Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 53
Filtrar
1.
Artigo em Inglês | MEDLINE | ID: mdl-38432776

RESUMO

DNA alterations in gametes, which may occur either spontaneously or as a result of exposure to genotoxicants, can lead to constitutional chromosomal anomalies in the offspring. Alcohol is an established genotoxicant. The goal of this hypothesis-testing longitudinal cohort study was to evaluate the effect of significant/sustained maternal alcohol exposure on clinically diagnosed constitutional chromosomal anomalies among children diagnosed with fetal alcohol syndrome (FAS). De-identified eligibility and claim healthcare records, prospectively generated from the 1990-2012 Florida Medicaid system within the Independent Healthcare Research Database (IHRD), were analyzed. Children examined were continuously eligible with ≥ 8 outpatient office visits during the 96-month period following birth. Among these children, 377 were diagnosed with FAS and 137,135 were not. The incidence rate of chromosomal anomalies involving segregation (trisomy 13, 18, or 21, n = 625), microdeletions (microdeletion syndromes, n = 39), and point mutations (sickle-cell anemia/cystic fibrosis, n = 2570) were examined using frequency risk ratio (RR) and logistic regression (adjusted odds ratio (aOR) for sex, race, residence, socioeconomic/environmental exposure status, and birth date) models. The incidence rates of chromosomal anomalies involving segregation (RR=5.92, aOR=5.85) and microdeletions (RR=41.6, aOR=34.1) were significantly increased in the FAS cohort as compared to the non-diagnosed cohort, but there was no difference in the incidence rate of point mutations (RR=1.14, aOR=1.29). Maternal toxicant exposure should be considered in the etiology of constitutional chromosomal anomaly in offspring.


Assuntos
Transtornos Cromossômicos , Transtornos do Espectro Alcoólico Fetal , Criança , Estados Unidos , Feminino , Gravidez , Humanos , Estudos Longitudinais , Transtornos do Espectro Alcoólico Fetal/epidemiologia , Transtornos do Espectro Alcoólico Fetal/genética , Estudos de Coortes , Aberrações Cromossômicas
2.
Free Radic Biol Med ; 208: 272-284, 2023 11 01.
Artigo em Inglês | MEDLINE | ID: mdl-37541454

RESUMO

The breast cancer 1 (Brca1) susceptibility gene regulates the repair of reactive oxygen species (ROS)-mediated DNA damage, which is implicated in neurodevelopmental disorders. Alcohol (ethanol, EtOH) exposure during pregnancy causes fetal alcohol spectrum disorders (FASD), including abnormal brain function, associated with enhanced ROS-initiated DNA damage. Herein, oxidative DNA damage in fetal brains and neurodevelopmental disorders were enhanced in saline-exposed +/- vs. +/+ Brca1 littermates. A single EtOH exposure during gestation further enhanced oxidative DNA damage, altered the expression of developmental/DNA damage response genes in fetal brains, and resulted in neurodevelopmental disorders, all of which were BRCA1-dependent. Pretreatment with the ROS inhibitor phenylbutylnitrone (PBN) blocked DNA damage and some neurodevelopmental disorders in both saline- and EtOH-exposed progeny, corroborating a ROS-dependent mechanism. Fetal BRCA1 protects against altered gene expression and neurodevelopmental disorders caused by both physiological and EtOH-enhanced levels of ROS formation. BRCA1 deficiencies may enhance the risk for FASD.


Assuntos
Transtornos do Espectro Alcoólico Fetal , Neoplasias , Transtornos do Neurodesenvolvimento , Gravidez , Humanos , Feminino , Espécies Reativas de Oxigênio/metabolismo , Etanol/toxicidade , Transtornos do Espectro Alcoólico Fetal/genética , Transtornos do Espectro Alcoólico Fetal/metabolismo , Transtornos do Neurodesenvolvimento/induzido quimicamente , Transtornos do Neurodesenvolvimento/genética , Expressão Gênica , Proteína BRCA1/genética
3.
Cells ; 12(2)2023 01 15.
Artigo em Inglês | MEDLINE | ID: mdl-36672262

RESUMO

Fetal alcohol spectrum disorders (FASDs) are associated with systemic inflammation and neurodevelopmental abnormalities. Several candidate genes were found to be associated with fetal alcohol exposure (FAE)-associated behaviors, but a sex-specific complete transcriptomic analysis was not performed at the adult stage. Recent studies have shown that they are regulated at the developmental stage. However, the sex-specific role of RNA in FAE offspring brain development and function has not been studied yet. Here, we carried out the first systematic RNA profiling by utilizing a high-throughput transcriptomic (RNA-seq) approach in response to FAE in the brain cortex of male and female offspring at adulthood (P60). Our RNA-seq data analysis suggests that the changes in RNA expression in response to FAE are marked sex-specific. We show that the genes Muc3a, Pttg1, Rec8, Clcnka, Capn11, and pnp2 exhibit significantly higher expression in the male offspring than in the female offspring at P60. FAE female mouse brain sequencing data also show an increased expression of Eno1, Tpm3, and Pcdhb2 compared to male offspring. We performed a pathway analysis using a commercial software package (Ingenuity Pathway Analysis). We found that the sex-specific top regulator genes (Rictor, Gaba, Fmri, Mlxipl) are highly associated with eIF2 (translation initiation), synaptogenesis (the formation of synapses between neurons in the nervous system), sirtuin (metabolic regulation), and estrogen receptor (involved in obesity, aging, and cancer) signaling. Taken together, our transcriptomic results demonstrate that FAE differentially alters RNA expression in the adult brain in a sex-specific manner.


Assuntos
Etanol , Transtornos do Espectro Alcoólico Fetal , Gravidez , Animais , Camundongos , Humanos , Masculino , Feminino , Etanol/metabolismo , Perfilação da Expressão Gênica , Transtornos do Espectro Alcoólico Fetal/genética , Córtex Cerebral/metabolismo , Fatores de Transcrição/metabolismo , RNA
4.
Alcohol Clin Exp Res ; 45(2): 329-337, 2021 02.
Artigo em Inglês | MEDLINE | ID: mdl-33296097

RESUMO

BACKGROUND: Fetal alcohol spectrum disorder (FASD) is characterized by severe clinical impairment, considerable social burden, and high mortality and morbidity, which are due to various malformations, sepsis, and cancer. As >50% of deaths from FASD occur during the first year of life, we hypothesized that there is the acceleration of biological aging in FASD. Several recent studies have established genome-wide DNA methylation (DNAm) profiles as "epigenetic clocks" that can estimate biological aging, and FASD has been associated with differential DNAm patterns. Therefore, we tested this hypothesis using epigenetic clocks. METHODS: We investigated 5 DNAm-based measures of epigenetic age (HorvathAge, HannumAge, SkinBloodAge, PhenoAge, and GrimAge) and telomere length (DNAmTL) using 4 independent publicly available DNAm datasets; 2 datasets were derived from buccal epithelium, and the other 2 datasets were derived from peripheral blood. RESULTS: Compared with controls, children with FASD exhibited an acceleration of GrimAge in 1 buccal and 2 blood datasets. No significant difference was found in other DNAm ages and DNAmTL. Meta-analyses showed a significant acceleration of GrimAge in the blood samples but not in the buccal samples. CONCLUSIONS: This study provides novel evidence regarding accelerated epigenetic aging in children with FASD.


Assuntos
Envelhecimento/genética , Envelhecimento/metabolismo , Epigênese Genética/fisiologia , Transtornos do Espectro Alcoólico Fetal/genética , Transtornos do Espectro Alcoólico Fetal/metabolismo , Adolescente , Criança , Estudos de Coortes , Bases de Dados Factuais , Feminino , Humanos , Masculino , Mucosa Bucal/metabolismo , Gravidez
5.
PLoS One ; 15(11): e0242276, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-33196678

RESUMO

Alcohol (ethanol, EtOH) consumption during pregnancy can result in fetal alcohol spectrum disorders (FASDs), which are characterized by prenatal and postnatal growth restriction and craniofacial dysmorphology. Recently, cell-derived extracellular vesicles, including exosomes and microvesicles containing several species of RNAs (exRNAs), have emerged as a mechanism of cell-to-cell communication. However, EtOH's effects on the biogenesis and function of non-coding exRNAs during fetal development have not been explored. Therefore, we studied the effects of maternal EtOH exposure on the composition of exosomal RNAs in the amniotic fluid (AF) using rat fetal alcohol exposure (FAE) model. Through RNA-Seq analysis we identified and verified AF exosomal miRNAs with differential expression levels specifically associated with maternal EtOH exposure. Uptake of purified FAE AF exosomes by rBMSCs resulted in significant alteration of molecular markers associated with osteogenic differentiation of rBMSCs. We also determined putative functional roles for AF exosomal miRNAs (miR-199a-3p, miR-214-3p and let-7g) that are dysregulated by FAE in osteogenic differentiation of rBMSCs. Our results demonstrate that FAE alters AF exosomal miRNAs and that exosomal transfer of dysregulated miRNAs has significant molecular effects on stem cell regulation and differentiation. Our results further suggest the usefulness of assessing molecular alterations in AF exRNAs to study the mechanisms of FAE teratogenesis that should be further investigated by using an in vivo model.


Assuntos
Líquido Amniótico/metabolismo , Diferenciação Celular/efeitos dos fármacos , Etanol/farmacologia , Exossomos/metabolismo , MicroRNAs/metabolismo , Líquido Amniótico/efeitos dos fármacos , Animais , Células Cultivadas , Modelos Animais de Doenças , Feminino , Transtornos do Espectro Alcoólico Fetal/genética , Transtornos do Espectro Alcoólico Fetal/metabolismo , Transtornos do Espectro Alcoólico Fetal/patologia , Células-Tronco Mesenquimais/citologia , Células-Tronco Mesenquimais/efeitos dos fármacos , Células-Tronco Mesenquimais/metabolismo , Osteogênese/efeitos dos fármacos , Gravidez , Ratos , Ratos Sprague-Dawley
6.
J Neurodev Disord ; 12(1): 15, 2020 05 16.
Artigo em Inglês | MEDLINE | ID: mdl-32416732

RESUMO

BACKGROUND: Fetal alcohol spectrum disorders (FASD) are common, seen in 1-5% of the population in the USA and Canada. Children diagnosed with FASD are not likely to remain with their biological parents, facing early maternal separation and foster placements throughout childhood. METHODS: We model FASD in mice via prenatal alcohol exposure and further induce early life stress through maternal separation. We use RNA-seq followed by clustering of expression profiles through weighted gene co-expression network analysis (WGCNA) to analyze transcriptomic changes that result from the treatments. We use reverse transcription qPCR to validate these changes in the mouse hippocampus. RESULTS: We report an association between adult hippocampal gene expression and prenatal ethanol exposure followed by postnatal separation stress that is related to behavioral changes. Expression profile clustering using WGCNA identifies a set of transcripts, module 19, associated with anxiety-like behavior (r = 0.79, p = 0.002) as well as treatment group (r = 0.68, p = 0.015). Genes in this module are overrepresented by genes involved in transcriptional regulation and other pathways related to neurodevelopment. Interestingly, one member of this module, Polr2a, polymerase (RNA) II (DNA directed) polypeptide A, is downregulated by the combination of prenatal ethanol and postnatal stress in an RNA-Seq experiment and qPCR validation (q = 2e-12, p = 0.004, respectively). CONCLUSIONS: Together, transcriptional control in the hippocampus is implicated as a potential underlying mechanism leading to anxiety-like behavior via environmental insults. Further research is required to elucidate the mechanism involved and use this insight towards early diagnosis and amelioration strategies involving children born with FASD.


Assuntos
Transtornos do Espectro Alcoólico Fetal/genética , Perfilação da Expressão Gênica , Hipocampo/metabolismo , Privação Materna , Processamento Pós-Transcricional do RNA , Consumo de Bebidas Alcoólicas/genética , Animais , Animais Recém-Nascidos , Ansiedade , Canadá , Modelos Animais de Doenças , Etanol/administração & dosagem , Etanol/metabolismo , Feminino , Transtornos do Espectro Alcoólico Fetal/metabolismo , Regulação da Expressão Gênica , Camundongos , Gravidez , Efeitos Tardios da Exposição Pré-Natal/genética , Efeitos Tardios da Exposição Pré-Natal/metabolismo , Transcriptoma
7.
Epigenomics ; 11(7): 767-785, 2019 05.
Artigo em Inglês | MEDLINE | ID: mdl-30873861

RESUMO

Aim: Fetal alcohol spectrum disorder (FASD) involves prenatal growth delay, impaired facial and CNS development and causes severe clinical, social-economic burdens. Here, we aim to detect DNA-methylation aberrations associated with FASD and potential FASD diagnostic and prognostic biomarkers. Patients & methods: The FASD diagnosis was established according to golden-standard protocols in a discovery and independent replication cohort. Genome-wide differential methylation association and replication analyses were performed. Results: We identified several loci that were robustly associated with FASD or one of its sub phenotypes. Our findings were evaluated using previously reported genome-wide surveys. Conclusion: We have detected robust FASD associated differentially methylated positions and differentially methylated regions for FASD in general and for FASD subphenotypes, in other words on growth delay, impaired facial and CNS development.


Assuntos
Metilação de DNA , Transtornos do Espectro Alcoólico Fetal/diagnóstico , Adolescente , Biomarcadores/metabolismo , Estudos de Casos e Controles , Criança , Pré-Escolar , Proteínas Associadas à Distrofina/genética , Feminino , Transtornos do Espectro Alcoólico Fetal/genética , Loci Gênicos , Estudo de Associação Genômica Ampla , Humanos , Lactente , Recém-Nascido , Masculino , Neuropeptídeos/genética , Proteínas Nucleares/genética , Fenótipo , Prognóstico , Receptores do Fator de Necrose Tumoral/genética , Proteína Gli2 com Dedos de Zinco/genética
8.
Exp Dermatol ; 27(1): 91-93, 2018 01.
Artigo em Inglês | MEDLINE | ID: mdl-28833556

RESUMO

The precise molecular basis of retinoid embryopathy is yet unknown. This hypothesis predicts that isotretinoin (13-cis retinoic acid), the prodrug of all-trans retinoic acid (ATRA), exaggerates neural crest cell (NCC) apoptosis via upregulation of the pro-apoptotic transcription factor p53, the guardian of the genome. Increased p53 signalling is associated with Treacher Collins-, CHARGE- and fetal alcohol syndrome, which exhibit dysmorphic craniofacial features resembling retinoid embryopathy. In addition, developmental studies of NCC homeostasis in the zebrafish support the pivotal role of p53. Translational evidence implies that isotretinoin-stimulated overactivation of p53 during embryogenesis represents the molecular basis of isotretinoin's teratogenicity.


Assuntos
Isotretinoína/toxicidade , Teratogênicos/toxicidade , Proteína Supressora de Tumor p53/metabolismo , Animais , Apoptose , Síndrome CHARGE/genética , Modelos Animais de Doenças , Desenvolvimento Embrionário , Transtornos do Espectro Alcoólico Fetal/genética , Homeostase , Humanos , Disostose Mandibulofacial/genética , Crista Neural/citologia , Transdução de Sinais , Pesquisa Translacional Biomédica , Tretinoína/química , Regulação para Cima , Peixe-Zebra
9.
PLoS One ; 12(7): e0180873, 2017.
Artigo em Inglês | MEDLINE | ID: mdl-28723918

RESUMO

Prenatal ethanol exposure can produce structural and functional deficits in the brain and result in Fetal Alcohol Spectrum Disorder (FASD). In rodent models acute exposure to a high concentration of alcohol causes increased apoptosis in the developing brain. A single causal molecular switch that signals for this increase in apoptosis has yet to be identified. The protein p53 has been suggested to play a pivotal role in enabling cells to engage in pro-apoptotic processes, and thus figures prominently as a hub molecule in the intracellular cascade of responses elicited by alcohol exposure. In the present study we examined the effect of ethanol-induced cellular and molecular responses in primary somatosensory cortex (SI) and hippocampus of 7-day-old wild-type (WT) and p53-knockout (KO) mice. We quantified apoptosis by active caspase-3 immunohistochemistry and ApopTag™ labeling, then determined total RNA expression levels in laminae of SI and hippocampal subregions. Immunohistochemical results confirmed increased incidence of apoptotic cells in both regions in WT and KO mice following ethanol exposure. The lack of p53 was not protective in these brain regions. Molecular analyses revealed a heterogeneous response to ethanol exposure that varied depending on the subregion, and which may go undetected using a global approach. Gene network analyses suggest that the presence or absence of p53 alters neuronal function and synaptic modifications following ethanol exposure, in addition to playing a classic role in cell cycle signaling. Thus, p53 may function in a way that underlies the intellectual and behavioral deficits observed in FASD.


Assuntos
Etanol/farmacologia , Transtornos do Espectro Alcoólico Fetal/genética , Expressão Gênica/efeitos dos fármacos , Proteína Supressora de Tumor p53/genética , Animais , Apoptose/efeitos dos fármacos , Caspase 3/metabolismo , Modelos Animais de Doenças , Transtornos do Espectro Alcoólico Fetal/metabolismo , Redes Reguladoras de Genes , Hipocampo/efeitos dos fármacos , Hipocampo/metabolismo , Camundongos , Camundongos Knockout , Córtex Somatossensorial/efeitos dos fármacos , Córtex Somatossensorial/metabolismo , Proteína Supressora de Tumor p53/metabolismo
10.
J Psychiatry Neurosci ; 42(2): 113-121, 2017 03.
Artigo em Inglês | MEDLINE | ID: mdl-28234207

RESUMO

BACKGROUND: Attention-deficit/hyperactivity disorder (ADHD) is often accompanied by impaired response inhibition; both have been associated with aberrant dopamine signalling. Given that prenatal exposure to alcohol or smoking is known to affect dopamine-rich brain regions, we hypothesized that individuals carrying the ADHD risk alleles of the dopamine receptor D4 (DRD4) and dopamine transporter (DAT1) genes may be especially sensitive to their effects. METHODS: Functional MRI data, information on prenatal adversities and genetic data were available for 239 adolescents and young adults participating in the multicentre ADHD cohort study NeuroIMAGE (average age 17.3 yr). We analyzed the effects of DRD4 and DAT1, prenatal exposure to alcohol and smoking and their interactions on ADHD severity, response inhibition and neural activity. RESULTS: We found no significant gene × environment interaction effects. We did find that the DRD4 7-repeat allele was associated with less superior frontal and parietal brain activity and with greater activity in the frontal pole and occipital cortex. Prenatal exposure to smoking was also associated with lower superior frontal activity, but with greater activity in the parietal lobe. Further, those exposed to alcohol had more activity in the lateral orbitofrontal cortex, and the DAT1 risk variant was associated with lower cerebellar activity. LIMITATIONS: Retrospective reports of maternal substance use and the cross-sectional study design restrict causal inference. CONCLUSION: While we found no evidence of gene × environment interactions, the risk factors under investigation influenced activity of brain regions associated with response inhibition, suggesting they may add to problems with inhibiting behaviour.


Assuntos
Transtorno do Deficit de Atenção com Hiperatividade/fisiopatologia , Encéfalo/fisiopatologia , Proteínas da Membrana Plasmática de Transporte de Dopamina/genética , Inibição Psicológica , Efeitos Tardios da Exposição Pré-Natal , Receptores de Dopamina D4/genética , Adolescente , Transtorno do Deficit de Atenção com Hiperatividade/diagnóstico por imagem , Transtorno do Deficit de Atenção com Hiperatividade/genética , Transtorno do Deficit de Atenção com Hiperatividade/psicologia , Encéfalo/diagnóstico por imagem , Estudos Transversais , Feminino , Transtornos do Espectro Alcoólico Fetal/diagnóstico por imagem , Transtornos do Espectro Alcoólico Fetal/genética , Transtornos do Espectro Alcoólico Fetal/fisiopatologia , Transtornos do Espectro Alcoólico Fetal/psicologia , Seguimentos , Interação Gene-Ambiente , Predisposição Genética para Doença , Humanos , Imageamento por Ressonância Magnética , Masculino , Gravidez , Efeitos Tardios da Exposição Pré-Natal/diagnóstico por imagem , Efeitos Tardios da Exposição Pré-Natal/genética , Efeitos Tardios da Exposição Pré-Natal/fisiopatologia , Efeitos Tardios da Exposição Pré-Natal/psicologia , Estudos Retrospectivos , Índice de Gravidade de Doença , Fumar/efeitos adversos
11.
Hepatology ; 65(2): 678-693, 2017 02.
Artigo em Inglês | MEDLINE | ID: mdl-28114741

RESUMO

Exposure to genotoxins such as ethanol-derived acetaldehyde leads to DNA damage and liver injury and promotes the development of cancer. We report here a major role for the transforming growth factor ß/mothers against decapentaplegic homolog 3 adaptor ß2-Spectrin (ß2SP, gene Sptbn1) in maintaining genomic stability following alcohol-induced DNA damage. ß2SP supports DNA repair through ß2SP-dependent activation of Fanconi anemia complementation group D2 (Fancd2), a core component of the Fanconi anemia complex. Loss of ß2SP leads to decreased Fancd2 levels and sensitizes ß2SP mutants to DNA damage by ethanol treatment, leading to phenotypes that closely resemble those observed in animals lacking both aldehyde dehydrogenase 2 and Fancd2 and resemble human fetal alcohol syndrome. Sptbn1-deficient cells are hypersensitive to DNA crosslinking agents and have defective DNA double-strand break repair that is rescued by ectopic Fancd2 expression. Moreover, Fancd2 transcription in response to DNA damage/transforming growth factor ß stimulation is regulated by the ß2SP/mothers against decapentaplegic homolog 3 complex. CONCLUSION: Dysfunctional transforming growth factor ß/ß2SP signaling impacts the processing of genotoxic metabolites by altering the Fanconi anemia DNA repair pathway. (Hepatology 2017;65:678-693).


Assuntos
Proteína do Grupo de Complementação D2 da Anemia de Fanconi/genética , Instabilidade Genômica/genética , Prenhez , Espectrina/genética , Fator de Crescimento Transformador beta2/genética , Análise de Variância , Animais , Animais Recém-Nascidos , Dano ao DNA/genética , Reparo do DNA/genética , Etanol/farmacologia , Feminino , Transtornos do Espectro Alcoólico Fetal/genética , Transtornos do Espectro Alcoólico Fetal/patologia , Humanos , Imuno-Histoquímica , Peroxidação de Lipídeos/genética , Camundongos , Camundongos Transgênicos , Gravidez , Reação em Cadeia da Polimerase em Tempo Real/métodos , Transdução de Sinais
12.
PLoS One ; 11(11): e0165081, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-27828986

RESUMO

Fetal alcohol spectrum disorders (FASD) are difficult to diagnose since many heavily exposed infants, at risk for intellectual disability, do not exhibit craniofacial dysmorphology or growth deficits. Consequently, there is a need for biomarkers that predict disability. In both animal models and human studies, alcohol exposure during pregnancy resulted in significant alterations in circulating microRNAs (miRNAs) in maternal blood. In the current study, we asked if changes in plasma miRNAs in alcohol-exposed pregnant mothers, either alone or in conjunction with other clinical variables, could predict infant outcomes. Sixty-eight pregnant women at two perinatal care clinics in western Ukraine were recruited into the study. Detailed health and alcohol consumption histories, and 2nd and 3rd trimester blood samples were obtained. Birth cohort infants were assessed by a geneticist and classified as unexposed (UE), heavily prenatally exposed and affected (HEa) or heavily exposed but apparently unaffected (HEua). MiRNAs were assessed in plasma samples using qRT-PCR arrays. ANOVA models identified 11 miRNAs that were all significantly elevated in maternal plasma from the HEa group relative to HEua and UE groups. In a random forest analysis classification model, a combination of high variance miRNAs, smoking history and socioeconomic status classified membership in HEa and UE groups, with a misclassification rate of 13%. The RFA model also classified 17% of the HEua group as UE-like, whereas 83% were HEa-like, at least at one stage of pregnancy. Collectively our data indicate that maternal plasma miRNAs predict infant outcomes, and may be useful to classify difficult-to-diagnose FASD subpopulations.


Assuntos
Consumo de Bebidas Alcoólicas/efeitos adversos , Transtornos do Espectro Alcoólico Fetal/genética , MicroRNAs/genética , Efeitos Tardios da Exposição Pré-Natal/genética , Adulto , Animais , Biomarcadores/sangue , Estudos de Coortes , Feminino , Transtornos do Espectro Alcoólico Fetal/diagnóstico , Transtornos do Espectro Alcoólico Fetal/etiologia , Humanos , Lactente , Recém-Nascido , Masculino , MicroRNAs/sangue , MicroRNAs/classificação , Assistência Perinatal , Valor Preditivo dos Testes , Gravidez , Efeitos Tardios da Exposição Pré-Natal/diagnóstico , Efeitos Tardios da Exposição Pré-Natal/etiologia , Prognóstico , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Sensibilidade e Especificidade , Ucrânia , Adulto Jovem
13.
PLoS One ; 11(3): e0149976, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-26930486

RESUMO

Fetal alcohol spectrum disorder is a collective term representing fetal abnormalities associated with maternal alcohol consumption. Prenatal alcohol exposure and related anomalies are well characterized, but the molecular mechanism behind this phenomenon is not well characterized. In this present study, our aim is to profile important genes that regulate cellular development during fetal development. Human embryonic carcinoma cells (NCCIT) are cultured to form embryoid bodies and then treated in the presence and absence of ethanol (50 mM). We employed RNA sequencing to profile differentially expressed genes in the ethanol-treated embryoid bodies from NCCIT vs. EB, NCCIT vs. EB+EtOH and EB vs. EB+EtOH data sets. A total of 632, 205 and 517 differentially expressed genes were identified from NCCIT vs. EB, NCCIT vs. EB+EtOH and EB vs. EB+EtOH, respectively. Functional annotation using bioinformatics tools reveal significant enrichment of differential cellular development and developmental disorders. Furthermore, a group of 42, 15 and 35 transcription factor-encoding genes are screened from all of the differentially expressed genes obtained from NCCIT vs. EB, NCCIT vs. EB+EtOH and EB vs. EB+EtOH, respectively. We validated relative gene expression levels of several transcription factors from these lists by quantitative real-time PCR. We hope that our study substantially contributes to the understanding of the molecular mechanism underlying the pathology of alcohol-mediated anomalies and ease further research.


Assuntos
Corpos Embrioides/metabolismo , Etanol/farmacologia , Perfilação da Expressão Gênica , Regulação da Expressão Gênica no Desenvolvimento/efeitos dos fármacos , Análise de Sequência de RNA/métodos , Técnicas de Cultura de Células/métodos , Diferenciação Celular/efeitos dos fármacos , Diferenciação Celular/genética , Depressores do Sistema Nervoso Central/farmacologia , Células-Tronco de Carcinoma Embrionário/metabolismo , Feminino , Transtornos do Espectro Alcoólico Fetal/genética , Humanos , Gravidez , Efeitos Tardios da Exposição Pré-Natal/genética , Reação em Cadeia da Polimerase Via Transcriptase Reversa
14.
Addict Biol ; 21(1): 23-34, 2016 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-25581210

RESUMO

Alcohol exposure during fetal and early postnatal development can lead to an increased incidence of later life adult-onset diseases. Examples include central nervous system dysfunction, depression, anxiety, hyperactivity, and an inability to deal with stressful situations, increased infection and cancer. Direct effects of alcohol leading to developmental abnormalities often involve epigenetic modifications of genes that regulate cellular functions. Epigenetic marks carried over from the parents are known to undergo molecular programming events that happen early in embryonic development by a wave of DNA demethylation, which leaves the embryo with a fresh genomic composition. The proopiomelanocortin (Pomc) gene controls neuroendocrine-immune functions and is imprinted by fetal alcohol exposure. Recently, this gene has been shown to be hypermethylated through three generations. Additionally, the alcohol epigenetic marks on the Pomc gene are maintained in the male but not in the female germline during this transgenerational transmission. These data suggest that the male-specific chromosome might be involved in transmitting alcohol epigenetic marks through multiple generations.


Assuntos
Epigênese Genética/genética , Transtornos do Espectro Alcoólico Fetal/genética , Exposição Paterna , Efeitos Tardios da Exposição Pré-Natal/genética , Pró-Opiomelanocortina/genética , Espermatozoides/metabolismo , Animais , Metilação de DNA/genética , Feminino , Regulação da Expressão Gênica , Células Germinativas , Humanos , Masculino , Gravidez
15.
Asian J Androl ; 17(6): 985-90, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-26262775

RESUMO

Although paternal ethanol (EtOH) abuse has been shown to affect the growth and behavior of offspring, the exact molecular and mechanistic basis remains largely unclear. Methylation alterations in imprinted genes may be related to well-documented teratogenic effects of ethanol. Here we show that chronic paternal ethanol exposure increases the susceptibility to abnormal behavior in offspring through male game epigenetic alteration. In our study, different doses of ethanol (0, 1.1, 3.3 g kg-1 ) were administered intra-gastrically to male mice and decreased sperm motility was found in the highest ethanol-exposed group compared with the controls. Data also showed a dose-dependent increase in deaf mice of the paternally ethanol-exposed groups. The methylation of H19, Peg3, Ndn and Snrpn was assessed in paternal spermatozoa and in the cerebral cortices of deaf mice. EtOH affected methylation of Peg3 (CpG 3, 7 and 9) in paternal spermatozoa and in the cerebral cortices of deaf mice, but the level of mRNA expression did not change, suggesting that other gene regulation may be involved in these processes. Overall, chronic paternal ethanol exposure could alter the methylation of imprinted genes in sire spermatozoa that could also be passed on to offspring, giving rise to developmental disorders. Our results provide possible epigenetic evidence for a paternal ethanol exposure contribution to Fetal Alcohol Syndrome (FAS).


Assuntos
Anti-Infecciosos Locais/farmacologia , Córtex Cerebral/efeitos dos fármacos , Metilação de DNA/efeitos dos fármacos , Etanol/farmacologia , Impressão Genômica/efeitos dos fármacos , Audição/efeitos dos fármacos , Exposição Paterna , RNA Mensageiro/efeitos dos fármacos , Espermatozoides/efeitos dos fármacos , Animais , Córtex Cerebral/metabolismo , Surdez/genética , Transtornos do Espectro Alcoólico Fetal/genética , Expressão Gênica/efeitos dos fármacos , Audição/genética , Fatores de Transcrição Kruppel-Like/efeitos dos fármacos , Fatores de Transcrição Kruppel-Like/genética , Masculino , Camundongos , Proteínas do Tecido Nervoso/efeitos dos fármacos , Proteínas do Tecido Nervoso/genética , Proteínas Nucleares/efeitos dos fármacos , Proteínas Nucleares/genética , RNA Longo não Codificante/efeitos dos fármacos , RNA Longo não Codificante/genética , RNA Mensageiro/metabolismo , Espermatozoides/metabolismo , Proteínas Centrais de snRNP/efeitos dos fármacos , Proteínas Centrais de snRNP/genética
16.
Pediatr Rev ; 36(1): 14-21, 2015 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-25554107

RESUMO

Epigenetic mechanisms are external modifications of DNA that cause changes in gene function and are involved in many diseases. Specific examples of pediatric diseases with a known or suspected epigenetic component include Beckwith-Wiedemann syndrome, childhood leukemia, allergies, asthma, fetal alcohol spectrum disorders, childhood obesity, and type 2 diabetes mellitus. Currently, epigenetically active treatments are being used to treat childhood leukemia. Potential epigenetically active treatments and preventive regimens are under study for other diseases. Pediatricians need to be aware of the epigenetic basis of disease to help inform clinical decision making in the future.


Assuntos
Epigênese Genética/fisiologia , Predisposição Genética para Doença , Síndrome de Beckwith-Wiedemann/genética , Criança , Transtornos do Espectro Alcoólico Fetal/genética , Humanos , Leucemia Mieloide Aguda/genética
17.
Biochem Biophys Res Commun ; 453(3): 338-44, 2014 Oct 24.
Artigo em Inglês | MEDLINE | ID: mdl-25264105

RESUMO

Fetal alcohol syndrome (FAS) is a birth defect due to maternal alcohol consumption during pregnancy. Because mesenchymal stem cells (MSCs) are the main somatic stem cells in adults and may contribute to tissue homeostasis and repair in adulthood, we investigated whether early life ethanol exposure affects MSCs and contributes to the propensity for disease onset in later life. Using a rodent model of FAS, we found that ethanol exposure (5.25g/kg/day) from postnatal days 4 to 9 in rat pups (mimic of human third trimester) caused long-term anomalies in bone marrow-derived MSCs. MSCs isolated from ethanol-exposed animals were prone to neural induction but resistant to osteogenic and adipogenic inductions compared to their age-matched controls. The altered differentiation may contribute to the severe trabecular bone loss seen in ethanol-exposed animals at 3months of age as well as overt growth retardation. Expression of alkaline phosphatase, osteocalcin, aP2, and PPARγ were substantially inhibited, but BDNF was up-regulated in MSCs isolated from ethanol-exposed 3month-old animals. Several signaling pathways were distorted in ethanol-exposed MSCs via altered trimethylation at histone 3 lysine 27. These results demonstrate that early life ethanol exposure can have long-term impacts in rat MSCs by both genetic and epigenetic mechanisms.


Assuntos
Epigênese Genética/efeitos dos fármacos , Etanol/toxicidade , Células-Tronco Mesenquimais/efeitos dos fármacos , Efeitos Tardios da Exposição Pré-Natal , Animais , Sequência de Bases , Primers do DNA , Modelos Animais de Doenças , Feminino , Transtornos do Espectro Alcoólico Fetal/etiologia , Transtornos do Espectro Alcoólico Fetal/genética , Masculino , Reação em Cadeia da Polimerase , Gravidez , Ratos
18.
Toxicol Lett ; 228(3): 140-6, 2014 Aug 04.
Artigo em Inglês | MEDLINE | ID: mdl-24857828

RESUMO

Prenatal alcohol exposure leads to congenital heart abnormal development, its mechanisms are still unknown. Recent reports have associated alcohol exposure with histone H3 acetylation. In the present study, we have performed the experiments to test the hypothesis that histone H3K14 acetylation is the key role in the fetal heart leads to over-expression of cardiac specific genes DHAND and EHAND caused by prenatal alcohol exposure. Seventy pregnant C57BL/6 mice were divided randomly into seven groups (n=10). They were the untreated group, dimethyl sulfoxide group, alcohol exposure group, curcumin treatment group, both alcohol and curcumin treatment group, SAHA treatment group, both alcohol and SAHA treatment group. Fetal mouse hearts were collected on embryonic day 14.5. The changes of HATs activities, the acetylation levels of histone H3K14 (H3K14ac), the expression levels of cardiac specific genes DHAND and EHAND, and structure of chromatin were determined. Our data indicates that curcumin and SAHA significantly reduces and increases the activities of HATs and the levels of histone H3K14ac in fetal hearts, respectively. The expression of DHAND and EHAND is significantly down-regulated and up-regulated in the groups treated with curcumin and SAHA. Furthermore, our results from ChIP assays have shown that the histone H3K14ac connects with the DHAND and EHAND genes are significantly inhibited by curcumin and simulated by SAHA. Our study suggests that prenatal alcohol exposure causes the over-expression of DHAND and EHAND by increasing H3K14ac in mice.


Assuntos
Fatores de Transcrição Hélice-Alça-Hélice Básicos/metabolismo , Etanol , Transtornos do Espectro Alcoólico Fetal/enzimologia , Coração Fetal/metabolismo , Histonas/metabolismo , Acetilação , Animais , Fatores de Transcrição Hélice-Alça-Hélice Básicos/genética , Sítios de Ligação , Imunoprecipitação da Cromatina , Feminino , Transtornos do Espectro Alcoólico Fetal/etiologia , Transtornos do Espectro Alcoólico Fetal/genética , Coração Fetal/efeitos dos fármacos , Regulação Neoplásica da Expressão Gênica , Inibidores de Histona Desacetilases/farmacologia , Camundongos , Camundongos Endogâmicos C57BL , Gravidez , Regiões Promotoras Genéticas , Regulação para Cima
19.
Alcohol Clin Exp Res ; 38(3): 694-703, 2014 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-24164477

RESUMO

BACKGROUND: Fetal alcohol spectrum disorders (FASD) are a highly variable set of phenotypes caused by fetal alcohol exposure. Numerous factors influence FASD phenotypes, including genetics. The zebrafish is a powerful vertebrate model system with which to identify these genetic factors. Many zebrafish mutants are housed at the Zebrafish International Resource Center (ZIRC). These mutants are readily accessible and an excellent source to screen for ethanol (EtOH)-sensitive developmental structural mutants. METHODS: We screened mutants obtained from ZIRC for sensitivity to EtOH teratogenesis. Embryos were treated with 1% EtOH (41 mM tissue levels) from 6 hours postfertilization onward. Levels of apoptosis were evaluated at 24 hours postfertilization. At 4 days postfertilization, the craniofacial skeleton, peripheral axon projections, and sensory neurons of neuromasts were examined. Fish were genotyped to determine whether there were phenotype/genotype correlations. RESULTS: Five of 20 loci interacted with EtOH. Notable among these was that vangl2, involved in convergent extension movements of the embryonic axis, interacted strongly with EtOH. Untreated vangl2 mutants had normal craniofacial morphology, while severe midfacial defects including synophthalmia and narrowing of the palatal skeleton were found in all EtOH-treated mutants and a low percentage of heterozygotes. The cell cycle gene, plk1, also interacted strongly with EtOH. Untreated mutants have slightly elevated levels of apoptosis and loss of ventral craniofacial elements. Exposure to EtOH results in extensive apoptosis along with loss of neural tissue and the entire craniofacial skeleton. Phenotypes of hinfp, mars, and foxi1 mutants were also exacerbated by EtOH. CONCLUSIONS: Our results provide insight into the gene-EtOH interactions that may underlie EtOH teratogenesis. They support previous findings that EtOH disrupts elongation of the embryonic axis. Importantly, these results show that the zebrafish is an efficient model with which to test for gene-EtOH interactions. Understanding these interactions will be crucial to understanding of the FASD variation.


Assuntos
Depressores do Sistema Nervoso Central/efeitos adversos , Anormalidades Craniofaciais/induzido quimicamente , Etanol/efeitos adversos , Transtornos do Espectro Alcoólico Fetal/genética , Peixe-Zebra/genética , Animais , Anormalidades Craniofaciais/genética , Genes cdc , Proteínas de Membrana/genética , Fenótipo , Fatores de Transcrição/genética , Proteínas de Peixe-Zebra/genética
20.
J Appl Toxicol ; 34(1): 66-75, 2014 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-23378141

RESUMO

Fetal alcohol spectrum disorder (FASD) is a set of developmental malformations caused by excess alcohol consumption during pregnancy. Using an in vitro system, we examined the role that chronic ethanol (EtOH) exposure plays in gene expression changes during the early stage of embryonic differentiation. We demonstrated that EtOH affected the cell morphology, cell cycle progression and also delayed the down-regulation of OCT4 and NANOG during differentiation. Gene expression profiling and pathway analysis demonstrated that EtOH deregulates many genes and pathways that are involved in early embryogenesis. Follow-up analyzes revealed that EtOH exposure to embryoid bodies (EBs) induced the expression of an organizer-specific gene, goosecoid (GSC), in comparison to controls. Moreover, EtOH treatment altered several important genes that are involved in embryonic structure formation, nervous system development, and placental and embryonic vascularization, which are all common processes that FASD can disrupt. Specifically, EtOH treatment let to a reduction in ALDOC, ENO2 and CDH1 expression, whereas EtOH treatment induced the expression of PTCH1, EGLN1, VEGFA and DEC2 in treated EBs. We also found that folic acid (FA) treatment was able to correct the expression of the majority of genes deregulated by EtOH exposure during early embryo development. Finally, the present study identified a gene set including GSC, which was deregulated by EtOH exposure that may contribute to the etiology of fetal alcohol syndrome (FAS). We also reported that EtOH-induced GSC expression is mediated by Nodal signaling, which may provide a new avenue for analyzing the molecular mechanisms behind EtOH teratogenicity in FASD individuals.


Assuntos
Etanol/efeitos adversos , Transtornos do Espectro Alcoólico Fetal/genética , Proteína Goosecoid/metabolismo , Ciclo Celular/efeitos dos fármacos , Diferenciação Celular/efeitos dos fármacos , Células Cultivadas , Análise por Conglomerados , Regulação para Baixo , Desenvolvimento Embrionário/efeitos dos fármacos , Feminino , Expressão Gênica/efeitos dos fármacos , Perfilação da Expressão Gênica , Proteína Goosecoid/genética , Proteínas de Homeodomínio/genética , Proteínas de Homeodomínio/metabolismo , Humanos , Análise em Microsséries , Proteína Homeobox Nanog , Fator 3 de Transcrição de Octâmero/genética , Fator 3 de Transcrição de Octâmero/metabolismo , Placenta/efeitos dos fármacos , Gravidez , Reprodutibilidade dos Testes , Transdução de Sinais
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA