Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 25
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Clin Epigenetics ; 13(1): 134, 2021 06 30.
Artigo em Inglês | MEDLINE | ID: mdl-34193245

RESUMO

BACKGROUND: X-chromosome inactivation (XCI) is a mechanism in which one of two X chromosomes in females is randomly inactivated in order to compensate for imbalance of gene dosage between sexes. However, about 15% of genes on the inactivated X chromosome (Xi) escape from XCI. The methylation level of the promoter region of the escape gene is lower than that of the inactivated genes. Dxz4 and/or Firre have critical roles for forming the three-dimensional (3D) structure of Xi. In mice, disrupting the 3D structure of Xi by deleting both Dxz4 and Firre genes led to changing of the escape genes list. To estimate the impact for escape genes by X-chromosome rearrangements, including DXZ4 and FIRRE, we examined the methylation status of escape gene promoters in patients with various X-chromosome rearrangements. RESULTS: To detect the breakpoints, we first performed array-based comparative genomic hybridization and whole-genome sequencing in four patients with X-chromosome rearrangements. Subsequently, we conducted array-based methylation analysis and reduced representation bisulfite sequencing in the four patients with X-chromosome rearrangements and controls. Of genes reported as escape genes by gene expression analysis using human hybrid cells in a previous study, 32 genes showed hypomethylation of the promoter region in both male controls and female controls. Three patients with X-chromosome rearrangements had no escape genes with abnormal methylation of the promoter region. One of four patients with the most complicated rearrangements exhibited abnormal methylation in three escape genes. Furthermore, in the patient with the deletion of the FIRRE gene and the duplication of DXZ4, most escape genes remained hypomethylated. CONCLUSION: X-chromosome rearrangements are unlikely to affect the methylation status of the promoter regions of escape genes, except for a specific case with highly complex rearrangements, including the deletion of the FIRRE gene and the duplication of DXZ4.


Assuntos
Metilação de DNA/genética , Inativação do Cromossomo X/genética , Adulto , Feminino , Genes Ligados ao Cromossomo X/genética , Genes Ligados ao Cromossomo X/fisiologia , Humanos , Masculino
2.
Cell Rep ; 29(8): 2422-2437.e8, 2019 11 19.
Artigo em Inglês | MEDLINE | ID: mdl-31747610

RESUMO

Palmitoylation is a reversible post-translational lipid modification that facilitates vesicular transport and subcellular localization of modified proteins. This process is catalyzed by ZDHHC enzymes that are implicated in several neurological and neurodevelopmental disorders. Loss-of-function mutations in ZDHHC9 have been identified in patients with X-linked intellectual disability (XLID) and associated with increased epilepsy risk. Loss of Zdhhc9 function in hippocampal cultures leads to shorter dendritic arbors and fewer inhibitory synapses, altering the ratio of excitatory-to-inhibitory inputs formed onto Zdhhc9-deficient cells. While Zdhhc9 promotes dendrite outgrowth through the palmitoylation of the GTPase Ras, it promotes inhibitory synapse formation through the palmitoylation of another GTPase, TC10. Zdhhc9 knockout mice exhibit seizure-like activity together with increased frequency and amplitude of both spontaneous and miniature excitatory and inhibitory postsynaptic currents. These findings present a plausible mechanism for how the loss of ZDHHC9 function may contribute to XLID and epilepsy.


Assuntos
Aciltransferases/metabolismo , Dendritos/metabolismo , Genes Ligados ao Cromossomo X/fisiologia , Deficiência Intelectual/metabolismo , Sinapses/metabolismo , Aciltransferases/genética , Animais , Células Cultivadas , Epilepsia/genética , Epilepsia/metabolismo , Genes Ligados ao Cromossomo X/genética , Hipocampo/metabolismo , Humanos , Deficiência Intelectual/genética , Lipoilação/genética , Lipoilação/fisiologia , Camundongos , Camundongos Knockout , Sinapses/genética , Proteínas ras/metabolismo , Proteínas rho de Ligação ao GTP/genética , Proteínas rho de Ligação ao GTP/metabolismo
3.
Science ; 363(6432): 1210-1213, 2019 03 15.
Artigo em Inglês | MEDLINE | ID: mdl-30872523

RESUMO

We report the reproductive strategy of the nematode Mesorhabditis belari This species produces only 9% males, whose sperm is necessary to fertilize and activate the eggs. However, most of the fertilized eggs develop without using the sperm DNA and produce female individuals. Only in 9% of eggs is the male DNA utilized, producing sons. We found that mixing of parental genomes only gives rise to males because the Y-bearing sperm of males are much more competent than the X-bearing sperm for penetrating the eggs. In this previously unrecognized strategy, asexual females produce few sexual males whose genes never reenter the female pool. Here, production of males is of interest only if sons are more likely to mate with their sisters. Using game theory, we show that in this context, the production of 9% males by M. belari females is an evolutionary stable strategy.


Assuntos
Óvulo/fisiologia , Partenogênese , Rhabditoidea/fisiologia , Razão de Masculinidade , Animais , Evolução Biológica , Feminino , Teoria dos Jogos , Genes Ligados ao Cromossomo X/fisiologia , Genes Ligados ao Cromossomo Y/fisiologia , Masculino , Interações Espermatozoide-Óvulo/fisiologia , Espermatozoides/fisiologia
4.
Nat Commun ; 9(1): 2555, 2018 07 02.
Artigo em Inglês | MEDLINE | ID: mdl-29967448

RESUMO

Although sex biases in disease presentation are well documented, the mechanisms mediating vulnerability or resilience to diseases are unknown. In utero insults are more likely to produce detrimental health outcomes for males versus females. In our mouse model of prenatal stress, male offspring experience long-term dysregulation of body weight and hypothalamic pituitary adrenal stress axis dysfunction, endophenotypes of male-biased neurodevelopmental disorders. Placental function is critical for healthy fetal development, and we previously showed that sex differences in placental O-linked N-acetylglucosamine transferase (OGT) mediate the effects of prenatal stress on neurodevelopmental programming. Here we show that one mechanism whereby sex differences in OGT confer variation in vulnerability to prenatal insults is by establishing sex-specific trophoblast gene expression patterns and via regulation of the canonically repressive epigenetic modification, H3K27me3. We demonstrate that high levels of H3K27me3 in the female placenta create resilience to the altered hypothalamic programming associated with prenatal stress exposure.


Assuntos
Histonas/metabolismo , N-Acetilglucosaminiltransferases/fisiologia , Placenta/metabolismo , Efeitos Tardios da Exposição Pré-Natal/patologia , Estresse Fisiológico/fisiologia , Animais , Peso Corporal , Modelos Animais de Doenças , Embrião de Mamíferos , Epigênese Genética/fisiologia , Feminino , Desenvolvimento Fetal/fisiologia , Perfilação da Expressão Gênica , Genes Ligados ao Cromossomo X/fisiologia , Código das Histonas/fisiologia , Humanos , Sistema Hipotálamo-Hipofisário/fisiologia , Masculino , Camundongos , Gravidez , Efeitos Tardios da Exposição Pré-Natal/etiologia , Restrição Física , Fatores Sexuais , Trofoblastos/metabolismo
5.
Hum Mol Genet ; 27(R2): R242-R249, 2018 08 01.
Artigo em Inglês | MEDLINE | ID: mdl-29701779

RESUMO

The X chromosome is unique in the genome. In this review we discuss recent advances in our understanding of the genetics and epigenetics of the X chromosome. The X chromosome shares limited conservation with its ancestral homologue the Y chromosome and the resulting difference in X-chromosome dosage between males and females is largely compensated for by X-chromosome inactivation. The process of inactivation is initiated by the long non-coding RNA X-inactive specific transcript (XIST) and achieved through interaction with multiple synergistic silencing pathways. Identification of Xist-interacting proteins has given insight into these processes yet the cascade of events from initiation to maintenance have still to be resolved. In particular, the initiation of inactivation in humans has been challenging to study as: it occurs very early in development; most human embryonic stem cell lines already have an inactive X; and the process seems to differ from mouse. Another difference between human and mouse X inactivation is the larger number of human genes that escape silencing. In humans over 20% of X-linked genes continue to be expressed from the otherwise inactive X chromosome. We are only beginning to understand how such escape occurs but there is growing recognition that escapees contribute to sexually dimorphic traits. The unique biology and epigenetics of the X chromosome have often led to its exclusion from disease studies, yet the X constitutes 5% of the genome and is an important contributor to disease, often in a sex-specific manner.


Assuntos
Cromossomos Humanos X/genética , Cromossomos Humanos X/metabolismo , Inativação do Cromossomo X/fisiologia , Animais , Cromossomos Humanos X/fisiologia , Feminino , Inativação Gênica/fisiologia , Genes Reguladores/genética , Genes Ligados ao Cromossomo X/genética , Genes Ligados ao Cromossomo X/fisiologia , Humanos , Masculino , Camundongos , RNA Longo não Codificante/genética , Cromossomo X/genética , Cromossomo X/metabolismo , Cromossomo X/fisiologia , Inativação do Cromossomo X/genética
6.
Biol Reprod ; 96(2): 469-477, 2017 02 01.
Artigo em Inglês | MEDLINE | ID: mdl-28203736

RESUMO

Cell junctions are necessary for spermatogenesis, and there are numerous types of junctions in testis, such as blood­testis barrier, intercellular bridge, and ectoplasmic specialization (ES). The details of their functions and construction are still unknown. To identify a novel protein essential to the function of a cell junction, we enriched testis membrane protein and analyzed it using a proteomics approach. Here, we report a novel ES protein, which is encoded on the X chromosome and an ortholog of hypothetical human protein KIAA1210. KIAA1210 is expressed in testis predominantly, localized to the sex body in spermatocyte, acrosome, and near ES. Moreover, KIAA1210 possesses a topoisomerase 2 (TOP2)-associated protein PAT1 domain, a herpes simplex virus 1 (HSV-1) large tegument protein UL36 hypothetical domain, and a provisional DNA translocase FtsK domain. Using IP-proteomics with specific antibody to KIAA1210, we identified proteins including TOP2 isoforms as components of a complex with KIAA1210, in cell junctions in testis. The interaction between KIAA1210 and TOP2 was confirmed by two different proteomic analyses. Furthermore, immunofluorescence showed that KIAA1210 and TOP2B co-localize around the sex body in spermatocyte, apical ES, and residual bodies in elongated spermatids. Our findings suggest that KIAA1210 may be essential cell junction protein that interacts with TOP2B to regulate the dynamic change of chromatin structures during spermiogenesis.


Assuntos
Acrossomo/metabolismo , Regulação da Expressão Gênica/fisiologia , Genes Ligados ao Cromossomo X/fisiologia , Proteínas de Membrana/metabolismo , Testículo/fisiologia , Sequência de Aminoácidos , Animais , Perfilação da Expressão Gênica , Masculino , Proteínas de Membrana/genética , Camundongos , Transporte Proteico
7.
PLoS One ; 9(8): e103659, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-25093841

RESUMO

In Drosophila melanogaster males, the expression of X-linked genes is regulated by mechanisms that operate on a chromosomal scale. One such mechanism, male-specific lethal complex-dependent X-linked dosage compensation, is thought to broadly enhance the expression of male X-linked genes through two-fold transcriptional upregulation. The evolutionary consequences of this form of dosage compensation are not well understood, particularly with regard to genes more highly expressed in males. It has been observed the X chromosome arrangement of these male-biased genes is non-random, consistent with what one might expect if there is a selective advantage for male-biased genes to avoid dosage compensation. Separately, it has been noted that the male-specific lethal complex and its dosage compensation mechanism appear absent in some male tissues, thus providing a control for the selection hypothesis. Here we utilized publicly available datasets to reassess the arrangement of X-linked male-biased expressed genes after accounting for expression in tissues not dosage compensated by the male-specific lethal complex. Our results do not corroborate previous observations supporting organismal-wide detrimental effects by dosage compensation on X-linked male-biased expressed genes. We instead find no evidence that dosage compensation has played a role in the arrangement of dosage compensated male-biased genes on the X chromosome.


Assuntos
Mecanismo Genético de Compensação de Dose , Drosophila melanogaster/genética , Complexos Multiproteicos/fisiologia , Cromossomo X/fisiologia , Animais , Proteínas Cromossômicas não Histona/metabolismo , Mapeamento Cromossômico , Cromossomos de Insetos/fisiologia , DNA Helicases/metabolismo , Proteínas de Ligação a DNA/metabolismo , Mecanismo Genético de Compensação de Dose/genética , Proteínas de Drosophila/genética , Proteínas de Drosophila/metabolismo , Genes Ligados ao Cromossomo X/fisiologia , Histona Acetiltransferases/metabolismo , Masculino , Complexos Multiproteicos/metabolismo , Proteínas Nucleares/metabolismo , Diferenciação Sexual/genética , Fatores de Transcrição/metabolismo , Ativação Transcricional
8.
Brain Res ; 1562: 23-38, 2014 May 08.
Artigo em Inglês | MEDLINE | ID: mdl-24661915

RESUMO

The cerebral cortex and hippocampus are important for the control of cognitive functions and social behaviors, many of which are sexually dimorphic and tightly regulated by gonadal steroid hormones via activation of their respective nuclear receptors. As different levels of sex steroid hormones are present between the sexes during early development and their receptors act as transcription factors to regulate gene expression, we hypothesize that sexually dimorphic gene expression in the developing mouse cortex and hippocampus might result in sex differences in brain structures and neural circuits governing distinct behaviors between the sexes as adults. To test our hypothesis, we used gene expression microarrays to identify 90 candidate genes differentially expressed in the neonatal cortex/hippocampus between male and female mice, including 55 male-biased and 35 female-biased genes. Among these genes, sexually dimorphic expression of eight sex chromosome genes was confirmed by reverse transcription with quantitative PCR (RT-qPCR), including three located on the X chromosome (Xist, Eif2s3x, and Kdm6a), three on the Y chromosome (Ddx3y, Eif2s3y, and Kdm5d), and two in the pseudoautosomal region of the X and Y chromosomes (Erdr1 and Mid1). In addition, five autosomal genes (Cd151, Dab2, Klk8, Meg3, and Prkdc) were also validated for their sexually dimorphic expression in the neonatal mouse cortex/hippocampus. Gene Ontology annotation analysis suggests that many of these sexually dimorphic genes are involved in histone modifications, cell proliferation/death, androgen/estrogen signaling pathways, and synaptic organization, and these biological processes have been implicated in differential neural development, cognitive function, and neurological diseases between the sexes.


Assuntos
Córtex Cerebral/metabolismo , Expressão Gênica , Hipocampo/metabolismo , Caracteres Sexuais , Animais , Animais Recém-Nascidos , Córtex Cerebral/crescimento & desenvolvimento , Análise por Conglomerados , Feminino , Perfilação da Expressão Gênica , Genes Ligados ao Cromossomo X/fisiologia , Genes Ligados ao Cromossomo Y/fisiologia , Hipocampo/crescimento & desenvolvimento , Masculino , Camundongos Endogâmicos C57BL , Análise em Microsséries , Reação em Cadeia da Polimerase em Tempo Real
9.
Neurosci Lett ; 559: 163-8, 2014 Jan 24.
Artigo em Inglês | MEDLINE | ID: mdl-24287374

RESUMO

A gene-environment (GxE) interaction is implicated in both the pathophysiology and treatment of major depressive disorder (MDD). This study modeled the effects of genetic vulnerability by using the Flinders sensitive line (FSL), a rat model of depression and its control counterpart-the Flinders resistant line (FRL). The effects of environmental vulnerability (e.g., early-life stress) were modeled by using maternal separation. Rats (n=105) were drawn from four groups reflecting experimental crossing of strain (FSL vs. FRL) and early-life stress (high vs. low) to assess the effects of two antidepressants (escitalopram or nortriptyline) compared to vehicle. Quantitative in vitro autoradiography was performed using [(125)I]MPPI (5-HT1A) and [(125)I]CYP (5-HT1B) in prefrontal cortex (PFC) and hippocampus. Stringent, Bonferroni-corrected statistical analyses showed significant strain-by-rearing-by-treatment (three-way) interactions in PFC 5-HT1A and hippocampal 5-HT1B receptors. Either vulnerability reduced serotonergic binding; no additive effects were associated with the two vulnerabilities. Both antidepressants increased hippocampal 5-HT1B receptor binding; however, only nortriptyline selectively increased PFC 5-HT1A receptor binding. Taken together, our findings demonstrate that antidepressant effects on the serotonergic system are shaped by a GxE interaction that depends on antidepressant class and brain region.


Assuntos
Antidepressivos/farmacologia , Química Encefálica/genética , Depressão/genética , Interação Gene-Ambiente , Receptor 5-HT1A de Serotonina/genética , Receptor 5-HT1B de Serotonina/genética , Meio Social , Animais , Antidepressivos/uso terapêutico , Depressão/tratamento farmacológico , Modelos Animais de Doenças , Feminino , Genes Ligados ao Cromossomo X/fisiologia , Masculino , Privação Materna , Ligação Proteica/genética , Ratos , Ratos Transgênicos , Receptor 5-HT1A de Serotonina/metabolismo , Receptor 5-HT1B de Serotonina/metabolismo , Especificidade da Espécie
10.
Rev Neurol (Paris) ; 169(6-7): 515-8, 2013.
Artigo em Francês | MEDLINE | ID: mdl-23557641

RESUMO

INTRODUCTION: Fragile X associated Tremor/Ataxia Syndrome (FXTAS) is related to premutation expansions of the FMR1 gene, including 55 to 200 CGG repeats, whereas full expansions, over 200 repeats, cause Fragile X mental retardation. FXTAS is observed in about one-third of men with premutation, generally in their 1950s and over, and less commonly in women. It is characterized by action tremor, ataxia, cognitive, parkinsonism, behavioral difficulties, autonomic dysfunction and peripheral neuropathy. Brain magnetic resonance imaging (MRI) often shows symmetric increases in T2-weighted signal intensity in the middle cerebellar peduncles. The diagnosis of FXTAS in a patient raises important family issues. CASE REPORT: A 47-year-old male patient complained of an abnormal hearing sensation and dizziness for several months. Neurological examination was normal. Brain MRI showed the common signal intensity in middle cerebellar peduncles and bilateral punctual increases in T2-weighted signal intensity in the cerebral white matter. Genetic analysis showed 87CGG repeats, in favor of a possible FXTAS. At the time of diagnosis, fragile X syndrome was subsequently suspected and confirmed in his 10-month-old grandson. DISCUSSION: Due to X-linked inheritance and to the specific related mutational mechanism, the diagnosis of FXTAS in a patient raises major issues for relatives over several generations, including males and females, who should be considered as obligate or potential premutation carriers. Premutated females are not only at risk of transmitting a full mutation to their children but also of developing Fragile X related premature ovarian failure (FXPOI) that may influence their choices in family planning. CONCLUSION: The diagnosis of FXATS in a patient should induce delivery of extensive information and genetic counseling for potential carrier relatives.


Assuntos
Família/psicologia , Síndrome do Cromossomo X Frágil/diagnóstico , Aconselhamento Genético/psicologia , Síndrome do Cromossomo X Frágil/genética , Genes Ligados ao Cromossomo X/fisiologia , Testes Genéticos , Humanos , Masculino , Pessoa de Meia-Idade , Linhagem , Risco
11.
Eur J Oral Sci ; 121(2): 63-8, 2013 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-23489894

RESUMO

As part of an international consortium, case-parent trios were collected for a genome-wide association study of isolated, non-syndromic oral clefts, including cleft lip (CL), cleft palate (CP), and cleft lip and palate (CLP). Non-syndromic oral clefts have a complex and heterogeneous etiology. Risk is influenced by genes and environmental factors, and differs markedly by gender. Family-based association tests (FBAT) were used on 14,486 single nucleotide polymorphisms (SNPs) spanning the X chromosome, stratified by type of cleft and racial group. Significant results, even after multiple-comparisons correction, were obtained for the Duchenne muscular dystrophy (DMD) gene, the largest single gene in the human genome, among CL/P (i.e., both CL and CLP combined) trios. When stratified into groups of European and Asian ancestry, stronger signals were obtained for Asian subjects. Although conventional sliding-window haplotype analysis showed no increase in significance, selected combinations of the 25 most significant SNPs in the DMD gene identified four SNPs together that attained genome-wide significance among Asian CL/P trios, raising the possibility of interaction between distant SNPs within the DMD gene.


Assuntos
Povo Asiático/genética , Fenda Labial/genética , Fissura Palatina/genética , Genes Ligados ao Cromossomo X/fisiologia , Marcadores Genéticos , Distrofia Muscular de Duchenne/genética , População Branca/genética , Adulto , Feminino , Estudo de Associação Genômica Ampla/métodos , Haplótipos/genética , Haplótipos/fisiologia , Humanos , Masculino , Polimorfismo de Nucleotídeo Único , Análise de Componente Principal , Risco
12.
Proc Natl Acad Sci U S A ; 109(29): 11752-7, 2012 Jul 17.
Artigo em Inglês | MEDLINE | ID: mdl-22753487

RESUMO

Susumu Ohno proposed in 1967 that, during the origin of mammalian sex chromosomes from a pair of autosomes, per-allele expression levels of X-linked genes were doubled to compensate for the degeneration of their Y homologs. This conjecture forms the foundation of the current evolutionary model of sex chromosome dosage compensation, but has been tested in mammals only indirectly via a comparison of expression levels between X-linked and autosomal genes in the same genome. The test results have been controversial, because examinations of different gene sets led to different conclusions that either support or refute Ohno's hypothesis. Here we resolve this uncertainty by directly comparing mammalian X-linked genes with their one-to-one orthologs in species that diverged before the origin of the mammalian sex chromosomes. Analyses of RNA sequencing data and proteomic data provide unambiguous evidence for expression halving (i.e., no change in per-allele expression level) of X-linked genes during evolution, with the exception of only ∼5% of genes that encode members of large protein complexes. We conclude that Ohno's hypothesis is rejected for the vast majority of genes, reopening the search for the evolutionary force driving the origin of chromosome-wide X inactivation in female mammals.


Assuntos
Mecanismo Genético de Compensação de Dose/genética , Evolução Molecular , Genes Ligados ao Cromossomo X/genética , Mamíferos/genética , Inativação do Cromossomo X/genética , Animais , Galinhas/genética , Biologia Computacional , Genes Ligados ao Cromossomo X/fisiologia , Humanos , Proteômica , Análise de Sequência de RNA , Especificidade da Espécie
13.
PLoS One ; 6(2): e17358, 2011 Feb 28.
Artigo em Inglês | MEDLINE | ID: mdl-21386982

RESUMO

BACKGROUND: The evolution of female choice mechanisms favouring males of their own kind is considered a crucial step during the early stages of speciation. However, although the genomics of mate choice may influence both the likelihood and speed of speciation, the identity and location of genes underlying assortative mating remain largely unknown. METHODS AND FINDINGS: We used mate choice experiments and gene expression analysis of female Drosophila melanogaster to examine three key components influencing speciation. We show that the 1,498 genes in Zimbabwean female D. melanogaster whose expression levels differ when mating with more (Zimbabwean) versus less (Cosmopolitan strain) preferred males include many with high expression in the central nervous system and ovaries, are disproportionately X-linked and form a number of clusters with low recombination distance. Significant involvement of the brain and ovaries is consistent with the action of a combination of pre- and postcopulatory female choice mechanisms, while sex linkage and clustering of genes lead to high potential evolutionary rate and sheltering against the homogenizing effects of gene exchange between populations. CONCLUSION: Taken together our results imply favourable genomic conditions for the evolution of reproductive isolation through mate choice in Zimbabwean D. melanogaster and suggest that mate choice may, in general, act as an even more important engine of speciation than previously realized.


Assuntos
Drosophila melanogaster/genética , Drosophila melanogaster/fisiologia , Genes Ligados ao Cromossomo X/fisiologia , Comportamento Sexual Animal/fisiologia , Isolamento Social , Animais , Comportamento de Escolha/fisiologia , Mapeamento Cromossômico , Análise por Conglomerados , Feminino , Regulação da Expressão Gênica , Estudos de Associação Genética , Masculino , Modelos Biológicos , Recombinação Genética/fisiologia , Caracteres Sexuais , Cromossomo X/genética , Cromossomo X/fisiologia
14.
Semin Neurol ; 31(5): 484-93, 2011 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-22266886

RESUMO

Hereditary spastic paraplegias (HSPs) are clinically and genetically highly heterogeneous. The key symptom of spastic paraparesis of lower limbs can be complicated by a variety of signs and symptoms including cognitive impairment, optic atrophy, cerebellar ataxia, peripheral nerve involvement, or seizures. At least 48 loci have been identified, termed SPG1-SPG48. Ten genes for autosomal dominant HSP are currently known, SPG4 being by far the most common subtype accounting for ∼50% of cases. SPG3 is especially common in young-onset cases. Autosomal recessive HSP seems to be even more heterogeneous. The known 12 autosomal recessive HSP genes collectively explain about one third of cases only. The most common causes for pure autosomal recessive HSP are SPG7 and SPG5. Mental retardation and thin corpus callosum on magnetic resonance imaging point toward SPG11 and SPG15. The authors provide an overview on clinical, neurophysiologic, and neuroradiologic characteristics of the more common HSP subtypes. More details are given in the tables for quick reference, and a genetic testing strategy is proposed.


Assuntos
Genes Dominantes/fisiologia , Genes Recessivos/fisiologia , Genes Ligados ao Cromossomo X/fisiologia , Paraplegia Espástica Hereditária/genética , Agenesia do Corpo Caloso/genética , Estudos de Associação Genética , Doenças Genéticas Ligadas ao Cromossomo X , Testes Genéticos/métodos , Humanos , Deficiência Intelectual/genética , Mutação
15.
Int J Biochem Cell Biol ; 42(11): 1784-7, 2010 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-20678582

RESUMO

The FOXP3 (forkhead box P3) gene is a member of forkhead winged helix family transcription factors and functions as both a transcriptional activator and a repressor. FOXP3 dysfunction is responsible for an X-linked autoimmune syndrome: immune dysregulation, polyendopathy, enterophathy, X-linked syndrome. In addition to its role as an essential transcription factor in regulatory T cells, the FOXP3 gene is an epithelial cell-intrinsic tumor suppressor for breast and prostate cancers. We will focus on the FOXP3 signalling pathway in epithelial cells and discuss how genetic and/or epigenetic inactivation of the FOXP3 contributes to the malignant transformation of cells.


Assuntos
Fatores de Transcrição Forkhead/metabolismo , Genes Ligados ao Cromossomo X/genética , Animais , Neoplasias da Mama/genética , Neoplasias da Mama/metabolismo , Feminino , Fatores de Transcrição Forkhead/genética , Genes Supressores de Tumor/fisiologia , Genes Ligados ao Cromossomo X/fisiologia , Humanos , Masculino , Neoplasias da Próstata/genética , Neoplasias da Próstata/metabolismo
16.
Cytogenet Genome Res ; 124(3-4): 298-311, 2009.
Artigo em Inglês | MEDLINE | ID: mdl-19556782

RESUMO

The effect of ectopic expression of male specific lethal 2 (msl2) on chromatin modification and gene expression was studied in Drosophila diploid females and metafemales (3X;2A). Results show that ectopic expression of MSL2 in transgenic msl2 females and metafemales sequesters the MOF histone acetylase to the X, which occurs concordantly with an increase of histone acetylation. Gene expression studies indicate that the X-linked genes are not affected by direct targeting of the MSL complex and the resulting increased H4Lys16 acetylation on the X chromosomes, suggesting one function of the MSL complex is to nullify the effect of a high level of histone acetylation. These results are not consistent with the hypothesis that the presence of the MSL complex conditions a two-fold upregulation. Autosomal gene expression is generally decreased in ectopically expressed MSL2 females, which correlates with the reduced autosomal histone acetylation. Metafemales show dosage compensation of X-linked genes with some autosomal reductions in expression. Interestingly, in metafemales with ectopically expressed MSL2, the autosomal expression is returned to a more normal level. There is a lower autosomal level of histone acetylation compared to the normal metafemales, suggesting a nullifying effect on the negative dosage effect of the X chromosome as previously hypothesized to occur in normal males.


Assuntos
Proteínas de Ligação a DNA/metabolismo , Mecanismo Genético de Compensação de Dose , Proteínas de Drosophila/metabolismo , Drosophila melanogaster/metabolismo , Histona Acetiltransferases/metabolismo , Proteínas Nucleares/metabolismo , Fatores de Transcrição/metabolismo , Animais , Animais Geneticamente Modificados/genética , Animais Geneticamente Modificados/metabolismo , Proteínas de Ligação a DNA/genética , Proteínas de Drosophila/genética , Drosophila melanogaster/genética , Drosophila melanogaster/ultraestrutura , Feminino , Expressão Gênica/fisiologia , Genes Ligados ao Cromossomo X/fisiologia , Histona Acetiltransferases/genética , Masculino , Proteínas Nucleares/genética , Fatores de Transcrição/genética , Cromossomo X/metabolismo , Cromossomo X/ultraestrutura
17.
PLoS One ; 3(12): e3915, 2008.
Artigo em Inglês | MEDLINE | ID: mdl-19104663

RESUMO

BACKGROUND: The fish medaka is the first vertebrate capable of full spermatogenesis in vitro from self-renewing spermatogonial stem cells to motile test-tube sperm. Precise staging and molecular dissection of this process has been hampered by the lack of suitable molecular markers. METHODOLOGY AND PRINCIPAL FINDINGS: We have generated a normalized medaka testis cDNA library and obtained 7040 high quality sequences representing 3641 unique gene clusters. Among these, 1197 unique clusters are homologous to known genes, and 2444 appear to be novel genes. Ontology analysis shows that the 1197 gene products are implicated in diverse molecular and cellular processes. These genes include markers for all major types of testicular somatic and germ cells. Furthermore, markers were identified for major spermatogenic stages ranging from spermatogonial stem cell self-renewal to meiosis entry, progression and completion. Intriguingly, the medaka testis expresses at least 13 homologs of the 33 mouse X-chromosomal genes that are enriched in the testis. More importantly, we show that key components of several signaling pathways known to be important for testicular function in mammals are well represented in the medaka testicular EST collection. CONCLUSIONS/SIGNIFICANCE: Medaka exhibits a considerable similarity in testicular gene expression to mammals. The medaka testicular EST collection we obtained has wide range coverage and will not only consolidate our knowledge on the comparative analysis of known genes' functions in the testis but also provide a rich resource to dissect molecular events and mechanism of spermatogenesis in vivo and in vitro in medaka as an excellent vertebrate model.


Assuntos
Clonagem Molecular/métodos , Etiquetas de Sequências Expressas , Peixes/genética , Mamíferos/genética , Oryzias/genética , Testículo/metabolismo , Animais , Sequência Conservada , Biblioteca Gênica , Genes Ligados ao Cromossomo X/fisiologia , Masculino , Camundongos , Modelos Biológicos , Família Multigênica , Especificidade de Órgãos/genética , Oryzias/metabolismo , Análise de Sequência de DNA
18.
Trends Genet ; 24(10): 478-81, 2008 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-18774619

RESUMO

It has been shown that selective pressure to compensate for the silencing of the sex chromosomes during male meiosis resulted in many X-linked genes being duplicated as functional retrogenes on autosomes. The silencing of male sex chromosomes was probably stratified during evolution, in accordance with their stratified diversification. Here I show that the timing of the retrocopying events is associated with the timing of the X-Y differentiation of the region of the X chromosome housing the parental copy of the gene.


Assuntos
Evolução Molecular , Retroelementos/genética , Cromossomos Sexuais/genética , Animais , Genes Ligados ao Cromossomo X/fisiologia , Humanos , Mamíferos/genética , Modelos Biológicos , Filogenia
19.
Pediatr Int ; 50(2): 172-4, 2008 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-18353053

RESUMO

BACKGROUND: 45,X Turner syndrome (TS) female subjects have visuospatial skill and social cognition deficits that may arise from X-linked imprinting. The aim of the present study was to compare phenotypic characteristics and neurocognitive pattern of 12 monosomic TS girls, according to X-linked imprinting. METHODS: Microsatellite markers were used to determine the parental origin of the missing chromosome X. Wechsler Intelligence Scale for Children-Revised (WISC-R) was administered as measures of general intellectual functioning. The results were compared in TS patients with maternally derived X chromosome (Xm) and paternally derived X chromosome (Xp). RESULTS: Six out of 12 patients (50%) had Xm, and the other six (50%) had Xp chromosome. There was no difference in the total, verbal and performance IQ score between the TS subgroups with Xm and Xp. When the WISC-R subtest score patterns were compared, the mean arithmetic scores were significantly poorer in the Xm TS than in the Xp TS. CONCLUSION: In monosomic TS cases, paternal imprinting may predict arithmetic ability, on the other hand, reductionist consideration defined by genetic imprinting is not sufficient to confirm this. Further studies should be undertaken to clarify this situation.


Assuntos
Cromossomos Humanos X/genética , Genes Ligados ao Cromossomo X/fisiologia , Síndrome de Turner/genética , Síndrome de Turner/psicologia , Adolescente , Criança , Cognição/fisiologia , Feminino , Humanos , Fenótipo , Síndrome de Turner/patologia
20.
Genes Dev ; 22(5): 682-91, 2008 Mar 01.
Artigo em Inglês | MEDLINE | ID: mdl-18316482

RESUMO

Meiotic silencing of sex chromosomes may cause their depletion of meiosis-specific genes during evolution. Here, we challenge this hypothesis by reporting the identification of TEX11 as the first X-encoded meiosis-specific factor in mice. TEX11 forms discrete foci on synapsed regions of meiotic chromosomes and appears to be a novel constituent of meiotic nodules involved in recombination. Loss of TEX11 function causes chromosomal asynapsis and reduced crossover formation, leading to elimination of spermatocytes, respectively, at the pachytene and anaphase I stages. Specifically, TEX11-deficient spermatocytes with asynapsed autosomes undergo apoptosis at the pachytene stage, while those with only asynapsed sex chromosomes progress. However, cells that survive the pachytene stage display chromosome nondisjunction at the first meiotic division, resulting in cell death and male infertility. TEX11 interacts with SYCP2, which is an integral component of the synaptonemal complex lateral elements. Thus, TEX11 promotes initiation and/or maintenance of synapsis and formation of crossovers, and may provide a physical link between these two meiotic processes.


Assuntos
Pareamento Cromossômico/genética , Troca Genética/genética , Genes Ligados ao Cromossomo X/fisiologia , Infertilidade Masculina/genética , Proteínas/fisiologia , Animais , Apoptose , Proteínas de Ciclo Celular , Proteínas Cromossômicas não Histona/metabolismo , Proteínas de Ligação a DNA , Feminino , Masculino , Camundongos , Camundongos Mutantes , Proteínas/genética , Espermatócitos/citologia , Espermatócitos/metabolismo , Espermatócitos/fisiologia , Cromossomo X/genética , Cromossomo Y/genética
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...