Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 34
Filtrar
1.
Front Nutr ; 10: 1190392, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-37565037

RESUMEN

Introduction: In humans, adversity in childhood exerts enduring effects on brain and increases the vulnerability to psychiatric diseases. It also leads to a higher risk of eating disorders and obesity. Maternal separation (MS) in mice has been used as a proxy of stress during infancy. We hypothesized that MS in mice affects motivation to obtain palatable food in adulthood and changes gene expression in reward system. Methods: Male and female pups from C57Bl/6J and C3H/HeN mice strains were subjected to a daily MS protocol from postnatal day (PND) 2 to PND14. At adulthood, their motivation for palatable food reward was assessed in operant cages. Results: Compared to control mice, male and female C3H/HeN mice exposed to MS increased their instrumental response for palatable food, especially when the effort required to obtain the reward was high. Importantly, this effect is shown in animals fed ad libitum. Transcriptional analysis revealed 375 genes differentially expressed in the nucleus accumbens of male MS C3H/HeN mice compared to the control group, some of these being associated with the regulation of the reward system (e.g., Gnas, Pnoc). Interestingly, C57Bl/6J mice exposed to MS did not show alterations in their motivation to obtain a palatable reward, nor significant changes in gene expression in the nucleus accumbens. Conclusion: MS produces long-lasting changes in motivation for palatable food in C3H/HeN mice, but has no impact in C57Bl/6J mice. These behavioral alterations are accompanied by drastic changes in gene expression in the nucleus accumbens, a key structure in the regulation of motivational processes.

2.
J Cachexia Sarcopenia Muscle ; 12(4): 1064-1078, 2021 08.
Artículo en Inglés | MEDLINE | ID: mdl-34196129

RESUMEN

BACKGROUND: Knowledge of age-related DNA methylation changes in skeletal muscle is limited, yet this tissue is severely affected by ageing in humans. METHODS: We conducted a large-scale epigenome-wide association study meta-analysis of age in human skeletal muscle from 10 studies (total n = 908 muscle methylomes from men and women aged 18-89 years old). We explored the genomic context of age-related DNA methylation changes in chromatin states, CpG islands, and transcription factor binding sites and performed gene set enrichment analysis. We then integrated the DNA methylation data with known transcriptomic and proteomic age-related changes in skeletal muscle. Finally, we updated our recently developed muscle epigenetic clock (https://bioconductor.org/packages/release/bioc/html/MEAT.html). RESULTS: We identified 6710 differentially methylated regions at a stringent false discovery rate <0.005, spanning 6367 unique genes, many of which related to skeletal muscle structure and development. We found a strong increase in DNA methylation at Polycomb target genes and bivalent chromatin domains and a concomitant decrease in DNA methylation at enhancers. Most differentially methylated genes were not altered at the mRNA or protein level, but they were nonetheless strongly enriched for genes showing age-related differential mRNA and protein expression. After adding a substantial number of samples from five datasets (+371), the updated version of the muscle clock (MEAT 2.0, total n = 1053 samples) performed similarly to the original version of the muscle clock (median of 4.4 vs. 4.6 years in age prediction error), suggesting that the original version of the muscle clock was very accurate. CONCLUSIONS: We provide here the most comprehensive picture of DNA methylation ageing in human skeletal muscle and reveal widespread alterations of genes involved in skeletal muscle structure, development, and differentiation. We have made our results available as an open-access, user-friendly, web-based tool called MetaMeth (https://sarah-voisin.shinyapps.io/MetaMeth/).


Asunto(s)
Metilación de ADN , Proteómica , Adolescente , Adulto , Anciano , Anciano de 80 o más Años , Islas de CpG , Epigénesis Genética , Femenino , Humanos , Masculino , Persona de Mediana Edad , Músculo Esquelético , Adulto Joven
3.
Nutrients ; 12(6)2020 May 28.
Artículo en Inglés | MEDLINE | ID: mdl-32481497

RESUMEN

Nutritional changes during developmental windows are of particular concern in offspring metabolic disease. Questions are emerging concerning the role of maternal weight changes before conception, particularly for weight loss, in the development of diet-related disorders. Understanding the physiological pathways affected by the maternal trajectories in the offspring is therefore essential, but a broad overview is still lacking. We recently reported both metabolic and behavioral negative outcomes in offspring born to obese or weight-loss mothers and fed a control of high-fat diet, suggesting long-term modeling of metabolic pathways needing to be further characterized. Using non-targeted LC-HRMS, we investigated the impact of maternal and post-weaning metabolic status on the adult male offspring's metabolome in three tissues involved in energy homeostasis: liver, hypothalamus and olfactory bulb. We showed that post-weaning diet interfered with the abundance of several metabolites, including 1,5-anhydroglucitol, saccharopine and ßhydroxybutyrate, differential in the three tissues. Moreover, maternal diet had a unique impact on the abundance of two metabolites in the liver. Particularly, anserine abundance, lowered by maternal obesity, was normalized by a preconceptional weight loss, whatever the post-weaning diet. This study is the first to identify a programming long-term effect of maternal preconception obesity on the offspring metabolome.


Asunto(s)
Encéfalo/metabolismo , Dieta , Hígado/metabolismo , Fenómenos Fisiologicos Nutricionales Maternos/fisiología , Intercambio Materno-Fetal/fisiología , Metaboloma , Obesidad Materna/metabolismo , Efectos Tardíos de la Exposición Prenatal/metabolismo , Destete , Ácido 3-Hidroxibutírico/metabolismo , Animales , Anserina/metabolismo , Desoxiglucosa/metabolismo , Metabolismo Energético , Femenino , Homeostasis , Lisina/análogos & derivados , Lisina/metabolismo , Masculino , Ratones Endogámicos C57BL , Embarazo
4.
Semin Cell Dev Biol ; 97: 172-180, 2020 01.
Artículo en Inglés | MEDLINE | ID: mdl-31587964

RESUMEN

The literature about Developmental Origins of Health and Disease (DOHaD) studies is considerably growing. Maternal and paternal environment, during all the development of the individual from gametogenesis to weaning and beyond, as well as the psychosocial environment in childhood and teenage, can shape the adult and the elderly person's susceptibility to her/his own environment and diseases. This non-conventional, non-genetic, inheritance is underlain by several mechanisms among which epigenetics is obviously central, due to the notion of memory of early decisional events during development even when this stimulus is gone, that is implied in Waddington's developmental concept. This review first summarizes the different mechanisms by which the environment can model the epigenome: receptor signalling, energy metabolism and signal mechanotransduction from extracellular matrix to chromatin. Then an overview of the epigenetic changes in response to maternal environment during the vulnerability time windows, gametogenesis, early development, placentation and foetal growth, and postnatal period, is described, with the specific example of overnutrition and food deprivation. The implication of epigenetics in DOHaD is obvious, however the precise causal chain from early environment to the epigenome modifications to the phenotype still needs to be deciphered.


Asunto(s)
Enfermedad/genética , Epigenoma/genética , Epigenómica/métodos , Predisposición Genética a la Enfermedad , Humanos , Padres , Fenotipo , Transducción de Señal
5.
Mol Reprod Dev ; 87(1): 124-134, 2020 01.
Artículo en Inglés | MEDLINE | ID: mdl-31746511

RESUMEN

Highly differentiated mature spermatozoa carry not only genetic but also epigenetic information that is to be transmitted to the embryo. DNA methylation is one epigenetic actor associated with sperm nucleus compaction, gene silencing, and prepatterning of embryonic gene expression. Therefore, the stability of this mark toward reproductive biotechnologies is a major issue in animal production. The present work explored the impact of hormonal induction of spermiation and sperm cryopreservation in two cyprinids, the goldfish (Carassius auratus) and the zebrafish (Danio rerio), using LUminometric Methylation Assay (LUMA). We showed that while goldfish hormonal treatment did increase sperm production, it did not alter global DNA methylation of spermatozoa. Different sperm samples repeatedly collected from the same males for 2 months also showed the same global DNA methylation level. Similarly, global DNA methylation was not affected after cryopreservation of goldfish spermatozoa with methanol, whereas less efficient cryoprotectants (dimethylsulfoxide and 1,2-propanediol) decreased DNA methylation. In contrast, cryopreservation of zebrafish spermatozoa with methanol induced a slight, but significant, increase in global DNA methylation. In the less compact nuclei, that is, goldfish fin somatic cells, cryopreservation did not change global DNA methylation regardless of the choice of cryoprotectant. To conclude, global DNA methylation is a robust parameter with respect to biotechnologies such as hormonal induction of spermiation and sperm cryopreservation, but it can be altered when the best sperm manipulation conditions are not met.


Asunto(s)
Criopreservación/métodos , Metilación de ADN/efectos de los fármacos , Domperidona/farmacología , Carpa Dorada/genética , Hormona Liberadora de Gonadotropina/farmacología , Preservación de Semen/métodos , Espermatozoides , Pez Cebra/genética , Animales , Crioprotectores/farmacología , Dimetilsulfóxido/farmacología , Combinación de Medicamentos , Femenino , Fertilización In Vitro/métodos , Masculino , Metanol/farmacología , Oocitos , Propilenglicol/farmacología , Motilidad Espermática/efectos de los fármacos
6.
Nutrients ; 11(5)2019 Apr 26.
Artículo en Inglés | MEDLINE | ID: mdl-31035463

RESUMEN

According to the "developmental origins of health and disease" (DOHaD) concept, maternal obesity predisposes the offspring to non-communicable diseases in adulthood. While a preconceptional weight loss (WL) is recommended for obese women, its benefits on the offspring have been poorly addressed. We evaluated whether preconceptional WL was able to reverse the adverse effects of maternal obesity in a mouse model, exhibiting a modification of foetal growth and of the expression of genes encoding epigenetic modifiers in liver and placenta. We tracked metabolic and olfactory behavioural trajectories of offspring born to control, obese or WL mothers. After weaning, the offspring were either put on a control diet (CD) or a high-fat (HFD). After only few weeks of HFD, the offspring developed obesity, metabolic alterations and olfactory impairments, independently of maternal context. However, male offspring born to obese mother gained even more weight under HFD than their counterparts born to lean mothers. Preconceptional WL normalized the offspring metabolic phenotypes but had unexpected effects on olfactory performance: a reduction in olfactory sensitivity, along with a lack of fasting-induced, olfactory-based motivation. Our results confirm the benefits of maternal preconceptional WL for male offspring metabolic health but highlight some possible adverse outcomes on olfactory-based behaviours.


Asunto(s)
Metabolismo Energético/fisiología , Obesidad/metabolismo , Olfato/fisiología , Pérdida de Peso , Animales , Dieta Alta en Grasa/efectos adversos , Femenino , Fertilización , Masculino , Ratones , Ratones Endogámicos C57BL , Madres , Embarazo
7.
FASEB J ; 32(5): 2768-2778, 2018 05.
Artículo en Inglés | MEDLINE | ID: mdl-29295860

RESUMEN

According to the Developmental Origin of Health and Disease (DOHaD) concept, maternal obesity and accelerated growth in neonates program obesity later in life. White adipose tissue (WAT) has been the focus of developmental programming events, although underlying mechanisms remain elusive. In rodents, WAT development primarily occurs during lactation. We previously reported that adult rat offspring from dams fed a high-fat (HF) diet exhibited fat accumulation and decreased peroxisome proliferator-activated receptor γ (PPARγ) mRNA levels in WAT. We hypothesized that PPARγ down-regulation occurs via epigenetic malprogramming which takes place during adipogenesis. We therefore examined epigenetic modifications in the PPARγ1 and PPARγ2 promoters in perirenal (pWAT) and inguinal fat pads of HF offspring at weaning (postnatal d 21) and in adulthood. Postnatal d 21 is a period characterized by active epigenomic remodeling in the PPARγ2 promoter (DNA hypermethylation and depletion in active histone modification H3ac and H3K4me3) in pWAT, consistent with increased DNA methyltransferase and DNA methylation activities. Adult HF offspring exhibited sustained hypermethylation and histone modification H3ac of the PPARγ2 promoter in both deposits, correlated with persistent decreased PPARγ2 mRNA levels. Consistent with the DOHaD hypothesis, retained epigenetic marks provide a mechanistic basis for the cellular memory linking maternal obesity to a predisposition for later adiposity.-Lecoutre, S., Pourpe, C., Butruille, L., Marousez, L., Laborie, C., Guinez, C., Lesage, J., Vieau, D., Eeckhoute, J., Gabory, A., Oger, F., Eberlé, D., Breton, C. Reduced PPARγ2 expression in adipose tissue of male rat offspring from obese dams is associated with epigenetic modifications.


Asunto(s)
Tejido Adiposo/metabolismo , Metilación de ADN , Epigénesis Genética , Obesidad/metabolismo , PPAR gamma/biosíntesis , Regiones Promotoras Genéticas , Tejido Adiposo/patología , Adiposidad/genética , Animales , Femenino , Histonas/genética , Histonas/metabolismo , Masculino , Obesidad/genética , PPAR gamma/genética , Procesamiento Proteico-Postraduccional , ARN Mensajero/biosíntesis , ARN Mensajero/genética , Ratas , Ratas Wistar
8.
Mol Metab ; 6(8): 922-930, 2017 08.
Artículo en Inglés | MEDLINE | ID: mdl-28752055

RESUMEN

OBJECTIVE: According to the Developmental Origin of Health and Disease (DOHaD) concept, maternal obesity and accelerated growth in neonates predispose offspring to white adipose tissue (WAT) accumulation. In rodents, adipogenesis mainly develops during lactation. The mechanisms underlying the phenomenon known as developmental programming remain elusive. We previously reported that adult rat offspring from high-fat diet-fed dams (called HF) exhibited hypertrophic adipocyte, hyperleptinemia and increased leptin mRNA levels in a depot-specific manner. We hypothesized that leptin upregulation occurs via epigenetic malprogramming, which takes place early during development of WAT. METHODS: As a first step, we identified in silico two potential enhancers located upstream and downstream of the leptin transcription start site that exhibit strong dynamic epigenomic remodeling during adipocyte differentiation. We then focused on epigenetic modifications (methylation, hydroxymethylation, and histone modifications) of the promoter and the two potential enhancers regulating leptin gene expression in perirenal (pWAT) and inguinal (iWAT) fat pads of HF offspring during lactation (postnatal days 12 (PND12) and 21 (PND21)) and in adulthood. RESULTS: PND12 is an active period for epigenomic remodeling in both deposits especially in the upstream enhancer, consistent with leptin gene induction during adipogenesis. Unlike iWAT, some of these epigenetic marks were still observable in pWAT of weaned HF offspring. Retained marks were only visible in pWAT of 9-month-old HF rats that showed a persistent "expandable" phenotype. CONCLUSIONS: Consistent with the DOHaD hypothesis, persistent epigenetic remodeling occurs at regulatory regions especially within intergenic sequences, linked to higher leptin gene expression in adult HF offspring in a depot-specific manner.


Asunto(s)
Epigénesis Genética , Leptina/genética , Obesidad/genética , Complicaciones del Embarazo/genética , Tejido Adiposo Blanco/metabolismo , Animales , Metilación de ADN , Femenino , Código de Histonas , Leptina/metabolismo , Masculino , Embarazo , Ratas , Ratas Wistar , Regulación hacia Arriba
9.
J Endocrinol ; 230(1): 39-53, 2016 07.
Artículo en Inglés | MEDLINE | ID: mdl-27122310

RESUMEN

According to the Developmental Origin of Health and Disease (DOHaD) concept, alterations of nutrient supply in the fetus or neonate result in long-term programming of individual body weight (BW) setpoint. In particular, maternal obesity, excessive nutrition, and accelerated growth in neonates have been shown to sensitize offspring to obesity. The white adipose tissue may represent a prime target of metabolic programming induced by maternal obesity. In order to unravel the underlying mechanisms, we have developed a rat model of maternal obesity using a high-fat (HF) diet (containing 60% lipids) before and during gestation and lactation. At birth, newborns from obese dams (called HF) were normotrophs. However, HF neonates exhibited a rapid weight gain during lactation, a key period of adipose tissue development in rodents. In males, increased BW at weaning (+30%) persists until 3months of age. Nine-month-old HF male offspring was normoglycemic but showed mild glucose intolerance, hyperinsulinemia, and hypercorticosteronemia. Despite no difference in BW and energy intake, HF adult male offspring was predisposed to fat accumulation showing increased visceral (gonadal and perirenal) depots weights and hyperleptinemia. However, only perirenal adipose tissue depot exhibited marked adipocyte hypertrophy and hyperplasia with elevated lipogenic (i.e. sterol-regulated element binding protein 1 (Srebp1), fatty acid synthase (Fas), and leptin) and diminished adipogenic (i.e. peroxisome proliferator-activated receptor gamma (Pparγ), 11ß-hydroxysteroid dehydrogenase type 1 (11ß-Hds1)) mRNA levels. By contrast, very few metabolic variations were observed in HF female offspring. Thus, maternal obesity and accelerated growth during lactation program offspring for higher adiposity via transcriptional alterations of visceral adipose tissue in a depot- and sex-specific manner.


Asunto(s)
Tejido Adiposo/metabolismo , Lactancia/metabolismo , Fenómenos Fisiologicos Nutricionales Maternos/fisiología , Obesidad/metabolismo , Efectos Tardíos de la Exposición Prenatal/metabolismo , Aumento de Peso/fisiología , Animales , Peso Corporal , Corticosterona/sangre , Femenino , Intolerancia a la Glucosa/metabolismo , Hiperinsulinismo/metabolismo , Masculino , PPAR gamma/genética , PPAR gamma/metabolismo , Embarazo , Ratas , Factores Sexuales , Proteína 1 de Unión a los Elementos Reguladores de Esteroles/genética , Proteína 1 de Unión a los Elementos Reguladores de Esteroles/metabolismo
10.
Development ; 143(6): 962-71, 2016 Mar 15.
Artículo en Inglés | MEDLINE | ID: mdl-26980793

RESUMEN

The H19 locus controls fetal growth by regulating expression of several genes from the imprinted gene network (IGN). H19 is fully repressed after birth, except in skeletal muscle. Using loss-of-function H19(Δ3) mice, we investigated the function of H19 in adult muscle. Mutant muscles display hypertrophy and hyperplasia, with increased Igf2 and decreased myostatin (Mstn) expression. Many imprinted genes are expressed in muscle stem cells or satellite cells. Unexpectedly, the number of satellite cells was reduced by 50% in H19(Δ3) muscle fibers. This reduction occurred after postnatal day 21, suggesting a link with their entry into quiescence. We investigated the biological function of these mutant satellite cells in vivo using a regeneration assay induced by multiple injections of cardiotoxin. Surprisingly, despite their reduced number, the self-renewal capacity of these cells is fully retained in the absence of H19. In addition, we observed a better regeneration potential of the mutant muscles, with enhanced expression of several IGN genes and genes from the IGF pathway.


Asunto(s)
Redes Reguladoras de Genes , Impresión Genómica , Músculos/fisiología , ARN Largo no Codificante/metabolismo , Regeneración/genética , Animales , Cardiotoxinas/toxicidad , Recuento de Células , Proliferación Celular/efectos de los fármacos , Eliminación de Gen , Redes Reguladoras de Genes/efectos de los fármacos , Impresión Genómica/efectos de los fármacos , Hiperplasia , Hipertrofia , Masculino , Ratones Endogámicos C57BL , Músculos/efectos de los fármacos , Músculos/patología , Mioblastos/efectos de los fármacos , Mioblastos/patología , ARN Largo no Codificante/genética , Regeneración/efectos de los fármacos , Células Satélite del Músculo Esquelético/patología
11.
Clin Epigenetics ; 8: 22, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-26925174

RESUMEN

BACKGROUND: Maternal obesity impacts fetal growth and pregnancy outcomes. To counteract the deleterious effects of obesity on fertility and pregnancy issue, preconceptional weight loss is recommended to obese women. Whether this weight loss is beneficial/detrimental for offspring remains poorly explored. Epigenetic mechanisms could be affected by maternal weight changes, perturbing expression of key developmental genes in the placenta or fetus. Our aim was to investigate the effects of chronic maternal obesity on feto-placental growth along with the underlying epigenetic mechanisms. We also tested whether preconceptional weight loss could alleviate these effects. RESULTS: Female mice were fed either a control diet (CTRL group), a high-fat diet (obese (OB) group), or a high-fat diet switched to a control diet 2 months before conception (weight loss (WL) group). At mating, OB females presented an obese phenotype while WL females normalized metabolic parameters. At embryonic day 18.5 (E18.5), fetuses from OB females presented fetal growth restriction (FGR; -13 %) and 28 % of the fetuses were small for gestational age (SGA). Fetuses from WL females normalized this phenotype. The expression of 60 epigenetic machinery genes and 32 metabolic genes was measured in the fetal liver, placental labyrinth, and junctional zone. We revealed 23 genes altered by maternal weight trajectories in at least one of three tissues. The fetal liver and placental labyrinth were more responsive to maternal obesity than junctional zone. One third (18/60) of the epigenetic machinery genes were differentially expressed between at least two maternal groups. Interestingly, genes involved in the histone acetylation pathway were particularly altered (13/18). In OB group, lysine acetyltransferases and Bromodomain-containing protein 2 were upregulated, while most histone deacetylases were downregulated. In WL group, the expression of only a subset of these genes was normalized. CONCLUSIONS: This study highlights the high sensitivity of the epigenetic machinery gene expression, and particularly the histone acetylation pathway, to maternal obesity. These obesity-induced transcriptional changes could alter the placental and the hepatic epigenome, leading to FGR. Preconceptional weight loss appears beneficial to fetal growth, but some effects of previous obesity were retained in offspring phenotype.


Asunto(s)
Epigénesis Genética/genética , Desarrollo Fetal/genética , Obesidad/complicaciones , Complicaciones del Embarazo/genética , Pérdida de Peso/genética , Acetilación , Animales , Dieta Alta en Grasa/efectos adversos , Epigénesis Genética/fisiología , Femenino , Desarrollo Fetal/fisiología , Retardo del Crecimiento Fetal/etiología , Retardo del Crecimiento Fetal/genética , Retardo del Crecimiento Fetal/fisiopatología , Expresión Génica/genética , Expresión Génica/fisiología , Histonas/metabolismo , Hígado/metabolismo , Ratones , Ratones Endogámicos C57BL , Obesidad/genética , Obesidad/fisiopatología , Placenta/metabolismo , Embarazo , Complicaciones del Embarazo/fisiopatología , Pérdida de Peso/fisiología
12.
Med Sci (Paris) ; 32(1): 27-34, 2016 Jan.
Artículo en Francés | MEDLINE | ID: mdl-26850604

RESUMEN

According to the new paradigm of the Developpemental Origins of Health and Disease (DOHaD), the environmental factors to which an individual is exposed throughout his life can leave an epigenetic footprint on the genome. A crucial period is the early development, where the epigenome is particularly sensitive to the effects of the environment, and during which the individual builds up his health capital that will enable him to respond more or less well to the vagaries of life. The research challenge is to decipher the modes of action and the epigenetic mechanisms put into play by environmental factors that lead to increased disease susceptibility or resilience. The challenge for health is to translate these scientific discoveries into action through, among others, the establishment of preventive recommendations to slow down the growing incidence of non communicable diseases.


Asunto(s)
Enfermedad/etiología , Ambiente , Epigénesis Genética/fisiología , Salud , Efectos Tardíos de la Exposición Prenatal/etiología , Susceptibilidad a Enfermedades/etiología , Femenino , Interacción Gen-Ambiente , Humanos , Embarazo , Efectos Tardíos de la Exposición Prenatal/genética
13.
Med Sci (Paris) ; 32(1): 35-44, 2016 Jan.
Artículo en Francés | MEDLINE | ID: mdl-26850605

RESUMEN

The existence of non-genetic and non-cultural mechanisms that transfer information on the memory of parental exposures to various environments, determining the reactivity of the following generations to their environments during their life, are of growing interest. Yet fundamental questions remain about the nature, the roles and relative importance of epigenetic marks and processes, non-coding RNAs, or other mechanisms, and their persistence over generations. A model incorporating the various transmission systems, their cross-talks and windows of susceptibility to the environment as a function of sex/gender of parent and offspring, has yet to be built.


Asunto(s)
Ambiente , Epigénesis Genética/fisiología , Efectos Tardíos de la Exposición Prenatal/etiología , Metilación de ADN/fisiología , Susceptibilidad a Enfermedades/etiología , Composición Familiar , Femenino , Interacción Gen-Ambiente , Humanos , Embarazo , Efectos Tardíos de la Exposición Prenatal/genética , ARN no Traducido/fisiología
14.
Med Sci (Paris) ; 32(1): 66-73, 2016 Jan.
Artículo en Francés | MEDLINE | ID: mdl-26850609

RESUMEN

Located at the feto-maternal interface, the placenta is involved in exchange, endocrine and immune functions, which impact fetal development. In contact with the maternal environment, this organ is sensitive to metabolic disorders as over-nutrition, obesity or diabetes. The alteration of blood parameters associated with these pathologies affects placental histology, vascularization and nutrient transfers and, according to the types of troubles, induces local inflammation or hypoxia. These placental changes lead to disturbance of development and fetal growth, which increase the risk of pathologies in offspring in adulthood. The placenta thus appears as a crucial player in the fetal programming.


Asunto(s)
Diabetes Mellitus/fisiopatología , Obesidad/fisiopatología , Placenta/fisiología , Complicaciones del Embarazo/fisiopatología , Femenino , Desarrollo Fetal/fisiología , Humanos , Intercambio Materno-Fetal/fisiología , Embarazo , Embarazo en Diabéticas/fisiopatología , Efectos Tardíos de la Exposición Prenatal/metabolismo , Efectos Tardíos de la Exposición Prenatal/fisiopatología
15.
Med Sci (Paris) ; 32(1): 93-9, 2016 Jan.
Artículo en Francés | MEDLINE | ID: mdl-26850613

RESUMEN

The human newborn is highly dependent on parental care for its survival but also for the healthy development of its brain. A large body of literature demonstrates the impact of early life adversity, even during the prenatal period, on the adult's health. The susceptibility to neuropsychiatric diseases is often potentiated by early stress. If there is an agreement that a critical developmental period exists, the mechanisms underlying the long term effects of early life adversity are still poorly understood. Recent studies in animals highlight the involvement of epigenetic processes in the transmission of such vulnerabilities, notably via modifications in germ cells, which can be transmitted in the next generations.


Asunto(s)
Susceptibilidad a Enfermedades , Trastornos Mentales/etiología , Efectos Tardíos de la Exposición Prenatal/psicología , Estrés Psicológico/complicaciones , Adulto , Animales , Encéfalo/embriología , Encéfalo/crecimiento & desarrollo , Modelos Animales de Enfermedad , Susceptibilidad a Enfermedades/psicología , Epigénesis Genética , Femenino , Humanos , Recién Nacido , Trastornos Mentales/epidemiología , Embarazo , Efectos Tardíos de la Exposición Prenatal/epidemiología , Estrés Psicológico/epidemiología
16.
Biol Aujourdhui ; 209(2): 175-87, 2015.
Artículo en Francés | MEDLINE | ID: mdl-26514387

RESUMEN

The environment, defined broadly by all that is external to the individual, conditions the phenotype during development, particularly the susceptibility to develop non-communicable diseases. This notion, called Developmental Origins of Health and Disease (DOHaD), is based on numerous epidemiological studies as well as animal models. Thus, parental nutrition and obesity can predispose the offspring to develop metabolic and cardiovascular diseases in adulthood. The known underlying mechanisms include an altered development of tissues that adapt to maternal metabolic condition, and a placental dysfunction, which in turn impacts fetal growth and development. Epigenetic mechanisms modulate gene expression without affecting the DNA sequence itself. The main epigenetic marks are DNA methylation and histone post-translational modifications. These marks are erased and set-up during gametogenesis and development in order to ensure cellular identity. Therefore, they can lead to a memorisation of early environment and induce long-term alteration of cell and tissue functions, which will condition the susceptibility to non-communicable diseases. The placenta is a programming agent of adult disease. The environment, such as smoking or psychosocial stress, is able to modify epigenetic processes in placenta, such as small RNA expression and DNA methylation. We showed that placenta is sensitive to maternal obesity and maternal nutrition, in terms of histology, transcription and epigenetic marks. A clear sexual dimorphism is remarkable in the placental response to maternal environment. In adulthood, the phenotype is also different between males and females. Epigenetic mechanisms could underlie this differential response of males and females to the same environment. The DOHaD can no longer be ignored in Biology of Reproduction. The prevention of non-communicable diseases must take this new paradigm into account. Research will allow a better comprehension of the mechanisms of this early conditioning and the marked sexual dimorphism it is associated to.


Asunto(s)
Desarrollo Embrionario , Epigénesis Genética , Fenómenos Fisiologicos Nutricionales Maternos , Placenta/fisiología , Adulto , Animales , Enfermedades Cardiovasculares/embriología , Enfermedades Cardiovasculares/fisiopatología , Metilación de ADN , Dieta Alta en Grasa/efectos adversos , Susceptibilidad a Enfermedades , Desarrollo Embrionario/genética , Femenino , Trastornos Nutricionales en el Feto/etiología , Trastornos Nutricionales en el Feto/prevención & control , Histonas/metabolismo , Humanos , Recién Nacido , Masculino , Desnutrición/fisiopatología , Síndrome Metabólico/embriología , Síndrome Metabólico/fisiopatología , Ratones , Modelos Biológicos , Obesidad/embriología , Obesidad/fisiopatología , Placenta/fisiopatología , Embarazo , Complicaciones del Embarazo/fisiopatología , Efectos Tardíos de la Exposición Prenatal , Fenómenos Fisiologicos de la Nutrición Prenatal , Procesamiento Proteico-Postraduccional , Conejos , Caracteres Sexuales
17.
J Exp Biol ; 218(Pt 1): 50-8, 2015 Jan 01.
Artículo en Inglés | MEDLINE | ID: mdl-25568451

RESUMEN

The recent and rapid worldwide increase in non-communicable diseases challenges the assumption that genetic factors are the primary contributors to such diseases. A new concept of the 'developmental origins of health and disease' (DOHaD) is at stake and therefore requires a paradigm shift. Maternal obesity and malnutrition predispose offspring to develop metabolic syndrome, a vicious cycle leading to transmission to subsequent generation(s), with differences in response and susceptibility according to the sex of the individual. The placenta is a programming agent of adult health and disease. Adaptations of placental phenotype in response to maternal diet and metabolic status alter fetal nutrient supply. This implies important epigenetic changes that are, however, still poorly documented in DOHaD studies, particularly concerning overnutrition. The aim of this review is to discuss the emerging knowledge on the relationships between the effect of maternal nutrition or metabolic status on placental function and the risk of diseases later in life, with a specific focus on epigenetic mechanisms and sexual dimorphism. Explaining the sex-specific causal variables and how males versus females respond and adapt to environmental perturbations should help physicians and patients to anticipate disease susceptibility.


Asunto(s)
Enfermedad/genética , Epigénesis Genética , Salud , Fenómenos Fisiologicos Nutricionales Maternos , Placenta/metabolismo , Animales , Femenino , Embarazo , Caracteres Sexuales
18.
PLoS One ; 9(11): e111556, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-25369064

RESUMEN

Once daily milking (ODM) induces a reduction in milk production when compared to twice daily milking (TDM). Unilateral ODM of one udder half and TDM of the other half, enables the study of underlying mechanisms independently of inter-individual variability (same genetic background) and of environmental factors. Our results show that in first-calf heifers three CpG, located 10 kb upstream from the CSN1S1 gene were methylated to 33, 34 and 28%, respectively, after TDM but these levels were higher after ODM, 38, 38 and 33%, respectively. These methylation levels were much lower than those observed in the mammary gland during pregnancy (57, 59 and 50%, respectively) or in the liver (74, 78 and 61%, respectively). The methylation level of a fourth CpG (CpG4), located close by (29% during TDM) was not altered after ODM. CpG4 methylation reached 39.7% and 59.5%, during pregnancy or in the liver, respectively. CpG4 is located within a weak STAT5 binding element, arranged in tandem with a second high affinity STAT5 element. STAT5 binding is only marginally modulated by CpG4 methylation, but it may be altered by the methylation levels of the three other CpG nearby. Our results therefore shed light on mechanisms that help to explain how milk production is almost, but not fully, restored when TDM is resumed (15.1 ± 0.2 kg/day instead of 16.2 ± 0.2 kg/day, p<0.01). The STAT5 elements are 100 bp away from a region transcribed in the antisense orientation, in the mammary gland during lactation, but not during pregnancy or in other reproductive organs (ovary or testes). We now need to clarify whether the transcription of this novel RNA is a consequence of STAT5 interacting with the CSN1S1 distal region, or whether it plays a role in the chromatin structure of this region.


Asunto(s)
Caseínas/genética , Metilación de ADN , Lactancia , Leche/química , Fragmentos de Péptidos/genética , Animales , Secuencia de Bases , Bovinos , Industria Lechera , Femenino , Glándulas Mamarias Animales/ultraestructura , Datos de Secuencia Molecular , Familia de Multigenes , Transcripción Genética
19.
PLoS One ; 8(6): e66816, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-23826145

RESUMEN

According to the developmental origins of health and diseases (DOHaD), and in line with the findings of many studies, obesity during pregnancy is clearly a threat to the health and well-being of the offspring, later in adulthood. We previously showed that 20% of male and female inbred mice can cope with the obesogenic effects of a high-fat diet (HFD) for 20 weeks after weaning, remaining lean. However the feeding of a control diet (CD) to DIO mice during the periconceptional/gestation/lactation period led to a pronounced sex-specific shift (17% to 43%) from susceptibility to resistance to HFD, in the female offspring only. Our aim in this study was to determine how, in the context of maternal obesity and T2D, a CD could increase resistance on female fetuses. Transcriptional analyses were carried out with a custom-built mouse liver microarray and by quantitative RT-PCR for muscle and adipose tissue. Both global DNA methylation and levels of pertinent histone marks were assessed by LUMA and western blotting, and the expression of 15 relevant genes encoding chromatin-modifying enzymes was analyzed in tissues presenting global epigenetic changes. Resistance was associated with an enhancement of hepatic pathways protecting against steatosis, the unexpected upregulation of neurotransmission-related genes and the modulation of a vast imprinted gene network. Adipose tissue displayed a pronounced dysregulation of gene expression, with an upregulation of genes involved in lipid storage and adipocyte hypertrophy or hyperplasia in obese mice born to lean and obese mothers, respectively. Global DNA methylation, several histone marks and key epigenetic regulators were also altered. Whether they were themselves lean (resistant) or obese (sensitive), the offspring of lean and obese mice clearly differed in terms of several metabolic features and epigenetic marks suggesting that the effects of a HFD depend on the leanness or obesity of the mother.


Asunto(s)
Dieta , Epigénesis Genética , Perfilación de la Expresión Génica , Obesidad/genética , Transcripción Genética , Tejido Adiposo/metabolismo , Tejido Adiposo/patología , Animales , Diabetes Mellitus Experimental/genética , Dieta Alta en Grasa , Femenino , Regulación de la Expresión Génica , Redes Reguladoras de Genes/genética , Impresión Genómica/genética , Hígado/metabolismo , Hígado/patología , Masculino , Ratones , Fenotipo , Embarazo , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Transmisión Sináptica/genética
20.
Biol Sex Differ ; 4(1): 5, 2013 Mar 21.
Artículo en Inglés | MEDLINE | ID: mdl-23514128

RESUMEN

Sex differences occur in most non-communicable diseases, including metabolic diseases, hypertension, cardiovascular disease, psychiatric and neurological disorders and cancer. In many cases, the susceptibility to these diseases begins early in development. The observed differences between the sexes may result from genetic and hormonal differences and from differences in responses to and interactions with environmental factors, including infection, diet, drugs and stress. The placenta plays a key role in fetal growth and development and, as such, affects the fetal programming underlying subsequent adult health and accounts, in part for the developmental origin of health and disease (DOHaD). There is accumulating evidence to demonstrate the sex-specific relationships between diverse environmental influences on placental functions and the risk of disease later in life. As one of the few tissues easily collectable in humans, this organ may therefore be seen as an ideal system for studying how male and female placenta sense nutritional and other stresses, such as endocrine disruptors. Sex-specific regulatory pathways controlling sexually dimorphic characteristics in the various organs and the consequences of lifelong differences in sex hormone expression largely account for such responses. However, sex-specific changes in epigenetic marks are generated early after fertilization, thus before adrenal and gonad differentiation in the absence of sex hormones and in response to environmental conditions. Given the abundance of X-linked genes involved in placentogenesis, and the early unequal gene expression by the sex chromosomes between males and females, the role of X- and Y-chromosome-linked genes, and especially those involved in the peculiar placenta-specific epigenetics processes, giving rise to the unusual placenta epigenetic landscapes deserve particular attention. However, even with recent developments in this field, we still know little about the mechanisms underlying the early sex-specific epigenetic marks resulting in sex-biased gene expression of pathways and networks. As a critical messenger between the maternal environment and the fetus, the placenta may play a key role not only in buffering environmental effects transmitted by the mother but also in expressing and modulating effects due to preconceptional exposure of both the mother and the father to stressful conditions.

SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...