Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 31
Filtrar
1.
Reproduction ; 165(4): 347-362, 2023 04 01.
Artigo em Inglês | MEDLINE | ID: mdl-36633493

RESUMO

In brief: Maternal obesity can impair metabolism in the embryo and the resulting offspring. This study shows that metabolic disruptions through α-ketoglutarate may link altered metabolism with epigenetic changes in embryos. Abstract: Maternal obesity can impair offspring metabolic health; however, the precise mechanism underpinning programming is unknown. Ten-Eleven translocase (TET) enzymes demethylate DNA using the TCA cycle intermediary α-ketoglutarate and may be involved in programming offspring health. Whether TETs are disrupted by maternal obesity is unknown. Five to six week-old C57Bl/6 female mice were fed a control diet (CD; 6% fat, n = 175) or a high-fat diet (HFD; 21% fat, n = 158) for 6 weeks. After superovulation, oocytes were collected for metabolic assessment, or females were mated and zygotes were cultured for embryo development, fetal growth, and assessment of global DNA methylation (5-methylcytosine (5mC), 5-hydroxymethylcytosine (5hmC), 5-formylcytosine (5fC), and 5-carboxycytosine (5caC)) in the two-cell embryo. Zygotes collected from superovulated CBAF1 females were cultured in media containing α-ketoglutarate (0, 1.4, 3.5, or 14.0 mM) or with 2-hydroxyglutarate (2HG) (0 or 20 mM), a competitive inhibitor of α-ketoglutarate, with methylation and blastocyst differentiation assessed. After HFD, oocytes showed increased pyruvate oxidation and intracellular ROS, with no changes in Tet3 expression, while two-cell embryo global 5hmC DNA methylation was reduced and 5fC increased. Embryos cultured with 1.4 mM α-ketoglutarate had decreased two-cell 5mC, while 14.0 mM α-ketoglutarate increased the 5hmC:5mC ratio. In contrast, supplementation with 20 mM 2HG increased 5mC and decreased 5fC:5mC and 5caC:5mC ratios. α-ketoglutarate up to 3.5 mM did not alter embryo development, while culturing in 14.0 mM α-ketoglutarate blocked development at the two-cell. Culture with 2HG delayed embryo development past the four-cell and decreased blastocyst total cell number. In conclusion, disruptions in metabolic intermediates in the preimplantation embryo may provide a link between maternal obesity and programming offspring for ill health.


Assuntos
Metilação de DNA , Obesidade Materna , Animais , Feminino , Humanos , Camundongos , Gravidez , 5-Metilcitosina/metabolismo , Citosina/metabolismo , Dieta Hiperlipídica , Ácidos Cetoglutáricos/farmacologia , Obesidade Materna/metabolismo , Zigoto/metabolismo
2.
Mol Reprod Dev ; 84(9): 914-925, 2017 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-28452160

RESUMO

The reproductive tract environment at conception programs the developmental trajectory of the embryo, sets the course of pregnancy, and impacts offspring phenotype and health. Despite the fundamental importance of this stage of reproduction, the rate-limiting regulatory mechanisms operating locally to control fertility and fecundity are incompletely understood. Emerging studies highlight roles for microRNAs (miRNAs) in regulating reproductive and developmental processes and in modulating the quality and strength of the female immune response. Since endometrial receptivity and robust placentation require specific adaptation of the immune response, we hypothesize that miRNAs participate in establishing pregnancy through effects on key gene networks in immune cells. Our recent studies investigated miRNAs that are induced in the peri-conception environment, focusing on miRNAs that have immune-regulatory roles-particularly miR-223, miR-155, and miR-146a. Genetic mouse models deficient in individual miRNAs are proving informative in defining roles for these miRNAs in the generation and stabilization of regulatory T cells (Treg cells) that confer adaptive immune tolerance. Overlapping and redundant functions between miRNAs that target multiple genes, combined with multiple miRNAs targeting individual genes, indicate complex and sensitive regulatory networks. Although to date most data on miRNA regulation of reproductive events are from mice, conserved functions of miRNAs across species imply similar biological pathways operate in all mammals. Understanding the regulation and roles of miRNAs in the peri-conception immune response will advance our knowledge of how environmental determinants act at conception, and could have practical applications for animal breeding as well as human fertility.


Assuntos
Fertilidade/imunologia , Tolerância Imunológica , MicroRNAs/imunologia , Placentação/imunologia , Linfócitos T Reguladores/imunologia , Animais , Feminino , Humanos , Camundongos , Gravidez
3.
Am J Physiol Endocrinol Metab ; 308(9): E805-21, 2015 May 01.
Artigo em Inglês | MEDLINE | ID: mdl-25690453

RESUMO

Obesity and type 2 diabetes are increasingly prevalent across all demographics. Paternal obesity in humans and rodents can program obesity and impair insulin sensitivity in female offspring. It remains to be determined whether these perturbed offspring phenotypes can be improved through targeted lifestyle interventions in the obese father. Using a mouse model, we demonstrate that diet or exercise interventions for 8 wk (2 rounds of spermatogenesis) in obese founder males restores insulin sensitivity and normalized adiposity in female offspring. Founder diet and/or exercise also normalizes abundance of X-linked sperm microRNAs that target genes regulating cell cycle and apoptosis, pathways central to oocyte and early embryogenesis. Additionally, obesity-associated comorbidities, including inflammation, glucose intolerance, stress, and hypercholesterolemia, were good predictors for sperm microRNA abundance and offspring phenotypes. Interventions aimed at improving paternal metabolic health during specific windows prior to conception can partially normalize aberrant epigenetic signals in sperm and improve the metabolic health of female offspring.


Assuntos
Fenômenos Fisiológicos da Nutrição Animal , Pai , Síndrome Metabólica/prevenção & controle , MicroRNAs/genética , Obesidade , Condicionamento Físico Animal/fisiologia , Espermatozoides/metabolismo , Animais , Dieta , Feminino , Fertilização/fisiologia , Infertilidade Masculina/genética , Infertilidade Masculina/prevenção & controle , Masculino , Síndrome Metabólica/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Obesos , Obesidade/genética , Gravidez , Efeitos Tardios da Exposição Pré-Natal/genética , Efeitos Tardios da Exposição Pré-Natal/metabolismo , Transcriptoma
4.
Biol Reprod ; 92(5): 124, 2015 May.
Artigo em Inglês | MEDLINE | ID: mdl-25715796

RESUMO

The periconceptual environment represents a critical window for programming fetal growth trajectories and susceptibility to disease; however, the underlying mechanism responsible for programming remains elusive. This study demonstrates a causal link between reduction of precompaction embryonic mitochondrial function and perturbed offspring growth trajectories and subsequent metabolic dysfunction. Incubation of embryos with carbonyl cyanide 4-(trifluoromethoxy) phenylhydrazone (FCCP), which uncouples mitochondrial oxidative phosphorylation, significantly reduced mitochondrial membrane potential and ATP production in 8-cell embryos and the number of inner cell mass cells within blastocysts; however, blastocyst development was unchanged. This perturbed embryonic mitochondrial function was concomitant with reduced birth weight in female offspring following embryo transfer, which persisted until weaning. FCCP-treated females also exhibited increased adiposity at 4 wk, increased adiposity gain between 4 and 14 wk, glucose intolerance at 8 wk, and insulin resistance at 14 wk. Although FCCP-treated males also exhibited reduced glucose tolerance, but their insulin sensitivity and adiposity gain between 4 and 14 wk was unchanged. To our knowledge, this is one of the first studies to demonstrate that reducing mitochondrial function and, thus, decreasing ATP output in the precompacting embryo can influence offspring phenotype. This is of great significance as a large proportion of patients requiring assisted reproductive technologies are of advanced maternal age or have a high body mass index, both of which have been independently linked with perturbed early embryonic mitochondrial function.


Assuntos
Carbonil Cianeto p-Trifluormetoxifenil Hidrazona/toxicidade , Fase de Clivagem do Zigoto/efeitos dos fármacos , Mitocôndrias/efeitos dos fármacos , Trifosfato de Adenosina/genética , Trifosfato de Adenosina/metabolismo , Adiposidade/efeitos dos fármacos , Animais , Peso ao Nascer , Técnicas de Cultura Embrionária , Transferência Embrionária , Desenvolvimento Embrionário/efeitos dos fármacos , Feminino , Regulação da Expressão Gênica no Desenvolvimento/efeitos dos fármacos , Tamanho da Ninhada de Vivíparos , Masculino , Metaloproteases/genética , Metaloproteases/metabolismo , Camundongos , Mitocôndrias/metabolismo , Gravidez , Efeitos Tardios da Exposição Pré-Natal
5.
J Assist Reprod Genet ; 32(5): 725-35, 2015 May.
Artigo em Inglês | MEDLINE | ID: mdl-25854657

RESUMO

PURPOSE: To investigate the impacts that a paternal high fat diet (HFD) has on embryology, ovarian/cumulus cell gene expression and COC metabolism from female offspring, using a mouse model. METHODS: Founder male mice were either fed a control diet (CD) or a HFD for 12 weeks. The HFD induced obesity but not diabetes, and founder males were then mated to normal weight CD fed female mice. Female offspring were maintained on a CD, super-ovulated, mated and the resultant zygotes were cultured to the blastocyst stage for embryo morphology, blastocyst cell number and apoptosis assessment. Ovaries and cumulus cells from offspring were collected for gene expression analysis of selected genes that maintain chromatin remodeling and endoplasmic reticulum (ER), metabolic and inflammatory homeostasis. Cumulus/oocyte complexes were also investigated for glucose uptake and lipid accumulation. RESULTS: Female offspring sired by obese fathers produced embryos with delayed development and impaired quality, displayed increases in ovarian expression of Glut1, Glut3 and Glut4, and an increase in cumulus cell expression of Glut4. Interestingly their COCs did take up more glucose, but did accumulate more lipid. CONCLUSIONS: A paternal HFD is associated with subfertility in female offspring despite the offspring being fed a CD and this subfertility is concomitant with ovarian/cumulus cell molecular alterations and increased lipid accumulation.


Assuntos
Blastocisto/patologia , Células do Cúmulo/metabolismo , Dieta Hiperlipídica/efeitos adversos , Obesidade/fisiopatologia , Oócitos/metabolismo , Ovário/metabolismo , Animais , Blastocisto/metabolismo , Células do Cúmulo/patologia , Embrião de Mamíferos/citologia , Embrião de Mamíferos/metabolismo , Feminino , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Obesos , Obesidade/etiologia , Oócitos/patologia , Ovário/patologia , Gravidez , Reação em Cadeia da Polimerase em Tempo Real , Sêmen/química
6.
FASEB J ; 27(10): 4226-43, 2013 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-23845863

RESUMO

Obesity is highly prevalent, and its incidence is increasing. The previous study showing a major effect of paternal obesity on metabolic health of offspring is confounded by comorbidity with diabetes. Therefore, we investigated the effect of diet-induced paternal obesity, in the absence of diabetes, on the metabolic health of two resultant generations and the molecular profiles of the testes and sperm. Founder (F0) male C57BL6 mice were fed either a high-fat diet (HFD) or a control diet (CD); n = 10/diet for a period of 10 wk. Testis expression of mRNA/microRNAs was analyzed by microarray and qPCR and sperm microRNA abundance by qPCR. Two subsequent generations were generated by mating F0 and then F1 mice to CD mice, and their metabolic health was investigated. All mice, other than F0 males, were maintained on a CD. HFD feeding induced paternal obesity with a 21% increase in adiposity, but not overt diabetes, and initiated intergenerational transmission of obesity and insulin resistance in two generations of offspring. This distinct phenotypic constellation is either partially or fully transmitted to both female and male F1 offspring and further transmitted through both parental lineages to the F2 generation, with a heightened effect on female F1 offspring (+67% in adiposity) and their F2 sons (+24% in adiposity). Founder male obesity altered the testes expression of 414 mRNAs by microarray and 11 microRNAs by qPCR, concomitant with alterations in sperm microRNA content and a 25% reduction in global methylation of germ cell DNA. Diet-induced paternal obesity modulates sperm microRNA content and germ cell methylation status, which are potential signals that program offspring health and initiate the transmission of obesity and impaired metabolic health to future generations. This study implicates paternal obesity in the transgenerational amplification of obesity and type 2 diabetes in humans.


Assuntos
MicroRNAs/metabolismo , Obesidade/metabolismo , Espermatozoides/metabolismo , Testículo/metabolismo , Transcriptoma/fisiologia , Animais , Metabolismo Energético , Feminino , Regulação da Expressão Gênica/fisiologia , Teste de Tolerância a Glucose , Resistência à Insulina , Masculino , Camundongos , Camundongos Endogâmicos C57BL , MicroRNAs/genética , Obesidade/genética , Espécies Reativas de Oxigênio , Fatores Sexuais
7.
Ann Nutr Metab ; 64(3-4): 231-8, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-25300265

RESUMO

BACKGROUND: The global rates of male overweight/obesity are rising, approaching 70% of the total adult population in Western nations. Overweight/obesity increases the risk of chronic diseases; however, there is increasing awareness that male obesity negatively impacts fertility, subsequent pregnancy, and the offspring health burden. Developmental programming is well defined in mothers; however, it is becoming increasingly evident that developmental programming can be paternally initiated and mediated through paternal obesity. KEY MESSAGES: Both human and rodent models have established that paternal obesity impairs sex hormones, basic sperm function, and molecular composition. This results in perturbed embryo development and health and an increased subsequent offspring disease burden in both sexes. The reversibility of obesity-induced parental programming has only recently received attention. Promising results in animal models utilizing diet and exercise interventions have shown improvements in sperm function and molecular composition, resulting in restorations of both embryo and fetal health and subsequent male offspring fertility. The direct mode for paternal inheritance is likely mediated via spermatozoa. We propose two main theories for the origin of male obesity-induced paternal programming: (1) accumulation of sperm DNA damage resulting in de novo mutations in the embryo and (2) changes in sperm epigenetic marks (microRNA, methylation, or acetylation) altering the access, transcription, and translation of paternally derived genes during early embryogenesis. CONCLUSIONS: Paternal overweight/obesity induces paternal programming of offspring phenotypes likely mediated through genetic and epigenetic changes in spermatozoa. These programmed changes to offspring health appear to be partially restored via diet/exercise interventions in obese fathers preconception, which have been shown to improve aspects of sperm DNA integrity. However, the majority of data surrounding paternal obesity and offspring phenotypes have come from rodent models; therefore, we contend that it will be increasingly important to study population-based data to determine the likely mode of inheritance in humans.


Assuntos
Dieta/efeitos adversos , Saúde da Família , Promoção da Saúde , Atividade Motora , Obesidade/etiologia , Obesidade/prevenção & controle , Cooperação do Paciente , Animais , Criança , Fenômenos Fisiológicos da Nutrição Infantil , Dano ao DNA , Epigênese Genética , Pai , Feminino , Desenvolvimento Fetal , Humanos , Estilo de Vida , Masculino , Política Nutricional , Obesidade/epidemiologia , Obesidade/patologia , Comportamento Paterno , Gravidez , Espermatozoides/patologia
8.
Am J Hum Genet ; 87(2): 173-88, 2010 Aug 13.
Artigo em Inglês | MEDLINE | ID: mdl-20655035

RESUMO

Copy number variants and indels in 251 families with evidence of X-linked intellectual disability (XLID) were investigated by array comparative genomic hybridization on a high-density oligonucleotide X chromosome array platform. We identified pathogenic copy number variants in 10% of families, with mutations ranging from 2 kb to 11 Mb in size. The challenge of assessing causality was facilitated by prior knowledge of XLID-associated genes and the ability to test for cosegregation of variants with disease through extended pedigrees. Fine-scale analysis of rare variants in XLID families leads us to propose four additional genes, PTCHD1, WDR13, FAAH2, and GSPT2, as candidates for XLID causation and the identification of further deletions and duplications affecting X chromosome genes but without apparent disease consequences. Breakpoints of pathogenic variants were characterized to provide insight into the underlying mutational mechanisms and indicated a predominance of mitotic rather than meiotic events. By effectively bridging the gap between karyotype-level investigations and X chromosome exon resequencing, this study informs discussion of alternative mutational mechanisms, such as noncoding variants and non-X-linked disease, which might explain the shortfall of mutation yield in the well-characterized International Genetics of Learning Disability (IGOLD) cohort, where currently disease remains unexplained in two-thirds of families.


Assuntos
Cromossomos Humanos X/genética , Variações do Número de Cópias de DNA/genética , Mutação INDEL/genética , Deficiência Intelectual/genética , Quebra Cromossômica , Segregação de Cromossomos/genética , Estudos de Coortes , Doença/genética , Feminino , Rearranjo Gênico/genética , Genes Ligados ao Cromossomo X/genética , Humanos , Masculino , Análise de Sequência com Séries de Oligonucleotídeos , Linhagem , Reprodutibilidade dos Testes , Retroelementos/genética , Deleção de Sequência/genética
9.
Reprod Fertil Dev ; 23(5): 691-701, 2011.
Artigo em Inglês | MEDLINE | ID: mdl-21635818

RESUMO

Environmental stress results in perturbations to mitochondrial function in the preimplantation embryo and hinders subsequent embryo and possibly offspring development. Global gene expression in fetal mouse brain was investigated following targeted mitochondrial inhibition by amino-oxyacetate (AOA) from the 2-cell to the blastocyst stage. Blastocysts were transferred to pseudopregnant recipients and RNA extracted from Day 18 fetal brains for microarray interrogation. Exposure to 5 µM AOA during preimplantation embryo development induced differential expression of 166 genes (>1.25 fold) in the fetal brain, relative to control medium-cultured embryos. Altered expression pathways included carbohydrate metabolism, neurological development, cellular proliferation and death, DNA replication, recombination and repair. Of 28 genes exhibiting the greatest change in expression, qPCR confirmed that 16 were significantly altered. Targeted qPCR assessment of a further 20 genes associated with methylation, acetylation and mitochondrial dysfunction revealed that three were significantly altered (Immp1l, Nars2, Sat2) and Dmap1 exhibited a sex-specific response to AOA exposure. Only 2/48 genes had significantly altered expression by qPCR (Nola3, Timm8b) in fetal brains exposed to 50 µM AOA embryo culture, excluding an AOA dose-dependent response. It was concluded that perturbation of mitochondrial function induced by 5 µM AOA during preimplantation embryo development alters gene expression in the neonatal brain in a manner that suggests that proper brain development may be compromised.


Assuntos
Ácido Amino-Oxiacético/farmacologia , Blastocisto/efeitos dos fármacos , Encéfalo/efeitos dos fármacos , Regulação da Expressão Gênica no Desenvolvimento/efeitos dos fármacos , Mitocôndrias/efeitos dos fármacos , RNA Mensageiro/metabolismo , Transcrição Gênica/efeitos dos fármacos , Animais , Blastocisto/metabolismo , Encéfalo/embriologia , Encéfalo/metabolismo , Técnicas de Cultura Embrionária , Transferência Embrionária , Epigênese Genética/efeitos dos fármacos , Perfilação da Expressão Gênica/métodos , Redes Reguladoras de Genes/efeitos dos fármacos , Idade Gestacional , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Endogâmicos CBA , Mitocôndrias/metabolismo , Análise de Sequência com Séries de Oligonucleotídeos , Reação em Cadeia da Polimerase
10.
Reprod Fertil Dev ; 23(7): 929-39, 2011.
Artigo em Inglês | MEDLINE | ID: mdl-21871212

RESUMO

Male obesity is associated with reduced sperm function and increased incidence of sperm DNA damage; however, the underlying molecular mechanisms have not yet been identified. Mammalian SIRT6 protein is involved in caloric-dependant DNA damage repair in other tissue types, yet a possible role for SIRT6 in male obesity and subfertility has not been investigated previously. To assess SIRT6 levels and activity in the testes, male mice (n=12 per diet) were fed either a control diet (CD; 6% fat) or a high-fat diet (HFD; 21% fat) for 16 weeks before the collection of testes and spermatozoa. SIRT6 protein was localised to the nucleus of transitional spermatids and the acrosome of mature spermatozoa, with levels significantly decreased in HFD-fed male mice (P<0.05). This decrease in SIRT6 protein was associated with transitional spermatids having increased levels of acetylated H3K9 in the nucleus (P<0.01) and increased DNA damage (P<0.001). We propose a role for SIRT6 in spermiogenesis and potentially protamination processes, which are known to be compromised by male obesity.


Assuntos
Obesidade/metabolismo , Sirtuínas/metabolismo , Espermatogênese , Espermatozoides/metabolismo , Testículo/metabolismo , Acetilação , Acrossomo/metabolismo , Acrossomo/ultraestrutura , Animais , Núcleo Celular/metabolismo , Núcleo Celular/ultraestrutura , Dano ao DNA , Gorduras na Dieta/efeitos adversos , Regulação para Baixo , Epididimo/metabolismo , Epididimo/ultraestrutura , Infertilidade Masculina/etiologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Obesidade/patologia , Obesidade/fisiopatologia , Transporte Proteico , RNA Mensageiro/metabolismo , Distribuição Aleatória , Sirtuínas/genética , Capacitação Espermática , Espermátides/metabolismo , Espermátides/ultraestrutura , Espermatozoides/ultraestrutura , Testículo/ultraestrutura
11.
Am J Med Genet B Neuropsychiatr Genet ; 156(2): 204-14, 2011 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-21302349

RESUMO

We report two rare genetic aberrations in a schizophrenia patient that may act together to confer disease susceptibility. A previously unreported balanced t(9;17)(q33.2;q25.3) translocation was observed in two schizophrenia-affected members of a small family with diverse psychiatric disorders. The proband also carried a 1.5 Mbp microduplication at 16p13.1 that could not be investigated in other family members. The duplication has been reported to predispose to schizophrenia, autism and mental retardation, with incomplete penetrance and variable expressivity. The t(9;17) (q33.2;q25.3) translocation breakpoint occurs within the open reading frames of KIAA1618 on 17q25.3, and TTLL11 (tyrosine tubulin ligase like 11) on 9q33.2, causing no change in the expression level of KIAA1618 but leading to loss of expression of one TTLL11 allele. TTLL11 belongs to a family of enzymes catalyzing polyglutamylation, an unusual neuron-specific post-translational modification of microtubule proteins, which modulates microtubule development and dynamics. The 16p13.1 duplication resulted in increased expression of NDE1, encoding a DISC1 protein partner mediating DISC1 functions in microtubule dynamics. We hypothesize that concomitant TTLL11-NDE1 deregulation may increase mutation load, among others, also on the DISC1 pathway, which could contribute to disease pathogenesis through multiple effects on neuronal development, synaptic plasticity, and neurotransmission. Our data illustrate the difficulties in interpreting the contribution of multiple potentially pathogenic changes likely to emerge in future next-generation sequencing studies, where access to extended families will be increasingly important.


Assuntos
Cromossomos Humanos Par 16 , Cromossomos Humanos Par 17/genética , Cromossomos Humanos Par 9/genética , Esquizofrenia/genética , Duplicações Segmentares Genômicas , Translocação Genética , Adulto , Alelos , Família , Humanos , Masculino , Mutação/genética , Proteínas do Tecido Nervoso/genética , Linhagem , Processamento de Proteína Pós-Traducional , Esquizofrenia/metabolismo , Esquizofrenia/patologia
12.
Endocrinology ; 162(10)2021 10 01.
Artigo em Inglês | MEDLINE | ID: mdl-34170298

RESUMO

Paternal experiences and exposures before conception can influence fetal development and offspring phenotype. The composition of seminal plasma contributes to paternal programming effects through modulating the female reproductive tract immune response after mating. To investigate whether paternal obesity affects seminal plasma immune-regulatory activity, C57Bl/6 male mice were fed an obesogenic high-fat diet (HFD) or control diet (CD) for 14 weeks. Although HFD consumption caused only minor changes to parameters of sperm quality, the volume of seminal vesicle fluid secretions was increased by 65%, and the concentrations and total content of immune-regulatory TGF-ß isoforms were decreased by 75% to 80% and 43% to 55%, respectively. Mating with BALB/c females revealed differences in the strength and properties of the postmating immune response elicited. Transcriptional analysis showed >300 inflammatory genes were similarly regulated in the uterine endometrium by mating independently of paternal diet, and 13 were dysregulated by HFD-fed compared with CD-fed males. Seminal vesicle fluid factors reduced in HFD-fed males, including TGF-ß1, IL-10, and TNF, were among the predicted upstream regulators of differentially regulated genes. Additionally, the T-cell response induced by mating with CD-fed males was blunted after mating with HFD-fed males, with 27% fewer CD4+ T cells, 26% fewer FOXP3+CD4+ regulatory T cells (Treg) cells, and 19% fewer CTLA4+ Treg cells, particularly within the NRP1+ thymic Treg cell population. These findings demonstrate that an obesogenic HFD alters the composition of seminal vesicle fluid and impairs seminal plasma capacity to elicit a favorable pro-tolerogenic immune response in females at conception.


Assuntos
Plasma/metabolismo , Sêmen/metabolismo , Adiposidade , Animais , Composição Corporal , Citocinas/metabolismo , Dieta Hiperlipídica , Feminino , Subpopulações de Linfócitos , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Camundongos Nus , Fenótipo , Gravidez , Prenhez , Isoformas de Proteínas , Reprodução , Sêmen/fisiologia , Espermatozoides/fisiologia , Linfócitos T/citologia , Linfócitos T Reguladores/imunologia , Fator de Crescimento Transformador beta1/metabolismo , Útero/patologia
13.
Hum Mutat ; 31(8): 889-900, 2010 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-20506206

RESUMO

The Aristaless-related homeobox gene (ARX) is one of the most frequently mutated genes in a spectrum of X-chromosome phenotypes with intellectual disability (ID) as their cardinal feature. To date, close to 100 families and isolated cases have been reported to carry 44 different mutations, the majority of these (59%) being a result of polyalanine tract expansions. At least 10 well-defined clinical entities, including Ohtahara, Partington, and Proud syndromes, X-linked infantile spasms, X-linked lissencephaly with ambiguous genitalia, X-linked myoclonic epilepsy and nonsyndromic intellectual disability have been ascertained from among the patients with ARX mutations. The striking intra- and interfamilial pleiotropy together with genetic heterogeneity (same clinical entities associated with different ARX mutations) are becoming a hallmark of ARX mutations. Although males are predominantly affected, some mutations associated with malformation phenotypes in males also show a phenotype in carrier females. Recent progress in the study of the effect of ARX mutations through sophisticated animal (mice) and cellular models begins to provide crucial insights into the molecular function of ARX and associated molecular pathology, thus guiding future inquiries into therapeutic interventions.


Assuntos
Proteínas de Homeodomínio/genética , Deficiência Intelectual/genética , Deficiência Intelectual/patologia , Fatores de Transcrição/genética , Proteína Duplacortina , Genes Ligados ao Cromossomo X , Estudos de Associação Genética , Humanos , Ilhotas Pancreáticas/metabolismo , Mutação/genética
14.
Twin Res Hum Genet ; 13(2): 168-78, 2010 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-20397747

RESUMO

Rett syndrome (RTT) is a severe neurodevelopmental disorder affecting females almost exclusively and is characterized by a wide spectrum of clinical manifestations. Mutations in the X-linked methyl-CpG-binding protein 2 (MECP2) gene have been found in up to 95% of classical RTT cases and a lesser proportion of atypical cases. Recently, mutations in another X-linked gene, CDKL5 (cyclin-dependent kinase-like 5) have been found to cause atypical RTT, in particular the early onset seizure (Hanefeld variant) and one female with autism. In this study we screened several cohorts of children for CDKL5 mutations, totaling 316 patients, including individuals with a clinical diagnosis of RTT but who were negative for MECP2 mutations (n=102), males with X-linked mental retardation (n=9), patients with West syndrome (n=52), patients with autism (n=59), patients with epileptic encephalopathy (n=33), patients with Aicardi syndrome (n=7) and other patients with intellectual disability with or without seizures (n=54). In all, seven polymorphic variations and four de novo mutations (c.586C>T [p.S196L]; c.58G>C [p.G20R]; c.2504delC [p.P835fs]; deletion of exons 1-3) were identified, and in all instances of the latter the clinical phenotype was that of an epileptic encephalopathy. These results suggest that pathogenic CDKL5 mutations are unlikely to be identified in the absence of severe early-onset seizures and highlight the importance of screening for large intragenic and whole gene deletions.


Assuntos
Transtornos Cognitivos/genética , Quinase 5 Dependente de Ciclina/genética , Mutação , Síndrome de Rett/diagnóstico , Síndrome de Rett/genética , Convulsões/genética , Sequência de Aminoácidos , Transtornos Cognitivos/diagnóstico , Transtornos Cognitivos/enzimologia , Estudos de Coortes , Quinase 5 Dependente de Ciclina/metabolismo , Feminino , Testes Genéticos , Humanos , Masculino , Dados de Sequência Molecular , Síndrome de Rett/enzimologia , Convulsões/diagnóstico , Convulsões/enzimologia
15.
Am J Med Genet A ; 149A(8): 1655-60, 2009 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-19606478

RESUMO

The Aristaless Related Homeobox (ARX) gene is a Q(50) paired homeobox gene. These genes are important regulators of essential events during vertebrate embryogenesis, including the development of the central and peripheral nervous system. Mutations in ARX have been identified in at least 82 different families and sporadic cases, and are responsible for at least 8 clinically distinct disorders. The recurrent 24 bp duplication (dup) mutation, c.429_452dup(24 bp), is the most frequent ARX mutation, which accounts for 45% of all cases reported to date. Here we report a novel de novo, familial dup mutation of 27 bp, c.430_456dup(27 bp), which involves the same region of the ARX gene in exon 2, as the dup24 bp mutation. The female progenitor of this dup27 bp allele exhibits mosaicism, likely resulting from a postmitotic de novo mutation event early in embryonic development. Three males with the dup27 bp mutation presented with infantile spasms, two of whom died early in life. Their phenotype appeared more severe, when compared to the spectrum of clinical presentations associated with the dup24 bp mutation. We propose that this might be at least partly due to the single, extra alanine residue (A) (21A in dup27 vs. 20A in dup24), which takes polyalanine tract 2 of ARX beyond the maximum, naturally occurring limit of 20A found in the human genome.


Assuntos
Pareamento de Bases/genética , Duplicação Gênica , Proteínas de Homeodomínio/genética , Mosaicismo/embriologia , Fatores de Transcrição/genética , Zigoto/metabolismo , Adulto , Sequência de Aminoácidos , Sequência de Bases , Pré-Escolar , Análise Mutacional de DNA , Características da Família , Feminino , Proteínas de Homeodomínio/química , Humanos , Lactente , Masculino , Dados de Sequência Molecular , Linhagem , Fenótipo , Síndrome , Fatores de Transcrição/química
16.
Am J Med Genet A ; 149A(7): 1482-6, 2009 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-19507262

RESUMO

Pathogenic variations of the ARX (aristaless-related homeobox) gene are associated with marked phenotypic pleiotropy. These phenotypes are X-linked neurological disorders that include brain and genital malformation and non-malformation syndromes. Typically, malformation phenotypes result from pathogenic variations that are predicted to truncate the ARX protein, or alter residues in the highly conserved homeodomain. While non-malformation phenotypes tend to be caused by pathogenic variations that are predicted to expand the first two polyalanine tracts of ARX, or alter residues outside of the homeodomain. The most common pathogenic variation of the ARX gene is a duplication of 24 bp, c.429_452 dup, which leads to an expansion of the second polyalanine tract of the ARX protein from 12 to 20 alanine residues. This pathogenic variation is associated with both sporadic and familial nonsyndromic mental retardation. Syndromic manifestations include mental retardation with hand dystonia (Partington syndrome), infantile spasms (West syndrome) and/or other epileptic seizures. Here, we report on a novel pathogenic variant of a tandem 33 bp duplication that is predicted to result in an expansion of polyalanine tract 2 in two brothers with mental retardation, epilepsy, dystonia, and the novel feature of intermittent hyperventilation. This pathogenic variation is predicted to result in a "non-homogeneous" polyalanine tract expansion that is longer than predicted expansion caused by the common 24 bp duplication. The location of the novel 33 bp duplication in the same region as the common 24 bp duplication supports this region as the ARX variation "hot spot."


Assuntos
Duplicação Gênica , Proteínas de Homeodomínio/genética , Deficiência Intelectual/genética , Irmãos , Fatores de Transcrição/genética , Adolescente , Sequência de Bases , Criança , Humanos , Masculino , Dados de Sequência Molecular , Linhagem , Síndrome
17.
Nutrients ; 11(9)2019 Sep 12.
Artigo em Inglês | MEDLINE | ID: mdl-31547309

RESUMO

Male obesity, which often co-presents with micronutrient deficiencies, is associated with sub-fertility. Here we investigate whether short-term dietary supplementation of micronutrients (zinc, selenium, lycopene, vitamins E and C, folic acid, and green tea extract) to obese mice for 12 days (designed to span the epididymal transit) could improve sperm quality and fetal outcomes. Five-week-old C57BL6 males were fed a control diet (CD, n = 24) or high fat diet (HFD, n = 24) for 10 weeks before allocation to the 12-day intervention of maintaining their original diets (CD, n = 12, HFD n = 12) or with micronutrient supplementation (CD + S, n = 12, HFD + S, n = 12). Measures of sperm quality (motility, morphology, capacitation, binding), sperm oxidative stress (DCFDA, MSR, and 8OHdG), early embryo development (2-cell cleavage, 8OHdG), and fetal outcomes were assessed. HFD + S males had reduced sperm intracellular reactive oxygen species (ROS) concentrations and 8OHdG lesions, which resulted in reduced 8OHdG lesions in the male pronucleus, increased 2-cell cleavage rates, and partial restoration of fetal weight similar to controls. Sub-fertility associated with male obesity may be restored with very short-term micronutrient supplementation that targets the timing of the transit of sperm through the epididymis, which is the developmental window where sperm are the most susceptible to oxidative damage.


Assuntos
Suplementos Nutricionais , Infertilidade Masculina/metabolismo , Micronutrientes/farmacologia , Obesidade/metabolismo , Estresse Oxidativo/efeitos dos fármacos , Animais , Dieta Hiperlipídica , Modelos Animais de Doenças , Desenvolvimento Embrionário/efeitos dos fármacos , Infertilidade Masculina/etiologia , Infertilidade Masculina/terapia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Obesos , Obesidade/complicações , Obesidade/terapia , Motilidade dos Espermatozoides/efeitos dos fármacos , Espermatozoides/efeitos dos fármacos
18.
Reprod Biomed Soc Online ; 9: 48-63, 2019 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-32021914

RESUMO

This study examined the nature and accuracy of information available across online platforms for couples trying to conceive. A consumer simulation-based investigation of English websites and social media (Facebook, Twitter, Instagram) was undertaken using common search terms identified in a pilot study. Claims about fertility and pregnancy health were then extracted from the results and analysed thematically. The accuracy of each claim was assessed independently by six fertility and conception experts, rated on a scale of 1 (not factual) to 4 (highly factual), with scores collated to produce a median rating. Claims with a median score < 3 were classified as inaccurate. The use of the terms 'trying to conceive' and '#TTC' were common identifiers on online platforms. Claims were extracted predominantly from websites (n = 89) rather than social media, with Twitter and Instagram comprising commercial elements and Facebook focused on community-based support. Thematic analysis revealed three major themes among the claims across all platforms: conception behaviour and monitoring, lifestyle and exposures, and medical. Fact-checking by the experts revealed that 40% of the information assessed was inaccurate, and that inaccuracies were more likely to be present in the conception behaviour and monitoring advice, the topics most amenable to modification. Since online information is a readily accessible and commonly utilized resource, there is opportunity for improved dissemination of evidence-based material to reach interested couples. Further cross-disciplinary and consumer-based research, such as a user survey, is required to understand how best to provide the 'trying to conceive' community with accurate information.

19.
Hum Mutat ; 28(2): 207-8, 2007 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-17221867

RESUMO

The EuroMRX family cohort consists of about 400 families with non-syndromic and 200 families with syndromic X-linked mental retardation (XLMR). After exclusion of Fragile X (Fra X) syndrome, probands from these families were tested for mutations in the coding sequence of 90 known and candidate XLMR genes. In total, 73 causative mutations were identified in 21 genes. For 42% of the families with obligate female carriers, the mental retardation phenotype could be explained by a mutation. There was no difference between families with (lod score >2) or without (lod score <2) significant linkage to the X chromosome. For families with two to five affected brothers (brother pair=BP families) only 17% of the MR could be explained. This is significantly lower (P=0.0067) than in families with obligate carrier females and indicates that the MR in about 40% (17/42) of the BP families is due to a single genetic defect on the X chromosome. The mutation frequency of XLMR genes in BP families is lower than can be expected on basis of the male to female ratio of patients with MR or observed recurrence risks. This might be explained by genetic risk factors on the X chromosome, resulting in a more complex etiology in a substantial portion of XLMR patients. The EuroMRX effort is the first attempt to unravel the molecular basis of cognitive dysfunction by large-scale approaches in a large patient cohort. Our results show that it is now possible to identify 42% of the genetic defects in non-syndromic and syndromic XLMR families with obligate female carriers.


Assuntos
Deficiência Intelectual Ligada ao Cromossomo X/genética , Mutação , Estudos de Coortes , Análise Mutacional de DNA , Feminino , Genes , Humanos , Escore Lod , Masculino , Deficiência Intelectual Ligada ao Cromossomo X/diagnóstico , Fenótipo
20.
Nutrients ; 9(2)2017 Feb 09.
Artigo em Inglês | MEDLINE | ID: mdl-28208792

RESUMO

Paternal obesity programs metabolic syndrome in offspring. Low-impact exercise in obese  males improves the metabolic health of female offspring, however whether this occurred in male  offspring remained unknown. C57BL/6NHsd (Harlan) mice were fed a control diet (CD; 6% fat, n =  7) or a high-fat diet (HFD; 21% fat, n = 16) for 18 weeks. After 9 weeks, HFD-fed mice either remained  sedentary (HH, n = 8) or undertook low-moderate exercise (HE, n = 8) for another 9 weeks. Male  offspring were assessed for glucose/insulin tolerance, body composition, plasma lipids, pancreatic  islet cell morphology and microRNA expression. Founder HH induced glucose intolerance, insulin  insensitivity, and hyperlipidaemia in male offspring (p < 0.05). Metabolic health was fully restored  in male offspring by founder exercise to control levels. Founder HH reduced pancreatic ß-cell area  and islet cell size in male offspring, and altered the expression of 13 pancreatic microRNAs (p <  0.05). Founder HE led to partial restoration of pancreatic islet cell morphology and the expression  of two pancreatic microRNAs (let7d-5p, 194-5p) in male offspring. Founder HE reduced male  offspring adiposity, increased muscle mass, reduced plasma free fatty acids (FFAs), and further  altered pancreatic microRNAs (35 vs. HH; 32 vs. CD) (p < 0.05). Low-impact exercise in obese fathers  prior to conception, without dietary change, may be a viable intervention strategy to reduce the illeffects of obesity-induced paternal programming in male offspring.


Assuntos
Ilhotas Pancreáticas/citologia , MicroRNAs/metabolismo , Obesidade/fisiopatologia , Obesidade/terapia , Condicionamento Físico Animal/métodos , Animais , Glicemia/fisiologia , Reprogramação Celular , Técnicas de Reprogramação Celular/métodos , Dieta Hiperlipídica/efeitos adversos , Pai , Ácidos Graxos não Esterificados/sangue , Feminino , Insulina/fisiologia , Ilhotas Pancreáticas/fisiopatologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Obesidade/genética , Herança Paterna
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA