Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 25
Filtrar
1.
Biochim Biophys Acta Mol Basis Dis ; 1870(5): 167175, 2024 Apr 16.
Artigo em Inglês | MEDLINE | ID: mdl-38626828

RESUMO

Loss of prolyl endopeptidase-like (PREPL) encoding a serine hydrolase with (thio)esterase activity leads to the recessive metabolic disorder Congenital Myasthenic Syndrome-22 (CMS22). It is characterized by severe neonatal hypotonia, feeding problems, growth retardation, and hyperphagia leading to rapid weight gain later in childhood. The phenotypic similarities with Prader-Willi syndrome (PWS) are striking, suggesting that similar pathways are affected. The aim of this study was to identify changes in the hypothalamic-pituitary axis in mouse models for both disorders and to examine mitochondrial function in skin fibroblasts of patients and knockout cell lines. We have demonstrated that Prepl is downregulated in the brains of neonatal PWS-IC-p/+m mice. In addition, the hypothalamic-pituitary axis is similarly affected in both Prepl-/- and PWS-IC-p/+m mice resulting in defective orexigenic signaling and growth retardation. Furthermore, we demonstrated that mitochondrial function is altered in PREPL knockout HEK293T cells and can be rescued with the supplementation of coenzyme Q10. Finally, PREPL-deficient and PWS patient skin fibroblasts display defective mitochondrial bioenergetics. The mitochondrial dysfunction in PWS fibroblasts can be rescued by overexpression of PREPL. In conclusion, we provide the first molecular parallels between CMS22 and PWS, raising the possibility that PREPL substrates might become therapeutic targets for treating both disorders.

2.
J Clin Med ; 11(14)2022 Jul 12.
Artigo em Inglês | MEDLINE | ID: mdl-35887798

RESUMO

Prader−Willi syndrome (PWS) is a complex, rare genetic disorder caused by a loss of expression of paternally expressed genes on chromosome 15q11.2-q13. The most common underlying genotypes are paternal deletion (DEL) and maternal uniparental disomy (mUPD). DELs can be subdivided into type 1 (DEL-1) and (smaller) type 2 deletions (DEL-2). Most research has focused on behavioral, cognitive and psychological differences between the different genotypes. However, little is known about physical health problems in relation to genetic subtypes. In this cross-sectional study, we compare physical health problems and other clinical features among adults with PWS caused by DEL (N = 65, 12 DEL-1, 27 DEL-2) and mUPD (N = 65). A meta-analysis, including our own data, showed that BMI was 2.79 kg/m2 higher in adults with a DEL (p = 0.001). There were no significant differences between DEL-1 and DEL-2. Scoliosis was more prevalent among adults with a DEL (80% vs. 58%; p = 0.04). Psychotic episodes were more prevalent among adults with an mUPD (44% vs. 9%; p < 0.001). In conclusion, there were no significant differences in physical health outcomes between the genetic subtypes, apart from scoliosis and BMI. The differences in health problems, therefore, mainly apply to the psychological domain.

3.
Genes (Basel) ; 12(6)2021 06 07.
Artigo em Inglês | MEDLINE | ID: mdl-34200226

RESUMO

Prader-Willi syndrome (PWS) is a rare genetic condition characterized by hypotonia, intellectual disability, and hypothalamic dysfunction, causing pituitary hormone deficiencies and hyperphagia, ultimately leading to obesity. PWS is most often caused by the loss of expression of a cluster of genes on chromosome 15q11.2-13. Patients with Prader-Willi-like syndrome (PWLS) display features of the PWS phenotype without a classical PWS genetic defect. We describe a 46-year-old patient with PWLS, including hypotonia, intellectual disability, hyperphagia, and pituitary hormone deficiencies. Routine genetic tests for PWS were normal, but a homozygous missense variant NM_003097.3(SNRPN):c.193C>T, p.(Arg65Trp) was identified. Single nucleotide polymorphism array showed several large regions of homozygosity, caused by high-grade consanguinity between the parents. Our functional analysis, the 'Pipeline for Rapid in silico, in vivo, in vitro Screening of Mutations' (PRiSM) screen, showed that overexpression of SNRPN-p.Arg65Trp had a dominant negative effect, strongly suggesting pathogenicity. However, it could not be confirmed that the variant was responsible for the phenotype of the patient. In conclusion, we present a unique homozygous missense variant in SNURF-SNRPN in a patient with PWLS. We describe the diagnostic trajectory of this patient and the possible contributors to her phenotype in light of the current literature on the genotype-phenotype relationship in PWS.


Assuntos
Proteínas Nucleares/genética , Síndrome de Prader-Willi/genética , Proteínas Centrais de snRNP/genética , Células Cultivadas , Feminino , Impressão Genômica , Células HEK293 , Homozigoto , Humanos , Pessoa de Meia-Idade , Mutação de Sentido Incorreto , Proteínas Nucleares/metabolismo , Fenótipo , Síndrome de Prader-Willi/diagnóstico , Proteínas Centrais de snRNP/metabolismo
4.
Hum Mol Genet ; 28(2): 220-229, 2019 01 15.
Artigo em Inglês | MEDLINE | ID: mdl-30260400

RESUMO

Angelman syndrome, Prader-Will syndrome and Dup15q syndrome map to a cluster of imprinted genes located at 15q11-q13. Imprinting at this domain is regulated by an imprinting control region consisting of two distinct elements, the Angelman syndrome imprinting center (AS-IC) and the Prader-Willi syndrome imprinting center (PWS-IC). Individuals inheriting deletions of the AS-IC exhibit reduced expression of the maternally expressed UBE3A gene and biallelic expression of paternal-only genes. We have previously demonstrated that AS-IC activity partly consists of providing transcription across the PWS-IC in oocytes, and that these transcripts are necessary for maternal imprinting of Snrpn. Here we report a novel mouse mutation that truncates transcripts prior to transiting the PWS-IC and results in a domain-wide imprinting defect. These results confirm a transcription-based model for imprint setting at this domain. The imprinting defect can be preempted by removal of the transcriptional block in oocytes, but not by its removal in early embryos. Imprinting defect mice exhibit several traits often found in individuals with Angelman syndrome imprinting defects.


Assuntos
Síndrome de Angelman/genética , Modelos Animais de Doenças , Impressão Genômica , Animais , Metilação de DNA , Éxons , Feminino , Regulação da Expressão Gênica , Masculino , Herança Materna , Camundongos , Mutação , Oócitos/metabolismo , Proteínas Centrais de snRNP/genética
5.
J Endocrinol ; 232(1): 123-135, 2017 01.
Artigo em Inglês | MEDLINE | ID: mdl-27799465

RESUMO

Prader-Willi syndrome (PWS), a neurodevelopmental disorder caused by loss of paternal gene expression from 15q11-q13, is characterised by growth retardation, hyperphagia and obesity. However, as single gene mutation mouse models for this condition display an incomplete spectrum of the PWS phenotype, we have characterised the metabolic impairment in a mouse model for 'full' PWS, in which deletion of the imprinting centre (IC) abolishes paternal gene expression from the entire PWS cluster. We show that PWS-ICdel mice displayed postnatal growth retardation, with reduced body weight, hyperghrelinaemia and marked abdominal leanness; proportionate retroperitoneal, epididymal/omental and inguinal white adipose tissue (WAT) weights being reduced by 82%, 84% and 67%, respectively. PWS-ICdel mice also displayed a 48% reduction in proportionate interscapular brown adipose tissue (isBAT) weight with significant 'beiging' of abdominal WAT, and a 2°C increase in interscapular surface body temperature. Maintenance of PWS-ICdel mice under thermoneutral conditions (30°C) suppressed the thermogenic activity in PWS-ICdel males, but failed to elevate the abdominal WAT weight, possibly due to a normalisation of caloric intake. Interestingly, PWS-ICdel mice also showed exaggerated food hoarding behaviour with standard and high-fat diets, but despite becoming hyperphagic when switched to a high-fat diet, PWS-ICdel mice failed to gain weight. This evidence indicates that, unlike humans with PWS, loss of paternal gene expression from the PWS cluster in mice results in abdominal leanness. Although reduced subcutaneous insulation may lead to exaggerated heat loss and thermogenesis, abdominal leanness is likely to arise from a reduced lipid storage capacity rather than increased energy utilisation in BAT.


Assuntos
Tecido Adiposo Branco/metabolismo , Peso Corporal/fisiologia , Mutação , Fenótipo , Síndrome de Prader-Willi/genética , Magreza/genética , Tecido Adiposo Marrom/metabolismo , Animais , Metilação de DNA , Modelos Animais de Doenças , Masculino , Camundongos , Deleção de Sequência , Termogênese/fisiologia
7.
Proc Natl Acad Sci U S A ; 112(22): 6871-5, 2015 Jun 02.
Artigo em Inglês | MEDLINE | ID: mdl-25378697

RESUMO

Clusters of imprinted genes are often controlled by an imprinting center that is necessary for allele-specific gene expression and to reprogram parent-of-origin information between generations. An imprinted domain at 15q11-q13 is responsible for both Angelman syndrome (AS) and Prader-Willi syndrome (PWS), two clinically distinct neurodevelopmental disorders. Angelman syndrome arises from the lack of maternal contribution from the locus, whereas Prader-Willi syndrome results from the absence of paternally expressed genes. In some rare cases of PWS and AS, small deletions may lead to incorrect parent-of-origin allele identity. DNA sequences common to these deletions define a bipartite imprinting center for the AS-PWS locus. The PWS-smallest region of deletion overlap (SRO) element of the imprinting center activates expression of genes from the paternal allele. The AS-SRO element generates maternal allele identity by epigenetically inactivating the PWS-SRO in oocytes so that paternal genes are silenced on the future maternal allele. Here we have investigated functional activities of the AS-SRO, the element necessary for maternal allele identity. We find that, in humans, the AS-SRO is an oocyte-specific promoter that generates transcripts that transit the PWS-SRO. Similar upstream promoters were detected in bovine oocytes. This result is consistent with a model in which imprinting centers become DNA methylated and acquire maternal allele identity in oocytes in response to transiting transcription.


Assuntos
Síndrome de Angelman/genética , Regulação da Expressão Gênica/genética , Impressão Genômica/genética , Modelos Biológicos , Síndrome de Prader-Willi/genética , Animais , Bovinos , Metilação de DNA , Primers do DNA/genética , Componentes do Gene , Humanos , Oócitos/metabolismo , Regiões Promotoras Genéticas/genética , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Análise de Sequência de RNA , Especificidade da Espécie , Proteínas Centrais de snRNP/genética , Proteínas Centrais de snRNP/metabolismo
8.
Reprod Sci ; 22(2): 250-7, 2015 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-25031318

RESUMO

Adenine nucleotide translocase (Ant) facilitates the exchange of adenosine triphosphate across the mitochondrial inner membrane and plays a critical role for bioenergetics in eukaryotes. Mice have 3 Ant paralogs, Ant1 (Slc25a4), Ant2 (Slc25a5), and Ant4 (Slc25a31), which are expressed in a tissue-dependent manner. We previously identified that Ant4 was expressed exclusively in testicular germ cells in adult mice and essential for spermatogenesis and subsequently male fertility. Further investigation into the process of spermatogenesis revealed that Ant4 was particularly highly expressed during meiotic prophase I and indispensable for normal progression of leptotene spermatocytes to the stages thereafter. In contrast, the expression and roles of Ant4 in female germ cells have not previously been elucidated. Here, we demonstrate that the Ant4 gene is expressed during embryonic ovarian development during which meiotic prophase I occurs. We confirmed embryonic ovary-specific Ant4 expression using a bacterial artificial chromosome transgene. In contrast to male, however, Ant4 null female mice were fertile although the litter size was slightly decreased. They showed apparently normal ovarian development which was morphologically indistinguishable from the control animals. These data indicate that Ant4 is a meiosis-specific gene expressed during both male and female gametogenesis however indispensable only during spermatogenesis and not oogenesis. The differential effects of Ant4 depletion within the processes of male and female gametogenesis may be explained by meiosis-specific inactivation of the X-linked Ant2 gene in male, a somatic paralog of the Ant4 gene.


Assuntos
Proteínas de Membrana Transportadoras/metabolismo , Oogênese , Ovário/metabolismo , Animais , Feminino , Fertilidade , Regulação da Expressão Gênica no Desenvolvimento , Genótipo , Tamanho da Ninhada de Vivíparos , Masculino , Prófase Meiótica I , Proteínas de Membrana Transportadoras/genética , Camundongos Endogâmicos C57BL , Camundongos Endogâmicos DBA , Camundongos Knockout , Ovário/embriologia , Fenótipo , Regiões Promotoras Genéticas , Espermatogênese
9.
Epigenetics ; 9(11): 1540-56, 2014 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-25482058

RESUMO

Reduced representation bisulfite sequencing (RRBS) was used to analyze DNA methylation patterns across the mouse brain genome in mice carrying a deletion of the Prader-Willi syndrome imprinting center (PWS-IC) on either the maternally- or paternally-inherited chromosome. Within the ~3.7 Mb imprinted Angelman/Prader-Willi syndrome (AS/PWS) domain, 254 CpG sites were interrogated for changes in methylation due to PWS-IC deletion. Paternally-inherited deletion of the PWS-IC increased methylation levels ~2-fold at each CpG site (compared to wild-type controls) at differentially methylated regions (DMRs) associated with 5' CpG island promoters of paternally-expressed genes; these methylation changes extended, to a variable degree, into the adjacent CpG island shores. Maternal PWS-IC deletion yielded little or no changes in methylation at these DMRs, and methylation of CpG sites outside of promoter DMRs also was unchanged upon maternal or paternal PWS-IC deletion. Using stringent ascertainment criteria, ~750,000 additional CpG sites were also interrogated across the entire mouse genome. This analysis identified 26 loci outside of the imprinted AS/PWS domain showing altered DNA methylation levels of ≥25% upon PWS-IC deletion. Curiously, altered methylation at 9 of these loci was a consequence of maternal PWS-IC deletion (maternal PWS-IC deletion by itself is not known to be associated with a phenotype in either humans or mice), and 10 of these loci exhibited the same changes in methylation irrespective of the parental origin of the PWS-IC deletion. These results suggest that the PWS-IC may affect DNA methylation at these loci by directly interacting with them, or may affect methylation at these loci through indirect downstream effects due to PWS-IC deletion. They further suggest the PWS-IC may have a previously uncharacterized function outside of the imprinted AS/PWS domain.


Assuntos
Encéfalo/fisiologia , Metilação de DNA , Impressão Genômica , Síndrome de Prader-Willi/genética , Síndrome de Angelman/genética , Animais , Antígenos de Neoplasias/genética , Proteínas de Transporte/genética , Ilhas de CpG , Feminino , Sequenciamento de Nucleotídeos em Larga Escala , Masculino , Camundongos Endogâmicos C57BL , Camundongos Mutantes , Proteínas de Neoplasias/genética , Regiões Promotoras Genéticas , Proteínas/genética , Ribonucleoproteínas/genética , Deleção de Sequência , Ubiquitina-Proteína Ligases
10.
Clin Transl Sci ; 6(5): 347-55, 2013 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-24127921

RESUMO

Prader-Willi syndrome (PWS) is a genetic disorder caused by deficiency of imprinted gene expression from the paternal chromosome 15q11-15q13 and clinically characterized by neonatal hypotonia, short stature, cognitive impairment, hypogonadism, hyperphagia, morbid obesity, and diabetes. Previous clinical studies suggest that a defect in energy metabolism may be involved in the pathogenesis of PWS. We focused our attention on the genes associated with energy metabolism and found that there were 95 and 66 mitochondrial genes differentially expressed in PWS muscle and brain, respectively. Assessment of enzyme activities of mitochondrial oxidative phosphorylation complexes in the brain, heart, liver, and muscle were assessed. We found the enzyme activities of the cardiac mitochondrial complexes II+‫III were up-regulated in the PWS imprinting center deletion mice compared to the wild-type littermates. These studies suggest that differential gene expression, especially of the mitochondrial genes may contribute to the pathophysiology of PWS.


Assuntos
Perfilação da Expressão Gênica , Regulação da Expressão Gênica , Impressão Genômica/genética , Mitocôndrias/genética , Mitocôndrias/patologia , Síndrome de Prader-Willi/genética , Deleção de Sequência/genética , Animais , Encéfalo/metabolismo , Encéfalo/patologia , Modelos Animais de Doenças , Complexo de Proteínas da Cadeia de Transporte de Elétrons/metabolismo , Redes Reguladoras de Genes/genética , Genoma/genética , Camundongos , Mitocôndrias/ultraestrutura , Músculos/metabolismo , Músculos/patologia , Análise de Sequência com Séries de Oligonucleotídeos , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Reprodutibilidade dos Testes
11.
Mamm Genome ; 24(5-6): 165-78, 2013 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-23609791

RESUMO

Prader-Willi syndrome (PWS) occurs in about 1 in 15,000 individuals and is a contiguous gene disorder causing developmental disability, hyperphagia usually with obesity, and behavioral problems, including an increased incidence of psychiatric illness. The genomic imprinting that regulates allele-specific expression of PWS candidate genes, the fact that multiple genes are typically inactivated, and the presence of many genes that produce functional RNAs rather than proteins has complicated the identification of the underlying genetic pathophysiology of PWS. Over 30 genetically modified mouse strains that have been developed and characterized have been instrumental in elucidating the genetic and epigenetic mechanisms for the regulation of PWS genes and in discovering their physiological functions. In 2011, a PWS Animal Models Working Group (AMWG) was established to generate discussions and facilitate exchange of ideas regarding the best use of PWS animal models. Here, we summarize the goals of the AMWG, describe current animal models of PWS, and make recommendations for strategies to maximize the utility of animal models and for the development and use of new animal models of PWS.


Assuntos
Modelos Animais de Doenças , Camundongos , Síndrome de Prader-Willi/genética , Animais , Humanos , Camundongos/genética , Camundongos/metabolismo , Síndrome de Prader-Willi/metabolismo
12.
PLoS One ; 8(2): e52390, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-23390487

RESUMO

The Angelman/Prader-Willi syndrome (AS/PWS) domain contains at least 8 imprinted genes regulated by a bipartite imprinting center (IC) associated with the SNRPN gene. One component of the IC, the PWS-IC, governs the paternal epigenotype and expression of paternal genes. The mechanisms by which imprinting and expression of paternal genes within the AS/PWS domain - such as MKRN3 and NDN - are regulated by the PWS-IC are unclear. The syntenic region in the mouse is organized and imprinted similarly to the human domain with the murine PWS-IC defined by a 6 kb interval within the Snrpn locus that includes the promoter. To identify regulatory elements that may mediate PWS-IC function, we mapped the location and allele-specificity of DNase I hypersensitive (DH) sites within the PWS-IC in brain cells, then identified transcription factor binding sites within a subset of these DH sites. Six major paternal-specific DH sites were detected in the Snrpn gene, five of which map within the 6 kb PWS-IC. We postulate these five DH sites represent functional components of the murine PWS-IC. Analysis of transcription factor binding within multiple DH sites detected nuclear respiratory factors (NRF's) and YY1 specifically on the paternal allele. NRF's and YY1 were also detected in the paternal promoter region of the murine Mrkn3 and Ndn genes. These results suggest that NRF's and YY1 may facilitate PWS-IC function and coordinately regulate expression of paternal genes. The presence of NRF's also suggests a link between transcriptional regulation within the AS/PWS domain and regulation of respiration. 3C analyses indicated Mkrn3 lies in close proximity to the PWS-IC on the paternal chromosome, evidence that the PWS-IC functions by allele-specific interaction with its distal target genes. This could occur by allele-specific co-localization of the PWS-IC and its target genes to transcription factories containing NRF's and YY1.


Assuntos
Síndrome de Angelman/genética , Regulação da Expressão Gênica , Fatores Nucleares Respiratórios/genética , Síndrome de Prader-Willi/genética , Elementos Reguladores de Transcrição , Fator de Transcrição YY1/genética , Proteínas Centrais de snRNP/genética , Alelos , Síndrome de Angelman/metabolismo , Síndrome de Angelman/patologia , Animais , Sequência de Bases , Sítios de Ligação , Desoxirribonuclease I/metabolismo , Loci Gênicos , Impressão Genômica , Humanos , Camundongos , Camundongos Transgênicos , Dados de Sequência Molecular , Fatores Nucleares Respiratórios/metabolismo , Síndrome de Prader-Willi/metabolismo , Síndrome de Prader-Willi/patologia , Ligação Proteica , Sintenia , Transcrição Gênica , Fator de Transcrição YY1/metabolismo , Proteínas Centrais de snRNP/metabolismo
13.
Methods Mol Biol ; 925: 61-6, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-22907490

RESUMO

Primordial germ cells (PGCs) play essential roles in both reproduction and development. In this chapter, we describe a method used in our laboratory for the immunopurification of PGCs from the mouse embryo. After dissection and disruption of the fetal gonad, PGCs are identified by a monoclonal antibody recognizing an epitope characteristic of pluripotent stem cells. After reaction with a paramagnetic bead-linked secondary antibody, the cell mixture is applied to a strong magnetic field. PGCs are recovered by release from the magnetic field. Purity is assessed by the alkaline phosphatase activity inherent to PGCs.


Assuntos
Separação Celular/métodos , Células-Tronco Embrionárias/citologia , Imunoprecipitação/métodos , Campos Magnéticos , Óvulo/citologia , Fosfatase Alcalina/metabolismo , Animais , Embrião de Mamíferos/citologia , Células-Tronco Embrionárias/metabolismo , Feminino , Camundongos , Microesferas
14.
Proc Natl Acad Sci U S A ; 109(9): 3446-50, 2012 Feb 28.
Artigo em Inglês | MEDLINE | ID: mdl-22331910

RESUMO

Imprinted gene expression associated with Prader-Willi syndrome (PWS) and Angelman syndrome (AS) is controlled by two imprinting centers (ICs), the PWS-IC and the AS-IC. The PWS-IC operates in cis to activate transcription of genes that are expressed exclusively from the paternal allele. We have created a conditional allele of the PWS-IC to investigate its developmental activity. Deletion of the paternal PWS-IC in the embryo before implantation abolishes expression of the paternal-only genes in the neonatal brain. Surprisingly, deletion of the PWS-IC in early brain progenitors does not affect the subsequent imprinted status of PWS/AS genes in the newborn brain. These results indicate that the PWS-IC functions to protect the paternal epigenotype at the epiblast stage of development but is dispensable thereafter.


Assuntos
Regulação da Expressão Gênica no Desenvolvimento , Impressão Genômica , Síndrome de Prader-Willi/genética , Alelos , Animais , Blastocisto , Encéfalo/embriologia , Metilação de DNA , Modelos Animais de Doenças , Desenvolvimento Embrionário/genética , Feminino , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Mutantes , Proteínas do Tecido Nervoso/biossíntese , Proteínas do Tecido Nervoso/genética , Neurogênese/genética , Síndrome de Prader-Willi/fisiopatologia , Regiões Promotoras Genéticas/genética , RNA Nucleolar Pequeno/biossíntese , RNA Nucleolar Pequeno/genética , Deleção de Sequência , Fatores de Tempo , Transcrição Gênica , Proteínas Centrais de snRNP/biossíntese , Proteínas Centrais de snRNP/genética
15.
Hum Mol Genet ; 20(17): 3461-6, 2011 Sep 01.
Artigo em Inglês | MEDLINE | ID: mdl-21659337

RESUMO

The human chromosomal 15q11-15q13 region is subject to both maternal and paternal genomic imprinting. Absence of paternal gene expression from this region results in Prader-Willi syndrome (PWS), while absence of maternal gene expression leads to Angelman syndrome. Transcription of paternally expressed genes in the region depends upon an imprinting center termed the PWS-IC. Imprinting defects in PWS can be caused by microdeletions and the smallest commonly deleted region indicates that the PWS-IC lies within a region of 4.3 kb. The function and location of the PWS-IC is evolutionarily conserved, but delineation of the PWS-IC in mouse has proven difficult. The first targeted mutation of the PWS-IC, a deletion of 35 kb spanning Snrpn exon 1, exhibited a complete PWS-IC deletion phenotype. Pups inheriting this mutation paternally showed a complete loss of paternal gene expression and died neonatally. A reported deletion of 4.8 kb showed only a reduction in paternal gene expression and incomplete penetrance of neonatal lethality, suggesting that some PWS-IC function had been retained. Here, we report that a 6 kb deletion spanning Snrpn exon 1 exhibits a complete PWS-IC deletion phenotype. Pups inheriting this mutation paternally lack detectable expression of all PWS genes and paternal silencing of Ube3a, exhibit maternal DNA methylation imprints at Ndn and Mkrn3 and suffer failure to thrive leading to a fully penetrant neonatal lethality.


Assuntos
Impressão Genômica/genética , Síndrome de Prader-Willi/genética , Animais , Southern Blotting , Linhagem Celular , Metilação de DNA/genética , Humanos , Camundongos , Mutação/genética
16.
PLoS Genet ; 7(12): e1002422, 2011 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-22242001

RESUMO

The Prader-Willi syndrome (PWS [MIM 17620]) and Angelman syndrome (AS [MIM 105830]) locus is controlled by a bipartite imprinting center (IC) consisting of the PWS-IC and the AS-IC. The most widely accepted model of IC function proposes that the PWS-IC activates gene expression from the paternal allele, while the AS-IC acts to epigenetically inactivate the PWS-IC on the maternal allele, thus silencing the paternally expressed genes. Gene order and imprinting patterns at the PWS/AS locus are well conserved from human to mouse; however, a murine AS-IC has yet to be identified. We investigated a potential regulatory role for transcription from the Snrpn alternative upstream exons in silencing the maternal allele using a murine transgene containing Snrpn and three upstream exons. This transgene displayed appropriate imprinted expression and epigenetic marks, demonstrating the presence of a functional AS-IC. Transcription of the upstream exons from the endogenous locus correlates with imprint establishment in oocytes, and this upstream exon expression pattern was conserved on the transgene. A transgene bearing targeted deletions of each of the three upstream exons exhibited loss of imprinting upon maternal transmission. These results support a model in which transcription from the Snrpn upstream exons directs the maternal imprint at the PWS-IC.


Assuntos
Síndrome de Angelman/genética , Impressão Genômica , Síndrome de Prader-Willi/genética , Proteínas Centrais de snRNP/genética , Alelos , Animais , Metilação de DNA , Epigênese Genética/genética , Éxons , Regulação da Expressão Gênica , Loci Gênicos , Humanos , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Oócitos/metabolismo , RNA Mensageiro Estocado/genética , Transcrição Gênica
17.
Eur J Neurosci ; 31(1): 156-64, 2010 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-20092561

RESUMO

The genes in the imprinted cluster on human chromosome 15q11-q13 are known to contribute to psychiatric conditions such as schizophrenia and autism. Major disruptions of this interval leading to a lack of paternal allele expression give rise to Prader-Willi syndrome (PWS), a neurodevelopmental disorder with core symptoms of a failure to thrive in infancy and, on emergence from infancy, learning disabilities and over-eating. Individuals with PWS also display a number of behavioural problems and an increased incidence of neuropsychiatric abnormalities, which recent work indicates involve aspects of frontal dysfunction. To begin to examine the contribution of genes in this interval to relevant psychological and behavioural phenotypes, we exploited the imprinting centre (IC) deletion mouse model for PWS (PWS-IC(+/-)) and the five-choice serial reaction time task (5-CSRTT), which is primarily an assay of visuospatial attention and response control that is highly sensitive to frontal manipulations. Locomotor activity, open-field behaviour and sensorimotor gating were also assessed. PWS-IC(+/-) mice displayed reduced locomotor activity, increased acoustic startle responses and decreased prepulse inhibition of startle responses. In the 5-CSRTT, the PWS-IC(+/-) mice showed deficits in discriminative response accuracy, increased correct reaction times and increased omissions. Task manipulations confirmed that these differences were likely to be due to impaired attention. Our data recapitulate several aspects of the PWS clinical condition, including findings consistent with frontal abnormalities, and may indicate novel contributions of the imprinted genes found in 15q11-q13 to behavioural and cognitive function generally.


Assuntos
Transtornos Cognitivos/genética , Comportamento Exploratório , Atividade Motora/genética , Síndrome de Prader-Willi/genética , Animais , Atenção , Peso Corporal , Encéfalo/fisiopatologia , Transtornos Cognitivos/metabolismo , Transtornos Cognitivos/fisiopatologia , Discriminação Psicológica , Modelos Animais de Doenças , Comportamento Exploratório/fisiologia , Insuficiência de Crescimento/genética , Insuficiência de Crescimento/fisiopatologia , Feminino , Impressão Genômica , Masculino , Camundongos , Camundongos Transgênicos , Atividade Motora/fisiologia , Testes Neuropsicológicos , Síndrome de Prader-Willi/fisiopatologia , Síndrome de Prader-Willi/psicologia , Tempo de Reação , Reflexo de Sobressalto/genética , Reflexo de Sobressalto/fisiologia , Deleção de Sequência
18.
Neurogenetics ; 11(2): 145-51, 2010 May.
Artigo em Inglês | MEDLINE | ID: mdl-19894069

RESUMO

Mutations affecting a cluster of coordinately regulated imprinted genes located at 15q11-q13 underlie both Prader-Willi syndrome (PWS) and Angelman syndrome (AS). Disruption of the predominately maternally expressed UBE3A locus is sufficient to meet diagnostic criteria for AS. However, AS patients with a deletion of the entire PWS/AS locus often have more severe traits than patients with point mutations in UBE3A suggesting that other genes contribute to the syndrome. ATP10A resides 200 kb telomeric to UBE3A and is of uncertain imprinted status. An initial report indicated bialleleic expression of the murine Atp10a in all tissues, but a subsequent report suggests that Atp10a is predominantly maternally expressed in the hippocampus and olfactory bulb. To resolve this discrepancy, we investigated Atp10a allelic expression in the brain, DNA methylation status, and sensitivity to mutations of the PWS imprinting center, an element required for imprinted gene expression in the region. We report that Atp10a is biallelically expressed in both the newborn and adult brain, and Atp10a allelic expression is insensitive to deletion or mutation of the PWS imprinting center. The CpG island associated with Atp10a is hypomethylated, a result consistent with the notion that Atp10a is not an imprinted gene.


Assuntos
Adenosina Trifosfatases/genética , Impressão Genômica , Proteínas de Membrana Transportadoras/genética , Família Multigênica , Síndrome de Angelman/genética , Animais , Ilhas de CpG , Metilação de DNA , Feminino , Humanos , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Polimorfismo Genético , Síndrome de Prader-Willi/genética , Análise de Sequência de DNA
19.
Hum Mol Genet ; 18(22): 4227-38, 2009 Nov 15.
Artigo em Inglês | MEDLINE | ID: mdl-19656775

RESUMO

Imprinting, non-coding RNA and chromatin organization are modes of epigenetic regulation that modulate gene expression and are necessary for mammalian neurodevelopment. The only two known mammalian clusters of genes encoding small nucleolar RNAs (snoRNAs), SNRPN through UBE3A(15q11-q13/7qC) and GTL2(14q32.2/12qF1), are neuronally expressed, localized to imprinted loci and involved in at least five neurodevelopmental disorders. Deficiency of the paternal 15q11-q13 snoRNA HBII-85 locus is necessary to cause the neurodevelopmental disorder Prader-Willi syndrome (PWS). Here we show epigenetically regulated chromatin decondensation at snoRNA clusters in human and mouse brain. An 8-fold allele-specific decondensation of snoRNA chromatin was developmentally regulated specifically in maturing neurons, correlating with HBII-85 nucleolar accumulation and increased nucleolar size. Reciprocal mouse models revealed a genetic and epigenetic requirement of the 35 kb imprinting center (IC) at the Snrpn-Ube3a locus for transcriptionally regulated chromatin decondensation. PWS human brain and IC deletion mouse Purkinje neurons showed significantly decreased nucleolar size, demonstrating the essential role of the 15q11-q13 HBII-85 locus in neuronal nucleolar maturation. These results are relevant to understanding the molecular pathogenesis of multiple human neurodevelopmental disorders, including PWS and some causes of autism.


Assuntos
Nucléolo Celular/química , Montagem e Desmontagem da Cromatina , Impressão Genômica , Neurônios/metabolismo , Síndrome de Prader-Willi/genética , RNA Nucleolar Pequeno/genética , Adulto , Animais , Nucléolo Celular/genética , Nucléolo Celular/metabolismo , Cromatina/metabolismo , Cromossomos de Mamíferos/genética , Cromossomos de Mamíferos/metabolismo , Humanos , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Pessoa de Meia-Idade , Neurônios/química , Síndrome de Prader-Willi/metabolismo , RNA Nucleolar Pequeno/metabolismo , Ubiquitina-Proteína Ligases/genética , Ubiquitina-Proteína Ligases/metabolismo , Proteínas Centrais de snRNP/genética , Proteínas Centrais de snRNP/metabolismo
20.
Mamm Genome ; 18(4): 255-62, 2007 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-17514346

RESUMO

Prader-Willi syndrome (PWS) and Angelman syndrome (AS) result from the disturbance of imprinted gene expression within human chromosome 15q11-q13. Some cases of PWS and AS are caused by microdeletions near the SNRPN gene that disrupt a regulatory element termed the imprinting center (IC). The IC has two functional components; an element at the promoter of SNRPN involved in PWS (PWS-IC) and an element 35 kilobases (kb) upstream of SNRPN involved in AS (AS-IC). To further understand the function of the IC, we sought to create a mouse model for AS-IC mutations. We have generated two deletions at a location analogous to that of the human AS-IC. Neither deletion produced an imprinting defect as indicated by DNA methylation and gene expression analyses. These results indicate that no elements critical for AS-IC function in mouse reside within the 12.8-kb deleted region and suggest that the specific location of the AS-IC is not conserved between human and mouse.


Assuntos
Autoantígenos/genética , Impressão Genômica/genética , Ribonucleoproteínas Nucleares Pequenas/genética , Deleção de Sequência/genética , Síndrome de Angelman/genética , Animais , Sequência de Bases , Metilação de DNA , Padrões de Herança/genética , Camundongos , Camundongos Endogâmicos C57BL , Proteínas Repressoras/genética , Ribonucleoproteínas/genética , Ubiquitina-Proteína Ligases/genética , Proteínas Centrais de snRNP
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA