Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 16 de 16
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Healthcare (Basel) ; 10(7)2022 Jul 05.
Artigo em Inglês | MEDLINE | ID: mdl-35885785

RESUMO

(1) Background: Aqua-walking in a natural environment is a health promoting physical activity that is gaining popularity and appropriate for a variety of populations, however, to date, there is little scientific evidence supporting the efficacy and safety of this activity for older adults. The objective was to propose a preliminary exploration of psychometric and metabolic responses to an acute Aqua walking session either during winter or summer in older adults Aqua walking exercisers. (2) Methods: Heart rate, body temperature, glycemia, and blood pressure were monitored in 37 (30 women, 7 men) participants aged 52 to 83 years old in two Aqua walking sessions (water at 13 °C and 18.5 °C, respectively). Anthropometry (body weight, waist, and hip circumferences), body composition, physical activity level, sedentary time, sleep quality, quality of life, physical self-perception, and perceived health and feelings on various parameters were also assessed. (3) Results: Present results revealed a greater quality of life, physical self-perception, and perceived health in aging Aqua walking exercisers compared to those found in the literature in younger populations. None of the metabolic or psychometric measurements were found to be different between classically calibrated Aqua walking sessions performed in winter compared to summer. By contrast, there was a time effect during the sessions for body temperature (p < 0.001), SBP (p = 0.17), perceived mental and physical well-being (p = 0.006 and p < 0.001, respectively), and anxiety (p < 0.001). Leg discomfort also showed a time effect (p = 0.0009) and interaction effect (p = 0.025). (4) Conclusion: Aqua walking appears here to be an accessible practice that can be performed all year long with a range of physical and mental benefits for older adults. Future studies should investigate the metabolic responses of Aqua walking in different populations.

2.
PLoS One ; 8(6): e66816, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-23826145

RESUMO

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.


Assuntos
Dieta , Epigênese Genética , Perfilação da Expressão Gênica , Obesidade/genética , Transcrição Gênica , Tecido Adiposo/metabolismo , Tecido Adiposo/patologia , Animais , Diabetes Mellitus Experimental/genética , Dieta Hiperlipídica , Feminino , Regulação da Expressão Gênica , Redes Reguladoras de Genes/genética , Impressão Genômica/genética , Fígado/metabolismo , Fígado/patologia , Masculino , Camundongos , Fenótipo , Gravidez , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Transmissão Sináptica/genética
3.
PLoS One ; 7(11): e47986, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-23144842

RESUMO

Males and females responses to gestational overnutrition set the stage for subsequent sex-specific differences in adult onset non communicable diseases. Placenta, as a widely recognized programming agent, contibutes to the underlying processes. According to our previous findings, a high-fat diet during gestation triggers sex-specific epigenetic alterations within CpG and throughout the genome, together with the deregulation of clusters of imprinted genes. We further investigated the impact of diet and sex on placental histology, transcriptomic and epigenetic signatures in mice. Both basal gene expression and response to maternal high-fat diet were sexually dimorphic in whole placentas. Numerous genes showed sexually dimorphic expression, but only 11 genes regardless of the diet. In line with the key role of genes belonging to the sex chromosomes, 3 of these genes were Y-specific and 3 were X-specific. Amongst all the genes that were differentially expressed under a high-fat diet, only 16 genes were consistently affected in both males and females. The differences were not only quantitative but remarkably qualitative. The biological functions and networks of genes dysregulated differed markedly between the sexes. Seven genes of the epigenetic machinery were dysregulated, due to effects of diet, sex or both, including the Y- and X-linked histone demethylase paralogues Kdm5c and Kdm5d, which could mark differently male and female epigenomes. The DNA methyltransferase cofactor Dnmt3l gene expression was affected, reminiscent of our previous observation of changes in global DNA methylation. Overall, this striking sexual dimorphism of programming trajectories impose a considerable revision of the current dietary interventions protocols.


Assuntos
Dieta Hiperlipídica/efeitos adversos , Epigênese Genética , Expressão Gênica , Placenta/metabolismo , Efeitos Tardios da Exposição Pré-Natal/genética , Animais , DNA (Citosina-5-)-Metiltransferases/genética , DNA (Citosina-5-)-Metiltransferases/metabolismo , Feminino , Regulação Enzimológica da Expressão Gênica , Histona Desmetilases , Masculino , Camundongos , Análise de Sequência com Séries de Oligonucleotídeos , Oxirredutases N-Desmetilantes/genética , Oxirredutases N-Desmetilantes/metabolismo , Placenta/fisiopatologia , Gravidez , Efeitos Tardios da Exposição Pré-Natal/metabolismo , Fenômenos Fisiológicos da Nutrição Pré-Natal , Caracteres Sexuais , Transcriptoma
4.
FASEB J ; 26(1): 460-7, 2012 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-21982950

RESUMO

Markers of prostate tumor recurrence after radical prostatectomy are lacking and highly demanded. The androgen receptor (AR) is a nuclear receptor that plays a pivotal role in normal and cancerous prostate tissue. AR interacts with a number of proteins modulating its stability, localization, and activity. To test the hypothesis that an increased expression of AR partners might foster tumor development, we immunopurified AR partners in human tumors xenografted into mice. One of the identified AR partners was the multifunctional enzyme carbamoyl-phosphate synthetase II, aspartate transcarbamylase, and dihydroorotase (CAD), which catalyzes the 3 initial steps of pyrimidine biosynthesis. We combined experiments in C4-2, LNCaP, 22RV1, and PC3 human prostate cell lines and analysis of frozen radical prostatectomy samples to study the CAD-AR interaction. We show here that in prostate tumor cells, CAD fosters AR translocation into the nucleus and stimulates its transcriptional activity. Notably, in radical prostatectomy specimens, CAD expression was not correlated with proliferation markers, but a higher CAD mRNA level was associated with local tumor extension (P=0.049) and cancer relapse (P=0.017). These results demonstrate an unsuspected function for a key metabolic enzyme and identify CAD as a potential predictive marker of cancer relapse.


Assuntos
Aspartato Carbamoiltransferase/metabolismo , Biomarcadores Tumorais/metabolismo , Carbamoil Fosfato Sintase (Glutamina-Hidrolizante)/metabolismo , Di-Hidro-Orotase/metabolismo , Recidiva Local de Neoplasia/diagnóstico , Neoplasias da Próstata/diagnóstico , Receptores Androgênicos/metabolismo , Androgênios/metabolismo , Animais , Aspartato Carbamoiltransferase/genética , Carbamoil Fosfato Sintase (Glutamina-Hidrolizante)/genética , Linhagem Celular Tumoral , Núcleo Celular/metabolismo , Citosol/metabolismo , Di-Hidro-Orotase/genética , Humanos , Masculino , Camundongos , Recidiva Local de Neoplasia/metabolismo , Transplante de Neoplasias , Valor Preditivo dos Testes , Neoplasias da Próstata/metabolismo , Pirimidinas/biossíntese , RNA Interferente Pequeno/farmacologia , Receptores Androgênicos/genética , Transcrição Gênica/fisiologia , Transplante Heterólogo
5.
Cancer Res ; 71(24): 7649-58, 2011 Dec 15.
Artigo em Inglês | MEDLINE | ID: mdl-22037878

RESUMO

The antitumor effects of pharmacologic inhibitors of angiogenesis are hampered in patients by the rapid development of tumor resistance, notably through increased invasiveness and accelerated metastasis. Here, we reevaluated the role of the endogenous antiangiogenic thrombospondin 1 (TSP1) in prostate carcinomas in which angiogenesis is an active process. In xenografted tumors, we observed that TSP1 altogether inhibited angiogenesis and fostered tumor development. Our results show that TSP1 is a potent stimulator of prostate tumor cell migration. This effect required CD36, which also mediates TSP1 antiangiogenic activity, and was mimicked by an antiangiogenic TSP1-derived peptide. As suspected for pharmacologic inhibitors of angiogenesis, the TSP1 capacities to increase hypoxia and to trigger cell migration are thus inherently linked. Importantly, although antiangiogenic TSP1 increases hypoxia in vivo, our data show that, in turn, hypoxia induced TSP1, thus generating a vicious circle in prostate tumors. In radical prostatectomy specimens, we found TSP1 expression significantly associated with invasive tumors and with tumors which eventually recurred. TSP1 may thus help select patients at risk of prostate-specific antigen relapse. Together, the data suggest that intratumor disruption of the hypoxic cycle through TSP1 silencing will limit tumor invasion.


Assuntos
Movimento Celular , Neovascularização Patológica/genética , Neoplasias da Próstata/genética , Interferência de RNA , Trombospondina 1/genética , Animais , Antígenos CD36/metabolismo , Cálcio/metabolismo , Hipóxia Celular , Linhagem Celular Tumoral , Ensaio de Imunoadsorção Enzimática , Regulação Neoplásica da Expressão Gênica , Humanos , Hipóxia , Masculino , Camundongos , Camundongos Nus , Invasividade Neoplásica , Recidiva Local de Neoplasia , Neovascularização Patológica/metabolismo , Neovascularização Patológica/patologia , Orquiectomia , Peptídeos/farmacologia , Neoplasias da Próstata/metabolismo , Neoplasias da Próstata/patologia , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Canais de Cátion TRPV/metabolismo , Trombospondina 1/química , Trombospondina 1/metabolismo , Carga Tumoral/efeitos dos fármacos , Ensaios Antitumorais Modelo de Xenoenxerto/métodos
6.
PLoS One ; 6(6): e21184, 2011.
Artigo em Inglês | MEDLINE | ID: mdl-21698161

RESUMO

BACKGROUND: Daily variations in lipid concentrations in both gut lumen and blood are detected by specific sensors located in the gastrointestinal tract and in specialized central areas. Deregulation of the lipid sensors could be partly involved in the dysfunction of glucose homeostasis. The study aimed at comparing the effect of Medialipid (ML) overload on insulin secretion and sensitivity when administered either through the intestine or the carotid artery in mice. METHODOLOGY/PRINCIPAL FINDINGS: An indwelling intragastric or intracarotid catheter was installed in mice and ML or an isocaloric solution was infused over 24 hours. Glucose and insulin tolerance and vagus nerve activity were assessed. Some mice were treated daily for one week with the anti-lipid peroxidation agent aminoguanidine prior to the infusions and tests. The intestinal but not the intracarotid infusion of ML led to glucose and insulin intolerance when compared with controls. The intestinal ML overload induced lipid accumulation and increased lipid peroxidation as assessed by increased malondialdehyde production within both jejunum and duodenum. These effects were associated with the concomitant deregulation of vagus nerve. Administration of aminoguanidine protected against the effects of lipid overload and normalized glucose homeostasis and vagus nerve activity. CONCLUSIONS/SIGNIFICANCE: Lipid overload within the intestine led to deregulation of gastrointestinal lipid sensing that in turn impaired glucose homeostasis through changes in autonomic nervous system activity.


Assuntos
Glucose/metabolismo , Homeostase , Intestinos/efeitos dos fármacos , Peroxidação de Lipídeos , Animais , Guanidinas/farmacologia , Resistência à Insulina , Mucosa Intestinal/metabolismo , Peroxidação de Lipídeos/efeitos dos fármacos , Camundongos , Estresse Oxidativo
7.
PLoS One ; 5(12): e14398, 2010 Dec 21.
Artigo em Inglês | MEDLINE | ID: mdl-21200436

RESUMO

BACKGROUND: Changes in imprinted gene dosage in the placenta may compromise the prenatal control of nutritional resources. Indeed monoallelic behaviour and sensitivity to changes in regional epigenetic state render imprinted genes both vulnerable and adaptable. METHODS AND FINDINGS: We investigated whether a high-fat diet (HFD) during pregnancy modified the expression of imprinted genes and local and global DNA methylation patterns in the placenta. Pregnant mice were fed a HFD or a control diet (CD) during the first 15 days of gestation. We compared gene expression patterns in total placenta homogenates, for male and female offspring, by the RT-qPCR analysis of 20 imprinted genes. Sexual dimorphism and sensitivity to diet were observed for nine genes from four clusters on chromosomes 6, 7, 12 and 17. As assessed by in situ hybridization, these changes were not due to variation in the proportions of the placental layers. Bisulphite-sequencing analysis of 30 CpGs within the differentially methylated region (DMR) of the chromosome 17 cluster revealed sex- and diet-specific differential methylation of individual CpGs in two conspicuous subregions. Bioinformatic analysis suggested that these differentially methylated CpGs might lie within recognition elements or binding sites for transcription factors or factors involved in chromatin remodelling. Placental global DNA methylation, as assessed by the LUMA technique, was also sexually dimorphic on the CD, with lower methylation levels in male than in female placentae. The HFD led to global DNA hypomethylation only in female placenta. Bisulphite pyrosequencing showed that neither B1 nor LINE repetitive elements could account for these differences in DNA methylation. CONCLUSIONS: A HFD during gestation triggers sex-specific epigenetic alterations within CpG and throughout the genome, together with the deregulation of clusters of imprinted genes important in the control of many cellular, metabolic and physiological functions potentially involved in adaptation and/or evolution. These findings highlight the importance of studying both sexes in epidemiological protocols and dietary interventions.


Assuntos
Ração Animal , Metilação de DNA , Gorduras na Dieta/metabolismo , Regulação da Expressão Gênica , Impressão Genômica , Placenta/metabolismo , Alelos , Animais , Epigênese Genética , Feminino , Dosagem de Genes , Masculino , Camundongos , Gravidez , Prenhez , Fatores Sexuais
8.
Curr Top Med Chem ; 9(12): 1117-29, 2009.
Artigo em Inglês | MEDLINE | ID: mdl-19860712

RESUMO

Synthetic small interfering RNAs (siRNAs) open promising new therapeutic perspectives in acute and chronic pathologies. A number of experiments in mice demonstrated the ability of naked siRNAs injected under a normal pressure to trigger gene silencing in vivo, translating into a measurable phenotype. We focus in this review on the information that we can gain from these experiments, and discuss how the specificity of the gene silencing in vivo can be controlled. Because the activity of most drugs increases with the dosing, we are prone to consider that increasing the concentration of siRNAs within cells enhances the efficiency and the duration of the silencing. However, because RNAi is a saturable process, and because increasing the siRNA concentration into cells can induce undesirable side effects, this must be demonstrated. We compare in this review the methods used to quantify and study the biodistribution of siRNAs in living animals, and discuss how these methods can help in designing for each model and each siRNA the most adequate protocol to silence a cognate target gene in vivo.


Assuntos
Sistemas de Liberação de Medicamentos , RNA Interferente Pequeno , Animais , Inativação Gênica , Camundongos
9.
Pediatr Res ; 63(4): 340-7, 2008 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-18356736

RESUMO

There is accumulating evidence for nongenetic transgenerational inheritance with conspicuous marked sexual dimorphism for both the modes of transmission and the effects. Given the critical spatiotemporal windows, the role of the sex chromosomes, the regulatory pathways underlying sexual differentiation during gonad and brain development, and other developmental processes, as well as the lifelong impact of sex hormones, it is not surprising that most of the common diseases, which often take root in early development, display some degree of sex bias. The flexibility of epigenetic marks may make it possible for environmental and nutritional factors, or endocrine disruptors to alter-during a particular spatiotemporal window in a sex-specific manner-the sex-specific methylation or demethylation of specific CpGs and histone/chromatin modifications underlying sex-specific expression of a substantial proportion of genes. Thus, finely tuned developmental program aspects, specific to one sex, may be more sensitive to specific environmental challenges, particularly during developmental programming and gametogenesis, but also throughout the individual's life under the influence of sex steroid hormones. This review highlights the importance of studying both sexes in epidemiologic protocols or dietary interventions both in humans and in experimental models in animals.


Assuntos
Impressão Genômica/genética , Caracteres Sexuais , Animais , Epigênese Genética , Feminino , Células Germinativas , Humanos , Padrões de Herança , Masculino , Processos de Determinação Sexual
10.
Epigenetics ; 2(3): 137-46, 2007 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-17965598

RESUMO

Epigenetic misprogramming during development is widely thought to have a persistent effect on the health of the offspring and may even be transmitted to the next generation. However, little is known about the stochastically, genetically and environmentally triggered epimutations occurring during an individual's lifetime. They may result from replication-dependent, replication-independent or DNA repair events. The rhythmic, circadian induction of a substantial proportion of genes by a network of clock genes, one of which is a histone acetyl transferase, nuclear receptors and transcription factors is controlled by chromatin remodeling. The associated circadian epigenetic patterns must be transient, sensitive to environmental cues and reversible. Links have been found between circadian rhythms and major components of energy homeostasis, thermogenesis and hunger-satiety, rest-activity rhythms and the sleep-wake cycle. Thus poorly adapted behavior or lifestyle and desynchronized cues may disturb the modulation of gene expression. This functional asynchrony may ultimately lead to persistence of aberrant and unphased "locking"or "leakage" of gene expression and inadapted responses in the body as a whole.


Assuntos
Regulação da Temperatura Corporal/genética , Montagem e Desmontagem da Cromatina/genética , Ritmo Circadiano/genética , Metabolismo Energético/genética , Epigênese Genética , Síndrome Metabólica/genética , Animais , Reparo do DNA/genética , Histona Acetiltransferases/genética , Histona Acetiltransferases/metabolismo , Humanos , Síndrome Metabólica/metabolismo , Mutação
11.
Obesity (Silver Spring) ; 15(8): 1996-2005, 2007 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-17712117

RESUMO

OBJECTIVE: The aim of this study was to assess the suitability of A/J and C57BL/6J mice of both sexes as models of some components of the human metabolic syndrome (MetS) under nutritional conditions more comparable with the actual worldwide diet responsible for the increased incidence of the MetS. RESEARCH METHODS: We fed large cohorts (n = 515) of two strains of mice, A/J and the C57BL/6J, and of both sexes a high-fat diet (HFD; 60% fat) that, in contrast with most previous reports using saturated fats, was enriched in mono- and polyunsaturated fatty acids, thus more closely mimicking most Western diets, or a control diet (10% fat), for 20 weeks. RESULTS: In sharp contrast to previous reports, weight gain and hyperleptinemia were similar in both strains and sexes. Hyperinsulinemia, glucose tolerance, insulin resistance, and hypercholesterolemia were observed, although with important differences between strains and sexes. A/J males displayed severely impaired glucose tolerance and insulin resistance. However, in contrast with C57BL6/J mice, which displayed overt type 2 diabetes, A/J mice of both sexes remained normoglycemic. DISCUSSION: With important differences in magnitude and time course, the phenotypic and metabolic characteristics of both strains and both sexes on this HFD demonstrate that these models are very useful for identifying the mechanisms underlying progression or resistance to subsequent type 2 diabetes.


Assuntos
Diabetes Mellitus Tipo 2/metabolismo , Gorduras na Dieta/administração & dosagem , Síndrome Metabólica/metabolismo , Obesidade/metabolismo , Animais , Glicemia/metabolismo , Composição Corporal/fisiologia , Peso Corporal/fisiologia , Colesterol/sangue , Estudos de Coortes , Diabetes Mellitus Tipo 2/sangue , Gorduras na Dieta/metabolismo , Modelos Animais de Doenças , Ingestão de Alimentos/fisiologia , Ácidos Graxos Monoinsaturados/administração & dosagem , Ácidos Graxos Monoinsaturados/metabolismo , Feminino , Teste de Tolerância a Glucose , Insulina/sangue , Leptina/sangue , Masculino , Síndrome Metabólica/sangue , Camundongos , Camundongos Endogâmicos C57BL , Obesidade/sangue , Gravidez , Organismos Livres de Patógenos Específicos , Triglicerídeos/sangue
12.
Clin Chem Lab Med ; 45(3): 321-7, 2007.
Artigo em Inglês | MEDLINE | ID: mdl-17378726

RESUMO

The phenotype of an individual is the result of complex interactions between genotype, epigenome and current, past and ancestral environment, leading to lifelong remodelling of our epigenomes. Various replication-dependent and -independent epigenetic mechanisms are involved in developmental programming, lifelong stochastic and environmental deteriorations, circadian deteriorations, and transgenerational effects. Several types of sequences can be targets of a host of environmental factors and can be associated with specific epigenetic signatures and patterns of gene expression. Depending on the nature and intensity of the insult, the critical spatiotemporal windows and developmental or lifelong processes involved, these epigenetic alterations can lead to permanent changes in tissue and organ structure and function, or to reversible changes using appropriate epigenetic tools. Given several encouraging trials, prevention and therapy of age- and lifestyle-related diseases by individualised tailoring of optimal epigenetic diets or drugs are conceivable. However, these interventions will require intense efforts to unravel the complexity of these epigenetic, genetic and environment interactions and to evaluate their potential reversibility with minimal side effects.


Assuntos
Epigênese Genética , Estado Nutricional , Ritmo Circadiano , Dieta , Meio Ambiente , Regulação da Expressão Gênica , Humanos , Fenótipo
13.
Am J Physiol Endocrinol Metab ; 292(4): E1095-100, 2007 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-17164437

RESUMO

With the worldwide epidemic of metabolic syndrome (MetS), the proportion of women that are overweight/obese and overfed during pregnancy has increased. The resulting abnormal uterine environment may have deleterious effects on fetal metabolic programming and lead to MetS in adulthood. A balanced/restricted diet and/or physical exercise often improve metabolic abnormalities in individuals with obesity and type 2 diabetes mellitus (T2D). We investigated whether reducing fat intake during the periconceptual/gestation/lactation period in mothers with high-fat diet (HFD)-induced obesity could be used to modify fetal/neonatal MetS programming positively, thereby preventing MetS. First generation (F1) C57BL/6J female mice with HFD-induced obesity and T2D were crossed with F1 males on control diet (CD). These F1 females were switched to a CD during the periconceptual/gestation/lactation period. At weaning, both male and female second generation (F2) mice were fed a HFD. Weight, caloric intake, lipid parameters, glucose, and insulin sensitivity were assessed. Sensitivity/resistance to the HFD differed significantly between generations and sexes. A similar proportion of the F1 and F2 males (80%) developed hyperphagia, obesity, and T2D. In contrast, a significantly higher proportion of the F2 females (43%) than of the previous F1 generation (17%) were resistant (P<0.01). Despite having free access to the HFD, these female mice were no longer hyperphagic and remained lean, with normal insulin sensitivity and glycemia but mild hypercholesterolemia and glucose intolerance, thus displaying a "satiety phenotype." This suggests that an appropriate dietary fatty acid profile and intake during the periconceptual/gestation/lactation period helps the female offspring to cope with deleterious intrauterine conditions.


Assuntos
Animais Recém-Nascidos/fisiologia , Dieta com Restrição de Gorduras , Gorduras na Dieta/administração & dosagem , Desenvolvimento Fetal , Síndrome Metabólica/prevenção & controle , Obesidade/fisiopatologia , Complicações na Gravidez/fisiopatologia , Animais , Gorduras na Dieta/farmacologia , Suscetibilidade a Doenças , Ingestão de Energia , Feminino , Fertilização , Glucose/metabolismo , Teste de Tolerância a Glucose , Homeostase , Hiperfagia/prevenção & controle , Insulina/metabolismo , Lactação , Metabolismo dos Lipídeos , Masculino , Síndrome Metabólica/etiologia , Camundongos , Camundongos Endogâmicos C57BL , Obesidade/etiologia , Obesidade/metabolismo , Gravidez , Complicações na Gravidez/etiologia , Fatores Sexuais , Aumento de Peso
14.
Diabetes ; 54(7): 1899-906, 2005 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-15983188

RESUMO

Human epidemiological studies and appropriately designed dietary interventions in animal models have provided considerable evidence to suggest that maternal nutritional imbalance and metabolic disturbances, during critical time windows of development, may have a persistent effect on the health of the offspring and may even be transmitted to the next generation. We now need to explain the mechanisms involved in generating such responses. The idea that epigenetic changes associated with chromatin remodeling and regulation of gene expression underlie the developmental programming of metabolic syndrome is gaining acceptance. Epigenetic alterations have been known to be of importance in cancer for approximately 2 decades. This has made it possible to decipher epigenetic codes and machinery and has led to the development of a new generation of drugs now in clinical trials. Although less conspicuous, epigenetic alterations have also been progressively shown to be relevant to common diseases such as atherosclerosis and type 2 diabetes. Imprinted genes, with their key roles in controlling feto-placental nutrient supply and demand and their epigenetic lability in response to nutrients, may play an important role in adaptation/evolution. The combination of these various lines of research on epigenetic programming processes has highlighted new possibilities for the prevention and treatment of metabolic syndrome.


Assuntos
Dieta , Síndrome Metabólica/epidemiologia , Síndrome Metabólica/genética , Fenômenos Fisiológicos da Nutrição/fisiologia , Animais , Promoção da Saúde , Humanos , Síndrome Metabólica/fisiopatologia , Modelos Animais , Neoplasias/genética
15.
Med Sci (Paris) ; 21(4): 396-404, 2005 Apr.
Artigo em Francês | MEDLINE | ID: mdl-15811305

RESUMO

The importance of epigenetic alterations has been acknowledged in cancer for about two decades by an increasing number of molecular oncologists who contributed to deciphering the epigenetic codes and machinery and opened the road for a new generation of drugs now in clinical trials. However, the relevance of epigenetics to common diseases such as metabolic syndrome and cardiovascular disease was less conspicuous. This review focuses on converging data supporting the hypothesis that, in addition to "thrifty genotype" inheritance, individuals with metabolic syndrome (MetS)--combining disturbances in glucose and insulin metabolism, excess of predominantly abdominally distributed weight, mild dyslipidemia and hypertension, with the subsequent development of obesity, type 2 diabetes mellitus (T2D) and cardiovascular disease (CVD)--have suffered improper "epigenetic programming" during their fetal/postnatal development due to maternal inadequate nutrition and metabolic disturbances and also during their lifetime. Moreover, as seen for obesity and T2D, MetS tends to appear earlier in childhood, to be more severe from generation to generation and to affect more pregnant women. Thus, in addition to maternal effects, MetS patients may display "transgenerational effects" via the incomplete erasure of epigenetic marks endured by their parents and grandparents. We highlight the susceptibility of epigenetic mechanisms controlling gene expression to environmental influences due to their inherent malleability, emphasizing the participation of transposable elements and the potential role of imprinted genes during critical time windows in epigenetic programming, from the very beginning of development throughout life. Increasing our understanding on epigenetic patterns significance and small molecules (nutrients, drugs) that reverse epigenetic (in)activation should provide us with the means to "unlock" silenced (enhanced) genes, and to "convert" the obsolete human thrifty genotype into a "squandering" phenotype.


Assuntos
Dieta , Epigênese Genética , Síndrome Metabólica/genética , Humanos
16.
Med Sci (Paris) ; 21 Spec No: 44-52, 2005 Dec.
Artigo em Francês | MEDLINE | ID: mdl-16598905

RESUMO

The importance of epigenetic alterations has been acknowledged in cancer for about two decades by an increasing number of molecular oncologists who contributed to deciphering the epigenetic codes and machinery and opened the road for a new generation of drugs now in clinical trials. However, the relevance of epigenetics to common diseases such as metabolic syndrome and cardiovascular disease was less conspicuous. This review focuses on converging data supporting the hypothesis that, in addition to "thrifty genotype" inheritance, individuals with metabolic syndrome (MetS)--combining disturbances in glucose and insulin metabolism, excess of predominantly abdominally distributed weight, mild dyslipidemia and hypertension, with the subsequent development of obesity, type 2 diabetes mellitus (T2D) and cardiovascular disease (CVD)--have suffered improper "epigenetic programing" during their fetal/postnatal development due to maternal inadequate nutrition and metabolic disturbances and also during their life-time. Moreover, as seen for obesity and T2D, MetS tends to appear earlier in childhood, to be more severe from generation to generation and to affect more pregnant women. Thus, in addition to maternal effects, MetS patients may display "transgenerational effects" via the incomplete erasure of epigenetic marks endured by their parents and grandparents. We highlight the susceptibility of epigenetic mechanisms controlling gene expression to environmental influences due to their inherent malleability, emphasizing the participation of transposable elements and the potential role of imprinted genes during critical time windows in epigenetic programming, from the very beginning of development throughout life. Increasing our understanding on epigenetic patterns significance and small molecules (nutrients, drugs) that reverse epigenetic (in) activation should provide us with the means to he obsolete human thrifty genotype into a "squandering" phenotype.


Assuntos
Desenvolvimento Fetal/genética , Alimentos Infantis , Síndrome Metabólica/genética , Fenômenos Fisiológicos da Nutrição/fisiologia , Placenta/fisiologia , Animais , Feminino , Genômica , Humanos , Recém-Nascido , Síndrome Metabólica/fisiopatologia , Camundongos , Gravidez
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...