Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 19 de 19
Filtrar
1.
Pediatr Res ; 92(5): 1299-1308, 2022 11.
Artículo en Inglés | MEDLINE | ID: mdl-35121849

RESUMEN

BACKGROUND: An adverse maternal environment (AME) predisposes progeny towards cognitive impairment in humans and mice. Cognitive impairment associates with hippocampal dysfunction. An important regulator of hippocampal function is the hippocampal serotonergic system. Dysregulation of hippocampal serotonin receptor 2c (HTR2c) expression is linked with cognitive impairment. HTR2c contains multiple mRNA variants and isoforms that are epigenetically regulated including DNA methylation, histone modifications, and small nucleolar RNA MBII-52. We tested the hypotheses that AME increases HTR2c variant expression and alters epigenetic modifications along the HTR2c gene locus. METHODS: We create an AME through maternal Western diet and prenatal environmental stress in the mouse. We analyzed hippocampal HTR2c and variants' expression, DNA methylation and histone modifications along the gene locus, and MBII-52 levels in postnatal day 21 offspring. RESULTS: AME significantly increased the expressions of total HTR2c and full-length variants (V201 and V202) concurrently with an altered epigenetic profile along the HTR2c gene locus in male offspring hippocampi. Moreover, increased full-length variants' expression in AME males was in line with increased MBII-52 levels. CONCLUSIONS: AME affects male offspring hippocampal expression of HTR2c and full-length variants via epigenetic mechanisms. Altered hippocampal HTR2c expression may contribute to cognitive impairment seen in adult males in this model. IMPACT: The key message of our article is that an adverse maternal environment increases expression of total HTR2c mRNA and protein, alters proportions of HTR2c mRNA variants, and impacts HTR2c epigenetic modifications in male offspring hippocampi relative to controls. Our findings add to the literature by providing the first report of altered HTR2c mRNA variant expression in association with altered epigenetic modifications in the hippocampus of offspring mice exposed to an adverse maternal environment. Our findings suggest that an adverse maternal environment affects the expression of genes previously determined to regulate cognitive function through an epigenetic mechanism in a sex-specific manner.


Asunto(s)
Epigénesis Genética , Hipocampo , Efectos Tardíos de la Exposición Prenatal , Animales , Femenino , Masculino , Ratones , Embarazo , Metilación de ADN , Hipocampo/metabolismo , Efectos Tardíos de la Exposición Prenatal/metabolismo , ARN Mensajero/metabolismo
2.
Dev Neurosci ; 43(2): 95-105, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-33940573

RESUMEN

An adverse maternal environment (AME) predisposes adult offspring toward cognitive impairment in humans and mice. However, the underlying mechanisms remain poorly understood. Epigenetic changes in response to environmental exposure may be critical drivers of this change. Epigenetic regulators, including microRNAs, have been shown to affect cognitive function by altering hippocampal neurogenesis which is regulated in part by brain-derived neurotropic factor (BDNF). We sought to investigate the effects of AME on miR profile and their epigenetic characteristics, as well as neurogenesis and BDNF expression in mouse hippocampus. Using our mouse model of AME which is composed of maternal Western diet and prenatal environmental stress, we found that AME significantly increased hippocampal miR-10b-5p levels. We also found that AME significantly decreased DNA methylation and increased accumulations of active histone marks H3 lysine (K) 4me3, H3K14ac, and -H3K36me3 at miR-10b promoter. Furthermore, AME significantly decreased hippocampal neurogenesis by decreasing cell numbers of Ki67+ (proliferation marker), NeuroD1+ (neuronal differentiation marker), and NeuN+ (mature neuronal marker) in the dentate gyrus (DG) region concurrently with decreased hippocampal BDNF protein levels. We speculate that the changes in epigenetic profile at miR-10b promoter may contribute to upregulation of miR-10b-5p and subsequently lead to decreased BDNF levels in a model of impaired offspring hippocampal neurogenesis and cognition in mice.


Asunto(s)
Factor Neurotrófico Derivado del Encéfalo , MicroARNs , Animales , Factor Neurotrófico Derivado del Encéfalo/genética , Factor Neurotrófico Derivado del Encéfalo/metabolismo , Epigénesis Genética , Femenino , Hipocampo/metabolismo , Masculino , Ratones , MicroARNs/genética , Neurogénesis , Embarazo
3.
J Nutr ; 151(10): 3102-3112, 2021 10 01.
Artículo en Inglés | MEDLINE | ID: mdl-34486661

RESUMEN

BACKGROUND: The role of an adverse maternal environment (AME) in conjunction with a postweaning Western diet (WD) in the development of nonalcoholic fatty liver disease (NAFLD) in adult offspring has not been explored. Likewise, the molecular mechanisms associated with AME-induced NAFLD have not been studied. The fatty acid translocase or cluster of differentiation 36 (CD36) has been implicated to play a causal role in the pathogenesis of WD-induced steatosis. However, it is unknown if CD36 plays a role in AME-induced NAFLD. OBJECTIVE: This study was designed to evaluate the isolated and additive impact of AME and postweaning WD on the expression and DNA methylation of hepatic Cd36 in association with the development of NAFLD in a novel mouse model. METHODS: AME constituted maternal WD and maternal stress, whereas the control (Con) group had neither. Female C57BL/6J mice were fed a WD [40% fat energy, 29.1% sucrose energy, and 0.15% cholesterol (wt/wt)] 5 wk prior to pregnancy and throughout lactation. Non invasive variable stressors (random frequent cage changing, limited bedding, novel object, etc.) were applied to WD dams during the last third of pregnancy to produce an AME. Con dams consumed the control diet (CD) (10% fat energy, no sucrose or cholesterol) and were not exposed to stress. Male offspring were weaned onto either CD or WD, creating 4 experimental groups: Con-CD, Con-WD, AME-CD, and AME-WD, and evaluated for metabolic and molecular parameters at 120 d of age. RESULTS: AME and postweaning WD independently and additively increased the development of hepatic steatosis in adult male offspring. AME and WD independently and additively upregulated hepatic CD36 protein and mRNA expression and hypomethylated promoters 2 and 3 of the Cd36 gene. CONCLUSIONS: Using a mouse AME model together with postweaning WD, this study demonstrates a role for CD36 in AME-induced NAFLD in offspring and reveals 2 regions of environmentally induced epigenetic heterogeneity within Cd36.


Asunto(s)
Enfermedad del Hígado Graso no Alcohólico , Animales , Metilación de ADN , Dieta Alta en Grasa/efectos adversos , Dieta Occidental/efectos adversos , Femenino , Hígado/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Enfermedad del Hígado Graso no Alcohólico/etiología , Enfermedad del Hígado Graso no Alcohólico/metabolismo , Embarazo
4.
Physiol Genomics ; 51(9): 462-470, 2019 09 01.
Artículo en Inglés | MEDLINE | ID: mdl-31373541

RESUMEN

BACKGROUND: Fetal growth restriction (FGR) is a major risk factor for bronchopulmonary dysplasia (BPD). Maternal stress and poor diet are linked to FGR. Effect of perinatal stress on lung development remains unknown. OBJECTIVE: Using a murine model of adverse early life environment (AELE), we hypothesized that maternal exposure to perinatal environmental stress and high-fat diet (Western diet) lead to impaired lung development in the offspring. METHODS: Female mice were placed on either control diet or Western diet before conception. Those exposed to Western diet were also exposed to perinatal environmental stress, the combination referred to as AELE. Pups were either euthanized at postnatal day 21 (P21) or weaned to control diet and environment until adulthood (8-14 wk old). Lungs were harvested for histology, gene expression by quantitative RT-PCR, microRNA profiling, and immunoblotting. RESULTS: AELE increased the mean linear intercept and decreased the radial alveolar count and secondary septation in P21 and adult mice. Capillary count was also decreased in P21 and adult mice. AELE lungs had decreased vascular endothelial growth factor A (VEGFA), VEGF receptor 2, endothelial nitric oxide synthase, and hypoxia inducible factor-1α protein levels and increased expression of genes that regulate DNA methylation and upregulation of microRNAs that target genes involved in lung development at P21. CONCLUSION: AELE leads to impaired lung alveolar and vascular growth, which persists into adult age despite normalizing the diet and environment at P21. AELE also alters the expression of genes involved in lung remodeling.


Asunto(s)
Dieta Occidental/efectos adversos , Retardo del Crecimiento Fetal/fisiopatología , Pulmón/crecimiento & desarrollo , Organogénesis , Estrés Fisiológico/genética , Estrés Fisiológico/inmunología , Animales , Animales Recién Nacidos , Metilación de ADN/genética , Modelos Animales de Enfermedad , Femenino , Subunidad alfa del Factor 1 Inducible por Hipoxia/metabolismo , Pulmón/patología , Masculino , Ratones , Ratones Endogámicos C57BL , MicroARNs/genética , Óxido Nítrico Sintasa/metabolismo , Embarazo , Transcriptoma , Factor A de Crecimiento Endotelial Vascular/metabolismo , Receptor 2 de Factores de Crecimiento Endotelial Vascular/metabolismo
5.
Physiol Genomics ; 50(11): 973-981, 2018 11 01.
Artículo en Inglés | MEDLINE | ID: mdl-30240345

RESUMEN

Adverse early life environment (AELE) predisposes adult offspring toward anxiety disorders. Anxiety disorders are associated with prenatal injuries in key regions of the brain including prefrontal cortex (PFC), hippocampus (HP), and hypothalamus (HT). Injuries in these brain regions result in an impaired hypothalamus-pituitary-adrenal axis (HPA axis) and stress response. An important regulator of the stress response is FK506-binding protein 5 (FKBP5). FKBP5 is a cochaperone of the glucocorticoid receptor (GR) and inhibits GR-mediated regulatory feed-back on the HPA axis in response to stress. Human studies have shown that polymorphisms of FKBP5 are associated with higher FKBP5 levels. Increased FKBP5 leads to GR resistance and impaired negative feedback, which is associated with anxiety disorders. FKBP5 and its mRNA splice variants in the aforementioned brain regions have not been reported. We hypothesized that AELE will increase expression of FKBP5 and its mRNA splice variants in PFC, HP, and HT as well as increase anxiety in adult mice. AELE increased expression of FKBP5 and its mRNA variants in PFC, HP and HT at postnatal day 21. Additionally, AELE caused anxiety and increased GR abundance in association with these changes in FKBP5 expression. We speculate that these changes in FKBP5 mRNA variants affect HPA axis function and contributes to subsequent anxiety-like behavior later in life in AELE mice.


Asunto(s)
Ansiedad/etiología , Encéfalo/fisiología , Proteínas de Unión a Tacrolimus/genética , Animales , Animales Recién Nacidos , Ansiedad/genética , Conducta Animal , Peso Corporal , Corticosterona/sangre , Femenino , Sistema Hipotálamo-Hipofisario/fisiología , Masculino , Ratones Endogámicos C57BL , Microglía/fisiología , Sistema Hipófiso-Suprarrenal/fisiología , Embarazo , Isoformas de Proteínas/genética , ARN Mensajero/genética , Receptores de Glucocorticoides/genética , Receptores de Glucocorticoides/metabolismo , Estrés Psicológico/complicaciones , Proteínas de Unión a Tacrolimus/metabolismo
6.
FASEB J ; 29(4): 1176-84, 2015 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-25466885

RESUMEN

Intrauterine growth restriction (IUGR) decreases serum IGF-1 levels. Postnatal IGF-1 expression is transcriptionally regulated by growth hormone (GH) through growth hormone response elements (GHREs). We hypothesized that IUGR disrupts the normal developmental maturation of hepatic IGF-1 intron 2 growth hormone response element (IN2GHRE) histone methylation of key lysines and DNA methylation. We also evaluated a 5' distal weak enhancer (IGF-1 5'-upstream region growth hormone response element; 5URGHRE) as a GHRE specificity control. IUGR was induced through a well-characterized model of bilateral uterine artery ligation of the pregnant rat. Offspring livers were tested at d 0 and 21. Chromatin immunoprecipitation and bisulfite sequencing quantified epigenetic characteristics. We found that distinct age-related developmental patterns of histone and DNA methylation characterize each GHRE. Development increased H3K4 trimethylation (me3) in both GHREs. However, H3K9me3 decreased with age at IN2GHRE and increased with age at 5URGHRE. IUGR altered the developmental pattern of H3K4me3 and K9me3 around the GHREs in a sex-specific manner at d 21. Developmental and IUGR-induced DNA methylation occurred in a GHRE-, CpG site-, and sex-specific manner. We conclude that IUGR disrupts developmental epigenetics around distal GHREs on the rat hepatic IGF-1 gene.


Asunto(s)
Epigénesis Genética , Retardo del Crecimiento Fetal/genética , Factor I del Crecimiento Similar a la Insulina/genética , Animales , Animales Recién Nacidos , Sitios de Unión/genética , Islas de CpG , Metilación de ADN , Femenino , Retardo del Crecimiento Fetal/metabolismo , Regulación del Desarrollo de la Expresión Génica , Histonas/metabolismo , Hígado/metabolismo , Masculino , Embarazo , Ratas , Ratas Sprague-Dawley , Elementos de Respuesta , Factor de Transcripción STAT5/metabolismo
7.
Am J Physiol Regul Integr Comp Physiol ; 309(2): R119-27, 2015 Jul 15.
Artículo en Inglés | MEDLINE | ID: mdl-25972460

RESUMEN

Intrauterine growth restriction (IUGR) increases the risk for neurodevelopment delay and neuroendocrine reprogramming in both humans and rats. Neuroendocrine reprogramming involves the glucocorticoid receptor (GR) gene that is epigenetically regulated in the hippocampus. Using a well-characterized rodent model, we have previously shown that IUGR increases GR exon 1.7 mRNA variant and total GR expressions in male rat pup hippocampus. Epigenetic regulation of GR transcription may involve chromatin remodeling of the GR gene. A key chromatin remodeler is Brahma-related gene-1(Brg1), a member of the ATP-dependent SWItch/Sucrose NonFermentable (SWI/SNF) chromatin remodeling complex. Brg1 regulates gene expression by affecting nucleosome repositioning and recruiting transcriptional components to target promoters. We hypothesized that IUGR would increase hippocampal Brg1 expression and binding to GR exon 1.7 promoter, as well as alter nucleosome positioning over GR promoters in newborn male pups. Further, we hypothesized that IUGR would lead to accumulation of specificity protein 1 (Sp1) and RNA pol II at GR exon 1.7 promoter. Indeed, we found that IUGR increased Brg1 expression and binding to GR exon 1.7 promoter. We also found that increased Brg1 binding to GR exon 1.7 promoter was associated with accumulation of Sp1 and RNA pol II carboxy terminal domain pSer-5 (a marker of active transcription). Furthermore, the transcription start site of GR exon 1.7 was located within a nucleosome-depleted region. We speculate that changes in hippocampal Brg1 expression mediate GR expression and subsequently trigger neuroendocrine reprogramming in male IUGR rats.


Asunto(s)
Ensamble y Desensamble de Cromatina , ADN Helicasas/metabolismo , Retardo del Crecimiento Fetal/metabolismo , Hipocampo/metabolismo , Proteínas Nucleares/metabolismo , Regiones Promotoras Genéticas , Receptores de Glucocorticoides/metabolismo , Factores de Transcripción/metabolismo , Animales , Animales Recién Nacidos , Sitios de Unión , ADN Helicasas/genética , Modelos Animales de Enfermedad , Exones , Retardo del Crecimiento Fetal/genética , Retardo del Crecimiento Fetal/fisiopatología , Regulación del Desarrollo de la Expresión Génica , Hipocampo/crecimiento & desarrollo , Hipocampo/fisiopatología , Masculino , Proteínas Nucleares/genética , Nucleosomas/metabolismo , ARN Polimerasa II/metabolismo , Ratas , Receptores de Glucocorticoides/genética , Factor de Transcripción Sp1/metabolismo , Factores de Transcripción/genética , Sitio de Iniciación de la Transcripción , Transcripción Genética , Regulación hacia Arriba
8.
Clin Obstet Gynecol ; 56(3): 622-32, 2013 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-23787712

RESUMEN

Early life environment predicts future health. The initial precedents and research focus on this concept arose out of historical events. However, this concept continues to be relevant as evidenced by the recent Chinese famine and the evidence of racial disparities in the United States. The latter allows us to introduce the "life course model" and "weathering" as relevant epigenetic phenomena. We then review the molecular components of environmental epigenetics. We subsequently present glucocorticoid receptor biology as a paradigm that involves all of the components. Finally, we suggest that environmental epigenetics are a key component of the future of personalized medicine.


Asunto(s)
Metilación de ADN/fisiología , Epigénesis Genética/fisiología , Histonas/metabolismo , MicroARNs/fisiología , Efectos Tardíos de la Exposición Prenatal/genética , ARN Largo no Codificante/fisiología , Femenino , Trastornos Nutricionales en el Feto/genética , Regulación del Desarrollo de la Expresión Génica , Interacción Gen-Ambiente , Predisposición Genética a la Enfermedad , Humanos , Mortalidad Infantil/etnología , Recién Nacido , Nucleosomas/fisiología , Embarazo , Receptores de Glucocorticoides/genética , Inanición
9.
Nutrients ; 15(9)2023 Apr 30.
Artículo en Inglés | MEDLINE | ID: mdl-37432267

RESUMEN

Exposure to adverse early-life environments (AME) increases the incidence of developing adult-onset non-alcoholic fatty liver disease (NAFLD). DNA methylation has been postulated to link AME and late-onset diseases. This study aimed to investigate whether and to what extent the hepatic DNA methylome was perturbed prior to the development of NAFLD in offspring exposed to AME in mice. AME constituted maternal Western diet and late-gestational stress. Male offspring livers at birth (d0) and weaning (d21) were used for evaluating the DNA methylome and transcriptome using the reduced representation of bisulfite sequencing and RNA-seq, respectively. We found AME caused 5879 differentially methylated regions (DMRs) and zero differentially expressed genes (DEGs) at d0 and 2970 and 123, respectively, at d21. The majority of the DMRs were distal to gene transcription start sites and did not correlate with DEGs. The DEGs at d21 were significantly enriched in GO biological processes characteristic of liver metabolic functions. In conclusion, AME drove changes in the hepatic DNA methylome, which preceded perturbations in the hepatic metabolic transcriptome, which preceded the onset of NAFLD. We speculate that subtle impacts on dynamic enhancers lead to long-range regulatory changes that manifest over time as gene network alternations and increase the incidence of NAFLD later in life.


Asunto(s)
Enfermedad del Hígado Graso no Alcohólico , Masculino , Animales , Ratones , Embarazo , Femenino , Enfermedad del Hígado Graso no Alcohólico/genética , Epigenoma , Transcriptoma , Metilación de ADN
10.
Anat Rec (Hoboken) ; 306(1): 162-175, 2023 01.
Artículo en Inglés | MEDLINE | ID: mdl-35983908

RESUMEN

An adverse maternal environment (AME) and Western diet (WD) in early life predispose offspring toward cognitive impairment in humans and mice. Cognitive impairment associates with hippocampal dysfunction. An important regulator of hippocampal function is the hippocampal Nociceptin/Orphanin FQ (N/OFQ) system. Previous studies find links between dysregulation of hippocampal N/OFQ receptor (NOP) expression and impaired cognitive function. NOP is encoded by the opioid receptor-like 1 (Oprl1) gene that contains multiple mRNA variants and isoforms. Regulation of Oprl1 expression includes histone modifications within the promoter. We tested the hypothesis that an AME and a postweaning WD increase the expression of hippocampal Oprl1 and select variants concurrent with altered histone code in the promoter. We created an AME-WD model combining maternal WD and prenatal environmental stress plus postweaning WD in the mouse. We analyzed the hippocampal expression of Oprl1, Oprl1 variants, and histone modifications in the Oprl1 promoter in offspring at postnatal day (P) 21 and P100. An AME and an AME-WD significantly increased the total hippocampal expression of Oprl1 and variant V4 concurrently with an increased accumulation of active histone marks in the promoter of male offspring. We concluded that an AME and an AME-WD alter hippocampal Oprl1 expression in offspring through an epigenetic mechanism in a variant-specific and sex-specific manner. Altered hippocampal Oprl1 expression may contribute to cognitive impairment seen in adult males in this model. Epigenetic regulation of Oprl1 is a potential mechanism by which an AME and a WD may contribute to neurocognitive impairment in male offspring.


Asunto(s)
Epigénesis Genética , Animales , Humanos , Masculino , Ratones
11.
Adv Sci (Weinh) ; 10(28): e2206692, 2023 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-37587835

RESUMEN

Regulatory T (Treg) cells are inevitable to prevent deleterious immune responses to self and commensal microorganisms. Treg function requires continuous expression of the transcription factor (TF) FOXP3 and is divided into two major subsets: resting (rTregs) and activated (aTregs). Continuous T cell receptor (TCR) signaling plays a vital role in the differentiation of aTregs from their resting state, and in their immune homeostasis. The process by which Tregs differentiate, adapt tissue specificity, and maintain stable phenotypic expression at the transcriptional level is still inconclusivei. In this work, the role of BATF is investigated, which is induced in response to TCR stimulation in naïve T cells and during aTreg differentiation. Mice lacking BATF in Tregs developed multiorgan autoimmune pathology. As a transcriptional regulator, BATF is required for Treg differentiation, homeostasis, and stabilization of FOXP3 expression in different lymphoid and non-lymphoid tissues. Epigenetically, BATF showed direct regulation of Treg-specific genes involved in differentiation, maturation, and tissue accumulation. Most importantly, FOXP3 expression and Treg stability require continuous BATF expression in Tregs, as it regulates demethylation and accessibility of the CNS2 region of the Foxp3 locus. Considering its role in Treg stability, BATF should be considered an important therapeutic target in autoimmune disease.


Asunto(s)
Enfermedades Autoinmunes , Linfocitos T Reguladores , Ratones , Animales , Diferenciación Celular , Factores de Transcripción Forkhead/genética , Factores de Transcripción Forkhead/metabolismo , Receptores de Antígenos de Linfocitos T/metabolismo
12.
Physiol Rep ; 8(8): e14407, 2020 04.
Artículo en Inglés | MEDLINE | ID: mdl-32333646

RESUMEN

Adverse maternal environment (AME) and high-fat diet in early childhood increase the risk of cognitive impairment and depression later in life. Cognitive impairment associates with hippocampal dysfunction. A key regulator of hippocampal function is the glucocorticoid receptor. Increased hippocampal GR expression associates with cognitive impairment and depression. Transcriptional control of GR relies in part upon the DNA methylation status at multiple alternative initiation sites that are tissue specific, with exon 1.7 being hippocampal specific. Increased exon 1.7 expression associates with upregulated hippocampal GR expression in early life stress animal models. However, the effects of AME combined with postweaning western diet (WD) on offspring behaviors and the expression of GR exon 1 variants in the hippocampus are unknown. We hypothesized that AME and postweaning WD would impair cognitive function and cause depression-like behavior in offspring in conjunction with dysregulated hippocampal expression of total GR and exon 1.7 variant in mice. We found that AME-WD impaired learning and memory in male adult offspring concurrently with increased hippocampal expression of total GR and GR 1.7. We also found that increased GR 1.7 expression was associated with decreased DNA methylation at the GR 1.7 promoter. We speculate that decreased DNA methylation at the GR 1.7 promoter plays a role in AME-WD induced increase of GR in the hippocampus. This increased GR expression may subsequently contribute to hippocampus dysfunction and lead to the cognitive impairment seen in this model.


Asunto(s)
Disfunción Cognitiva/genética , Disfunción Cognitiva/metabolismo , Metilación de ADN , Dieta Occidental/efectos adversos , Hipocampo/metabolismo , Exposición Materna/efectos adversos , Receptores de Glucocorticoides/genética , Animales , Disfunción Cognitiva/patología , Femenino , Desarrollo Fetal , Hipocampo/efectos de los fármacos , Hipocampo/patología , Masculino , Intercambio Materno-Fetal , Ratones , Ratones Endogámicos C57BL , Embarazo , Efectos Tardíos de la Exposición Prenatal , Regiones Promotoras Genéticas , Distribución Aleatoria , Receptores de Glucocorticoides/biosíntesis , Receptores de Glucocorticoides/metabolismo
13.
Birth Defects Res ; 110(20): 1551-1555, 2018 12 01.
Artículo en Inglés | MEDLINE | ID: mdl-30576090

RESUMEN

BACKGROUND: Cardiac fibrosis is a cardinal feature of multiple types of cardiovascular disease, which lead to heart failure. Multiple studies connect adverse maternal environment (AME) with cardiac fibrosis. AME does not always result in fibrosis, though. An additional "insult", such as an adult Western diet (WD), is frequently necessary. The additive effects of AME and adult WD on cardiac fibrosis is not well-understood. AME can also alter DNA methylation. DNA methyltransferase (DNMT) and ten-eleven translocation (TET) are methylation modifying genes that regulate DNA methylation, but it is unknown if AME changes cardiac gene expression of DNMT and TET. We sought to use a model of AME and adult WD to investigate the development of cardiac fibrosis and cardiac mRNA expression of DNMT and TET genes. METHODS: We exposed dams to WD or control diet (CD) 5 weeks before pregnancy and through lactation. We added environmental stressors during the last third of pregnancy to dams on WD to create AME. Dams on CD experienced no added stressors to create control maternal environment (CME). Male offspring were weaned at Postnatal Week 3 (W3) and placed on WD or CD to create four groups: CME-CD, CME-WD, AME-CD, and AME-WD. RESULTS: AME-WD increased cardiac fibrosis in adulthood (p < .05), whereas AME-CD and CME-WD did not. TET1-3 and DNMT3a mRNA levels decreased in AME versus CME offspring (p < .01). CONCLUSION: AME increases susceptibility to cardiac fibrosis in adult male mice. Early-life changes to TET expression may mediate susceptibility to fibrosis, but further testing is needed.


Asunto(s)
Fibrosis/etiología , Exposición Materna/efectos adversos , Miocardio/patología , Animales , Enfermedades Cardiovasculares/etiología , ADN (Citosina-5-)-Metiltransferasa 1/genética , ADN (Citosina-5-)-Metiltransferasa 1/fisiología , Metilación de ADN , Proteínas de Unión al ADN/genética , Proteínas de Unión al ADN/fisiología , Dieta Occidental/efectos adversos , Femenino , Fibrosis/genética , Corazón/fisiología , Masculino , Ratones , Ratones Endogámicos C57BL , Embarazo , Efectos Tardíos de la Exposición Prenatal/fisiopatología , Proteínas Proto-Oncogénicas/genética , Proteínas Proto-Oncogénicas/fisiología
14.
Int J Dev Neurosci ; 55: 56-65, 2016 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-27666383

RESUMEN

BACKGROUND: Adverse maternal lifestyle resulting in adverse early life environment (AELE) increases risks for neuropsychiatric disorders in offspring. Neuropsychiatric disorders are associated with impaired neurogenesis and neuro-inflammation in the hippocampus (HP). Microglia are neuro-inflammatory cells in the brain that regulate neurogenesis via toll-like receptors (TLR). TLR-9 is implicated in neurogenesis inhibition and is responsible for stress-related inflammatory responses. We hypothesized that AELE would increase microglia cell count and increase TLR-9 expression in juvenile mouse HP. These increases in microglia cell count and TLR-9 expression would be associated with decrease neural stem cell count and neuronal cell count. METHODS: We developed a mouse model of AELE combining Western diet and a stress environment. Stress environment consisted of random change from embryonic day 13 (E13) to E17 as well as static change in maternal environment from E13 to postnatal day 21(P21). At P21, we measured hippocampal cell numbers of microglia, neural stem cell and neuron, as well as hippocampal TLR-9 expression. RESULTS: AELE significantly increased total microglia number and TLR-9 expression in the hippocampus. Concurrently, AELE significantly decreased neural stem cell and neuronal numbers. CONCLUSIONS: AELE increased the neuro-inflammatory cellular response in the juvenile HP. We speculate that increased neuro-inflammatory responses may contribute to impaired neurogenesis seen in this model.


Asunto(s)
Ambiente , Hipocampo/patología , Microglía/patología , Células-Madre Neurales/fisiología , Efectos Tardíos de la Exposición Prenatal/patología , Factores de Edad , Análisis de Varianza , Animales , Animales Recién Nacidos , Antígenos CD/metabolismo , Proteínas de Unión al Calcio/genética , Proteínas de Unión al Calcio/metabolismo , Células Cultivadas , Dieta Occidental , Modelos Animales de Enfermedad , Femenino , Citometría de Flujo , Hipocampo/crecimiento & desarrollo , Masculino , Ratones , Ratones Endogámicos C57BL , Proteínas de Microfilamentos/genética , Proteínas de Microfilamentos/metabolismo , Nestina/metabolismo , Fosfopiruvato Hidratasa/metabolismo , Embarazo , Efectos Tardíos de la Exposición Prenatal/dietoterapia , ARN Mensajero/metabolismo , Receptor Toll-Like 9/genética , Receptor Toll-Like 9/metabolismo
15.
Epigenomics ; 7(7): 1173-84, 2015 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-26585860

RESUMEN

Children exposed to early-life adversity carry a greater risk of poor health and disease into adulthood. This increased disease risk is shadowed by changes in the epigenome. Epigenetics can change gene expression to modify disease risk; unfortunately, how epigenetics are changed by the environment is unclear. It is known that the environment modifies the microbiota, and recent data indicate that the microbiota and the epigenome interact and respond to each other. Specifically, the microbiome may alter the epigenome through the production of metabolites. Investigating the relationship between the microbiome and the epigenome may provide novel understanding of the impact of early-life environment on long-term health.


Asunto(s)
Epigénesis Genética/efectos de los fármacos , Genoma Humano , Microbiota/fisiología , Probióticos/farmacología , Procesamiento Proteico-Postraduccional/efectos de los fármacos , Colina/biosíntesis , Cromatina/química , Cromatina/efectos de los fármacos , Cromatina/metabolismo , Metilación de ADN , Resistencia a la Enfermedad/efectos de los fármacos , Resistencia a la Enfermedad/genética , Ácidos Grasos Volátiles/biosíntesis , Ácido Fólico/biosíntesis , Interacción Gen-Ambiente , Histonas/genética , Histonas/inmunología , Humanos , Isotiocianatos/metabolismo , Polifenoles/biosíntesis , Probióticos/metabolismo
16.
Clin Perinatol ; 41(4): 847-75, 2014 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-25459778

RESUMEN

Infant mortality rate (IMR) is a reference indicator for societal health status. Trend analysis of IMR highlights 2 challenges to overcome in the United States: (1) US IMR is higher than most industrialized countries and (2) there are persistent racial/ethnic disparities in birth outcomes, especially for blacks. Racial/ethnic infant mortality disparities result from the complex interplay of adverse social, economic, and environmental exposures. In this article, racial/ethnic disparities are discussed, highlighting trends, the role of epigenetics in understanding mechanisms, key domains of community action planning, and programs and policies addressing the racial gaps in adverse birth outcomes.


Asunto(s)
Negro o Afroamericano/estadística & datos numéricos , Epigénesis Genética/genética , Disparidades en el Estado de Salud , Disparidades en Atención de Salud/etnología , Mortalidad Infantil/etnología , Nacimiento Prematuro/etnología , Racismo , Población Blanca/estadística & datos numéricos , Negro o Afroamericano/genética , Femenino , Humanos , Lactante , Recién Nacido , Evaluación de Resultado en la Atención de Salud , Atención Perinatal/estadística & datos numéricos , Embarazo , Nacimiento Prematuro/epidemiología , Nacimiento Prematuro/genética , Atención Prenatal/estadística & datos numéricos , Clase Social , Estados Unidos/epidemiología
17.
Expert Rev Endocrinol Metab ; 9(6): 605-614, 2014 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-30736198

RESUMEN

Obesity and its consequences impact everyone. Obesity occurs because of an interaction between an obesogenic environment and genetics. In order to confront obesity, we must understand the contribution of each of these components. Environmental influences on obesity include our extrinsic environment, such as food deserts, as well as our intrinsic environment, like perinatal exposures. Epigenetics provides a biological mechanism to reveal the accumulation of extrinsic and intrinsic environmental exposures from fetal life to adulthood. Human and animal studies demonstrate changes in epigenetic modifications which are associated with an obesogenic environment. Furthermore, evidence exists in humans and animal models that suggest environmental epigenetics may serve as a biomarker or a target for intervention. To successfully target obesity, we must intervene on an environmental as well as genetic level. Combating food deserts for example will help to change the extrinsic environment, while targeting epigenetic modification remains a goal for changing our biology.

18.
Int J Dev Neurosci ; 38: 59-67, 2014 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-25107645

RESUMEN

Intrauterine growth restriction (IUGR) programs neurodevelopmental impairment and long-term neurological morbidities. Neurological morbidities in IUGR infants are correlated with changes hippocampal volume. We previously demonstrated that IUGR alters hippocampal cellular composition in both neonatal and juvenile rat pups in association with altered hippocampal gene expression and epigenetic determinants. PPARγ signaling is important for neurodevelopment as well as epigenetic integrity in the brain via the PPARγ-Setd8-H4K20me(1) axis and Wnt signaling. We hypothesized that IUGR would decrease expression of PPARγ, Setd8, and H4K20me(1) in juvenile rat hippocampus. We further hypothesized that reduced PPARγ-Setd8-H4K20me(1) would be associated with reduced Wnt signaling genes Wnt3a and ß-catenin, and wnt target gene Axin2. To test our hypothesis we used a rat model of uteroplacental insufficiency-induced IUGR. We demonstrated that PPARγ localizes to oligodendrocytes, neurons and astrocytes within the juvenile rat hippocampus. We also demonstrated that IUGR reduces levels of PPARγ, Setd8 and H4K20me(1) in male and female juvenile rat hippocampus in conjunction with reduced Wnt signaling components in only male rats. We speculate that reduced PPARγ and Wnt signaling may contribute to altered hippocampal cellular composition which, in turn, may contribute to impaired neurodevelopment and subsequent neurocognitive impairment in IUGR offspring.


Asunto(s)
Retardo del Crecimiento Fetal/patología , Regulación del Desarrollo de la Expresión Génica/fisiología , Hipocampo/metabolismo , Hipocampo/fisiopatología , PPAR gamma/metabolismo , Vía de Señalización Wnt/fisiología , Actinas/genética , Actinas/metabolismo , Animales , Animales Recién Nacidos , Peso Corporal , Modelos Animales de Enfermedad , Femenino , N-Metiltransferasa de Histona-Lisina/genética , N-Metiltransferasa de Histona-Lisina/metabolismo , Histonas/metabolismo , Masculino , PPAR gamma/genética , Fosfopiruvato Hidratasa/metabolismo , ARN Mensajero , Ratas , Factores Sexuales
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA