Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 42
Filtrar
1.
Elife ; 102021 08 17.
Artigo em Inglês | MEDLINE | ID: mdl-34402430

RESUMO

Maternal loss of imprinting (LOI) at the H19/IGF2 locus results in biallelic IGF2 and reduced H19 expression and is associated with Beckwith--Wiedemann syndrome (BWS). We use mouse models for LOI to understand the relative importance of Igf2 and H19 mis-expression in BWS phenotypes. Here we focus on cardiovascular phenotypes and show that neonatal cardiomegaly is exclusively dependent on increased Igf2. Circulating IGF2 binds cardiomyocyte receptors to hyperactivate mTOR signaling, resulting in cellular hyperplasia and hypertrophy. These Igf2-dependent phenotypes are transient: cardiac size returns to normal once Igf2 expression is suppressed postnatally. However, reduced H19 expression is sufficient to cause progressive heart pathologies including fibrosis and reduced ventricular function. In the heart, H19 expression is primarily in endothelial cells (ECs) and regulates EC differentiation both in vivo and in vitro. Finally, we establish novel mouse models to show that cardiac phenotypes depend on H19 lncRNA interactions with Mirlet7 microRNAs.


Assuntos
Sistema Cardiovascular/metabolismo , Sistema Cardiovascular/patologia , RNA Longo não Codificante/genética , RNA Longo não Codificante/metabolismo , Animais , Síndrome de Beckwith-Wiedemann/genética , Síndrome de Beckwith-Wiedemann/metabolismo , Cardiomiopatias/metabolismo , Cardiomiopatias/patologia , Diferenciação Celular , Modelos Animais de Doenças , Células Endoteliais/metabolismo , Células Endoteliais/patologia , Fator de Crescimento Insulin-Like II/genética , Fator de Crescimento Insulin-Like II/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , MicroRNAs/metabolismo , Miócitos Cardíacos/metabolismo , Miócitos Cardíacos/patologia , Fenótipo , Transdução de Sinais
2.
Nucleic Acids Res ; 48(20): 11394-11407, 2020 11 18.
Artigo em Inglês | MEDLINE | ID: mdl-33053156

RESUMO

Genomic imprinting is an epigenetic process regulated by germline-derived DNA methylation that is resistant to embryonic reprogramming, resulting in parental origin-specific monoallelic gene expression. A subset of individuals affected by imprinting disorders (IDs) displays multi-locus imprinting disturbances (MLID), which may result from aberrant establishment of imprinted differentially methylated regions (DMRs) in gametes or their maintenance in early embryogenesis. Here we investigated the extent of MLID in a family harbouring a ZFP57 truncating variant and characterize the interactions between human ZFP57 and the KAP1 co-repressor complex. By ectopically targeting ZFP57 to reprogrammed loci in mouse embryos using a dCas9 approach, we confirm that ZFP57 recruitment is sufficient to protect oocyte-derived methylation from reprogramming. Expression profiling in human pre-implantation embryos and oocytes reveals that unlike in mice, ZFP57 is only expressed following embryonic-genome activation, implying that other KRAB-zinc finger proteins (KZNFs) recruit KAP1 prior to blastocyst formation. Furthermore, we uncover ZNF202 and ZNF445 as additional KZNFs likely to recruit KAP1 to imprinted loci during reprogramming in the absence of ZFP57. Together, these data confirm the perplexing link between KZFPs and imprint maintenance and highlight the differences between mouse and humans in this respect.


Assuntos
Metilação de DNA , Embrião de Mamíferos/metabolismo , Impressão Genômica , Células Germinativas/metabolismo , Oócitos/metabolismo , Proteínas Repressoras/metabolismo , Síndrome de Beckwith-Wiedemann/metabolismo , Estudos de Coortes , DNA (Citosina-5-)-Metiltransferases/metabolismo , DNA Metiltransferase 3A , Humanos , Análise em Microsséries , Mutação , Linhagem , Pseudo-Hipoparatireoidismo/metabolismo , RNA-Seq , Proteínas Repressoras/genética , Irmãos , Transcriptoma , Proteína 28 com Motivo Tripartido
3.
J Hum Genet ; 64(9): 937-943, 2019 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-31235774

RESUMO

Beckwith-Wiedemann syndrome (BWS) is a representative imprinting disorder. Gain of methylation at imprinting control region 1 (ICR1-GOM), leading to the biallelic expression of IGF2 and silencing of H19, is one of the causative alterations in BWS. Twenty percent of BWS patients with ICR1-GOM have genetic defects in ICR1. Evidence of methylation anticipation in familial BWS patients with ICR1 genetic defects has been reported. However, the precise methylation pattern and extent of anticipation in these patients remain elusive. In addition, although age-related IGF2-DMR0 hypomethylation has been reported in the normal population, the period of its occurrence is unknown. In this study, we analyzed 10 sites (IGF2-DMR0, IGF2-DMR2, CTCF binding sites 1-7, and the H19 promoter) within the IGF2/H19 domain in familial BWS patients harboring a pathogenic variant in ICR1. We found that sites near the variant had relatively higher methylation in the first affected generation and observed methylation anticipation through maternal transmission in the next generation. The extent of anticipation was greater at sites far from the variant than nearby sites. The extended and severe GOM might be due to the insufficient erasure/demethylation of pre-acquired ICR1-GOM in primordial germ cells or during the preimplantation stage. In the normal population, age-related IGF2-DMR0 hypomethylation occurred; it became established by young adulthood and continued to old age. Further studies are needed to clarify (1) the precise mechanism of anticipation in patients with familial BWS and (2) the mechanism and biological significance of constitutive hypomethylation of IGF2-DMR0 and/or other imprinted differentially methylated regions.


Assuntos
Síndrome de Beckwith-Wiedemann/genética , Metilação de DNA/genética , Inativação Gênica , Fator de Crescimento Insulin-Like II/genética , Linhagem , RNA Longo não Codificante/genética , Elementos de Resposta , Adulto , Síndrome de Beckwith-Wiedemann/metabolismo , Criança , Pré-Escolar , Feminino , Humanos , Lactente , Fator de Crescimento Insulin-Like II/biossíntese , Masculino , Pessoa de Meia-Idade , RNA Longo não Codificante/biossíntese
4.
Eur J Hum Genet ; 27(6): 903-908, 2019 06.
Artigo em Inglês | MEDLINE | ID: mdl-30778172

RESUMO

Beckwith-Wiedemann syndrome (BWS; OMIM #130650) is an imprinting disorder caused by genetic or epigenetic alterations of one or both imprinting control regions on chromosome 11p15.5. Hypomethylation of the centromeric imprinting control region (KCNQ1OT1:TSS-DMR, ICR2) is the most common molecular cause of BWS and is present in about half of the cases. Based on a BWS family with a maternal deletion of the 5' part of KCNQ1 we have recently hypothesised that transcription of KCNQ1 is a prerequisite for the establishment of methylation at the KCNQ1OT1:TSS-DMR in the oocyte. Further evidence for this hypothesis came from a mouse model where methylation failed to be established when a poly(A) truncation cassette was inserted into this locus to prevent transcription through the DMR. Here we report on a family where a balanced translocation disrupts the KCNQ1 gene in intron 9. Maternal inheritance of this translocation is associated with hypomethylation of the KCNQ1OT1:TSS-DMR and BWS. This finding strongly supports our previous hypothesis that transcription of KCNQ1 is required for establishing the maternal methylation imprint at the KCNQ1OT1:TSS-DMR.


Assuntos
Síndrome de Beckwith-Wiedemann , Metilação de DNA , Impressão Genômica , Íntrons , Transcrição Gênica , Translocação Genética , Animais , Síndrome de Beckwith-Wiedemann/genética , Síndrome de Beckwith-Wiedemann/metabolismo , Feminino , Humanos , Canal de Potássio KCNQ1/genética , Canal de Potássio KCNQ1/metabolismo , Masculino , Camundongos
5.
J Clin Invest ; 129(1): 209-214, 2019 01 02.
Artigo em Inglês | MEDLINE | ID: mdl-30352048

RESUMO

The loss of insulin-secreting ß cells is characteristic among type I and type II diabetes. Stimulating proliferation to expand sources of ß cells for transplantation remains a challenge because adult ß cells do not proliferate readily. The cell cycle inhibitor p57 has been shown to control cell division in human ß cells. Expression of p57 is regulated by the DNA methylation status of the imprinting control region 2 (ICR2), which is commonly hypomethylated in Beckwith-Wiedemann syndrome patients who exhibit massive ß cell proliferation. We hypothesized that targeted demethylation of the ICR2 using a transcription activator-like effector protein fused to the catalytic domain of TET1 (ICR2-TET1) would repress p57 expression and promote cell proliferation. We report here that overexpression of ICR2-TET1 in human fibroblasts reduces p57 expression levels and increases proliferation. Furthermore, human islets overexpressing ICR2-TET1 exhibit repression of p57 with concomitant upregulation of Ki-67 while maintaining glucose-sensing functionality. When transplanted into diabetic, immunodeficient mice, the epigenetically edited islets show increased ß cell replication compared with control islets. These findings demonstrate that epigenetic editing is a promising tool for inducing ß cell proliferation, which may one day alleviate the scarcity of transplantable ß cells for the treatment of diabetes.


Assuntos
Síndrome de Beckwith-Wiedemann/metabolismo , Proliferação de Células , Inibidor de Quinase Dependente de Ciclina p57/biossíntese , Desmetilação do DNA , Loci Gênicos , Células Secretoras de Insulina/metabolismo , Regulação para Cima , Síndrome de Beckwith-Wiedemann/patologia , Fibroblastos/metabolismo , Fibroblastos/patologia , Humanos , Células Secretoras de Insulina/patologia , Antígeno Ki-67/biossíntese
6.
Clin Epigenetics ; 10(1): 114, 2018 08 30.
Artigo em Inglês | MEDLINE | ID: mdl-30165906

RESUMO

BACKGROUND: Beckwith-Wiedemann syndrome (BWS) is an imprinting disorder with a population frequency of approximately 1 in 10,000. The most common epigenetic defect in BWS is a loss of methylation (LOM) at the 11p15.5 imprinting centre, KCNQ1OT1 TSS-DMR, and affects 50% of cases. We hypothesised that genetic factors linked to folate metabolism may play a role in BWS predisposition via effects on methylation maintenance at KCNQ1OT1 TSS-DMR. RESULTS: Single nucleotide variants (SNVs) in the folate pathway affecting methylenetetrahydrofolate reductase (MTHFR), methionine synthase reductase (MTRR), 5-methyltetrahydrofolate-homocysteine S-methyltransferase (MTR), cystathionine beta-synthase (CBS) and methionine adenosyltransferase (MAT1A) were examined in 55 BWS patients with KCNQ1OT1 TSS-DMR LOM and in 100 unaffected cases. MTHFR rs1801133: C>T was more prevalent in BWS with KCNQ1OT1 TSS-DMR LOM (p < 0.017); however, the relationship was not significant when the Bonferroni correction for multiple testing was applied (significance, p = 0.0036). None of the remaining 13 SNVs were significantly different in the two populations tested. The DNMT1 locus was screened in 53 BWS cases, and three rare missense variants were identified in each of three patients: rs138841970: C>T, rs150331990: A>G and rs757460628: G>A encoding NP_001124295 p.Arg136Cys, p.His1118Arg and p.Arg1223His, respectively. These variants have population frequencies of less than 1 in 1000 and were absent from 100 control cases. Functional characterization using a hemimethylated DNA trapping assay revealed a reduced methyltransferase activity relative to wild-type DNMT1 for each variant ranging from 40 to 70% reduction in activity. CONCLUSIONS: This study is the first to examine folate pathway genetics in BWS and to identify rare DNMT1 missense variants in affected individuals. Our data suggests that reduced DNMT1 activity could affect maintenance of methylation at KCNQ1OT1 TSS-DMR in some cases of BWS, possibly via a maternal effect in the early embryo. Larger cohort studies are warranted to further interrogate the relationship between impaired MTHFR enzymatic activity attributable to MTHFR rs1801133: C>T, dietary folate intake and BWS.


Assuntos
Síndrome de Beckwith-Wiedemann/genética , DNA (Citosina-5-)-Metiltransferase 1/genética , Metilação de DNA , Ácido Fólico/metabolismo , Mutação de Sentido Incorreto , Síndrome de Beckwith-Wiedemann/metabolismo , Feminino , Impressão Genômica , Células HeLa , Humanos , Masculino , Redes e Vias Metabólicas , Metilenotetra-Hidrofolato Redutase (NADPH2)/genética , Polimorfismo de Nucleotídeo Único , Canais de Potássio de Abertura Dependente da Tensão da Membrana/genética
7.
Nucleic Acids Res ; 45(22): 12766-12779, 2017 Dec 15.
Artigo em Inglês | MEDLINE | ID: mdl-29244185

RESUMO

Imprinted genes occur in discrete clusters that are coordinately regulated by shared DNA elements called Imprinting Control Regions. H19 and Igf2 are linked imprinted genes that play critical roles in development. Loss of imprinting (LOI) at the IGF2/H19 locus on the maternal chromosome is associated with the developmental disorder Beckwith Wiedemann Syndrome (BWS) and with several cancers. Here we use comprehensive genetic and genomic analyses to follow muscle development in a mouse model of BWS to dissect the separate and shared roles for misexpression of Igf2 and H19 in the disease phenotype. We show that LOI results in defects in muscle differentiation and hypertrophy and identify primary downstream targets: Igf2 overexpression results in over-activation of MAPK signaling while loss of H19 lncRNA prevents normal down regulation of p53 activity and therefore results in reduced AKT/mTOR signaling. Moreover, we demonstrate instances where H19 and Igf2 misexpression work separately, cooperatively, and antagonistically to establish the developmental phenotype. This study thus identifies new biochemical roles for the H19 lncRNA and underscores that LOI phenotypes are multigenic so that complex interactions will contribute to disease outcomes.


Assuntos
Síndrome de Beckwith-Wiedemann/genética , Impressão Genômica , Fator de Crescimento Insulin-Like II/genética , Mutação , RNA Longo não Codificante/genética , Animais , Síndrome de Beckwith-Wiedemann/metabolismo , Diferenciação Celular/genética , Células Cultivadas , Modelos Animais de Doenças , Perfilação da Expressão Gênica/métodos , Humanos , Fator de Crescimento Insulin-Like II/metabolismo , Camundongos , Músculo Esquelético/metabolismo , Mioblastos/citologia , Mioblastos/metabolismo , RNA Longo não Codificante/metabolismo , Transdução de Sinais/genética
8.
Horm Res Paediatr ; 86(3): 206-211, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-27255538

RESUMO

BACKGROUND: Beckwith-Wiedemann syndrome (BWS) is an overgrowth syndrome with an increased risk of cancer. Most BWS patients show a molecular defect in the 11p15 region that contains imprinted genes. BWS has been associated with malignant neoplasms during infancy. Descriptions of benign tumors, especially in adult patients, are rarer. METHODS/RESULTS: We report the case of a BWS patient with pituitary adenoma caused by loss of methylation (LOM) at ICR2 (locus CDKN1C/KCNQ1OT1). The patient was referred to an endocrinology unit for suspicion of Cushing's disease due to a history of macroglossia and hemihyperplasia. Biological tests led to the diagnosis of ACTH-dependent hypercortisolism. MRI showed a microadenoma of the pituitary gland, confirming the diagnosis of Cushing's disease. DNA methylation analysis revealed LOM at ICR2 that was in a mosaic state in the patient's leukocytes, but was present in nearly all cells of the pituitary adenoma. The epigenetic defect was associated with a somatic USP8 mutation in the adenoma. CONCLUSION: Pituitary adenoma rarely occurs in patients with BWS. However, BWS should be considered in cases of pituitary adenoma with minor and/or major signs of BWS. The association between ICR2 LOM and USP8 mutation in the adenoma is questionable. © 2016 S. Karger AG, Basel.


Assuntos
Adenoma , Síndrome de Beckwith-Wiedemann , Síndrome de Cushing , Metilação de DNA , Endopeptidases , Complexos Endossomais de Distribuição Requeridos para Transporte , Epigênese Genética , Loci Gênicos , Neoplasias Hipofisárias , Ubiquitina Tiolesterase , Adenoma/genética , Adenoma/metabolismo , Adolescente , Adulto , Síndrome de Beckwith-Wiedemann/complicações , Síndrome de Beckwith-Wiedemann/genética , Síndrome de Beckwith-Wiedemann/metabolismo , Síndrome de Cushing/etiologia , Síndrome de Cushing/genética , Síndrome de Cushing/metabolismo , Inibidor de Quinase Dependente de Ciclina p57/genética , Inibidor de Quinase Dependente de Ciclina p57/metabolismo , Endopeptidases/genética , Endopeptidases/metabolismo , Complexos Endossomais de Distribuição Requeridos para Transporte/genética , Complexos Endossomais de Distribuição Requeridos para Transporte/metabolismo , Feminino , Humanos , Neoplasias Hipofisárias/genética , Neoplasias Hipofisárias/metabolismo , Canais de Potássio de Abertura Dependente da Tensão da Membrana/genética , Canais de Potássio de Abertura Dependente da Tensão da Membrana/metabolismo , Ubiquitina Tiolesterase/genética , Ubiquitina Tiolesterase/metabolismo
9.
Oxid Med Cell Longev ; 2016: 4782426, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-27239251

RESUMO

Oxidative stress is a distinctive sign in several genetic disorders characterized by cancer predisposition, such as Ataxia-Telangiectasia, Fanconi Anemia, Down syndrome, progeroid syndromes, Beckwith-Wiedemann syndrome, and Costello syndrome. Recent literature unveiled new molecular mechanisms linking oxidative stress to the pathogenesis of these conditions, with particular regard to mitochondrial dysfunction. Since mitochondria are one of the major sites of ROS production as well as one of the major targets of their action, this dysfunction is thought to be the cause of the prooxidant status. Deeper insight of the pathogenesis of the syndromes raises the possibility to identify new possible therapeutic targets. In particular, the use of mitochondrial-targeted agents seems to be an appropriate clinical strategy in order to improve the quality of life and the life span of the patients.


Assuntos
Síndrome de Beckwith-Wiedemann/metabolismo , Mitocôndrias/metabolismo , Doenças Mitocondriais/metabolismo , Neoplasias/metabolismo , Estresse Oxidativo , Síndrome de Prader-Willi/metabolismo , Fatores Etários , Animais , Anticarcinógenos/uso terapêutico , Antioxidantes/uso terapêutico , Síndrome de Beckwith-Wiedemann/complicações , Síndrome de Beckwith-Wiedemann/tratamento farmacológico , Síndrome de Beckwith-Wiedemann/genética , Transformação Celular Neoplásica/genética , Transformação Celular Neoplásica/metabolismo , Predisposição Genética para Doença , Humanos , Mitocôndrias/efeitos dos fármacos , Doenças Mitocondriais/tratamento farmacológico , Doenças Mitocondriais/genética , Neoplasias/genética , Neoplasias/prevenção & controle , Estresse Oxidativo/efeitos dos fármacos , Síndrome de Prader-Willi/complicações , Síndrome de Prader-Willi/tratamento farmacológico , Síndrome de Prader-Willi/genética , Fatores de Risco
10.
J Clin Invest ; 126(2): 527-42, 2016 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-26784546

RESUMO

Beckwith-Wiedemann syndrome (BWS) is a human stem cell disorder, and individuals with this disease have a substantially increased risk (~800-fold) of developing tumors. Epigenetic silencing of ß2-spectrin (ß2SP, encoded by SPTBN1), a SMAD adaptor for TGF-ß signaling, is causally associated with BWS; however, a role of TGF-ß deficiency in BWS-associated neoplastic transformation is unexplored. Here, we have reported that double-heterozygous Sptbn1+/- Smad3+/- mice, which have defective TGF-ß signaling, develop multiple tumors that are phenotypically similar to those of BWS patients. Moreover, tumorigenesis-associated genes IGF2 and telomerase reverse transcriptase (TERT) were overexpressed in fibroblasts from BWS patients and TGF-ß-defective mice. We further determined that chromatin insulator CCCTC-binding factor (CTCF) is TGF-ß inducible and facilitates TGF-ß-mediated repression of TERT transcription via interactions with ß2SP and SMAD3. This regulation was abrogated in TGF-ß-defective mice and BWS, resulting in TERT overexpression. Imprinting of the IGF2/H19 locus and the CDKN1C/KCNQ1 locus on chromosome 11p15.5 is mediated by CTCF, and this regulation is lost in BWS, leading to aberrant overexpression of growth-promoting genes. Therefore, we propose that loss of CTCF-dependent imprinting of tumor-promoting genes, such as IGF2 and TERT, results from a defective TGF-ß pathway and is responsible at least in part for BWS-associated tumorigenesis as well as sporadic human cancers that are frequently associated with SPTBN1 and SMAD3 mutations.


Assuntos
Síndrome de Beckwith-Wiedemann/metabolismo , Proteínas de Transporte/metabolismo , Proteínas dos Microfilamentos/metabolismo , Proteínas de Neoplasias/metabolismo , Neoplasias/metabolismo , Proteínas Repressoras/metabolismo , Fator de Crescimento Transformador beta/metabolismo , Animais , Síndrome de Beckwith-Wiedemann/genética , Fator de Ligação a CCCTC , Proteínas de Transporte/genética , Cromossomos Humanos Par 11/genética , Cromossomos Humanos Par 11/metabolismo , Inibidor de Quinase Dependente de Ciclina p57/genética , Inibidor de Quinase Dependente de Ciclina p57/metabolismo , Células Hep G2 , Humanos , Fator de Crescimento Insulin-Like II/genética , Fator de Crescimento Insulin-Like II/metabolismo , Canal de Potássio KCNQ1/genética , Canal de Potássio KCNQ1/metabolismo , Camundongos , Camundongos Knockout , Proteínas dos Microfilamentos/genética , Proteínas de Neoplasias/genética , Neoplasias/genética , Proteínas Repressoras/genética , Transdução de Sinais/genética , Proteína Smad3/genética , Proteína Smad3/metabolismo , Telomerase/biossíntese , Telomerase/genética , Telomerase/metabolismo , Fator de Crescimento Transformador beta/genética
11.
Genet Test Mol Biomarkers ; 19(12): 684-91, 2015 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-26505556

RESUMO

AIMS: To study the frequency of methylation abnormalities among Estonian patients selected according to published clinical diagnostic scoring systems for Silver-Russell syndrome (SRS) and Beckwith-Wiedemann syndrome (BWS). MATERIALS AND METHODS: Forty-eight patients with clinical suspicion of SRS (n = 20) or BWS (n = 28) were included in the study group, to whom methylation-specific multiplex ligation-dependant probe amplification analysis of 11p15 region was made. In addition, to patients with minimal diagnostic score for either SRS or BWS, multilocus methylation-specific single nucleotide primer extension assay was performed. RESULTS: Five (38%) SRS patients with positive clinical scoring had abnormal methylation pattern at chromosome 11p15, whereas in the BWS group, only one patient was diagnosed with imprinting control region 2 (ICR2) hypomethylation (8%). An unexpected hypomethylation of the PLAGL1 (6q24) and IGF2R (6q25) genes in the patient with the highest BWS scoring was found. CONCLUSIONS: Compared to BWS, diagnostic criteria used for selecting SRS patients gave us a similar detection rate of 11p15 imprinting disorders as seen in other studies. A more careful selection of patients with possible BWS should be considered to improve the detection of molecularly confirmed cases. Genome-wide multilocus methylation tests could be used in routine clinical practice as it increases the detection rates of imprinting disorders.


Assuntos
Síndrome de Beckwith-Wiedemann , Proteínas de Ciclo Celular , Metilação de DNA , Receptor IGF Tipo 2 , Síndrome de Silver-Russell , Fatores de Transcrição , Proteínas Supressoras de Tumor , Síndrome de Beckwith-Wiedemann/diagnóstico , Síndrome de Beckwith-Wiedemann/genética , Síndrome de Beckwith-Wiedemann/metabolismo , Proteínas de Ciclo Celular/genética , Proteínas de Ciclo Celular/metabolismo , Estônia , Feminino , Humanos , Lactente , Recém-Nascido , Masculino , Receptor IGF Tipo 2/genética , Receptor IGF Tipo 2/metabolismo , Síndrome de Silver-Russell/diagnóstico , Síndrome de Silver-Russell/genética , Síndrome de Silver-Russell/metabolismo , Fatores de Transcrição/genética , Fatores de Transcrição/metabolismo , Proteínas Supressoras de Tumor/genética , Proteínas Supressoras de Tumor/metabolismo
12.
J Clin Endocrinol Metab ; 100(11): 3963-6, 2015 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-26367199

RESUMO

CONTEXT: Several patients with Beckwith-Wiedemann Syndrome (BWS) with multiple imprinting defects found by genetic analysis have been described. However, only two cases have been described with both genetic and clinical signs and symptoms of multiple diseases caused by imprinting defects. CASE DESCRIPTION: The girl in this case presented at the age of 6 months with morbid obesity (body mass index, +7.5 SDS) and a large umbilical hernia. Genetic analysis showed BWS (hypomethylation of the KCNQ1OT1 gene). Calcium homeostasis was normal, and she had no signs of Albright hereditary osteodystrophy. At the age of 10 years, she presented with fatigue, and laboratory analyses showed marked hypocalcemia with signs of PTH resistance, but without evidence for Albright hereditary osteodystrophy, thus suggesting pseudohypoparathyroidism type 1B. Consistent with this diagnosis, methylation analysis of the GNAS complex revealed hypomethylation (about 20%) of the GNAS exon 1A, NESPAS, and GNASXL loci and hypermethylation (100% methylation) of the NESP locus. CONCLUSIONS: Imprinting defects at several different loci can occur in some patients, thus causing multiple different diseases. Symptoms of pseudohypoparathyroidism type 1B may be absent at diagnosis of BWS, yet prolonged subclinical hypocalcemia and/or hyperphosphatemia can have negative consequences (eg, intracerebral calcifications, myocardial dysfunction). We therefore suggest that patients with an imprinting disorder should be monitored for elevations in PTH, and epigenetic analysis of the GNAS complex locus should be considered.


Assuntos
Síndrome de Beckwith-Wiedemann/genética , Metilação de DNA , Regulação para Baixo , Epigênese Genética , Impressão Genômica , Pseudo-Hipoparatireoidismo/genética , Síndrome de Beckwith-Wiedemann/complicações , Síndrome de Beckwith-Wiedemann/metabolismo , Síndrome de Beckwith-Wiedemann/fisiopatologia , Cromograninas , Progressão da Doença , Éxons , Feminino , Subunidades alfa Gs de Proteínas de Ligação ao GTP/antagonistas & inibidores , Subunidades alfa Gs de Proteínas de Ligação ao GTP/genética , Subunidades alfa Gs de Proteínas de Ligação ao GTP/metabolismo , Loci Gênicos , Hérnia Umbilical/etiologia , Humanos , Hiperfosfatemia/etiologia , Hipocalcemia/etiologia , Lactente , Obesidade Infantil/etiologia , Canais de Potássio de Abertura Dependente da Tensão da Membrana/genética , Canais de Potássio de Abertura Dependente da Tensão da Membrana/metabolismo , Pseudo-Hipoparatireoidismo/complicações , Pseudo-Hipoparatireoidismo/metabolismo , Pseudo-Hipoparatireoidismo/fisiopatologia , RNA Antissenso/metabolismo , Pseudo-Hipoparatireoidismo
13.
Turk J Pediatr ; 56(2): 177-82, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-24911853

RESUMO

Beckwith-Wiedemann syndrome (BWS) is a congenital disorder of imprinting caused by epimutations and mutations affecting two imprinted loci on chromosome 11p15. Its clinical features are heterogeneous, including macrosomia, macroglossia, hemihyperplasia, abdominal wall defects, neonatal hypoglycemia, and increased risk of embryonal tumors such as Wilms tumor, adrenocortical carcinoma, hepatoblastoma, and neuroblastoma. The molecular and clinical heterogeneity of BWS makes the diagnosis challenging, but essential, since different etiologies of BWS have different clinical prognoses - most crucially, patients with gain of maternal methylation at imprinting control region type 1 (ICR1) are at significant risk of Wilms tumor or hepatoblastoma. We present three cases of BWS with different symptomatology and two different molecular diagnoses. The authors emphasize the importance of molecular studies in the long-term follow-up of children with BWS, including refinement of phenotype-genotype correlation and its connection with optimal management and tumor surveillance.


Assuntos
Síndrome de Beckwith-Wiedemann/genética , Cromossomos Humanos Par 11/genética , Testes Genéticos/métodos , Impressão Genômica , Mutação , Síndrome de Beckwith-Wiedemann/diagnóstico , Síndrome de Beckwith-Wiedemann/metabolismo , Metilação de DNA , Análise Mutacional de DNA , Feminino , Seguimentos , Humanos , Masculino , Fatores de Tempo
14.
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
15.
Pediatrics ; 133(4): e1082-6, 2014 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-24639276

RESUMO

Beckwith-Wiedemann syndrome (BWS) is caused by dysregulation of imprinted genes on chromosome 11.p15.5. The syndrome includes overgrowth, macroglossia, organomegaly, abdominal wall defects, hypoglycemia, and long-term malignancy risk. No patient who has BWS has been reported with hypopituitarism. We describe a patient who presented at birth with macrosomia, macroglossia, respiratory distress, jaundice, and hypoglycemia, and who was followed for 4.5 years. Genetic test for BWS was performed, which detected loss of maternal methylation on region KvDMR1 (11p15.5). The hypoglycemia was attributable to hyperinsulinism and was treated with diazoxide and chlorothiazide. She responded well, but the hypoglycemia returned after reducing the diazoxide. It was possible to stop the diazoxide after 2.5 years. On routine follow-up she was noted to be developing short stature. Baseline pituitary and growth hormone (GH) stimulation tests detected GH deficiency and secondary hypothyroidism. A brain MRI showed a small anterior pituitary gland. Thereafter, thyroxine and replacement therapy with GH were started, which resulted in a remarkable improvement in growth velocity. This is the first patient to be reported as having hypopituitarism and BWS. It is unclear if the BWS and the hypopituitarism are somehow connected; however, further investigations are necessary. Hypopituitarism explains the protracted hypoglycemia and the short stature. In our patient, GH therapy seems to be safe, but strict follow-up is required given the increased cancer risk related to BWS.


Assuntos
Síndrome de Beckwith-Wiedemann/complicações , Síndrome de Beckwith-Wiedemann/metabolismo , Hipopituitarismo/etiologia , Pré-Escolar , Seguimentos , Humanos , Recém-Nascido , Metilação , Canais de Potássio de Abertura Dependente da Tensão da Membrana/metabolismo
16.
Tumori ; 100(6): 590-9, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-25688491

RESUMO

Oxidative stress plays a key role in carcinogenesis. Oxidative damage to cell components can lead to the initiation, promotion and progression of cancer. Oxidative stress is also a distinctive sign in several genetic disorders characterized by a cancer predisposition such as ataxia-telangiectasia, Fanconi anemia, Down syndrome, Beckwith-Wiedemann syndrome and Costello syndrome. Taking into account the link between oxidative stress and cancer, the capacity of antioxidant agents to prevent or delay neoplastic development has been tested in various studies, both in vitro and in vivo, with interesting and promising results. In recent years, research has been conducted into the molecular mechanisms linking oxidative stress to the pathogenesis of the genetic syndromes we consider in this review, with the resulting identification of possible new therapeutic targets. The aim of this review is to focus on the oxidative mechanisms intervening in carcinogenesis in cancer-prone genetic disorders and to analyze the current status and future prospects of antioxidants.


Assuntos
Antioxidantes/uso terapêutico , Doenças Genéticas Inatas/complicações , Doenças Genéticas Inatas/metabolismo , Neoplasias/metabolismo , Neoplasias/prevenção & controle , Estresse Oxidativo , Ataxia Telangiectasia/complicações , Ataxia Telangiectasia/metabolismo , Síndrome de Beckwith-Wiedemann/complicações , Síndrome de Beckwith-Wiedemann/metabolismo , Carcinogênese/genética , Carcinogênese/metabolismo , Síndrome de Costello/complicações , Síndrome de Costello/metabolismo , Síndrome de Down/complicações , Síndrome de Down/metabolismo , Anemia de Fanconi/complicações , Anemia de Fanconi/metabolismo , Humanos , Lactente , Neoplasias/genética , Estresse Oxidativo/efeitos dos fármacos , Risco
17.
Epigenomics ; 5(3): 331-40, 2013 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-23750647

RESUMO

Genomic imprinting is a parent-of-origin allele-specific epigenetic process that is critical for normal development and health. The establishment and maintenance of normal imprinting is dependent on both cis-acting imprinting control centers, which are marked by differentially (parental allele specific) methylated marks, and trans mechanisms, which regulate the establishment and/or maintenance of the correct methylation epigenotype at the imprinting control centers. Studies of rare human imprinting disorders such as familial hydatidiform mole, Beckwith-Wiedemann syndrome and familial transient neonatal diabetes mellitus have enabled the identification of genetic (e.g., mutations in KHDC3L [C6ORF221], NLRP2 [NALP2], NLRP7 [NALP7] and ZFP57) and environmental (assisted reproductive technologies) factors that can disturb the normal trans mechanisms for imprinting establishment and/or maintenance. Here we review the clinical and molecular aspects of these imprinting disorders in order to demonstrate how the study of rare inherited disorders can illuminate the molecular characteristics of fundamental epigenetic processes, such as genomic imprinting.


Assuntos
Técnicas de Reprodução Assistida , Proteínas Adaptadoras de Transdução de Sinal/genética , Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Proteínas Reguladoras de Apoptose , Síndrome de Beckwith-Wiedemann/genética , Síndrome de Beckwith-Wiedemann/metabolismo , Síndrome de Beckwith-Wiedemann/patologia , Metilação de DNA , Feminino , Impressão Genômica , Humanos , Mola Hidatiforme/genética , Mola Hidatiforme/metabolismo , Mola Hidatiforme/patologia , Gravidez , Proteínas/genética , Proteínas/metabolismo , Síndrome de Silver-Russell/genética , Síndrome de Silver-Russell/metabolismo , Síndrome de Silver-Russell/patologia
18.
Pathology ; 44(6): 519-27, 2012 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-22772341

RESUMO

AIMS: Placental mesenchymal dysplasia (PMD) is a rare condition which is associated with the disparate fetal outcomes of Beckwith-Wiedemann syndrome (BWS), fetal growth restriction or intrauterine and neonatal death. We aimed to investigate the potential epigenetic/genetic anomalies associated with PMD and their relationship with the different causes of BWS. METHODS: Eight archival cases in which PMD, BWS or both were diagnosed were investigated by correlating morphology with p57 Kip2 expression, XY fluorescence in situ hybridisation (FISH) analysis and DNA genotyping. RESULTS: Placentae from BWS cases caused by aberrant IC2 methylation, leading to abnormal p57 Kip2 expression, did not show PMD but had a striking excess of extravillous trophoblast. PMD in the absence of BWS was caused by androgenetic/biparental mosaicism. The single case of BWS with PMD was due to mosaic uniparental disomy of 11p15.5. In the latter two aetiologies, our results indicate that the uniparental disomy is confined to the villous mesenchyme. CONCLUSIONS: These results suggest that the link between PMD and BWS is uniparental disomy of genes confined to the telomeric IC1 region of 11p15.5. A strong candidate gene is IGF2, a known growth factor of placental mesenchyme.


Assuntos
Síndrome de Beckwith-Wiedemann/patologia , Inibidor de Quinase Dependente de Ciclina p57/genética , Fator de Crescimento Insulin-Like II/genética , Placenta/patologia , Trofoblastos/patologia , Síndrome de Beckwith-Wiedemann/genética , Síndrome de Beckwith-Wiedemann/metabolismo , Cromossomos Humanos Par 11 , Inibidor de Quinase Dependente de Ciclina p57/metabolismo , Metilação de DNA , Epigenômica , Feminino , Estudos de Associação Genética , Genótipo , Humanos , Fator de Crescimento Insulin-Like II/metabolismo , Fenótipo , Placenta/metabolismo , Gravidez , Trofoblastos/metabolismo
19.
Nat Genet ; 44(7): 788-92, 2012 May 27.
Artigo em Inglês | MEDLINE | ID: mdl-22634751

RESUMO

IMAGe syndrome (intrauterine growth restriction, metaphyseal dysplasia, adrenal hypoplasia congenita and genital anomalies) is an undergrowth developmental disorder with life-threatening consequences. An identity-by-descent analysis in a family with IMAGe syndrome identified a 17.2-Mb locus on chromosome 11p15 that segregated in the affected family members. Targeted exon array capture of the disease locus, followed by high-throughput genomic sequencing and validation by dideoxy sequencing, identified missense mutations in the imprinted gene CDKN1C (also known as P57KIP2) in two familial and four unrelated patients. A familial analysis showed an imprinted mode of inheritance in which only maternal transmission of the mutation resulted in IMAGe syndrome. CDKN1C inhibits cell-cycle progression, and we found that targeted expression of IMAGe-associated CDKN1C mutations in Drosophila caused severe eye growth defects compared to wild-type CDKN1C, suggesting a gain-of-function mechanism. All IMAGe-associated mutations clustered in the PCNA-binding domain of CDKN1C and resulted in loss of PCNA binding, distinguishing them from the mutations of CDKN1C that cause Beckwith-Wiedemann syndrome, an overgrowth syndrome.


Assuntos
Hiperplasia Suprarrenal Congênita/genética , Inibidor de Quinase Dependente de Ciclina p57/genética , Retardo do Crescimento Fetal/genética , Doenças Genéticas Ligadas ao Cromossomo X/genética , Mutação , Osteocondrodisplasias/genética , Antígeno Nuclear de Célula em Proliferação/metabolismo , Hiperplasia Suprarrenal Congênita/metabolismo , Insuficiência Adrenal , Animais , Síndrome de Beckwith-Wiedemann/genética , Síndrome de Beckwith-Wiedemann/metabolismo , Linhagem Celular Transformada , Cromossomos Humanos Par 11 , Inibidor de Quinase Dependente de Ciclina p57/metabolismo , Drosophila , Éxons , Feminino , Retardo do Crescimento Fetal/metabolismo , Doenças Genéticas Ligadas ao Cromossomo X/metabolismo , Loci Gênicos , Predisposição Genética para Doença , Células HEK293 , Humanos , Hipoadrenocorticismo Familiar , Masculino , Osteocondrodisplasias/metabolismo , Antígeno Nuclear de Célula em Proliferação/genética , Ligação Proteica/genética , Estrutura Terciária de Proteína/genética
20.
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
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...