Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 42
Filtrar
1.
Nutrients ; 15(2)2023 Jan 16.
Artigo em Inglês | MEDLINE | ID: mdl-36678336

RESUMO

Perinatal nutrition is a key player in the susceptibility to developing metabolic diseases in adulthood, leading to the concept of "metabolic programming". The aim of this study was to assess the impact of maternal protein restriction during gestation and lactation on glucose homeostasis and eating behaviour in female offspring. Pregnant rats were fed a normal or protein-restricted (PR) diet and followed throughout gestation and lactation. Body weight, glucose homeostasis, and eating behaviour were evaluated in offspring, especially in females. Body weight gain was lower in PR dams during lactation only, despite different food and water intakes throughout gestation and lactation. Plasma concentration of leptin, adiponectin and triglycerides increased drastically before delivery in PR dams in relation to fat deposits. Although all pups had identical birth body weight, PR offspring body weight differed from control offspring around postnatal day 10 and remained lower until adulthood. Offspring glucose homeostasis was mildly impacted by maternal PR, although insulin secretion was reduced for PR rats at adulthood. Food intake, satiety response, and cerebral activation were examined after a lipid preload and demonstrated some differences between the two groups of rats. Maternal PR during gestation and lactation does induce extrauterine growth restriction, accompanied by alterations in maternal plasma leptin and adiponectin levels, which may be involved in programming the alterations in eating behaviour observed in females at adulthood.


Assuntos
Dieta com Restrição de Proteínas , Leptina , Animais , Feminino , Gravidez , Ratos , Adiponectina/metabolismo , Peso Corporal , Encéfalo/metabolismo , Dieta com Restrição de Proteínas/efeitos adversos , Glucose , Lactação/metabolismo , Fenômenos Fisiológicos da Nutrição Materna , Triglicerídeos , Aumento de Peso
2.
Front Microbiol ; 12: 672224, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34211445

RESUMO

Understanding the link between mother's obesity and regulation of the child's appetite is a prerequisite for the design of successful preventive strategies. Beyond the possible contributions of genetic heritage, family culture, and hormonal and metabolic environment during pregnancy, we investigate in the present paper the causal role of the transmission of the maternal microbiotas in obesity as microbiotas differ between lean and obese mothers, maternal microbiotas are the main determinants of a baby's gut colonization, and the intestinal microbiota resulting from the early colonization could impact the feeding behavior of the offspring with short- and long-term consequences on body weight. We thus investigated the potential role of vertical transfers of maternal microbiotas in programming the eating behavior of the offspring. Selectively bred obese-prone (OP)/obese-resistant (OR) Sprague-Dawley dams were used since differences in the cecal microbiota have been evidenced from males of that strain. Microbiota collected from vagina (at the end of gestation), feces, and milk (at postnatal days 1, 5, 10, and 15) of OP/OR dams were orally inoculated to conventional Fischer F344 recipient pups from birth to 15 days of age to create three groups of pups: F-OP, F-OR, and F-Sham group (that received the vehicle). We first checked microbiotal differences between inoculas. We then assessed the impact of transfer (from birth to adulthood) onto the intestinal microbiota of recipients rats, their growth, and their eating behavior by measuring their caloric intake, their anticipatory food reward responses, their preference for sweet and fat tastes in solutions, and the sensations that extend after food ingestion. Finally, we searched for correlation between microbiota composition and food intake parameters. We found that maternal transfer of microbiota differing in composition led to alterations in pups' gut microbiota composition that did not last until adulthood but were associated with specific eating behavior characteristics that were predisposing F-OP rats to higher risk of over consuming at subsequent periods of their life. These findings support the view that neonatal gut microbiotal transfer can program eating behavior, even without a significant long-lasting impact on adulthood microbiota composition.

3.
Int J Mol Sci ; 21(15)2020 Jul 30.
Artigo em Inglês | MEDLINE | ID: mdl-32751478

RESUMO

Lactation is a critical period during which maternal sub- or over-nutrition affect milk composition and offspring development that can have lasting health effects. The consequences of moderate high-fat, high-simple carbohydrate diet (WD) consumption by rat dams, during gestation and lactation, on milk composition and offspring blood lipidome and its growth, at weaning, were investigated by using a comprehensive lipidomic study on mass-spectrometric platform combined to targeted fatty- and free amino-acids analysis. This holistic approach allowed clear-cut differences in mature milk-lipidomic signature according to maternal diet with a similar content of protein, lactose and leptin. The lower WD-milk content in total fat and triglycerides (TGs), particularly in TGs-with saturated medium-chain, and higher levels in both sphingolipid (SL) and TG species with unsaturated long-chain were associated to a specific offspring blood-lipidome with decreased levels in TGs-containing saturated fatty acid (FA). The sexual-dimorphism in the FA-distribution in TG (higher TGs-rich in oleic and linoleic acids, specifically in males) and SL species (increased levels in very long-chain ceramides, specifically in females) could be associated with some differences that we observed between males and females like a higher total body weight gain in females and an increased preference for fatty taste in males upon weaning.


Assuntos
Dieta Hiperlipídica , Lipidômica , Lipídeos , Fenômenos Fisiológicos da Nutrição Materna , Leite/química , Animais , Feminino , Humanos , Recém-Nascido , Lactação , Lipídeos/análise , Lipídeos/sangue , Masculino , Gravidez , Ratos , Ratos Sprague-Dawley , Desmame
4.
Nutrients ; 12(5)2020 May 19.
Artigo em Inglês | MEDLINE | ID: mdl-32438566

RESUMO

Fetal brain development is closely dependent on maternal nutrition and metabolic status. Maternal protein restriction (PR) is known to be associated with alterations in the structure and function of the hypothalamus, leading to impaired control of energy homeostasis and food intake. The objective of this study was to identify the cellular and molecular systems underlying these effects during fetal development. We combined a global transcriptomic analysis on the fetal hypothalamus from a rat model of maternal PR with in vitro neurosphere culture and cellular analyses. Several genes encoding proteins from the mitochondrial respiratory chain complexes were overexpressed in the PR group and mitochondrial metabolic activity in the fetal hypothalamus was altered. The level of the N6-methyladenosine epitranscriptomic mark was reduced in the PR fetuses, and the expression of several genes involved in the writing/erasing/reading of this mark was indeed altered, as well as genes encoding several RNA-binding proteins. Additionally, we observed a higher number of neuronal-committed progenitors at embryonic day 17 (E17) in the PR fetuses. Together, these data strongly suggest a metabolic adaptation to the amino acid shortage, combined with the post-transcriptional control of protein expression, which might reflect alterations in the control of the timing of neuronal progenitor differentiation.


Assuntos
Dieta com Restrição de Proteínas/efeitos adversos , Feto/metabolismo , Hipotálamo/embriologia , Fenômenos Fisiológicos da Nutrição Materna/genética , Mitocôndrias/genética , Animais , Feminino , Desenvolvimento Fetal/genética , Hipotálamo/metabolismo , Gravidez , Ratos
5.
Nutrients ; 11(11)2019 Nov 08.
Artigo em Inglês | MEDLINE | ID: mdl-31717308

RESUMO

The pancreas has an essential role in the regulation of glucose homeostasis by secreting insulin, the only hormone with a blood glucose lowering effect in mammals. Several circulating molecules are able to positively or negatively influence insulin secretion. Among them, nutrients such as fatty acids or amino acids can directly act on specific receptors present on pancreatic beta cells. Dietary intake, especially excessive nutrient intake, is known to modify energy balance in adults, resulting in pancreatic dysfunction. However, gestation and lactation are critical periods for fetal development and pup growth and specific dietary nutrients are required for optimal growth. Feeding alterations during these periods will impact offspring development and increase the risk of developing metabolic disorders in adulthood, leading to metabolic programming. This review will focus on the influence of nutrient intake during gestation and lactation periods on pancreas development and function in offspring, highlighting the molecular mechanism of imprinting on this organ.


Assuntos
Dieta , Ilhotas Pancreáticas/metabolismo , Lactação , Fenômenos Fisiológicos da Nutrição Materna , Estado Nutricional , Valor Nutritivo , Efeitos Tardios da Exposição Pré-Natal , Animais , Desenvolvimento Infantil , Dieta/efeitos adversos , Comportamento Alimentar , Feminino , Desenvolvimento Fetal , Idade Gestacional , Humanos , Recém-Nascido , Ilhotas Pancreáticas/crescimento & desenvolvimento , Ilhotas Pancreáticas/patologia , Gravidez , Recomendações Nutricionais
6.
Nutrients ; 11(9)2019 Aug 21.
Artigo em Inglês | MEDLINE | ID: mdl-31438620

RESUMO

Oligosaccharides (OS) are commonly added to infant formulas, however, their physiological impact, particularly on adult health programming, is poorly described. In adult animals, OS modify microbiota and stimulate colonic fermentation and enteroendocrine cell (EEC) activity. Since neonatal changes in microbiota and/or EEC density could be long-lasting and EEC-derived peptides do regulate short-term food intake, we hypothesized that neonatal OS consumption could modulate early EECs, with possible consequences for adult eating behavior. Suckling rats were supplemented with fructo-oligosaccharides (FOS), beta-galacto-oligosaccharides/inulin (GOS/In) mix, alpha-galacto-oligosaccharides (αGOS) at 3.2 g/kg, or a control solution (CTL) between postnatal day (PND) 5 and 14/15. Pups were either sacrificed at PND14/15 or weaned at PND21 onto standard chow. The effects on both microbiota and EEC were characterized at PND14/15, and eating behavior at adulthood. Very early OS supplementation drastically impacted the intestinal environment, endocrine lineage proliferation/differentiation particularly in the ileum, and the density of GLP-1 cells and production of satiety-related peptides (GLP-1 and PYY) in the neonatal period. However, it failed to induce any significant lasting changes on intestinal microbiota, enteropeptide secretion or eating behavior later in life. Overall, the results did not demonstrate any OS programming effect on satiety peptides secreted by L-cells or on food consumption, an observation which is a reassuring outlook from a human perspective.


Assuntos
Envelhecimento/fisiologia , Comportamento Alimentar , Oligossacarídeos/química , Oligossacarídeos/farmacologia , Animais , Animais Recém-Nascidos , Bactérias/classificação , Bactérias/genética , Suplementos Nutricionais , Conteúdo Gastrointestinal , Masculino , Oligossacarídeos/administração & dosagem , RNA Ribossômico 16S , Ratos , Ratos Sprague-Dawley , Paladar
7.
Psychopharmacology (Berl) ; 236(5): 1583-1596, 2019 May.
Artigo em Inglês | MEDLINE | ID: mdl-31147734

RESUMO

RATIONALE: Intestinal permeability plays an important role in gut-brain axis communication. Recent studies indicate that intestinal permeability increases in neonate pups during maternal separation (MS). OBJECTIVES: The present study aims to determine whether pharmacological inhibition of myosin light chain kinase (MLCK), which regulates tight junction contraction and controls intestinal permeability, in stressed neonates, protects against the long-term effects of MS. METHODS: Male Wistar rats were exposed to MS (3 h per day from post-natal day (PND)2 to PND14) or left undisturbed and received daily intraperitoneal injection of a MLCK inhibitor (ML-7, 5 mg/kg) or vehicle during the same period. At adulthood, emotional behaviors, corticosterone response to stress, and gut microbiota composition were analyzed. RESULTS: ML-7 restored gut barrier function in MS rats specifically during the neonatal period. Remarkably, ML-7 prevented MS-induced sexual reward-seeking impairment and reversed the alteration of corticosterone response to stress at adulthood. The effects of ML-7 were accompanied by the normalization of the abundance of members of Lachnospiraceae, Clostridiales, Desulfovibrio, Bacteroidales, Enterorhabdus, and Bifidobacterium in the feces of MS rats at adulthood. CONCLUSIONS: Altogether, our work suggests that improvement of intestinal barrier defects during development may alleviate some of the long-term effects of early-life stress and provides new insight on brain-gut axis communication in a context of stress.


Assuntos
Azepinas/farmacologia , Microbioma Gastrointestinal/efeitos dos fármacos , Mucosa Intestinal/efeitos dos fármacos , Mucosa Intestinal/metabolismo , Privação Materna , Naftalenos/farmacologia , Estresse Psicológico/metabolismo , Animais , Animais Recém-Nascidos , Azepinas/uso terapêutico , Corticosterona/metabolismo , Relação Dose-Resposta a Droga , Feminino , Microbioma Gastrointestinal/fisiologia , Masculino , Quinase de Cadeia Leve de Miosina/farmacologia , Quinase de Cadeia Leve de Miosina/uso terapêutico , Naftalenos/uso terapêutico , Gravidez , Ratos , Ratos Wistar , Estresse Psicológico/tratamento farmacológico , Estresse Psicológico/psicologia , Fatores de Tempo
8.
Arch Oral Biol ; 96: 87-95, 2018 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-30205238

RESUMO

OBJECTIVE: Maternal diet has consequences on many organs of the offspring, but salivary glands have received little attention despite the importance of the saliva secretory function in oral health and control of food intake. The objective of this work was therefore to document in rats the impact of maternal high-fat/high-sugar diet (Western Diet) on submandibular glands of the progeny. DESIGN: Sprague-Dawley rat dams were fed either a Western diet or control diet during gestation and lactation and their pups were sacrificed 25 days after birth. The pups' submandibular gland protein content was characterized by means of 2D-electrophoresis followed by LC-MS/MS. Data were further analyzed by Gene Ontology enrichment analysis and protein-protein interactions mapping. The expression of two specific proteins was also evaluated using immunohistochemistry. RESULTS: Combining both male and female pups (n = 18), proteome analysis revealed that proteins involved in protein quality control (e.g. heat shock proteins, proteasome sub-units) and microtubule proteins were over-expressed in Western diet conditions, which may translate intense metabolic activity. A cluster of proteins controlling oxidative stress (e.g. Glutathione peroxidases, peroxiredoxin) and enhancement of the antioxidant activity molecular function were also characteristic of maternal Western diet as well as under-expression of annexin A5. The down-regulating effect of maternal Western diet on Annexin A5 expression was significant only for males (p < 0.05). CONCLUSIONS: A maternal Western diet modifies the protein composition of the offspring's salivary glands, which may have consequences on the salivary function.


Assuntos
Dieta Ocidental , Proteínas e Peptídeos Salivares/metabolismo , Glândula Submandibular/metabolismo , Animais , Feminino , Imuno-Histoquímica , Masculino , Estresse Oxidativo , Gravidez , Efeitos Tardios da Exposição Pré-Natal , Ratos , Ratos Sprague-Dawley
9.
J Nutr Biochem ; 55: 124-141, 2018 05.
Artigo em Inglês | MEDLINE | ID: mdl-29413487

RESUMO

Perinatal undernutrition affects not only fetal and neonatal growth but also adult health outcome, as suggested by the metabolic imprinting concept. However, the exact mechanisms underlying offspring metabolic adaptations are not yet fully understood. Specifically, it remains unclear whether the gestation or the lactation is the more vulnerable period to modify offspring metabolic flexibility. We investigated in a rodent model of intrauterine growth restriction (IUGR) induced by maternal protein restriction (R) during gestation which time window of maternal undernutrition (gestation, lactation or gestation-lactation) has more impact on the male offspring metabolomics phenotype. Plasma metabolome and hepatic lipidome of offspring were characterized through suckling period and at adulthood using liquid chromatography-high-resolution mass spectrometry. Multivariate analysis of these fingerprints highlighted a persistent metabolomics signature in rats suckled by R dams, with a clear-cut discrimination from offspring fed by control (C) dams. Pups submitted to a nutritional switch at birth presented a metabolomics signature clearly distinct from that of pups nursed by dams maintained on a consistent perinatal diet. Control rats suckled by R dams presented transiently higher branched-chain amino acid (BCAA) oxidation during lactation besides increased fatty acid (FA) ß-oxidation, associated with preserved insulin sensitivity and lesser fat accretion that persisted throughout their life. In contrast, IUGR rats displayed permanently impaired ß-oxidation, associated to increased glucose or BCAA oxidation at adulthood, depending on the fact that pups experienced slow postnatal or catch-up growth, as suckled by R or C dams, respectively. Taken together, these findings provide evidence for a significant contribution of the lactation period in metabolic programming.


Assuntos
Sangue/metabolismo , Lactação , Fígado/metabolismo , Fenômenos Fisiológicos da Nutrição Materna , Animais , Animais Lactentes , Antioxidantes/metabolismo , Proteínas Alimentares/administração & dosagem , Ácidos Graxos/metabolismo , Feminino , Retardo do Crescimento Fetal , Metabolismo dos Lipídeos , Fígado/fisiologia , Masculino , Metaboloma , Gravidez , Ratos Sprague-Dawley , Transcriptoma
10.
FASEB J ; 32(4): 2160-2171, 2018 04.
Artigo em Inglês | MEDLINE | ID: mdl-29242276

RESUMO

Butyrate can improve gut functions, whereas histone deacetylase inhibitors might alleviate neurocognitive alterations. Our aim was to assess whether oral butyrate could modulate brain metabolism and plasticity and if this would relate to gut function. Sixteen pigs were subjected to sodium butyrate (SB) supplementation via beverage water or water only [control (C)]. All pigs had blood sampled after 2 and 3 wk of treatment, and were subjected to a brain positron emission tomography after 3 wk. Animals were euthanized after 4 wk to sample pancreas, intestine, and brain for gut physiology and anatomy measurements, as well as hippocampal histology, Ki67, and doublecortin (DCX) immunohistochemistry. SB compared with C treatment triggered basal brain glucose metabolism changes in the nucleus accumbens and hippocampus ( P = 0.003), increased hippocampal granular cell layer volume ( P = 0.006), and neurogenesis (Ki67: P = 0.026; DCX: P = 0.029). After 2 wk of treatment, plasma levels of glucose, insulin, lactate, glucagon-like peptide 1, and peptide tyrosine tyrosine remained unchanged. After 3 wk, plasma levels of lactate were lower in SB compared with C animals ( P = 0.028), with no difference for glucose and insulin. Butyrate intake impacted very little gut anatomy and function. These results demonstrate that oral SB impacted brain functions with little effects on the gut.-Val-Laillet, D., Guérin, S., Coquery, N., Nogret, I., Formal, M., Romé, V., Le Normand, L., Meurice, P., Randuineau, G., Guilloteau, P., Malbert, C.-H., Parnet, P., Lallès, J.-P., Segain, J.-P. Oral sodium butyrate impacts brain metabolism and hippocampal neurogenesis, with limited effects on gut anatomy and function in pigs.


Assuntos
Ácido Butírico/farmacologia , Hipocampo/efeitos dos fármacos , Antagonistas dos Receptores Histamínicos/farmacologia , Intestinos/efeitos dos fármacos , Neurogênese , Administração Oral , Animais , Glicemia/metabolismo , Ácido Butírico/administração & dosagem , Ácido Butírico/efeitos adversos , Feminino , Hipocampo/crescimento & desenvolvimento , Hipocampo/metabolismo , Antagonistas dos Receptores Histamínicos/administração & dosagem , Antagonistas dos Receptores Histamínicos/efeitos adversos , Insulina/sangue , Intestinos/fisiologia , Ácido Láctico/sangue , Suínos
11.
Artigo em Inglês | MEDLINE | ID: mdl-28900415

RESUMO

Perinatal maternal consumption of energy dense food increases the risk of obesity in children. This is associated with an overconsumption of palatable food that is consumed for its hedonic property. The underlying mechanism that links perinatal maternal diet and offspring preference for fat is still poorly understood. In this study, we aim at studying the influence of maternal high-fat/high-sugar diet feeding [western diet (WD)] during gestation and lactation on the reward pathways controlling feeding in the rat offspring from birth to sexual maturity. We performed a longitudinal follow-up of WD and Control offspring at three critical time periods (childhood, adolescence, and adulthood) and focus on investigating the influence of perinatal exposure to palatable diet on (i) fat preference, (ii) gene expression profile, and (iii) neuroanatomical/architectural changes of the mesolimbic dopaminergic networks. We showed that WD feeding restricted to the perinatal period has a clear long-lasting influence on the organization of homeostatic and hedonic brain circuits but not on fat preference. We demonstrated a period specific evolution of the preference for fat that we correlated with specific brain molecular signatures. In offspring from WD fed dams, we observed during childhood the existence of fat preference associated with a higher expression of key gene involved in the dopamine (DA) systems; at adolescence, a high-fat preference for both groups, progressively reduced during the 3 days test for the WD group and associated with a reduced expression of key gene involved in the DA systems for the WD group that could suggest a compensatory mechanism to protect them from further high-fat exposure; and finally at adulthood, a preference for fat that was identical to control rats but associated with profound modification in key genes involved in the γ-aminobutyric acid network, serotonin receptors, and polysialic acid-NCAM-dependent remodeling of the hypothalamus. Altogether, these data reveal that maternal WD, restricted to the perinatal period, has no sustained impact on energy homeostasis and fat preference later in life even though a strong remodeling of the hypothalamic homeostatic and reward pathway involved in eating behavior occurred. Further functional experiments would be needed to understand the relevance of these circuits remodeling.

12.
Artigo em Inglês | MEDLINE | ID: mdl-28443064

RESUMO

Perinatal malnutrition is associated with low birth weight and an increased risk of developing metabolic syndrome in adulthood. Modification of food intake (FI) regulation was observed in adult rats born with intrauterine growth retardation induced by maternal dietary protein restriction during gestation and maintained restricted until weaning. Gastrointestinal peptides and particularly cholecystokinin (CCK) play a major role in short-term regulation of FI by relaying digestive signals to the hindbrain via the vagal afferent nerve (VAN). We hypothesized that vagal sensitivity to CCK could be affected in rats suffering from undernutrition [low protein (LP)] during fetal and postnatal life, leading to an altered gut-brain communication and impacting satiation. Our aim was to study short-term FI along with signals of appetite and satiation in adult LP rats compared to control rats. The dose-response to CCK injection was investigated on FI as well as the associated signaling pathways activated in nodose ganglia. We showed that LP rats have a reduced first-meal satiety ratio after a fasting period associated to a higher postprandial plasmatic CCK release, a reduced sensitivity to CCK when injected at low concentration and a reduced presence of CCK-1 receptor in nodose ganglia. Accordingly, the lower basal and CCK-induced phosphorylation of calcium/calmodulin-dependent protein kinase in nodose ganglia of LP rats could reflect an under-expressed vanilloid family of transient receptor potential cation channels on VAN. Altogether, the present data demonstrated a reduced vagal sensitivity to CCK in LP rats at adulthood, which could contribute to deregulation of FI reported in this model.

13.
Future Sci OA ; 3(1): FSO164, 2017 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-28344827

RESUMO

AIM: To investigate the effect of a protein restriction and a supplementation with methyl donor nutrients during fetal and early postnatal life on the expression and epigenetic state of imprinted genes from the IGF system. MATERIALS & METHODS: Pregnant female rats were fed a protein-restricted diet supplemented or not with methyl donor. RESULTS: Gene expression of the Igf2, H19, Igf1, Igf2r and Plagl1 genes in the liver of male offspring at birth and weaning was strongly influenced by maternal diet. Whereas the methylation profiles of the Igf2, H19 and Igf2r genes were remarkably stable, DNA methylation of Plagl1 promoter was slightly modified. CONCLUSION: DNA methylation of most, but not all, imprinted gene regulatory regions was resistant to methyl group nutritional supply.

14.
Med Sci (Paris) ; 32(1): 27-34, 2016 Jan.
Artigo em Francês | MEDLINE | ID: mdl-26850604

RESUMO

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.


Assuntos
Doença/etiologia , Meio Ambiente , Epigênese Genética/fisiologia , Saúde , Efeitos Tardios da Exposição Pré-Natal/etiologia , Suscetibilidade a Doenças/etiologia , Feminino , Interação Gene-Ambiente , Humanos , Gravidez , Efeitos Tardios da Exposição Pré-Natal/genética
15.
Med Sci (Paris) ; 32(1): 35-44, 2016 Jan.
Artigo em Francês | MEDLINE | ID: mdl-26850605

RESUMO

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.


Assuntos
Meio Ambiente , Epigênese Genética/fisiologia , Efeitos Tardios da Exposição Pré-Natal/etiologia , Metilação de DNA/fisiologia , Suscetibilidade a Doenças/etiologia , Características da Família , Feminino , Interação Gene-Ambiente , Humanos , Gravidez , Efeitos Tardios da Exposição Pré-Natal/genética , RNA não Traduzido/fisiologia
16.
Med Sci (Paris) ; 32(1): 85-92, 2016 Jan.
Artigo em Francês | MEDLINE | ID: mdl-26850612

RESUMO

Eating behavior is strongly regulated by intrinsic physiological factors and largely influenced by the individual and cultural environments. Excessive food intake and sedentary lifestyle are the main reasons for the global epidemic of obesity. The influence of family background on eating habits makes no doubt but the fact that the nutritional, metabolic and hormonal status of the parents before conception, and of the mother during gestation and lactation, may influence the child's future eating behavior is an innovative concept that opens the way for preventive policies. In the last decades, research on human cohorts and animal models have targeted biological mechanisms (neuroanatomical, epigenetic) that give some clues on how eating behavior can be formatted by early nutrition and related sensory experience.


Assuntos
Comportamento Alimentar , Efeitos Tardios da Exposição Pré-Natal , Fenômenos Fisiológicos da Nutrição Pré-Natal , Sensação/fisiologia , Adulto , Filhos Adultos , Animais , Criança , Comportamento Alimentar/fisiologia , Comportamento Alimentar/psicologia , Feminino , Humanos , Recém-Nascido , Masculino , Gravidez , Efeitos Tardios da Exposição Pré-Natal/fisiopatologia , Efeitos Tardios da Exposição Pré-Natal/psicologia
17.
J Nutr ; 146(3): 532-41, 2016 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-26865647

RESUMO

BACKGROUND: Intrauterine growth restriction (IUGR) results from either maternal undernutrition or impaired placental blood flow, exposing offspring to increased perinatal mortality and a higher risk of metabolic syndrome and cardiovascular disease during adulthood. l-Citrulline is a precursor of l-arginine and nitric oxide (NO), which regulates placental blood flow. Moreover, l-citrulline stimulates protein synthesis in other models of undernutrition. OBJECTIVE: The aim of the study was to determine whether l-citrulline supplementation would enhance fetal growth in a model of IUGR induced by maternal dietary protein restriction. METHODS: Pregnant rats were fed either a control (20% protein) or a low-protein (LP; 4% protein) diet. LP dams were randomly allocated to drink tap water either as such or supplemented with l-citrulline (2 g · kg(-1) · d(-1)), an isonitrogenous amount of l-arginine, or nonessential l-amino acids (NEAAs). On day 21 of gestation, dams received a 2-h infusion of l-[1-(13)C]-valine until fetuses were extracted by cesarean delivery. Isotope enrichments were measured in free amino acids and fetal muscle, liver, and placenta protein by GC-mass spectrometry. RESULTS: Fetal weight was ∼29% lower in the LP group (3.82 ± 0.06 g) than in the control group (5.41 ± 0.10 g) (P < 0.001). Regardless of supplementation, fetal weight remained below that of control fetuses. Yet, compared with the LP group, l-citrulline and l-arginine equally increased fetal weight to 4.15 ± 0.08 g (P < 0.05) and 4.13 ± 0.1 g (P < 0.05 compared with LP), respectively, whereas NEAA did not (4.05 ± 0.05 g; P = 0.07). Fetal muscle protein fractional synthesis rate was 35% lower in the LP fetuses (41% ± 11%/d) than in the control (61% ± 13%/d) fetuses (P < 0.001) and was normalized by l-citrulline (56% ± 4%/d; P < 0.05 compared with LP, NS compared with control) and not by other supplements. Urinary nitrite and nitrate excretion was lower in the LP group (6.4 ± 0.8 µmol/d) than in the control group (17.9 ± 1.1 µmol/d; P < 0.001) and increased in response to l-citrulline or l-arginine (12.1 ± 2.2 and 10.6 ± 0.9 µmol/d; P < 0.05), whereas they did not in the LP + NEAA group. CONCLUSION: l-Citrulline increases fetal growth in a model of IUGR, and the effect may be mediated by enhanced fetal muscle protein synthesis and/or increased NO production.


Assuntos
Citrulina/administração & dosagem , Suplementos Nutricionais , Desenvolvimento Fetal/efeitos dos fármacos , Retardo do Crescimento Fetal/tratamento farmacológico , Fenômenos Fisiológicos da Nutrição Materna , Animais , Arginina/metabolismo , Dieta com Restrição de Proteínas/efeitos adversos , Feminino , Peso Fetal/efeitos dos fármacos , Feto/efeitos dos fármacos , Feto/metabolismo , Óxido Nítrico/metabolismo , Estado Nutricional , Placenta/efeitos dos fármacos , Placenta/metabolismo , Gravidez , Biossíntese de Proteínas/efeitos dos fármacos , Ratos , Ratos Sprague-Dawley
18.
J Nutrigenet Nutrigenomics ; 8(4-6): 153-63, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-26629831

RESUMO

BACKGROUND/AIMS: Children born preterm are more likely than full-term infants to develop eating difficulties that can affect their growth. Although this behavior is certainly influenced by their fetal and postnatal history, a large individual variability exists that results from a complex interaction between genetic and environmental factors. We performed an original pilot study to identify common genetic variants associated with eating difficulties at 2 years of age in the POLYNUCA cohort of preterm infants. METHODS: Eating behavior was assessed using a parental questionnaire in a cohort of 234 very preterm infants (including 38 pairs of twins). Eighty-two common single nucleotide polymorphisms (SNPs) were selected in a total of 40 candidate genes involved in the regulation of energy homeostasis and food intake. RESULTS: Eating behavior was strongly correlated in monozygotic (r = 0.92, p = 0.001) but not dizygotic twins (r = 0.27, p = 0.14), suggesting a strong heritability of this trait. One SNP (rs11671975) in the insulin receptor (INSR) gene was significantly associated with eating behavior. This effect was maintained after adjustment for birth weight Z score and maternal education level, two factors that are associated with eating difficulties at 2 years of age. CONCLUSION: The INSR gene is potentially associated with eating difficulties in preterm infants.


Assuntos
Antígenos CD/genética , Transtornos da Alimentação e da Ingestão de Alimentos/genética , Recém-Nascido Prematuro , Polimorfismo de Nucleotídeo Único , Receptor de Insulina/genética , Fatores Etários , Pré-Escolar , Estudos de Coortes , Feminino , França , Estudos de Associação Genética , Humanos , Lactente , Recém-Nascido , Recém-Nascido Prematuro/crescimento & desenvolvimento , Masculino
19.
J Nutr ; 145(9): 2052-9, 2015 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-26180243

RESUMO

BACKGROUND: Dietary fibers have been associated with a reduction in appetite and energy intake. Although a few studies suggest that nonviscous fibers can exert such effects, likely through colonic fermentation, limited data are available. OBJECTIVE: The objective of this study was to determine whether α-galacto-oligosaccharides (α-GOSs), fermentable soluble fibers extracted from legumes, could reduce appetite, food intake, and inflammation in overweight subjects. METHODS: In 2 single-center, double-blind, randomized, placebo-controlled trials, 88 overweight adults [50% men and 50% women; 18-60 y old; body mass index (in kg/m(2)): 25-28] were supplemented for 14 d with tea that contained α-GOSs with different α-GOS dosages (6, 12, or 18 g α-GOSs/d), formulas (12 g α-GOSs/d with >80% of molecules with a degree of polymerization of 2, 3, or 4), or a control substance (glucose syrup). Appetite scores (5 appetite dimensions were assessed on visual analog scales during a preload test meal), food intake (test meal and 24-h food recall), and inflammatory markers [plasma lipopolysaccharide (LPS) and C-reactive protein (CRP)] were evaluated at day 0 (baseline) and day 15. RESULTS: Changes in appetite scores from day 0 to day 15 were significantly higher after α-GOS intake, with areas under the curve for the satiety score of +121 ± 108, +218 ± 218, and +306 ± 205 score · min for 6, 12, and 18 g α-GOSs/d, respectively, and -5 ± 64 score · min for the control group. We observed dose-dependent effects that did not vary by α-GOS composition. The administration of 6, 12, or 18 g α-GOSs/d significantly and dose-dependently increased the change in energy intake from day 0 to day 15 during a test meal (-13 ± 19, -26 ± 22, and -32 ± 22 kcal, respectively; +6 ± 21 kcal for the control group). Reductions in energy intake during lunch and dinner were also higher in the α-GOS groups in the dose-effect study. At day 15, LPS was dose-dependently reduced without an association with α-GOS composition (0.16 ± 0.02, 0.12 ± 0.08, and 0.08 ± 0.05 EU/mL for 6, 12, and 18 g α-GOSs/d, respectively, and 0.06 ± 0.04 EU/mL for the control group) and CRP was significantly lower in the α-GOS groups than in the control group in the formulation-effect study. CONCLUSIONS: Consumption of α-GOSs for 14 d dose-dependently reduced appetite, food intake, and inflammation in overweight adults with no impact of α-GOS composition. Consequently, α-GOSs appear to promote long-term weight loss and mitigate metabolic disorders.


Assuntos
Apetite/efeitos dos fármacos , Inflamação/tratamento farmacológico , Sobrepeso/tratamento farmacológico , Trissacarídeos/administração & dosagem , Adolescente , Adulto , Índice de Massa Corporal , Suplementos Nutricionais , Relação Dose-Resposta a Droga , Método Duplo-Cego , Ingestão de Energia , Feminino , Humanos , Masculino , Refeições , Pessoa de Meia-Idade , Saciação , Resultado do Tratamento , Redução de Peso , Adulto Jovem
20.
J Nutr Biochem ; 26(7): 784-95, 2015 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-25935308

RESUMO

Perinatal undernutrition affects not only fetal and neonatal growth but also adult health outcome, as suggested by the metabolic imprinting concept. Although maternal milk is the only channel through which nutrients are transferred from mother to offspring during the postnatal period, the impact of maternal undernutrition on milk composition is poorly understood. The present study investigates, in a rat model of nutritional programming, the effects of feeding an isocaloric, low-protein diet throughout gestation and lactation on milk composition and its possible consequences on offspring's growth and metabolic status. We used an integrated methodological approach that combined targeted analyses of macronutrients, free amino acid and fatty acid content throughout lactation, with an untargeted mass-spectrometric-based metabolomic phenotyping. Whereas perinatal dietary protein restriction failed to alter milk protein content, it dramatically decreased the concentration of most free amino acids at the end of lactation. Interestingly, a decrease of several amino acids involved in insulin secretion or gluconeogenesis was observed, suggesting that maternal protein restriction during the perinatal period may impact the insulinotrophic effect of milk, which may, in turn, account for the slower growth of the suckled male offspring. Besides, the decrease in sulfur amino acids may alter redox status in the offspring. Maternal undernutrition was also associated with an increase in milk total fatty acid content, with modifications in their pattern. Altogether, our results show that milk composition is clearly influenced by maternal diet and suggest that alterations in milk composition may play a role in offspring growth and metabolic programming.


Assuntos
Aminoácidos/metabolismo , Dieta com Restrição de Proteínas/efeitos adversos , Ácidos Graxos/metabolismo , Desenvolvimento Fetal , Lactação/metabolismo , Fenômenos Fisiológicos da Nutrição Materna , Leite/metabolismo , Adiposidade , Animais , Animais Lactentes , Feminino , Transtornos do Crescimento/etiologia , Masculino , Doenças Metabólicas/etiologia , Metabolômica/métodos , Proteínas do Leite/administração & dosagem , Proteínas do Leite/análise , Proteínas do Leite/biossíntese , Estresse Oxidativo , Gravidez , Distribuição Aleatória , Ratos Sprague-Dawley
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA