or body mass index (BMI) ≤-2SDS in toddlers; (6) protruding forehead at the age of 1-3 years. Subjects were considered to have likely SRS if they met at least four of these six criteria. Molecular investigations were performed blind to the clinical data. RESULTS: The 69 patients were classified into two groups (Likely-SRS (n=60), Unlikely-SRS (n=9)). Forty-six Likely-SRS patients (76.7%) displayed either 11p15 ICR1 hypomethylation (n=35; 58.3%) or maternal UPD of chromosome 7 (mUPD7) (n=11; 18.3%). Eight Unlikely-SRS patients had neither ICR1 hypomethylation nor mUPD7, whereas one patient had mUPD7. The clinical score and molecular results yielded four groups that differed significantly overall and for individual scoring system factors. Further molecular screening led identifying chromosomal abnormalities in Likely-SRS-double-negative and Unlikely-SRS groups. Four Likely-SRS-double negative patients carried a DLK1/GTL2 IG-DMR hypomethylation, a mUPD16; a mUPD20 and a de novo 1q21 microdeletion. CONCLUSIONS: This new scoring system is very sensitive (98%) for the detection of patients with SRS with demonstrated molecular abnormalities. Given its clinical and molecular heterogeneity, SRS could be considered as a spectrum.


Assuntos
Genótipo , Fenótipo , Projetos de Pesquisa/normas , Síndrome de Silver-Russell/genética , Síndrome de Silver-Russell/patologia , Peso ao Nascer/fisiologia , Índice de Massa Corporal , Testa/anormalidades , Crescimento/fisiologia , Humanos , Megalencefalia/patologia , Estudos Prospectivos
7.
J Med Genet ; 52(1): 53-60, 2015 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-25395389

RESUMO

BACKGROUND: The structural organisation of the human IGF2/ICR1/H19 11p15 domain is very complex, and the mechanisms underlying its regulation are poorly understood. The Imprinted Center Region 1 (ICR1) contains seven binding sites for the zinc-finger protein CTCF (CBS: CTCF Binding Sites); three additional differentially methylated regions (DMR) are located at the H19 promoter (H19DMR) and two in the IGF2 gene (DMR0 and DMR2), respectively. Loss of imprinting at the IGF2/ICR1/H19 domain results in two growth disorders with opposite phenotypes: Beckwith-Wiedemann syndrome and Russell Silver syndrome (RSS). Despite the IGF2/ICR1/H19 locus being widely studied, the extent of hypomethylation across the domain remains not yet addressed in patients with RSS. METHODS: We assessed a detailed investigation of the methylation status of the 11p15 ICR1 CBS1-7, IGF2DMR0 and H19DMR (H19 promoter) in a population of controls (n=50) and RSS carrying (n=104) or not (n=65) carrying a hypomethylation at the 11p15 ICR1 region. RESULTS: The methylation indexes (MI) were balanced at all regions in the control population and patients with RSS without any as yet identified molecular anomaly. Interestingly, patients with RSS with ICR1 hypomethylation showed uneven profiles of methylation among the CBSs and DMRs. Furthermore, normal MIs at CBS1 and CBS7 were identified in 9% of patients. CONCLUSIONS: The hypomethylation does not spread equally throughout the IGF2/ICR1/H19 locus, and some loci could have normal MI, which may lead to underdiagnosis of patients with RSS with ICR1 hypomethylation. The uneven pattern of methylation suggests that some CBSs may play different roles in the tridimensional chromosomal looping regulation of this locus.


Assuntos
Cromossomos Humanos Par 11/genética , Metilação de DNA/genética , Regulação da Expressão Gênica/genética , Fator de Crescimento Insulin-Like II/genética , RNA Longo não Codificante/genética , Síndrome de Silver-Russell/genética , Sequência de Bases , Humanos , Fator de Crescimento Insulin-Like II/metabolismo , Dados de Sequência Molecular , Paris , Análise de Componente Principal , RNA Longo não Codificante/metabolismo , Reação em Cadeia da Polimerase em Tempo Real , Análise de Sequência de DNA , Sulfitos
8.
J Med Genet ; 51(8): 502-11, 2014 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-24996904

RESUMO

BACKGROUND: Disruption of 11p15 imprinting results in two fetal growth disorders with opposite phenotypes: the Beckwith-Wiedemann (BWS; MIM 130650) and the Silver-Russell (SRS; MIM 180860) syndromes. DNA methylation defects account for 60% of BWS and SRS cases and, in most cases, occur without any identified mutation in a cis-acting regulatory sequence or a trans-acting factor. METHODS: We investigated whether 11p15 cis-acting sequence variants account for primary DNA methylation defects in patients with SRS and BWS with loss of DNA methylation at ICR1 and ICR2, respectively. RESULTS: We identified a 4.5 kb haplotype that, upon maternal transmission, is associated with a risk of ICR2 loss of DNA methylation in patients with BWS. This novel region is located within the second intron of the KCNQ1 gene, 170 kb upstream of the ICR2 imprinting centre and encompasses two CTCF binding sites. We showed that, within the 4.5 kb region, two SNPs (rs11823023 and rs179436) affect CTCF occupancy at DNA motifs flanking the CTCF 20 bp core motif. CONCLUSIONS: This study shows that genetic variants confer a risk of DNA methylation defect with a parent-of-origin effect and highlights the crucial role of CTCF for the regulation of genomic imprinting of the CDKN1C/KCNQ1 domain.


Assuntos
Síndrome de Beckwith-Wiedemann/genética , Canal de Potássio KCNQ1/genética , Sítios de Ligação/genética , Fator de Ligação a CCCTC , Metilação de DNA/genética , Feminino , Haplótipos/genética , Humanos , Íntrons/genética , Canal de Potássio KCNQ1/metabolismo , Masculino , Mutação/genética , Proteínas Repressoras/metabolismo
9.
Hum Mutat ; 35(10): 1211-20, 2014 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-25044976

RESUMO

Russell-Silver Syndrome (RSS) is a prenatal and postnatal growth retardation syndrome caused mainly by 11p15 ICR1 hypomethylation. Clinical presentation is heterogeneous in RSS patients with 11p15 ICR1 hypomethylation. We previously identified a subset of RSS patients with 11p15 ICR1 and multilocus hypomethylation. Here, we examine the relationships between IGF2 expression, 11p15 ICR1 methylation, and multilocus imprinting defects in various cell types from 39 RSS patients with 11p15 ICR1 hypomethylation in leukocyte DNA. 11p15 ICR1 hypomethylation was more pronounced in leukocytes than in buccal mucosa cells. Skin fibroblast IGF2 expression was correlated with the degree of ICR1 hypomethylation. Different tissue-specific multilocus methylation defects coexisted in 38% of cases, with some loci hypomethylated and others hypermethylated within the same cell type in some cases. Our new results suggest that tissue-specific epigenotypes may lead to clinical heterogeneity in RSS.


Assuntos
Cromossomos Humanos Par 11/genética , Metilação de DNA , Impressão Genômica , Fator de Crescimento Insulin-Like II/genética , Síndrome de Silver-Russell/genética , Adulto , Criança , Pré-Escolar , Epitélio/metabolismo , Fibroblastos/metabolismo , Expressão Gênica , Humanos , Recém-Nascido , Leucócitos/metabolismo , Mucosa Bucal/metabolismo , Especificidade de Órgãos , Pele/metabolismo
10.
Hum Mutat ; 34(8): 1172-80, 2013 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-23649963

RESUMO

Most patients with pseudohypoparathyroidism type 1b (PHP-1b) display a loss of imprinting (LOI) encompassing the GNAS locus resulting in PTH resistance. In other imprinting disorders, such as Russell-Silver or Beckwith-Wiedemann syndrome, we and others have shown that the LOI is not restricted to one imprinted locus but may affect other imprinted loci for some patients. Therefore, we hypothesized that patients with PHP-1b might present multilocus imprinting defects. We investigated, in 63 patients with PHP-1b, the methylation pattern of eight imprinted loci: GNAS, ZAC1, PEG1/MEST, ICR1, and ICR2 on chromosome 11p15, SNRPN, DLK1/GTL2 IG-DMR, and L3MBTL1. We found multilocus imprinting defects in four PHP-1b patients carrying broad LOI at the GNAS locus (1) simultaneous hypermethylation at L3MBTL1 differentially methylated region 3 (DMR3), and hypomethylation at PEG1/MEST DMR (n = 1), (2) hypermethylation at the L3MBTL1 (DMR3) (n = 1) and at the DLK1/GTL2 IG-DMR (n = 1), and (3) hypomethylation at the L3MBTL1 DMR3 (n = 1). We suggest that mechanisms underlying multilocus imprinting defects in PHP-1b differ from those of other imprinting disorders having only multilocus loss of methylation. Furthermore, our results favor the hypothesis of "epidominance", that is, the phenotype is controlled by the most severely affected imprinted locus.


Assuntos
Metilação de DNA , Subunidades alfa Gs de Proteínas de Ligação ao GTP/genética , Impressão Genômica , Polimorfismo de Nucleotídeo Único , Pseudo-Hipoparatireoidismo/genética , Cromograninas , Estudos de Coortes , Subunidades alfa Gs de Proteínas de Ligação ao GTP/química , Subunidades alfa Gs de Proteínas de Ligação ao GTP/metabolismo , Humanos , Pseudo-Hipoparatireoidismo/metabolismo
11.
Hum Mol Genet ; 19(5): 803-14, 2010 Mar 01.
Artigo em Inglês | MEDLINE | ID: mdl-20007505

RESUMO

The imprinted expression of the IGF2 and H19 genes is controlled by the imprinting control region 1 (ICR1) located at chromosome 11p15.5. This methylation-sensitive chromatin insulator works by binding the zinc-finger protein CTCF in a parent-specific manner. DNA methylation defects involving the ICR1 H19/IGF2 domain result in two growth disorders with opposite phenotypes: an overgrowth disorder, the Beckwith-Wiedemann syndrome (maternal ICR1 gain of methylation in 10% of BWS cases) and a growth retardation disorder, the Silver-Russell syndrome (paternal ICR1 loss of methylation in 60% of SRS cases). Although a few deletions removing part of ICR1 have been described in some familial BWS cases, little information is available regarding the mechanism of ICR1 DNA methylation defects. We investigated the CTCF gene and the ICR1 domain in 21 BWS patients with ICR1 gain of methylation and 16 SRS patients with ICR1 loss of methylation. We identified four constitutional ICR1 genetic defects in BWS patients, including a familial case. Three of those defects are newly identified imprinting defects consisting of small deletions and a single mutation, which do not involve one of the CTCF binding sites. Moreover, two of those defects affect OCT-binding sequences which are suggested to maintain the unmethylated state of the maternal allele. A single-nucleotide variation was identified in a SRS patient. Our data extends the spectrum of constitutive genetic ICR1 abnormalities and suggests that extensive and accurate analysis of ICR1 is required for appropriate genetic counseling in BWS patients with ICR1 gain of methylation.


Assuntos
Cromossomos Humanos Par 11/genética , Retardo do Crescimento Fetal/genética , Impressão Genômica , Fator de Crescimento Insulin-Like II/genética , Mutação , Fatores de Transcrição de Octâmero/metabolismo , RNA não Traduzido/genética , Sequência de Bases , Síndrome de Beckwith-Wiedemann/genética , Estudos de Coortes , Metilação de DNA , Feminino , Retardo do Crescimento Fetal/metabolismo , Humanos , Fator de Crescimento Insulin-Like II/metabolismo , Masculino , RNA Longo não Codificante , RNA não Traduzido/metabolismo
12.
Hum Mutat ; 32(2): 249-58, 2011 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-21280150

RESUMO

Many human syndromes involve a loss of imprinting (LOI) due to a loss (LOM) or a gain of DNA methylation (GOM). Most LOI occur as mosaics and can therefore be difficult to detect with conventional methods. The human imprinted 11p15 region is crucial for the control of fetal growth, and LOI at this locus is associated with two clinical disorders with opposite phenotypes: Beckwith-Wiedemann syndrome (BWS), characterized by fetal overgrowth and a high risk of tumors, and Russell-Silver syndrome (RSS), characterized by intrauterine and postnatal growth restriction. Until recently, we have been using Southern blotting for the diagnosis of RSS and BWS. We describe here a powerful quantitative technique, allele-specific methylated multiplex real-time quantitative PCR (ASMM RTQ-PCR), for the diagnosis of these two complex disorders. We first checked the specificity of the probes and primers used for ASMM RTQ-PCR. We then carried out statistical validation for this method, on both retrospective and prospective populations of patients. This analysis demonstrated that ASMM RTQ-PCR is more sensitive than Southern blotting for detecting low degree of LOI. Moreover, ASMM RTQ-PCR is a very rapid, reliable, simple, safe, and cost effective method.


Assuntos
Síndrome de Beckwith-Wiedemann/diagnóstico , Metilação de DNA , Impressão Genômica , Técnicas de Diagnóstico Molecular/métodos , Reação em Cadeia da Polimerase/métodos , Síndrome de Silver-Russell/diagnóstico , Síndrome de Beckwith-Wiedemann/genética , Cromossomos Humanos Par 11 , Humanos , Reprodutibilidade dos Testes , Sensibilidade e Especificidade , Síndrome de Silver-Russell/genética
13.
Hum Mutat ; 32(10): 1171-82, 2011 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-21780245

RESUMO

The imprinted 11p15 region is organized in two domains, each of them under the control of its own imprinting control region (ICR1 for the IGF2/H19 domain and ICR2 for the KCNQ1OT1/CDKN1C domain). Disruption of 11p15 imprinting results in two fetal growth disorders with opposite phenotypes: the Beckwith-Wiedemann (BWS) and the Silver-Russell (SRS) syndromes. Various 11p15 genetic and epigenetic defects have been demonstrated in BWS and SRS. Among them, isolated DNA methylation defects account for approximately 60% of patients. To investigate whether cryptic copy number variations (CNVs) involving only part of one of the two imprinted domains account for 11p15 isolated DNA methylation defects, we designed a single nucleotide polymorphism array covering the whole 11p15 imprinted region and genotyped 185 SRS or BWS cases with loss or gain of DNA methylation at either ICR1 or ICR2. We describe herein novel small gain and loss CNVs in six BWS or SRS patients, including maternally inherited cis-duplications involving only part of one of the two imprinted domains. We also show that ICR2 deletions do not account for BWS with ICR2 loss of methylation and that uniparental isodisomy involving only one of the two imprinted domains is not a mechanism for SRS or BWS.


Assuntos
Síndrome de Beckwith-Wiedemann/genética , Cromossomos Humanos Par 11 , Variações do Número de Cópias de DNA , Impressão Genômica , Síndrome de Silver-Russell/genética , Adulto , Sequência de Bases , Hibridização Genômica Comparativa , Metilação de DNA , Feminino , Humanos , Masculino , Linhagem , Polimorfismo de Nucleotídeo Único
14.
Hum Mol Genet ; 18(24): 4724-33, 2009 Dec 15.
Artigo em Inglês | MEDLINE | ID: mdl-19755383

RESUMO

Genomic imprinting plays an important role in mammalian development. Loss of imprinting (LOI) through loss (LOM) or gain (GOM) of methylation is involved in many human disorders and cancers. The imprinted 11p15 region is crucial for the control of foetal growth and LOI at this locus is implicated in two clinically opposite disorders: Beckwith Wiedemann syndrome (BWS) with foetal overgrowth associated with an enhanced tumour risk and Russell-Silver syndrome (RSS) with intrauterine and postnatal growth restriction. So far, only a few studies have assessed multilocus LOM in human imprinting diseases. To investigate multilocus LOI syndrome, we studied the methylation status of five maternally and two paternally methylated loci in a large series (n = 167) of patients with 11p15-related foetal growth disorders. We found that 9.5% of RSS and 24% of BWS patients showed multilocus LOM at regions other than ICR1 and ICR2 11p15, respectively. Moreover, over two third of multilocus LOM RSS patients also had LOM at a second paternally methylated locus, DLK1/GTL2 IG-DMR. No additional clinical features due to LOM of other loci were found suggesting an (epi)dominant effect of the 11p15 LOM on the clinical phenotype for this series of patients. Surprisingly, four patients displayed LOM at both ICR1 and ICR2 11p15. Three of them had a RSS and one a BWS phenotype. Our results show for the first time that multilocus LOM can also concern RSS patients. Moreover, LOM can involve both paternally and maternally methylated loci in the same patient.


Assuntos
Síndrome de Beckwith-Wiedemann/genética , Cromossomos Humanos Par 11/genética , Retardo do Crescimento Fetal/genética , Impressão Genômica , Síndrome de Silver-Russell/genética , Proteínas de Ligação ao Cálcio , Estudos de Coortes , Metilação de DNA , Feminino , Humanos , Peptídeos e Proteínas de Sinalização Intercelular/genética , Masculino , Proteínas de Membrana/genética , Proteínas/genética , RNA Longo não Codificante , Análise de Sequência de DNA
15.
Bull Acad Natl Med ; 194(2): 287-97; discussion 297-300, 2010 Feb.
Artigo em Francês | MEDLINE | ID: mdl-21166119

RESUMO

Epigenetic phenomena play a key role in regulating gene expression. One of the most widely studied epigenetic modification is DNA methylation at cytosine residues of CpG dinucleotides in gene promoters, transposons and imprinting control regions (ICR). Genomic imprinting refers to epigenetic marking of genes that results in monoallelic expression depending on the parental origin. Several genes encoding key hormones involved in embryonic and fetal growth are imprinted. There are two critical periods of epigenetic reprogramming: gametogenesis and early preimplantation development. Major reprogramming takes place in primordial germ cells, in which parental imprints are erased and totipotency is restored. Imprint marks are then re-established during spermatogenesis or oogenesis, depending on gender. Upon fertilization, genome-wide demethylation is followed by a wave of de novo methylation, both processes being resisted by imprinted loci. Disruption of imprinting can cause growth defects such as the Beckwith-Wiedemann overgrowth syndrome (BWS) and the Russell-Silver (RSS) intrauterine and postnatal growth retardation syndrome. These growth disorders are caused by abnormal DNA methylation in the 11p15 imprinted region encompassing many imprinted genes, such as IGF2. BWS has been linked to loss of methylation (LOM) in the centromeric ICR2/KCNQIOT1 region of the maternal allele, or gain of methylation in the telomeric ICR1/IGF2/H19 region of the maternal allele. This latter epigenetic defect is associated with an increased risk of tumors such as nephroblastoma. LOM in the telomeric ICR1 region of the paternal allele has been detected in RSS. Early embryogenesis is a critical period of epigenetic regulation, and is sensitive to environmental factors. Individuals conceived with the help of assisted reproductive technology (ART) are over-represented among BWS patients, suggesting that ART may favor altered imprinting at the imprinted centromeric 11p15 locus (LOM in the maternally methylated ICR2 region). The underlying cause of these imprinting defects, both spontaneous and ART-related, is unclear. However, recent data show that, in patients with BWS or RSS, including those conceived with the help of ART the DNA methylation defect involves imprinted loci other than 11p15. This suggests that unfaithful maintenance of DNA methylation marks following fertilization involves dysregulation of a trans-acting regulatory factor.


Assuntos
Anormalidades Congênitas/genética , Epigenômica , Impressão Genômica , Metilação de DNA , Humanos
16.
Sci Adv ; 5(2): eaau9425, 2019 02.
Artigo em Inglês | MEDLINE | ID: mdl-30801013

RESUMO

Imprinting disorders (IDs) often affect growth in humans, leading to diseases with overlapping features, regardless of the genomic region affected. IDs related to hypomethylation of the human 14q32.2 region and its DLK1/MEG3 domain are associated with Temple syndrome (TS14). TS14 is a rare type of growth retardation, the clinical signs of which overlap considerably with those of Silver-Russell syndrome (SRS), another ID related to IGF2 down-regulation at 11p15.5 region. We show that 14q32.2 hypomethylation affects expression, not only for genes at this locus but also for other imprinted genes, and especially lowers IGF2 levels at 11p15.5. Furthermore, expression of nonimprinted genes is also affected, some of which are also deregulated in SRS patients. These findings highlight the epigenetic regulation of gene expression at the DLK1/MEG3 domain. Expression profiling of TS14 and SRS patients highlights common signatures, which may account for the clinical overlap observed between TS14 and SRS.


Assuntos
Proteínas de Ligação ao Cálcio , Cromossomos Humanos Par 11 , Cromossomos Humanos Par 14 , Metilação de DNA , Epigênese Genética , Impressão Genômica , Proteínas de Membrana , Síndrome de Silver-Russell , Proteínas de Ligação ao Cálcio/biossíntese , Proteínas de Ligação ao Cálcio/genética , Cromossomos Humanos Par 11/genética , Cromossomos Humanos Par 11/metabolismo , Cromossomos Humanos Par 14/genética , Cromossomos Humanos Par 14/metabolismo , Feminino , Humanos , Masculino , Proteínas de Membrana/biossíntese , Proteínas de Membrana/genética , Síndrome de Silver-Russell/genética , Síndrome de Silver-Russell/metabolismo
17.
J Clin Endocrinol Metab ; 103(7): 2436-2446, 2018 07 01.
Artigo em Inglês | MEDLINE | ID: mdl-29659920

RESUMO

Context: Silver-Russell syndrome (SRS) (mainly secondary to 11p15 molecular disruption) and Temple syndrome (TS) (secondary to 14q32.2 molecular disruption) are imprinting disorders with phenotypic (prenatal and postnatal growth retardation, early feeding difficulties) and molecular overlap. Objective: To describe the clinical overlap between SRS and TS and extensively study the molecular aspects of TS. Patients: We retrospectively collected data on 28 patients with disruption of the 14q32.2 imprinted region, identified in our center, and performed extensive molecular analysis. Results: Seventeen (60.7%) patients showed loss of methylation of the MEG3/DLK1 intergenic differentially methylated region by epimutation. Eight (28.6%) patients had maternal uniparental disomy of chromosome 14 and three (10.7%) had a paternal deletion in 14q32.2. Most patients (72.7%) had a Netchine-Harbison SRS clinical scoring system ≥4/6, and consistent with a clinical diagnosis of SRS. The mean age at puberty onset was 7.2 years in girls and 9.6 years in boys; 37.5% had premature pubarche. The body mass index of all patients increased before pubarche and/or the onset of puberty. Multilocus analysis identified multiple methylation defects in 58.8% of patients. We identified four potentially damaging genetic variants in genes encoding proteins involved in the establishment or maintenance of DNA methylation. Conclusions: Most patients with 14q32.2 disruption fulfill the criteria for a clinical diagnosis of SRS. These clinical data suggest similar management of patients with TS and SRS, with special attention to their young age at the onset of puberty and early increase of body mass index.


Assuntos
Transtornos Cromossômicos/genética , Cromossomos Humanos Par 14/genética , Síndrome de Silver-Russell/genética , Adolescente , Adulto , Proteínas de Ligação ao Cálcio , Criança , Pré-Escolar , Deleção Cromossômica , Transtornos Cromossômicos/diagnóstico , Metilação de DNA/genética , Diagnóstico Diferencial , Feminino , Impressão Genômica/genética , Humanos , Peptídeos e Proteínas de Sinalização Intercelular/genética , Masculino , Proteínas de Membrana/genética , Fenótipo , Puberdade Precoce/genética , RNA Longo não Codificante/genética , Estudos Retrospectivos , Síndrome de Silver-Russell/diagnóstico , Síndrome , Dissomia Uniparental , Adulto Jovem
18.
J Clin Endocrinol Metab ; 92(8): 3148-54, 2007 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-17504900

RESUMO

CONTEXT: Russell-Silver syndrome (RSS), characterized by intrauterine and postnatal growth retardation, dysmorphic features, and frequent body asymmetry, spares cranial growth. Maternal uniparental disomy for chromosome 7 (mUPD7) is found in 5-10% of cases. We identified loss of methylation (LOM) of 11p15 Imprinting Center Region 1 (ICR1) domain (including IGF-II) as a mechanism leading to RSS. OBJECTIVE: The aim was to screen for 11p15 epimutation and mUPD7 in RSS and non-RSS small-for-gestational-age (SGA) patients and identify epigenetic-phenotypic correlations. STUDIED POPULATION AND METHODS: A total of 127 SGA patients were analyzed. Clinical diagnosis of RSS was established when the criterion of being SGA was associated with at least three of five criteria: postnatal growth retardation, relative macrocephaly, prominent forehead, body asymmetry, and feeding difficulties. Serum IGF-II was evaluated for 82 patients. RESULTS: Of the 127 SGA patients, 58 were diagnosed with RSS; 37 of these (63.8%) displayed partial LOM of the 11p15 ICR1 domain, and three (5.2%) had mUPD7. No molecular abnormalities were found in the non-RSS SGA group (n = 69). Birth weight, birth length, and postnatal body mass index (BMI) were lower in the abnormal 11p15 RSS group (ab-ICR1-RSS) than in the normal 11p15 RSS group [-3.4 vs.-2.6 SD score (SDS), -4.4 vs.-3.4 SDS, and -2.5 vs.-1.6 SDS, respectively; P < 0.05]. Among RSS patients, prominent forehead, relative macrocephaly, body asymmetry, and low BMI were significantly associated with ICR1 LOM. All ab-ICR1-RSS patients had at least four of five criteria of the scoring system. Postnatal IGF-II levels were within normal values. CONCLUSION: The 11p15 ICR1 epimutation is a major, specific cause of RSS exhibiting failure to thrive. We propose a clinical scoring system (including a BMI < -2 SDS), highly predictive of 11p15 ICR1 LOM, for the diagnosis of RSS.


Assuntos
Anormalidades Múltiplas/genética , Retardo do Crescimento Fetal/genética , Envelhecimento/metabolismo , Cromossomos Humanos Par 7/genética , DNA/genética , Face/anormalidades , Feminino , Impressão Genômica , Humanos , Recém-Nascido , Recém-Nascido Pequeno para a Idade Gestacional/fisiologia , Fator de Crescimento Insulin-Like II/metabolismo , Masculino , Metilação , Mutação/genética , Mutação/fisiologia , Fenótipo , Síndrome
19.
Curr Opin Endocrinol Diabetes Obes ; 21(1): 30-8, 2014 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-24322424

RESUMO

PURPOSE OF REVIEW: The imprinted human 11p15.5 region encompasses two imprinted domains important for the control of fetal growth: the H19/IGF2 domain in the telomeric region and the KCNQ1OT1/CDKN1C domain in the centromeric region. These two domains are differentially methylated and each is regulated by its own imprinting control region (ICR): ICR1 in the telomeric region and ICR2 in the centromeric region. Aberrant methylation of the 11p15.5 imprinted region, through genetic or epigenetic mechanisms, leads to two clinical syndromes, with opposite growth phenotypes: Russell-Silver Syndrome (RSS; with severe fetal and postnatal growth retardation) and Beckwith-Wiedemann Syndrome (BWS; an overgrowth syndrome). RECENT FINDINGS: In this review, we discuss the recently identified molecular abnormalities at 11p15.5 involved in RSS and BWS, which have led to the identification of cis-acting elements and trans-acting regulatory factors involved in the regulation of imprinting in this region. We also discuss the multilocus imprinting disorders identified in various human syndromes, their clinical outcomes and their impact on commonly identified metabolism disorders. SUMMARY: These new findings and progress in this field will have direct consequence for diagnostic and predictive tools, risk assessment and genetic counseling for these syndromes.


Assuntos
Síndrome de Beckwith-Wiedemann/genética , Testes Genéticos/métodos , Impressão Genômica , Síndrome de Silver-Russell/genética , Síndrome de Beckwith-Wiedemann/diagnóstico , Inibidor de Quinase Dependente de Ciclina p57/genética , Metilação de DNA , Epigênese Genética , Feminino , Aconselhamento Genético , Testes Genéticos/tendências , Humanos , Recém-Nascido , Fator de Crescimento Insulin-Like II/genética , Masculino , Canais de Potássio de Abertura Dependente da Tensão da Membrana/genética , Guias de Prática Clínica como Assunto , Gravidez , Medição de Risco , Síndrome de Silver-Russell/diagnóstico
20.
Curr Pharm Des ; 20(11): 1751-63, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-23888961

RESUMO

Genomic imprinting is among the most important epigenetic mechanisms whereby expression of a subset of genes is restricted to a single parental allele. Loss of imprinting (LOI) through hypo or hyper methylation is involved in various human syndromes. These LOI occur early during development and usually impair growth. Some imprinting syndromes are the consequences of genetic anomalies, such as uniparental disomies (UPD) or copy number variations (deletion or duplications) involving the imprinted domains; others are due to LOI at the imprinting control regions (ICR) regulating each domain. Imprinting disorders are phenotypically heterogeneous, although some share various common clinical features such that diagnosis may be difficult. Multilocus imprinting defects associated with several syndromes have been increasingly reported in recent years, although there are no obvious clinical differences between monolocus and multilocus LOI patients. Subsequently, some rare mutations of transacting factors have been identified in patients with multilocus imprinting defects but they do not explain the majority of the cases; this therefore implies that other factors are involved. By contrast, no mutation of a transacting factor has yet been identified in monolocus LOI. The effect of the environment on the regulation of imprinting is clearly illustrated by studies of assisted reproductive technology (ART). The regulation of imprinting is complex and involves a huge range of genetic and environmental factors; the identification of these factors will undoubtedly help to elucidate the regulation of imprinting and contribute to the understanding of imprinting disorders. This would be beneficial for diagnostics, clinical follow up and the development of treatment guidelines.


Assuntos
Retardo do Crescimento Fetal/genética , Impressão Genômica/genética , Transtornos do Crescimento/genética , Alelos , Animais , Variações do Número de Cópias de DNA , Epigênese Genética , Retardo do Crescimento Fetal/diagnóstico , Transtornos do Crescimento/diagnóstico , Humanos , Mutação , Guias de Prática Clínica como Assunto , Técnicas de Reprodução Assistida , Síndrome
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA