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

Medicinas Complementares
País/Região como assunto
Tipo de documento
Intervalo de ano de publicação
1.
PLoS Genet ; 19(10): e1010961, 2023 10.
Artigo em Inglês | MEDLINE | ID: mdl-37856383

RESUMO

Imprinted genes are subject to germline epigenetic modification resulting in parental-specific allelic silencing. Although genomic imprinting is thought to be important for maternal behaviour, this idea is based on serendipitous findings from a small number of imprinted genes. Here, we undertook an unbiased systems biology approach, taking advantage of the recent delineation of specific neuronal populations responsible for controlling parental care, to test whether imprinted genes significantly converge to regulate parenting behaviour. Using single-cell RNA sequencing datasets, we identified a specific enrichment of imprinted gene expression in a recognised "parenting hub", the galanin-expressing neurons of the preoptic area. We tested the validity of linking enriched expression in these neurons to function by focusing on MAGE family member L2 (Magel2), an imprinted gene not previously linked to parenting behaviour. We confirmed expression of Magel2 in the preoptic area galanin expressing neurons. We then examined the parenting behaviour of Magel2-null(+/p) mice. Magel2-null mothers, fathers and virgin females demonstrated deficits in pup retrieval, nest building and pup-directed motivation, identifying a central role for this gene in parenting. Finally, we show that Magel2-null mothers and fathers have a significant reduction in POA galanin expressing cells, which in turn contributes to a reduced c-Fos response in the POA upon exposure to pups. Our findings identify a novel imprinted gene that impacts parenting behaviour and, moreover, demonstrates the utility of using single-cell RNA sequencing data to predict gene function from expression and in doing so here, have identified a purposeful role for genomic imprinting in mediating parental behaviour.


Assuntos
Galanina , Poder Familiar , Feminino , Animais , Camundongos , Galanina/genética , Galanina/metabolismo , Hipotálamo/metabolismo , Impressão Genômica/genética , Fenótipo , Antígenos de Neoplasias/genética , Proteínas/genética
2.
Elife ; 112022 01 20.
Artigo em Inglês | MEDLINE | ID: mdl-35049495

RESUMO

Genomic imprinting refers to the mono-allelic and parent-specific expression of a subset of genes. While long recognized for their role in embryonic development, imprinted genes have recently emerged as important modulators of postnatal physiology, notably through hypothalamus-driven functions. Here, using mouse models of loss, gain and parental inversion of expression, we report that the paternally expressed Zdbf2 gene controls neonatal growth in mice, in a dose-sensitive but parent-of-origin-independent manner. We further found that Zdbf2-KO neonates failed to fully activate hypothalamic circuits that stimulate appetite, and suffered milk deprivation and diminished circulating Insulin Growth Factor 1 (IGF-1). Consequently, only half of Zdbf2-KO pups survived the first days after birth and those surviving were smaller. This study demonstrates that precise imprinted gene dosage is essential for vital physiological functions at the transition from intra- to extra-uterine life, here the adaptation to oral feeding and optimized body weight gain.


Assuntos
Proteínas de Ligação a DNA/genética , Ingestão de Alimentos/genética , Impressão Genômica/genética , Hipotálamo , Aumento de Peso/genética , Animais , Animais Recém-Nascidos/genética , Animais Recém-Nascidos/fisiologia , Feminino , Hipotálamo/metabolismo , Hipotálamo/fisiologia , Masculino , Camundongos , Camundongos Knockout , Gravidez
3.
FASEB J ; 31(12): 5149-5158, 2017 12.
Artigo em Inglês | MEDLINE | ID: mdl-28778973

RESUMO

Folic acid is an essential component of 1-carbon metabolism, which generates methyl groups for DNA methylation. Disruption of genomic imprinting leads to biallelic expression which may affect disease susceptibility possibly reflected in high levels of S-adenosyl-homocysteine (SAH) and low levels of S-adenosyl-methionine (SAM). We investigated the association between folic acid supplementation during pregnancy and loss of imprinting (LOI) of IGF2 and H19 genes in placentas and cord blood of 90 mother-child dyads in association with the methylenetetrahydrofolate reductase (MTHFR) genotype. Pyrosequencing was used to evaluate deviation from monoallelic expression among 47 placentas heterozygous for H19 and 37 placentas and cord blood tissues heterozygous for IGF2 and H19 methylation levels of 48 placentas. We detected relaxation of imprinting (ROI) and LOI of H19 in placentas not associated with differences in methylation levels of the H19ICR. Placentas retained monoallelic allele-specific gene expression of IGF2, but 32.4% of cord blood samples displayed LOI of IGF2 and 10.8% showed ROI. High SAH levels were significantly associated with low H19 methylation. An interesting positive association between SAM/SAH ratio and high H19 methylation levels was detected among infants with low B12 levels. Our data suggest profound differences in regulation of imprinting in placenta and cord blood; a lack of correlation of the methylome, transcriptome, and proteome; and a complex regulatory feedback network between free methyl groups and genomic imprinting at birth.-Tserga, A., Binder, A. M., Michels, K. B. Impact of folic acid intake during pregnancy on genomic imprinting of IGF2/H19 and 1-carbon metabolism.


Assuntos
Ácido Fólico/metabolismo , Impressão Genômica/genética , Fator de Crescimento Insulin-Like II/genética , Alelos , Metilação de DNA/genética , Metilação de DNA/fisiologia , Epigenômica , Feminino , Ácido Fólico/administração & dosagem , Genótipo , Humanos , Recém-Nascido , Fator de Crescimento Insulin-Like II/metabolismo , Metilenotetra-Hidrofolato Redutase (NADPH2)/genética , Placenta/metabolismo , Polimorfismo de Nucleotídeo Único/genética , Gravidez , Vitamina B 12/sangue
4.
Epigenetics Chromatin ; 10: 31, 2017.
Artigo em Inglês | MEDLINE | ID: mdl-28649282

RESUMO

BACKGROUND: Differential DNA methylation plays a critical role in the regulation of imprinted genes. The differentially methylated state of the imprinting control region is inherited via the gametes at fertilization, and is stably maintained in somatic cells throughout development, influencing the expression of genes across the imprinting cluster. In contrast, DNA methylation patterns are more labile at secondary differentially methylated regions which are established at imprinted loci during post-implantation development. To investigate the nature of these more variably methylated secondary differentially methylated regions, we adopted a hairpin linker bisulfite mutagenesis approach to examine CpG dyad methylation at differentially methylated regions associated with the murine Dlk1/Gtl2 imprinting cluster on both complementary strands. RESULTS: We observed homomethylation at greater than 90% of the methylated CpG dyads at the IG-DMR, which serves as the imprinting control element. In contrast, homomethylation was only observed at 67-78% of the methylated CpG dyads at the secondary differentially methylated regions; the remaining 22-33% of methylated CpG dyads exhibited hemimethylation. CONCLUSIONS: We propose that this high degree of hemimethylation could explain the variability in DNA methylation patterns at secondary differentially methylated regions associated with imprinted loci. We further suggest that the presence of 5-hydroxymethylation at secondary differentially methylated regions may result in hemimethylation and methylation variability as a result of passive and/or active demethylation mechanisms.


Assuntos
Metilação de DNA/genética , Desenvolvimento Embrionário/genética , Impressão Genômica/genética , Peptídeos e Proteínas de Sinalização Intercelular/genética , Animais , Proteínas de Ligação ao Cálcio , Ilhas de CpG/genética , DNA/genética , Fertilização , Camundongos , RNA Longo não Codificante/genética
5.
Curr Biol ; 25(5): 537-45, 2015 Mar 02.
Artigo em Inglês | MEDLINE | ID: mdl-25660546

RESUMO

BACKGROUND: The paternal allele of Ube3a is silenced by imprinting in neurons, and Angelman syndrome (AS) is a disorder arising from a deletion or mutation of the maternal Ube3a allele, which thereby eliminates Ube3a neuronal expression. Sleep disorders such as short sleep duration and increased sleep onset latency are very common in AS. RESULTS: We found a unique link between neuronal imprinting of Ube3a and circadian rhythms in two mouse models of AS, including enfeebled circadian activity behavior and slowed molecular rhythms in ex vivo brain tissues. As a consequence of compromised circadian behavior, metabolic homeostasis is also disrupted in AS mice. Unsilencing the paternal Ube3a allele restores functional circadian periodicity in neurons deficient in maternal Ube3a but does not affect periodicity in peripheral tissues that are not imprinted for uniparental Ube3a expression. The ubiquitin ligase encoded by Ube3a interacts with the central clock components BMAL1 and BMAL2. Moreover, inactivation of Ube3a expression elevates BMAL1 levels in brain regions that control circadian behavior of AS-model mice, indicating an important role for Ube3a in modulating BMAL1 turnover. CONCLUSIONS: Ube3a expression constitutes a direct mechanistic connection between symptoms of a human neurological disorder and the central circadian clock mechanism. The lengthened circadian period leads to delayed phase, which could explain the short sleep duration and increased sleep onset latency of AS subjects. Moreover, we report the pharmacological rescue of an AS phenotype, in this case, altered circadian period. These findings reveal potential treatments for sleep disorders in AS patients.


Assuntos
Síndrome de Angelman/fisiopatologia , Ritmo Circadiano/fisiologia , Impressão Genômica/genética , Neurônios/metabolismo , Ubiquitina-Proteína Ligases/genética , Fatores de Transcrição ARNTL/metabolismo , Análise de Variância , Síndrome de Angelman/genética , Animais , Peso Corporal/fisiologia , Cronoterapia/métodos , Ritmo Circadiano/efeitos dos fármacos , Ritmo Circadiano/genética , Deleção de Genes , Humanos , Fígado/metabolismo , Camundongos , Transtornos do Sono-Vigília/terapia
6.
Nat Commun ; 4: 2311, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-23921428

RESUMO

Imprinted, maternally silenced insulin-like growth factor-2 is expressed in both the foetus and placenta and has been shown to have roles in foetal and placental development in animal models. Here we compared mice engineered to be null for the placenta-specific P0 transcript (insulin-like growth factor-2-P0 KO) to mice with disruptions of all four insulin-like growth factor-2 transcripts, and therefore null for insulin-like growth factor-2 in both placenta and foetus (insulin-like growth factor-2-total KO). Both models lead to intrauterine growth restriction but dissociate between a situation where there is an imbalance between foetal demand and placental supply of nutrients (the insulin-like growth factor-2-P0 KO) and one where demand and supply is more balanced (the insulin-like growth factor-2-total KO). Increased reactivity to anxiety-provoking stimuli is manifested later in life only in those animals where there is a mismatch between placental supply and foetal demand for nutrients during gestation. Our findings further distinguish placental dysfunction from intrauterine growth restriction and reveal a role for the placenta in long-term programming of emotional behaviour.


Assuntos
Envelhecimento/patologia , Ansiedade/patologia , Fator de Crescimento Insulin-Like II/deficiência , Placenta/metabolismo , Estimulação Acústica , Animais , Animais Recém-Nascidos , Ansiedade/genética , Comportamento Animal , Peso ao Nascer , Peso Corporal , Modelos Animais de Doenças , Comportamento Exploratório , Feminino , Regulação da Expressão Gênica no Desenvolvimento , Impressão Genômica/genética , Hipocampo/metabolismo , Hipocampo/patologia , Fator de Crescimento Insulin-Like II/genética , Fator de Crescimento Insulin-Like II/metabolismo , Aprendizagem em Labirinto , Camundongos , Camundongos Knockout , Gravidez
7.
Neurology ; 80(4): 385-91, 2013 Jan 22.
Artigo em Inglês | MEDLINE | ID: mdl-23284065

RESUMO

OBJECTIVE: To identify brain regions with metabolic changes in DYT11 myoclonus-dystonia (DYT11-MD) relative to control subjects and to compare metabolic abnormalities in DYT11-MD with those found in other forms of hereditary dystonia and in posthypoxic myoclonus. METHODS: [(18)F]-fluorodeoxyglucose PET was performed in 6 subjects with DYT11-MD (age 30.5 ± 10.1 years) and in 6 nonmanifesting DYT11 mutation carriers (NM-DYT11; age 59.1 ± 8.9 years) representing the parental generation of the affected individuals. These data were compared to scan data from age-matched healthy control subjects using voxel-based whole brain searches and group differences were considered significant at p < 0.05 (corrected, statistical parametric mapping). As a secondary analysis, overlapping abnormalities were identified by comparisons to hereditary dystonias (DYT1, DYT6, dopa-responsive dystonia) and to posthypoxic myoclonus. RESULTS: We found significant DYT11 genotype-specific metabolic increases in the inferior pons and in the posterior thalamus as well as reductions in the ventromedial prefrontal cortex. Significant phenotype-related increases were present in the parasagittal cerebellum. This latter abnormality was shared with posthypoxic myoclonus, but not with other forms of dystonia. By contrast, all dystonia cohorts exhibited significant metabolic increases in the superior parietal lobule. CONCLUSIONS: The findings are consistent with a subcortical myoclonus generator in DYT11-MD, likely involving the cerebellum. By contrast, subtle increases in the superior parietal cortex relate to the additional presence of dystonic symptoms. Although reduced penetrance in DYT11-MD has been attributed to the maternal imprinting epsilon-sarcoglycan mutations, NM-DYT11 carriers showed significant metabolic abnormalities that are not explained by this genetic model.


Assuntos
Encéfalo/diagnóstico por imagem , Encéfalo/metabolismo , Distúrbios Distônicos , Sarcoglicanas/genética , Adulto , Idoso , Cerebelo/diagnóstico por imagem , Cerebelo/metabolismo , Distúrbios Distônicos/diagnóstico por imagem , Distúrbios Distônicos/genética , Distúrbios Distônicos/metabolismo , Metabolismo Energético/genética , Saúde da Família , Feminino , Fluordesoxiglucose F18 , Impressão Genômica/genética , Genótipo , Humanos , Masculino , Pessoa de Meia-Idade , Modelos Genéticos , Lobo Parietal/diagnóstico por imagem , Lobo Parietal/metabolismo , Penetrância , Fenótipo , Tomografia por Emissão de Pósitrons , Córtex Pré-Frontal/diagnóstico por imagem , Córtex Pré-Frontal/metabolismo , Tálamo/diagnóstico por imagem , Tálamo/metabolismo , Adulto Jovem
8.
Folia Histochem Cytobiol ; 50(2): 171-9, 2012 Jul 05.
Artigo em Inglês | MEDLINE | ID: mdl-22763974

RESUMO

The insulin-like growth factor-2 (Igf2)-H19 locus encodes important paternally imprinted genes that govern normal embryonic development. While Igf-2 encodes IGF2, which is an autocrine/paracrine mitogen,  transcription of H19 gives rise to non-coding mRNA that is a precursor of several microRNAs (miRNAs) that negatively affect cell proliferation. The proper imprinting of a differentially methylated region (DMR) within this locus, with methylation of the paternal chromosome and a lack of methylation on the maternal chromosome, regulates expression of both of these genes so that Igf2 is transcribed only from the paternal chromosome and H19 only from the maternal chromosome. There is growing evidence that this 'Yin-Yang' locus regulates embryonic development. Furthermore, recent evidence indicates that erasure of imprinting (hypomethylation) of the Igf2-H19 locus on both chromosomes, which leads to downregulation of Igf2 and upregulation of H19 expression, plays an important role in regulating quiescence of pluripotent stem cells in adult organisms, and may be involved in the regulation of lifespan. In contrast, hypermethylation of this locus on both chromosomes (loss of imprinting) results in Igf2 overexpression and is observed in several malignancies. In this review, we will discuss the biological consequences of changes in Igf2-H19 expression.


Assuntos
Transformação Celular Neoplásica/genética , Desenvolvimento Embrionário/genética , Impressão Genômica/genética , Fator de Crescimento Insulin-Like II/genética , Longevidade/genética , Células-Tronco Pluripotentes/citologia , RNA Longo não Codificante/genética , Células-Tronco Adultas/citologia , Células-Tronco Adultas/metabolismo , Animais , Humanos , Células-Tronco Pluripotentes/metabolismo
9.
PLoS One ; 7(1): e29753, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-22253771

RESUMO

The alternatively spliced trimeric G-protein subunit XLαs, which is involved in cAMP signalling, is encoded by the Gnasxl transcript of the imprinted Gnas locus. XLαs deficient mice show neonatal feeding problems, leanness, inertia and a high mortality rate. Mutants that survive to weaning age develop into healthy and fertile adults, which remain lean despite elevated food intake. The adult metabolic phenotype can be attributed to increased energy expenditure, which appears to be caused by elevated sympathetic nervous system activity. To better understand the changing phenotype of Gnasxl deficient mice, we compared XLαs expression in neonatal versus adult tissues, analysed its co-localisation with neural markers and characterised changes in the nutrient-sensing mTOR1-S6K pathway in the hypothalamus. Using a newly generated conditional Gnasxl lacZ gene trap line and immunohistochemistry we identified various types of muscle, including smooth muscle cells of blood vessels, as the major peripheral sites of expression in neonates. Expression in all muscle tissues was silenced in adults. While Gnasxl expression in the central nervous system was also developmentally silenced in some midbrain nuclei, it was upregulated in the preoptic area, the medial amygdala, several hypothalamic nuclei (e.g. arcuate, dorsomedial, lateral and paraventricular nuclei) and the nucleus of the solitary tract. Furthermore, expression was detected in the ventral medulla as well as in motoneurons and a subset of sympathetic preganglionic neurons of the spinal cord. In the arcuate nucleus of Gnasxl-deficient mice we found reduced activity of the nutrient sensing mTOR1-S6K signalling pathway, which concurs with their metabolic status. The expression in these brain regions and the hypermetabolic phenotype of adult Gnasxl-deficient mice imply an inhibitory function of XLαs in energy expenditure and sympathetic outflow. By contrast, the neonatal phenotype of mutant mice appears to be due to a transient role of XLαs in muscle tissues.


Assuntos
Encéfalo/crescimento & desenvolvimento , Encéfalo/metabolismo , Subunidades alfa Gs de Proteínas de Ligação ao GTP/deficiência , Subunidades alfa Gs de Proteínas de Ligação ao GTP/genética , Regulação da Expressão Gênica no Desenvolvimento , Impressão Genômica/genética , Transdução de Sinais/genética , Animais , Animais Recém-Nascidos , Sequência de Bases , Biomarcadores/metabolismo , Encéfalo/efeitos dos fármacos , Cromograninas , Subunidades alfa Gs de Proteínas de Ligação ao GTP/metabolismo , Regulação da Expressão Gênica no Desenvolvimento/efeitos dos fármacos , Inativação Gênica/efeitos dos fármacos , Marcação de Genes , Loci Gênicos/genética , Impressão Genômica/efeitos dos fármacos , Hipotálamo/efeitos dos fármacos , Hipotálamo/metabolismo , Insulina/farmacologia , Leptina/farmacologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Dados de Sequência Molecular , Músculos/efeitos dos fármacos , Músculos/metabolismo , Miócitos de Músculo Liso/efeitos dos fármacos , Miócitos de Músculo Liso/metabolismo , Neuropeptídeos/metabolismo , Fenótipo , Transdução de Sinais/efeitos dos fármacos , Medula Espinal/efeitos dos fármacos , Medula Espinal/metabolismo
10.
Nature ; 481(7380): 185-9, 2011 Dec 21.
Artigo em Inglês | MEDLINE | ID: mdl-22190039

RESUMO

Angelman syndrome is a severe neurodevelopmental disorder caused by deletion or mutation of the maternal allele of the ubiquitin protein ligase E3A (UBE3A). In neurons, the paternal allele of UBE3A is intact but epigenetically silenced, raising the possibility that Angelman syndrome could be treated by activating this silenced allele to restore functional UBE3A protein. Using an unbiased, high-content screen in primary cortical neurons from mice, we identify twelve topoisomerase I inhibitors and four topoisomerase II inhibitors that unsilence the paternal Ube3a allele. These drugs included topotecan, irinotecan, etoposide and dexrazoxane (ICRF-187). At nanomolar concentrations, topotecan upregulated catalytically active UBE3A in neurons from maternal Ube3a-null mice. Topotecan concomitantly downregulated expression of the Ube3a antisense transcript that overlaps the paternal copy of Ube3a. These results indicate that topotecan unsilences Ube3a in cis by reducing transcription of an imprinted antisense RNA. When administered in vivo, topotecan unsilenced the paternal Ube3a allele in several regions of the nervous system, including neurons in the hippocampus, neocortex, striatum, cerebellum and spinal cord. Paternal expression of Ube3a remained elevated in a subset of spinal cord neurons for at least 12 weeks after cessation of topotecan treatment, indicating that transient topoisomerase inhibition can have enduring effects on gene expression. Although potential off-target effects remain to be investigated, our findings suggest a therapeutic strategy for reactivating the functional but dormant allele of Ube3a in patients with Angelman syndrome.


Assuntos
Alelos , Inativação Gênica/efeitos dos fármacos , Neurônios/efeitos dos fármacos , Neurônios/metabolismo , Inibidores da Topoisomerase/farmacologia , Ubiquitina-Proteína Ligases/genética , Síndrome de Angelman/tratamento farmacológico , Síndrome de Angelman/genética , Animais , Células Cultivadas , Córtex Cerebral/citologia , Córtex Cerebral/efeitos dos fármacos , Córtex Cerebral/metabolismo , Avaliação Pré-Clínica de Medicamentos , Pai , Feminino , Impressão Genômica/efeitos dos fármacos , Impressão Genômica/genética , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Mães , Bibliotecas de Moléculas Pequenas/administração & dosagem , Bibliotecas de Moléculas Pequenas/química , Bibliotecas de Moléculas Pequenas/farmacologia , Inibidores da Topoisomerase/administração & dosagem , Inibidores da Topoisomerase/análise , Inibidores da Topoisomerase/farmacocinética , Topotecan/administração & dosagem , Topotecan/farmacocinética , Topotecan/farmacologia , Ubiquitina-Proteína Ligases/deficiência
11.
J Mol Endocrinol ; 47(2): R67-74, 2011 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-21798993

RESUMO

Genomic imprinting is an important and enigmatic form of gene regulation in mammals in which one copy of a gene is silenced in a manner determined by its parental history. Imprinted genes range from those with constitutive monoallelic silencing to those, typically more remote from imprinting control regions, that display developmentally regulated, tissue-specific or partial monoallelic expression. This diversity may make these genes, and the processes they control, more or less sensitive to factors that modify or disrupt epigenetic marks. Imprinted genes have important functions in development and physiology, including major endocrine/neuroendocrine axes. Owing to is central role in coordinating growth, metabolism and reproduction, as well as evidence from genetic and knockout studies, the hypothalamus may be a focus for imprinted gene action. Are there unifying principles that explain why a gene should be imprinted? Conflict between parental genomes over limiting maternal resources, but also co-adaptation between mothers and offspring, have been invoked to explain the evolution of imprinting. Recent reports suggest there may be many more genes imprinted in the hypothalamus than hitherto expected, and it will be important for these new candidates to be validated and to determine whether they conform to current notions of how imprinting is regulated. In fully evaluating the role of imprinted genes in the hypothalamus, much work needs to be done to identify the specific neuronal populations in which particular genes are expressed, establish whether there are pathways in common and whether imprinted genes are involved in long-term programming of hypothalamic functions.


Assuntos
Impressão Genômica/genética , Hipotálamo/metabolismo , Animais , Evolução Molecular , Humanos
12.
BMC Genomics ; 11: 649, 2010 Nov 22.
Artigo em Inglês | MEDLINE | ID: mdl-21092170

RESUMO

BACKGROUND: Genomic imprinting is an evolutionary conserved mechanism of epigenetic gene regulation in placental mammals that results in silencing of one of the parental alleles. In order to decipher interactions between allele-specific DNA methylation of imprinted genes and evolutionary conservation, we performed a genome-wide comparative investigation of genomic sequences and highly conserved elements of imprinted genes in human and mouse. RESULTS: Evolutionarily conserved elements in imprinted regions differ from those associated with autosomal genes in various ways. Whereas for maternally expressed genes strong divergence of protein-encoding sequences is most prominent, paternally expressed genes exhibit substantial conservation of coding and noncoding sequences. Conserved elements in imprinted regions are marked by enrichment of CpG dinucleotides and low (TpG+CpA)/(2·CpG) ratios indicate reduced CpG deamination. Interestingly, paternally and maternally expressed genes can be distinguished by differences in G+C and CpG contents that might be associated with unusual epigenetic features. Especially noncoding conserved elements of paternally expressed genes are exceptionally G+C and CpG rich. In addition, we confirmed a frequent occurrence of intronic CpG islands and observed a decelerated degeneration of ancient LINE-1 repeats. We also found a moderate enrichment of YY1 and CTCF binding sites in imprinted regions and identified several short sequence motifs in highly conserved elements that might act as additional regulatory elements. CONCLUSIONS: We discovered several novel conserved DNA features that might be related to allele-specific DNA methylation. Our results hint at reduced CpG deamination rates in imprinted regions, which affects mostly noncoding conserved elements of paternally expressed genes. Pronounced differences between maternally and paternally expressed genes imply specific modes of evolution as a result of differences in epigenetic features and a special response to selective pressure. In addition, our data support the potential role of intronic CpG islands as epigenetic key regulatory elements and suggest that evolutionary conserved LINE-1 elements fulfill regulatory functions in imprinted regions.


Assuntos
Sequência Conservada/genética , Impressão Genômica/genética , Animais , Sequência de Bases , Sítios de Ligação , Fator de Ligação a CCCTC , Ilhas de CpG/genética , DNA Intergênico/genética , Éxons/genética , Variação Genética , Humanos , Camundongos , Anotação de Sequência Molecular , Fases de Leitura Aberta/genética , Sequências Reguladoras de Ácido Nucleico/genética , Sequências Repetitivas de Ácido Nucleico/genética , Proteínas Repressoras/metabolismo , Fator de Transcrição YY1
13.
PLoS Genet ; 5(9): e1000663, 2009 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-19779546

RESUMO

Balanced maternal and paternal genome contributions are a requirement for successful seed development. Unbalanced contributions often cause seed abortion, a phenomenon that has been termed "triploid block." Misregulation of imprinted regulatory genes has been proposed to be the underlying cause for abnormalities in growth and structure of the endosperm in seeds with deviating parental contributions. We identified a mutant forming unreduced pollen that enabled us to investigate direct effects of unbalanced parental genome contributions on seed development and to reveal the underlying molecular mechanism of dosage sensitivity. We provide evidence that parent-of-origin-specific expression of the Polycomb group (PcG) gene MEDEA is causally responsible for seed developmental aberrations in Arabidopsis seeds with increased paternal genome contributions. We propose that imprinted expression of PcG genes is an evolutionary conserved mechanism to balance parental genome contributions in embryo nourishing tissues.


Assuntos
Proteínas de Arabidopsis/genética , Arabidopsis/genética , Genes de Plantas , Impressão Genômica/genética , Ploidias , Proteínas Repressoras/genética , Alelos , Sequência de Aminoácidos , Arabidopsis/citologia , Proteínas de Arabidopsis/química , Proteínas de Arabidopsis/metabolismo , Cruzamentos Genéticos , Diploide , Perfilação da Expressão Gênica , Regulação da Expressão Gênica de Plantas , Teste de Complementação Genética , Homozigoto , Proteínas de Domínio MADS/genética , Proteínas de Domínio MADS/metabolismo , Dados de Sequência Molecular , Mutação/genética , Fenótipo , Pólen/citologia , Pólen/genética , Proteínas do Grupo Polycomb , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Proteínas Repressoras/metabolismo , Sementes/genética , Sementes/crescimento & desenvolvimento
14.
PLoS One ; 4(2): e4352, 2009.
Artigo em Inglês | MEDLINE | ID: mdl-19194500

RESUMO

BACKGROUND: Genomic imprinting is an exception to Mendelian genetics in that imprinted genes are expressed monoallelically, dependent on parental origin. In mammals, imprinted genes are critical in numerous developmental and physiological processes. Aberrant imprinted gene expression is implicated in several diseases including Prader-Willi/Angelman syndromes and cancer. METHODOLOGY/PRINCIPAL FINDINGS: To identify novel imprinted genes, transcription profiling was performed on two uniparentally derived cell lines, androgenetic and parthenogenetic primary mouse embryonic fibroblasts. A maternally expressed transcript termed Imprinted RNA near Meg3/Gtl2 (Irm) was identified and its expression studied by Northern blotting and whole mounts in situ hybridization. The imprinted region that contains Irm has a parent of origin effect in three mammalian species, including the sheep callipyge locus. In mice and humans, both maternal and paternal uniparental disomies (UPD) cause embryonic growth and musculoskeletal abnormalities, indicating that both alleles likely express essential genes. To catalog all imprinted genes in this chromosomal region, twenty-five mouse mRNAs in a 1.96Mb span were investigated for allele specific expression. CONCLUSIONS/SIGNIFICANCE: Ten imprinted genes were elucidated. The imprinting of three paternally expressed protein coding genes (Dlk1, Peg11, and Dio3) was confirmed. Seven noncoding RNAs (Meg3/Gtl2, Anti-Peg11, Meg8, Irm/"Rian", AK050713, AK053394, and Meg9/Mirg) are characterized by exclusive maternal expression. Intriguingly, the majority of these noncoding RNA genes contain microRNAs and/or snoRNAs within their introns, as do their human orthologs. Of the 52 identified microRNAs that map to this region, six are predicted to regulate negatively Dlk1, suggesting an additional mechanism for interactions between allelic gene products. Since several previous studies relied heavily on in silico analysis and RT-PCR, our findings from Northerns and cDNA cloning clarify the genomic organization of this region. Our results expand the number of maternally expressed noncoding RNAs whose loss may be responsible for the phenotypes associated with mouse pUPD12 and human pUPD14 syndromes.


Assuntos
Cromossomos de Mamíferos/genética , Impressão Genômica/genética , Peptídeos e Proteínas de Sinalização Intercelular/genética , Iodeto Peroxidase/genética , Família Multigênica , Processamento Alternativo/genética , Animais , Anticorpos , Sequência de Bases , Proteínas de Ligação ao Cálcio , Centrômero/genética , Clonagem Molecular , DNA Complementar/genética , Embrião de Mamíferos/citologia , Embrião de Mamíferos/metabolismo , Feminino , Fibroblastos/metabolismo , Camundongos , MicroRNAs/genética , Dados de Sequência Molecular , Análise de Sequência com Séries de Oligonucleotídeos , Partenogênese/genética , Poliadenilação/genética , Proteínas/genética , RNA Longo não Codificante , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Telômero/genética
15.
Trends Endocrinol Metab ; 15(1): 12-20, 2004.
Artigo em Inglês | MEDLINE | ID: mdl-14693421

RESUMO

Prader-Willi syndrome (PWS) is a complex human genetic disease that arises from lack of expression of paternally inherited imprinted genes on chromosome 15q11-q13. Identification of the imprinting control centre, novel imprinted genes and distinct phenotypes in PWS patients and mouse models has increased interest in this human obesity syndrome. In this review I focus on: (i) the chromosomal region and candidate genes associated with PWS, and the possible links with individual PWS phenotypes identified using mouse models; (ii) the metabolic and hormonal phenotypes in PWS; (iii) postmortem studies of human PWS hypothalami; and (iv) current and potential advances in the management of PWS and its complications. This could have benefits for a wide spectrum of endocrine, paediatric and neuropsychiatric diseases.


Assuntos
Cromossomos Humanos Par 15 , Impressão Genômica/genética , Hipotálamo/fisiopatologia , Síndrome de Prader-Willi , Animais , Hormônios Gastrointestinais/metabolismo , Regulação da Expressão Gênica , Grelina , Humanos , Hiperfagia/complicações , Hiperfagia/genética , Leptina/metabolismo , Obesidade/genética , Hormônios Peptídicos/metabolismo , Fenótipo , Síndrome de Prader-Willi/genética , Síndrome de Prader-Willi/fisiopatologia , Síndrome de Prader-Willi/terapia , Serotonina/metabolismo
16.
Am J Hum Genet ; 69(6): 1389-94, 2001 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-11592036

RESUMO

The Prader-Willi syndrome (PWS)/Angelman syndrome (AS) region, on human chromosome 15q11-q13, exemplifies coordinate control of imprinted gene expression over a large chromosomal domain. Establishment of the paternal state of the region requires the PWS imprinting center (PWS-IC); establishment of the maternal state requires the AS-IC. Cytosine methylation of the PWS-IC, which occurs during oogenesis in mice, occurs only after fertilization in humans, so this modification cannot be the gametic imprint for the PWS/AS region in humans. Here, we demonstrate that the PWS-IC shows parent-specific complementary patterns of H3 lysine 9 (Lys9) and H3 lysine 4 (Lys4) methylation. H3 Lys9 is methylated on the maternal copy of the PWS-IC, and H3 Lys4 is methylated on the paternal copy. We suggest that H3 Lys9 methylation is a candidate maternal gametic imprint for this region, and we show how changes in chromatin packaging during the life cycle of mammals provide a means of erasing such an imprint in the male germline.


Assuntos
Cromossomos Humanos Par 15/genética , Metilação de DNA , Impressão Genômica/genética , Histonas/química , Histonas/genética , Lisina/metabolismo , Síndrome de Prader-Willi/genética , Alelos , Éxons/genética , Feminino , Células Germinativas/metabolismo , Humanos , Lisina/genética , Masculino , Modelos Genéticos , Síndrome de Prader-Willi/metabolismo , Regiões Promotoras Genéticas/genética
17.
Clin Genet ; 60(4): 255-63, 2001 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-11683768

RESUMO

The hypothalamus integrates physiological processes essential for survival and reproduction. Recent studies have shown that developmental events can affect these processes. Pathways required for the induction of the ventral midline of the hypothalamus or for the differentiation of specific hypothalamic lineages have the potential of causing endocrine and metabolic disorders, including obesity. Also, some genes with paternal monoallelic expression are involved in the development of hypothalamic centers that are critical physiological regulators. Developmental defects affecting the hypothalamus might represent a more frequent cause of clinical disorders than previously suspected.


Assuntos
Hipotálamo/embriologia , Hipotálamo/fisiopatologia , Núcleo Hipotalâmico Paraventricular/embriologia , Animais , Diferenciação Celular , Metabolismo Energético , Impressão Genômica/genética , Humanos , Hipotálamo/crescimento & desenvolvimento , Hipotálamo/metabolismo , Neurônios/citologia , Neurônios/metabolismo , Núcleo Hipotalâmico Paraventricular/citologia , Núcleo Hipotalâmico Paraventricular/crescimento & desenvolvimento , Núcleo Hipotalâmico Paraventricular/metabolismo , Fatores de Transcrição/metabolismo
18.
J Allergy Clin Immunol ; 106(5): 832-9, 2000 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-11080703

RESUMO

BACKGROUND: Against the background of the controversial discussion about an increase in allergic rhinitis in recent years, intraindividual longitudinal data is lacking for IgE-mediated seasonal allergic rhinitis (SAR). Little is known about the development of SAR in terms of prevalence and incidence rates from birth to school age. OBJECTIVE: In a prospective birth cohort, we investigated the development of sensitization and symptoms of SAR. SAR should be defined with high specificity, and associated risk factors should be determined. METHODS: Annual longitudinal data about seasonal allergic symptoms and sensitization was available for 587 children from birth to their seventh birthday. The definition of SAR was based on a combination of exposure-related symptoms and sensitization. RESULTS: Up to 7 years of age, SAR developed in 15% of the children. Incidence and prevalence of symptoms and sensitization were low during early childhood (<2%) and increased steadily with age. Children in which SAR had already developed in the second year all were born in spring or early summer, resulting in at least two seasons of pollen exposure before manifestation of SAR. Risk factors assessed by multiple logistic regression analysis were male sex (odds ratio [OR] = 2.4), atopic mothers (OR = 2.6) and fathers (OR = 3.6) having allergic rhinitis themselves, first-born child (OR = 2.0), early sensitization to food (OR = 3.3), and atopic dermatitis (OR = 2.5), whereas early wheezing was not associated with SAR. CONCLUSION: The development of SAR is characterized by a marked increase in prevalence and incidence after the second year of life. Our longitudinal data further indicate that in combination with the risk of allergic predisposition, at least 2 seasons of pollen allergen exposure are needed before allergic rhinitis becomes clinically manifest.


Assuntos
Rinite Alérgica Sazonal/fisiopatologia , Fatores Etários , Alérgenos/imunologia , Criança , Pré-Escolar , Estudos de Coortes , Feminino , Seguimentos , Impressão Genômica/genética , Impressão Genômica/imunologia , Alemanha/epidemiologia , Humanos , Imunoglobulina E/sangue , Incidência , Lactente , Estudos Longitudinais , Masculino , Pólen/imunologia , Prevalência , Estudos Prospectivos , Sons Respiratórios/imunologia , Sons Respiratórios/fisiopatologia , Rinite Alérgica Sazonal/epidemiologia , Rinite Alérgica Sazonal/genética , Rinite Alérgica Sazonal/imunologia , Fatores de Risco
19.
Mol Cell Biol ; 18(11): 6767-76, 1998 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-9774690

RESUMO

Genomic imprinting results in parent-specific monoallelic expression of a small number of genes in mammals. The identity of imprints is unknown, but much evidence points to a role for DNA methylation. The maternal alleles of the imprinted H19 gene are active and hypomethylated; the paternal alleles are inactive and hypermethylated. Roles for other epigenetic modifications are suggested by allele-specific differences in nuclease hypersensitivity at particular sites. To further analyze the possible epigenetic mechanisms determining monoallelic expression of H19, we have conducted in vivo dimethylsulfate and DNase I footprinting of regions upstream of the coding sequence in parthenogenetic and androgenetic embryonic stem cells. These cells carry only maternally and paternally derived alleles, respectively. We observed the presence of maternal-allele-specific dimethylsulfate and DNase I footprints at the promoter indicative of protein-DNA interactions at a CCAAT box and at binding sites for transcription factors Sp1 and AP-2. Also, at the boundary of a region further upstream for which existent differential methylation has been suggested to constitute an imprint, we observed a number of strand-specific dimethylsulfate reactivity differences specific to the maternal allele, along with an unusual chromatin structure in that both strands of maternally derived DNA were strongly hypersensitive to DNase I cutting over a distance of 100 nucleotides. We therefore reveal the existence of novel parent-specific epigenetic modifications, which in addition to DNA methylation, could constitute imprints or maintain monoallelic expression of H19.


Assuntos
Metilação de DNA , Impressão Genômica/genética , Proteínas Musculares/genética , RNA não Traduzido , Alelos , Animais , Sequência de Bases , Cromatina/química , Pegada de DNA , Proteínas de Ligação a DNA/genética , Desoxirribonuclease I/metabolismo , Regulação da Expressão Gênica/genética , Camundongos , Dados de Sequência Molecular , Podofilina/análogos & derivados , Podofilina/genética , Podofilotoxina/análogos & derivados , Regiões Promotoras Genéticas/genética , RNA Longo não Codificante , Sequências Repetitivas de Ácido Nucleico/genética , Células-Tronco , Ésteres do Ácido Sulfúrico/metabolismo , Fator de Transcrição AP-2 , Fatores de Transcrição/genética
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA