Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 30
Filtrar
1.
J Med Genet ; 51(8): 502-11, 2014 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-24996904

RESUMEN

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.


Asunto(s)
Síndrome de Beckwith-Wiedemann/genética , Canal de Potasio KCNQ1/genética , Sitios de Unión/genética , Factor de Unión a CCCTC , Metilación de ADN/genética , Femenino , Haplotipos/genética , Humanos , Intrones/genética , Canal de Potasio KCNQ1/metabolismo , Masculino , Mutación/genética , Proteínas Represoras/metabolismo
2.
Hum Mol Genet ; 23(21): 5763-73, 2014 Nov 01.
Artículo en Inglés | MEDLINE | ID: mdl-24916376

RESUMEN

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.


Asunto(s)
Síndrome de Beckwith-Wiedemann/genética , Síndrome de Beckwith-Wiedemann/metabolismo , Metilación de ADN , Impresión Genómica , Factor II del Crecimiento Similar a la Insulina/genética , Factor 3 de Transcripción de Unión a Octámeros/metabolismo , ARN Largo no Codificante/genética , Factores de Transcripción SOXB1/metabolismo , Secuencia de Bases , Síndrome de Beckwith-Wiedemann/diagnóstico , Sitios de Unión , Estudios de Casos y Controles , Cromosomas Humanos Par 11 , Femenino , Frecuencia de los Genes , Heterocigoto , Humanos , Masculino , Mutación , Motivos de Nucleótidos , Linaje , Fenotipo , Eliminación de Secuencia
3.
J Med Genet ; 50(1): 11-20, 2013 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-23240093

RESUMEN

The 11p15 region is organised into two independent imprinted domains controlled by imprinting control regions, which carry opposite germline imprints. Dysregulation of 11p15 genomic imprinting results in two human fetal growth disorders (Silver-Russell syndrome (SRS, MIM 180860) and Beckwith-Wiedemann syndrome (BWS, MIM 130650)) with opposite growth phenotypes. The mouse orthologous region on distal chromosome 7 (dist7) is well conserved in its organisation and its regulation. Targeted mutagenesis in mice has provided highly valuable clues in terms of the mechanisms involved in the regulation of genomic imprinting of the 11p15/dist7 imprinted region. On the other hand, the recent identification of unexpected genetic defects in BWS and SRS patients also brought new insights into the mechanisms of 11p15 imprinting regulation. However, some mouse models and human genetic defects show contradictions in term of growth phenotypes and parental transmission. In this review, we extensively analyse those various mouse and human models and more particularly models with mutations affecting the two imprinting centres, in order to improve our understanding of regulation of 11p15/dist7 genomic imprinting.


Asunto(s)
Cromosomas Humanos Par 11 , Cromosomas de los Mamíferos , Modelos Animales de Enfermedad , Impresión Genómica , Ratones , Animales , Duplicación Cromosómica , Inhibidor p57 de las Quinasas Dependientes de la Ciclina/genética , Epigénesis Genética , Humanos , Factor II del Crecimiento Similar a la Insulina/genética , Canal de Potasio KCNQ1/genética , ARN Largo no Codificante/genética
4.
Eur J Hum Genet ; 20(2): 240-3, 2012 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-21863054

RESUMEN

The imprinted expression of the IGF2 and H19 genes is controlled by the imprinting control region 1 (ICR1) located at chromosome 11p15.5. DNA methylation defects involving ICR1 result in two growth disorders with opposite phenotypes: an overgrowth disorder, the Beckwith-Wiedemann syndrome (maternal ICR1 hypermethylation in 10% of BWS cases) and a growth retardation disorder, the Silver-Russell syndrome (paternal ICR1 loss of methylation in 60% of SRS cases). In familial BWS, hypermethylation of ICR1 has been found in association with microdeletion of repetitive DNA motifs within ICR1 that bind the zinc finger protein CTCF; but more recently, ICR1 point mutations were described in BWS pedigrees. We present a case report of two brothers with BWS and prolonged post-pubertal growth resulting in very large stature. A maternally inherited point mutation was identified in ICR1 in both brothers, which altered binding of OCT transcription factors. The same mutation was present on the paternally inherited allele of their unaffected mother. This is a second report of a point mutation causing ICR1 hypermethylation by altering an OCT-binding motif. The atypical growth phenotype of the brothers may be connected to the unusual underlying cause of their BWS.


Asunto(s)
Síndrome de Beckwith-Wiedemann/genética , Impresión Genómica , Factor II del Crecimiento Similar a la Insulina/genética , Mutación , Factores de Transcripción de Octámeros/metabolismo , ARN no Traducido/genética , Alelos , Secuencia de Bases , Síndrome de Beckwith-Wiedemann/diagnóstico , Sitios de Unión/genética , Preescolar , Metilación de ADN , Orden Génico , Genotipo , Humanos , Lactante , Masculino , Linaje , Fenotipo , Regiones Promotoras Genéticas , ARN Largo no Codificante
5.
Hum Mutat ; 32(10): 1171-82, 2011 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-21780245

RESUMEN

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.


Asunto(s)
Síndrome de Beckwith-Wiedemann/genética , Cromosomas Humanos Par 11 , Variaciones en el Número de Copia de ADN , Impresión Genómica , Síndrome de Silver-Russell/genética , Adulto , Secuencia de Bases , Hibridación Genómica Comparativa , Metilación de ADN , Femenino , Humanos , Masculino , Linaje , Polimorfismo de Nucleótido Simple
6.
Hum Mutat ; 32(2): 249-58, 2011 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-21280150

RESUMEN

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.


Asunto(s)
Síndrome de Beckwith-Wiedemann/diagnóstico , Metilación de ADN , Impresión Genómica , Técnicas de Diagnóstico Molecular/métodos , Reacción en Cadena de la Polimerasa/métodos , Síndrome de Silver-Russell/diagnóstico , Síndrome de Beckwith-Wiedemann/genética , Cromosomas Humanos Par 11 , Humanos , Reproducibilidad de los Resultados , Sensibilidad y Especificidad , Síndrome de Silver-Russell/genética
7.
Bull Acad Natl Med ; 194(2): 287-97; discussion 297-300, 2010 Feb.
Artículo en Francés | MEDLINE | ID: mdl-21166119

RESUMEN

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.


Asunto(s)
Anomalías Congénitas/genética , Epigenómica , Impresión Genómica , Metilación de ADN , Humanos
8.
Hum Mol Genet ; 19(5): 803-14, 2010 Mar 01.
Artículo en Inglés | MEDLINE | ID: mdl-20007505

RESUMEN

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.


Asunto(s)
Cromosomas Humanos Par 11/genética , Retardo del Crecimiento Fetal/genética , Impresión Genómica , Factor II del Crecimiento Similar a la Insulina/genética , Mutación , Factores de Transcripción de Octámeros/metabolismo , ARN no Traducido/genética , Secuencia de Bases , Síndrome de Beckwith-Wiedemann/genética , Estudios de Cohortes , Metilación de ADN , Femenino , Retardo del Crecimiento Fetal/metabolismo , Humanos , Factor II del Crecimiento Similar a la Insulina/metabolismo , Masculino , ARN Largo no Codificante , ARN no Traducido/metabolismo
9.
Hum Mol Genet ; 18(24): 4724-33, 2009 Dec 15.
Artículo en Inglés | MEDLINE | ID: mdl-19755383

RESUMEN

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.


Asunto(s)
Síndrome de Beckwith-Wiedemann/genética , Cromosomas Humanos Par 11/genética , Retardo del Crecimiento Fetal/genética , Impresión Genómica , Síndrome de Silver-Russell/genética , Proteínas de Unión al Calcio , Estudios de Cohortes , Metilación de ADN , Femenino , Humanos , Péptidos y Proteínas de Señalización Intercelular/genética , Masculino , Proteínas de la Membrana/genética , Proteínas/genética , ARN Largo no Codificante , Análisis de Secuencia de ADN
10.
Best Pract Res Clin Endocrinol Metab ; 22(3): 403-14, 2008 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-18538282

RESUMEN

Silver-Russell syndrome (SRS) is a clinically heterogeneous syndrome characterized by intra-uterine and postnatal growth retardation with spared cranial growth, dysmorphic features and frequent body asymmetry. Various cytogenetic abnormalities have been described in a small number of SRS or SRS-like cases involving chromosomes 7, 8, 11, 15, 17 and 18. However, until recent data became available involving imprinted genes on chromosome 7 and chromosome 11p15, the molecular cause of the syndrome was unknown in most cases. Genomic imprinting is the best example of transcriptional control of genes by epigenetic modifications. Many imprinted genes play key roles in fetal and placental growth and behaviour. This is illustrated in SRS, which can now be considered as a new imprinting disease model. These new findings in the pathophysiology of SRS allow long-term follow-up studies to be performed based on molecular diagnosis. This could help to define appropriate clinical guidelines regarding growth and feeding difficulties.


Asunto(s)
Epigénesis Genética , Retardo del Crecimiento Fetal/genética , Trastornos del Crecimiento/genética , Cromosomas Humanos Par 11/genética , Cromosomas Humanos Par 7/genética , Impresión Genómica , Humanos , Recién Nacido , Recién Nacido Pequeño para la Edad Gestacional , Síndrome , Disomía Uniparental/genética
11.
Artículo en Inglés | MEDLINE | ID: mdl-18279777

RESUMEN

Fetal programming encompasses the role of developmental plasticity in response to environmental and nutritional signals during early life and its potential adverse consequences (risk of cardiovascular, metabolic and behavioural diseases) in later life. The first studies in this field highlighted an association between poor fetal growth and chronic adult diseases. However, environmental signals during early life may lead to adverse long-term effects independently of obvious effects on fetal growth. Adverse long-term effects reflect a mismatch between early (fetal and neonatal) environmental conditions and the conditions that the individual will confront later in life. The mechanisms underlying this risk remain unclear. However, experimental data in rodents and recent observations in humans suggest that epigenetic changes in regulatory genes and growth-related genes play a significant role in fetal programming. Improvements in our understanding of the biochemical and molecular mechanisms at play in fetal programming would make it possible to identify biomarkers for detecting infants at high risk of adult-onset diseases. Such improvements should also lead to the development of preventive and therapeutic strategies.


Asunto(s)
Epigénesis Genética , Desarrollo Fetal , Feto/fisiología , Animales , Femenino , Enfermedades Fetales/genética , Enfermedades Fetales/metabolismo , Regulación del Desarrollo de la Expresión Génica , Humanos , Lactante , Masculino , Polimorfismo Genético , Embarazo , Fenómenos Fisiologicos de la Nutrición Prenatal
12.
Ann Surg ; 247(1): 157-64, 2008 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-18156936

RESUMEN

OBJECTIVE: To determine the minimal common region of loss on 17p13 in a cohort of adrenocortical carcinomas (ACCs) (defined by a Weiss score > or =3) and adrenocortical adenomas (ACAs) (defined by a Weiss score <3) and subsequently to assess 3 genes in this region that could be involved in adrenocortical tumorigenesis. SUMMARY BACKGROUND DATA: Loss of heterozygosity (LOH) of 17p13 has been demonstrated to occur more frequently in ACCs compared with ACAs. METHODS: Using 12 microsatellite markers across 17p13, LOH analysis was performed on 37 paired blood and adrenocortical tumor samples (23 ACC and 14 ACA samples) to determine the minimal common region of loss for ACCs and ACAs. From this minimal region of loss, 3 genes were selected for quantitative real time reverse transcription polymerase chain reaction analysis on 20 ACCs and 30 ACAs. RESULTS: LOH at 17p13 was found in 74% of ACCs compared with 14% of ACAs. There was a 10.4-Mb common minimal region of loss in ACCs whereas no minimal region of loss in ACAs could be demonstrated. Expression of Acyl coenzyme-A dehydrogenase very long chain (ACADVL) and Arachidonate 15-lipoxygenase second type (ALOX15B) was significantly down-regulated in ACCs compared with ACAs whereas there was no difference in expression of Potassium channel tetramerization domain containing 11 (KCTD11) in ACCs and ACAs. CONCLUSIONS: We demonstrated a minimal common region of loss of 10.4-Mb on 17p13 in ACCs and within this region, we found that ACADVL and ALOX15B expression are good discriminators between ACCs and ACAs.


Asunto(s)
Acil-CoA Deshidrogenasas/genética , Neoplasias de la Corteza Suprarrenal/genética , Araquidonato 12-Lipooxigenasa/genética , Cromosomas Humanos Par 17 , Genes Supresores de Tumor , Pérdida de Heterocigocidad , Neoplasias de la Corteza Suprarrenal/enzimología , Anciano , Femenino , Regulación Neoplásica de la Expresión Génica , Marcadores Genéticos , Humanos , Masculino , Repeticiones de Microsatélite , Persona de Mediana Edad , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Estadísticas no Paramétricas
13.
Am J Med Genet A ; 143A(23): 2796-803, 2007 Dec 01.
Artículo en Inglés | MEDLINE | ID: mdl-17994567

RESUMEN

Beckwith-Wiedemann syndrome (BWS) is an overgrowth condition with tumor proclivity linked to a genetic imbalance of a complex imprinted region in 11p15.5. A female child with features fitting in with the BWS diagnostic framework and an apparent loss of imprinting (LOI) of the IGF2 gene in 11p15.5 was also reported to have a de novo chromosome 18q segmental deletion (Patient 1), thus pointing at the location of a possible trans-activating regulator element for maintenance of IGF2 imprinting and providing one of the few examples of locus heterogeneity of BWS. A second child with de novo 18q23 deletion and features of macroglossia, naevus flammeus, bilateral inguinal hernia and transient neonatal hypoglycemia, thus also fitting in with the BWS diagnostic framework, is here fully reported (Patient 2). In this child, an analysis of the BWS1 locus precluded any paternal isodisomy and showed a normal imprinting pattern (mono-allelic expression of IGF2 and normal H19 and CDKN1OT1/LIT1 methylation index). In Patients 1 and 2, deletions were shown to overlap, defining a minimal region of haplo-insufficiency of 3.8-5.6 Mb in 18q23. We conclude that this region provides a candidate location for an original macroglossia condition with strong overlap with BWS, but without obvious upstream functional relationship with the BWS1 locus in 11p15.5. Because this minimal region of haplo-insufficiency falls into a common region of deletion in 18q- syndrome, we inferred that this macroglossia condition would follow a recessive pattern of inheritance.


Asunto(s)
Síndrome de Beckwith-Wiedemann/genética , Cromosomas Humanos Par 18 , Macroglosia/genética , Femenino , Haplotipos , Humanos , Hibridación Fluorescente in Situ , Recién Nacido , Masculino , Mapeo Físico de Cromosoma
14.
J Clin Endocrinol Metab ; 92(8): 3148-54, 2007 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-17504900

RESUMEN

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.


Asunto(s)
Anomalías Múltiples/genética , Retardo del Crecimiento Fetal/genética , Envejecimiento/metabolismo , Cromosomas Humanos Par 7/genética , ADN/genética , Cara/anomalías , Femenino , Impresión Genómica , Humanos , Recién Nacido , Recién Nacido Pequeño para la Edad Gestacional/fisiología , Factor II del Crecimiento Similar a la Insulina/metabolismo , Masculino , Metilación , Mutación/genética , Mutación/fisiología , Fenotipo , Síndrome
15.
Clin Cancer Res ; 13(3): 844-50, 2007 Feb 01.
Artículo en Inglés | MEDLINE | ID: mdl-17289876

RESUMEN

PURPOSE: Allelic losses [loss of heterozygosity (LOH)] at the 17p13 locus are frequent (85%) in adrenocortical cancers. The tumor suppressor gene TP53 is located at 17p13. The aim of the study was to determine the frequency of TP53 somatic inactivating mutations in adrenocortical tumors with 17p13 LOH and their clinico-biological correlations. EXPERIMENTAL DESIGN: TP53 somatic mutations, intragenic LOH (VNTR1 marker), and p53 overexpression were studied in 36 adrenocortical tumors with 17p13 LOH determined by Southern blot. RESULTS: TP53 mutations were detected in 33% of the tumors, and VNTR1 LOH was present in 44% of the cases and did not always correlate with the presence of a TP53 mutation. Only the TP53-mutant tumors exhibit a strong nuclear immunoreactivity. TP53-mutant tumors were significantly larger than wild-type TP53 tumors (median tumor weight: 640 versus 185 g; P=0.02), were associated with a more advanced stage of tumor progression (MacFarlane stage IV; P=0.01), and had a shorter disease-free survival (P=0.03). CONCLUSIONS: The finding that only a minority of adrenocortical tumors with 17p13 LOH had either a VNTR1 LOH or a TP53 mutation indicates that TP53 might not be the only or major tumor suppressor gene at 17p13 involved in adrenocortical cancer progression. We suggest that a genetic instability of the 17p13 region, occurring early in adrenocortical cancer development, involves various genes located in this region. TP53 might be only one of them, and its alteration by the occurrence of inactivating mutation is associated with the development of more aggressive tumors.


Asunto(s)
Neoplasias de la Corteza Suprarrenal/genética , Cromosomas Humanos Par 17 , Genes p53 , Pérdida de Heterocigocidad , Repeticiones de Minisatélite/genética , Mutación , Adulto , Alelos , Progresión de la Enfermedad , Femenino , Humanos , Inmunohistoquímica , Masculino , Persona de Mediana Edad , ARN Neoplásico/química
16.
J Clin Endocrinol Metab ; 92(1): 148-54, 2007 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-17062775

RESUMEN

CONTEXT: Prognostic parameters of metastatic adrenocortical carcinoma (ACC) are poorly characterized. OBJECTIVE: The objective of the study was to describe the clinical presentation of metastatic ACC and determine prognostic factors for survival. DESIGN: This was a retrospective cohort study (1988-2004). SETTING: The study was conducted in an institutional practice. PATIENTS: Participants included 124 consecutive patients with metastatic ACC, 70 from Gustave-Roussy Institute (main cohort) and 54 patients from the Cochin Hospital (validation cohort). Clinical data concerning all patients, histopathologic slides of primary tumors (44 in the main cohort and 40 in the validation cohort), and molecular biology data on 15 primary tumors (main cohort) were analyzed. INTERVENTION: There was no intervention. MAIN OUTCOME: The main outcome was the specific survival after discovery of the first metastasis (Kaplan-Meier method). This included univariate analysis on the main cohort, confirmed on the validation cohort and then analyzed in a multivariate analysis. RESULTS: In the main cohort, overall median survival was 20 months. In univariate analysis, the presence of hepatic and bone metastases, the number of metastatic lesions and the number of tumoral organs at the time of the first metastasis, a high mitotic rate (>20 per 50 high-power field), and atypical mitoses in the primary tumor predicted survival (P = 0.05, 0.003, 0.046, 0.001, 0.01, and < 0.001, respectively). The number of tumoral organs and a high mitotic rate were confirmed on the validation cohort (P = 0.009 and 0.03, respectively). These two parameters were confirmed in multivariate analysis (P = 0.0058 and 0.049). CONCLUSION: Metastatic ACC is a heterogeneous disease with poor outcome. The combination of the number of tumoral organs at the time of the first metastasis and the mitotic rate can predict different outcomes.


Asunto(s)
Neoplasias de la Corteza Suprarrenal/mortalidad , Adolescente , Neoplasias de la Corteza Suprarrenal/genética , Neoplasias de la Corteza Suprarrenal/patología , Adulto , Anciano , Neoplasias Óseas/secundario , Estudios de Cohortes , Femenino , Humanos , Neoplasias Hepáticas/secundario , Pérdida de Heterocigocidad , Masculino , Persona de Mediana Edad , Análisis Multivariante , Pronóstico , Estudios Retrospectivos , Tasa de Supervivencia
17.
Cancer Res ; 65(17): 7622-7, 2005 Sep 01.
Artículo en Inglés | MEDLINE | ID: mdl-16140927

RESUMEN

Adrenocortical cancer is a rare cancer with a very poor prognosis. The genetic alterations identified to date in adrenocortical tumors are limited. Activating mutations of the Wnt signaling pathway have been observed in more frequent cancers, particularly digestive tract tumors. We investigated whether Wnt pathway activation is involved in adrenocortical tumorigenesis. In a series of 39 adrenocortical tumors, immunohistochemistry revealed abnormal cytoplasmic and/or nuclear accumulation of beta-catenin in 10 of 26 adrenocortical adenomas and in 11 of 13 adrenocortical carcinomas. An activating somatic mutation of the beta-catenin gene was shown in 7 of 26 adrenocortical adenomas and in 4 of 13 adrenocortical carcinomas; these mutations were observed only in adrenocortical tumors with abnormal beta-catenin accumulation and most were point mutations altering the Ser45 of exon 3 (in the consensus GSK3-beta/CK1 phosphorylation site). Functional studies showed that the activating Ser45 beta-catenin mutation found in the adrenocortical cancer H295R cell line leads to constitutive activation of T-cell factor-dependent transcription. This is the first molecular defect to be reported with the same prevalence in both benign (27%) and malignant (31%) adrenocortical tumors. beta-Catenin mutations are also the most frequent genetic defect currently known in adrenocortical adenomas. In adrenocortical adenomas, beta-catenin alterations are more frequent in nonfunctioning tumors, suggesting that beta-catenin pathway activation might be mostly involved in the development of nonsecreting adrenocortical adenomas and adrenocortical carcinomas. The very frequent and substantial accumulation of beta-catenin in adrenocortical carcinomas suggests that other alterations might also be involved. This finding may contribute to new therapeutic approaches targeting the Wnt pathway in malignant adrenocortical tumors, for which limited medical therapy is available.


Asunto(s)
Neoplasias de la Corteza Suprarrenal/genética , Carcinoma Corticosuprarrenal/genética , Proteínas del Citoesqueleto/genética , Péptidos y Proteínas de Señalización Intercelular/genética , Transactivadores/genética , Neoplasias de la Corteza Suprarrenal/metabolismo , Neoplasias de la Corteza Suprarrenal/patología , Carcinoma Corticosuprarrenal/metabolismo , Carcinoma Corticosuprarrenal/patología , Adulto , Anciano , Línea Celular Tumoral , Proteínas del Citoesqueleto/metabolismo , Femenino , Humanos , Inmunohistoquímica , Masculino , Persona de Mediana Edad , Mutación , Transducción de Señal , Transactivadores/metabolismo , Proteínas Wnt , beta Catenina
18.
Nat Genet ; 37(9): 1003-7, 2005 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-16086014

RESUMEN

Silver-Russell syndrome (SRS, OMIM 180860) is a congenital disorder characterized by severe intrauterine and postnatal growth retardation, dysmorphic facial features and body asymmetry. SRS is genetically heterogenous with maternal uniparental disomy with respect to chromosome 7 occurring in approximately 10% of affected individuals. Given the crucial role of the 11p15 imprinted region in the control of fetal growth, we hypothesized that dysregulation of genes at 11p15 might be involved in syndromic intrauterine growth retardation. We identified an epimutation (demethylation) in the telomeric imprinting center region ICR1 of the 11p15 region in several individuals with clinically typical SRS. This epigenetic defect is associated with, and probably responsible for, relaxation of imprinting and biallelic expression of H19 and downregulation of IGF2. These findings provide new insight into the pathogenesis of SRS and strongly suggest that the 11p15 imprinted region, in addition to those of 7p11.2-p13 and 7q31-qter, is involved in SRS.


Asunto(s)
Cromosomas Humanos Par 11/genética , Metilación de ADN , Impresión Genómica/genética , Trastornos del Crecimiento/genética , Mutación/genética , Telómero , Factor de Unión a CCCTC , Proteínas de Unión al ADN/genética , Retardo del Crecimiento Fetal , Trastornos del Crecimiento/fisiopatología , Humanos , Factor II del Crecimiento Similar a la Insulina , Datos de Secuencia Molecular , Regiones Promotoras Genéticas/genética , Proteínas/genética , ARN Largo no Codificante , ARN no Traducido/genética , Proteínas Represoras/genética , Síndrome
19.
Am J Med Genet C Semin Med Genet ; 137C(1): 4-11, 2005 Aug 15.
Artículo en Inglés | MEDLINE | ID: mdl-16010674

RESUMEN

Here, we report the clinical and molecular analysis of 75 patients with overgrowth and mental retardation, including 45 previously reported cases [Rio et al., 2003; Baujat et al., 2004]. Two groups are distinguished: group I corresponding to patients with recognizable overgrowth syndromes (Sotos syndrome (SS), Weaver syndrome (WS), Beckwith-Wiedemann syndrome, Simpson-Golabi-Behmel syndrome (SGBS), and del(22)(qter) syndrome) (60 cases) and group II corresponding to unclassified cases (15 patients). We investigated NSD1 and GPC3 deletions or mutations, 11p15 abnormalities, and 22qter deletions. Surprisingly, in Group I, two SS patients had 11p15 abnormalities and two patients with Beckwith-Wiedemann syndrome had NSD1 aberrations. In group II, two cases of del(22)(qter) were identified but neither NSD1, 11p15, nor GPC3 abnormalities were detected. These results emphasize the clinical and molecular overlap in overgrowth conditions.


Asunto(s)
Trastornos del Crecimiento/genética , Discapacidad Intelectual/genética , Síndrome de Beckwith-Wiedemann/genética , Cromosomas Humanos Par 11/genética , Cromosomas Humanos Par 22/genética , Análisis Mutacional de ADN , Glipicanos , Histona Metiltransferasas , N-Metiltransferasa de Histona-Lisina , Humanos , Péptidos y Proteínas de Señalización Intracelular/genética , Proteínas de la Membrana/genética , Repeticiones de Microsatélite/genética , Mutación/genética , Proteínas de Neoplasias/genética , Proteínas Nucleares/genética , Síndrome
20.
J Clin Endocrinol Metab ; 90(3): 1819-29, 2005 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-15613424

RESUMEN

The aim of this study was to identify predictor sets of genes whose over- or underexpression in human sporadic adrenocortical tumors would help to identify malignant vs. benign tumors and to predict postsurgical metastatic recurrence. For this, we analyzed the expression of 230 candidate genes using cDNA microarrays in a series of 57 well-characterized human sporadic adrenocortical tumors (33 adenomas and 24 carcinomas). We identified two clusters of genes (the IGF-II cluster containing eight genes, including IGF-II, and the steroidogenesis cluster containing six genes encoding steroidogenic enzymes plus eight other genes) whose combined levels of expression appeared to be good predictors of malignancy. This predictive value was as strong as that of the pathological score of Weiss. The analysis of the population of carcinomas (13 tumors) for genes whose expression would be strongly different between recurring and nonrecurring tumors allowed identification of 14 genes meeting these criteria. Among these genes, there are probably new markers of tumor evolution that will deserve additional validation on a larger scale. Taken together, these results show that the parallel analysis of the expression levels of a selected group of genes on microgram quantities of tumor RNA (a quantity that can be obtained from fine needle aspirations) appears as a complementary method to histopathology for the diagnosis and prognosis of evolution of adrenocortical carcinomas.


Asunto(s)
Neoplasias de la Corteza Suprarrenal/genética , Perfilación de la Expresión Génica , Análisis de Secuencia por Matrices de Oligonucleótidos , Adolescente , Neoplasias de la Corteza Suprarrenal/metabolismo , Neoplasias de la Corteza Suprarrenal/mortalidad , Adulto , Anciano , Biomarcadores de Tumor/genética , Supervivencia sin Enfermedad , Femenino , Regulación Neoplásica de la Expresión Génica , Marcadores Genéticos , Humanos , Masculino , Persona de Mediana Edad , ARN Mensajero/análisis , Esteroides/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA