Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 28
Filtrar
1.
Int J Obes (Lond) ; 46(5): 901-917, 2022 05.
Artigo em Inglês | MEDLINE | ID: mdl-35022547

RESUMO

In the last two decades, evidence from human and animal studies suggests that paternal obesity around the time of conception can have adverse effects on offspring health through developmental programming. This may make significant contributions to the current epidemic of obesity and related metabolic and reproductive complications like diabetes, cardiovascular disease, and subfertility/infertility. To date, changes in seminal fluid composition, sperm DNA methylation, histone composition, small non-coding RNAs, and sperm DNA damage have been proposed as potential underpinning mechanism to program offspring health. In this review, we discuss current human and rodent evidence on the impact of paternal obesity/overnutrition on offspring health, followed by the proposed mechanisms, with a focus on sperm DNA damage underpinning paternal programming. We also summarize the different intervention strategies implemented to minimize effects of paternal obesity. Upon critical review of literature, we find that obesity-induced altered sperm quality in father is linked with compromised offspring health. Paternal exercise intervention before conception has been shown to improve metabolic health. Further work to explore the mechanisms underlying benefits of paternal exercise on offspring are warranted. Conversion to healthy diets and micronutrient supplementation during pre-conception have shown some positive impacts towards minimizing the impact of paternal obesity on offspring. Pharmacological approaches e.g., metformin are also being applied. Thus, interventions in the obese father may ameliorate the potential detrimental impacts of paternal obesity on offspring.


Assuntos
Pai , Espermatozoides , Animais , Metilação de DNA , Epigênese Genética , Humanos , Masculino , Obesidade/metabolismo , Responsabilidade Social
2.
Eur J Nutr ; 61(7): 3741-3753, 2022 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-35708759

RESUMO

PURPOSE: Emerging evidence from rodent studies suggests that high-fat-diet (HFD)-induced obesity is characterized by increased oxidative damage in sperm and testis. However, interventions using micronutrient supplementation to mitigate oxidative damage in obesity have not been extensively studied. This study aimed to investigate the effect of an antioxidant-based micronutrient supplement (added folate, vitamin B6, choline, betaine, and zinc) on sperm and testicular oxidative damage in HFD-fed male Sprague Dawley rats. METHODS: Rats (3-weeks-old, 12/group) were weaned onto control (C) or HFD (H) or these diets with micronutrient supplement (CS; HS); sperm and testis were harvested at 30.5 weeks. To assess oxidative stress and antioxidant capacity in testis, levels of malondialdehyde (MDA), glutathione (GSH), folate and susceptibility index (SI) of pro-oxidative damage, mRNA expression of Nrf2, NFκB-p65, IL-6, IL-10 and TNF-α, in addition to superoxide-dismutase (SOD), catalase and glutathione-peroxidase (GPx) activities were measured. 8-hydroxy-2-deoxyguanosine (8-OHdG) were assessed in both sperm and testis. RESULTS: HFD-fed rats had significantly increased 8-OHdG content in sperm and testis, increased testicular SI, decreased testicular weight, SOD and GPx activity compared to control. Strikingly, supplementation of HFD appeared to significantly reduce 8-OHdG in sperm and testis (22% and 24.3%, respectively), reduce testicular SI and MDA content (28% and 40%, respectively), increase testicular weight (24%), SOD and GPX activity (30% and 70%, respectively) and GSH content (19%). Moreover, supplementation had significant impact to increase testicular folate content regardless of diet. Furthermore, an overall effect of supplementation to increase testicular mRNA expression of Nrf2 was observed across groups. Interestingly, testicular SI was positively correlated with sperm and testicular 8-OHdG and MDA content, suggesting a critical role of testicular antioxidant activity to combat oxidative damage in sperm and testis. CONCLUSION: Our findings suggest that antioxidant-based micronutrient supplement has the potential to interrupt HFD-induced sperm and testicular oxidative damage by improving testicular antioxidant capacity.


Assuntos
Antioxidantes , Testículo , 8-Hidroxi-2'-Desoxiguanosina , Animais , Antioxidantes/metabolismo , Antioxidantes/farmacologia , Dieta Hiperlipídica/efeitos adversos , Ácido Fólico/farmacologia , Glutationa/metabolismo , Masculino , Micronutrientes , Fator 2 Relacionado a NF-E2/metabolismo , Obesidade/metabolismo , Estresse Oxidativo , RNA Mensageiro/metabolismo , Ratos , Ratos Sprague-Dawley , Sêmen/metabolismo , Espermatozoides , Superóxido Dismutase/metabolismo
3.
Am J Physiol Endocrinol Metab ; 312(2): E117-E125, 2017 02 01.
Artigo em Inglês | MEDLINE | ID: mdl-27965204

RESUMO

Emerging evidence suggests that paternal obesity plays an important role in offspring health. Our previous work using a rodent model of diet-induced paternal obesity showed that female offspring from high-fat diet (HFD)-fed fathers develop glucose intolerance due to impairment of pancreatic insulin secretion. Here, we focused on the health outcomes of male offspring from HFD-fed fathers. Male Sprague-Dawley rats (3 wk old) were fed control (CD-F0) or HFD (HFD-F0) for 12 wk before mating with control-fed females. Male offspring were fed control diets for up to 8 wk or 6 mo. Although male offspring from HFD-F0 did not develop any obvious glucose metabolism defects in this study, surprisingly, a growth deficit phenotype was observed from birth to 6 mo of age. Male offspring from HFD-F0 had reduced birth weight compared with CD-F0, followed by reduced postweaning growth from 9 wk of age. This resulted in 10% reduction in body weight at 6 mo with significantly smaller fat pads and skeletal muscles. Reduced circulating levels of growth hormone (GH) and IGF-I were detected at 8 wk and 6 mo, respectively. Expression of adipogenesis markers was decreased in adipose tissue of HFD-F0 offspring at 8 wk and 6 mo, and expression of growth markers was decreased in muscle of HFD-F0 offspring at 8 wk. We propose that the reduced GH secretion at 8 wk of age altered the growth of male offspring from HFD-F0, resulting in smaller animals from 9 wk to 6 mo of age. Furthermore, increased muscle triglyceride content and expression of lipogenic genes were observed in HFD-F0 offspring, potentially increasing their metabolic risk.


Assuntos
Adiposidade , Pai , Crescimento e Desenvolvimento , Obesidade , Efeitos Tardios da Exposição Pré-Natal , Animais , Feminino , Masculino , Gravidez , Efeitos Tardios da Exposição Pré-Natal/metabolismo , Efeitos Tardios da Exposição Pré-Natal/fisiopatologia , Ratos , Ratos Sprague-Dawley
4.
FASEB J ; 28(4): 1830-41, 2014 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-24421403

RESUMO

We previously showed that paternal high-fat diet (HFD) consumption programs ß-cell dysfunction in female rat offspring, together with transcriptome alterations in islets. Here we investigated the retroperitoneal white adipose tissue (RpWAT) transcriptome using gene and pathway enrichment and pathway analysis to determine whether commonly affected network topologies exist between these two metabolically related tissues. In RpWAT, 5108 genes were differentially expressed due to a paternal HFD; the top 5 significantly enriched networks identified by pathway analysis in offspring of HFD fathers compared with those of fathers fed control diet were: mitochondrial and cellular response to stress, telomerase signaling, cell death and survival, cell cycle, cellular growth and proliferation, and cancer. A total of 187 adipose olfactory receptor genes were down-regulated. Interrogation against the islet transcriptome identified specific gene networks and pathways, including olfactory receptor genes that were similarly affected in both tissues (411 common genes, P<0.05). In particular, we highlight a common molecular network, cell cycle and cancer, with the same hub gene, Myc, suggesting early onset developmental changes that persist, shared responses to programmed systemic factors, or crosstalk between tissues. Thus, paternal HFD consumption triggers unique gene signatures, consistent with premature aging and chronic degenerative disorders, in both RpWAT and pancreatic islets of daughters.


Assuntos
Dieta Hiperlipídica , Gordura Intra-Abdominal/metabolismo , Ilhotas Pancreáticas/metabolismo , Efeitos Tardios da Exposição Pré-Natal/genética , Transcriptoma/genética , Animais , Análise por Conglomerados , Gorduras na Dieta/administração & dosagem , Feminino , Redes Reguladoras de Genes/efeitos dos fármacos , Redes Reguladoras de Genes/genética , Masculino , Análise de Sequência com Séries de Oligonucleotídeos , Gravidez , Ratos , Ratos Sprague-Dawley , Transcriptoma/efeitos dos fármacos
5.
Bioessays ; 35(9): 787-93, 2013 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-23881837

RESUMO

The contribution of inherited non-genetic factors to complex diseases is of great current interest. The ways in which mothers and fathers can affect their offspring's health clearly differ as a result of the intimate interactions between mother and offspring during pre- and postnatal life. There is, however, potential for some overlap in mechanisms, particularly epigenetic mechanisms. A small number of epidemiological studies and animal models have investigated the non-genetic contribution of the parents to offspring health. Discovering new mechanisms of disease inheritance is technically difficult, especially in genetically, socially and environmentally heterogeneous human populations. Therefore, rigorous experimental design, appropriate sample numbers and the use of high-throughput technologies are necessary to provide convincing evidence. Based on recent examples from the literature, here we propose several ways to improve human studies that aim to identify the underlying mechanisms of transgenerational inheritance of metabolic disease.


Assuntos
Epigênese Genética , Impressão Genômica/fisiologia , Doenças Metabólicas/genética , Animais , Índice de Massa Corporal , Metilação de DNA , Modelos Animais de Doenças , Feminino , Predisposição Genética para Doença , Sequenciamento de Nucleotídeos em Larga Escala , Humanos , Masculino , Fenômenos Fisiológicos da Nutrição Materna , Doenças Metabólicas/embriologia
6.
Hum Mol Genet ; 20(19): 3757-68, 2011 Oct 01.
Artigo em Inglês | MEDLINE | ID: mdl-21724553

RESUMO

The pathogenic agent responsible for the expanded repeat diseases, a group of neurodegenerative diseases that includes Huntington's disease is not yet fully understood. Expanded polyglutamine (polyQ) is thought to be the toxic agent in certain cases, however, not all expanded repeat disease genes can encode a polyQ sequence. Since a repeat-containing RNA intermediary is common to all of these diseases, hairpin-forming single-stranded RNA has been investigated as a potential common pathogenic agent. More recently, it has become apparent that most of the expanded repeat disease loci have transcription occurring from both strands, raising the possibility that the complementary repeat RNAs could form a double-stranded structure. In our investigation using Drosophila models of these diseases, we identified a fortuitous integration event that models bidirectional repeat RNA transcription with the resultant flies exhibiting inducible pathology. We therefore established further lines of Drosophila expressing independent complementary repeat RNAs and found that these are toxic. The Dicer pathway is essential for this toxicity and in neuronal cells accounts for metabolism of the high copy number (CAG.CUG)(100) double-stranded RNAs down to (CAG)(7) single-stranded small RNAs. We also observe significant changes to the microRNA profile in neurons. These data identify a novel pathway through which double-stranded repeat RNA is toxic and capable of eliciting symptoms common to neurodegenerative human diseases resulting from dominantly inherited expanded repeats.


Assuntos
Drosophila/genética , Doenças Neurodegenerativas/genética , Doenças Neurodegenerativas/patologia , RNA de Cadeia Dupla/química , RNA de Cadeia Dupla/metabolismo , Expansão das Repetições de Trinucleotídeos , Animais , Animais Geneticamente Modificados , Modelos Animais de Doenças , Drosophila/metabolismo , Proteínas de Drosophila/genética , Proteínas de Drosophila/metabolismo , Feminino , Humanos , Masculino , Doenças Neurodegenerativas/metabolismo , Neurônios/metabolismo , Conformação de Ácido Nucleico , RNA Helicases/genética , RNA Helicases/metabolismo , RNA de Cadeia Dupla/genética , Ribonuclease III/genética , Ribonuclease III/metabolismo
7.
J Nutr ; 141(1): 95-100, 2011 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-21106931

RESUMO

Methyl deficiencies have been implicated in metabolic programming during the periods of oocyte and embryo development. Semisynthetic methyl-deficient diets (MD) with no folic acid, 0.05% choline, and approximately one-half the recommended content of methionine were fed to female rats for 3 wk prior to mating and for the first 5 d of gestation. During the period of MD feeding, plasma homocysteine concentrations were approximately twice those of rats fed the complete (CON) diet. From d 5, both groups received a complete semipurified AIN diet until birth. On d 8, plasma homocysteine concentrations did not differ between the 2 groups. Thereafter, dams and offspring were fed a nonpurified diet for the remainder of the experiment. At 6 mo of age, the homeostatic model assessment (HOMA) index of the male MD offspring tended to be 32% higher (P = 0.053) and peak insulin during an oral glucose tolerance test (oGTT) was 39% higher (P < 0.05) compared with the male CON offspring. There was no difference in the response to an oGTT in the female offspring at 6 mo of age. The increased HOMA index of male MD offspring persisted to 12 mo of age. The peak glucose concentration during oGTT was 23% higher (P < 0.05) in MD compared with the CON males despite 39% greater (P < 0.05) peak insulin concentrations. This study shows that in rats, a physiologically relevant methyl-deficient diet fed during the period of oocyte maturation and preimplantation development programs gender-specific changes in glucose handling by the offspring.


Assuntos
Glucose/metabolismo , Homeostase , Desnutrição/metabolismo , Fenômenos Fisiológicos da Nutrição Materna/fisiologia , Animais , Glicemia/análise , Deficiência de Colina/metabolismo , Feminino , Teste de Tolerância a Glucose , Homocisteína/sangue , Insulina/sangue , Masculino , Metilação , Estado Nutricional , Ratos , Caracteres Sexuais
8.
Cells ; 10(7)2021 07 11.
Artigo em Inglês | MEDLINE | ID: mdl-34359921

RESUMO

Obesity increases the risk of metabolic disorders, partly through increased oxidative stress. Here, we examined the effects of a dietary micronutrient supplement (consisting of folate, vitamin B6, choline, betaine, and zinc) with antioxidant and methyl donor activities. Male Sprague Dawley rats (3 weeks old, 17/group) were weaned onto control (C) or high-fat diet (HFD) or same diets with added micronutrient supplement (CS; HS). At 14.5 weeks of age, body composition was measured by magnetic resonance imaging. At 21 weeks of age, respiratory quotient and energy expenditure was measured using Comprehensive Lab Animal Monitoring System. At 22 weeks of age, an oral glucose tolerance test (OGTT) was performed, and using fasting glucose and insulin values, Homeostasis Model Assessment of Insulin Resistance (HOMA-IR) was calculated as a surrogate measure of insulin resistance. At 30.5 weeks of age, blood and liver tissues were harvested. Liver antioxidant capacity, lipids and expression of genes involved in lipid metabolism (Cd36, Fabp1, Acaca, Fasn, Cpt1a, Srebf1) were measured. HFD increased adiposity (p < 0.001) and body weight (p < 0.001), both of which did not occur in the HS group. The animals fed HFD developed impaired fasting glucose, impaired glucose tolerance, and fasting hyperinsulinemia compared to control fed animals. Interestingly, HS animals demonstrated an improvement in fasting glucose and fasting insulin. Based on insulin release during OGTT and HOMA-IR, the supplement appeared to reduce the insulin resistance developed by HFD feeding. Supplementation increased hepatic glutathione content (p < 0.05) and reduced hepatic triglyceride accumulation (p < 0.001) regardless of diet; this was accompanied by altered gene expression (particularly of CPT-1). Our findings show that dietary micronutrient supplementation can reduce weight gain and adiposity, improve glucose metabolism, and improve hepatic antioxidant capacity and lipid metabolism in response to HFD intake.


Assuntos
Suplementos Nutricionais , Glucose/metabolismo , Metabolismo dos Lipídeos , Fígado/metabolismo , Micronutrientes , Obesidade/metabolismo , Animais , Antioxidantes/metabolismo , Dieta Hiperlipídica , Modelos Animais de Doenças , Regulação da Expressão Gênica , Leptina/sangue , Metabolismo dos Lipídeos/genética , Lipídeos/sangue , Obesidade/genética , Ratos Sprague-Dawley , Substâncias Reativas com Ácido Tiobarbitúrico/metabolismo
9.
Mol Cell Biol ; 27(7): 2777-90, 2007 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-17283065

RESUMO

The Sp/Krüppel-like factor (Sp/Klf) family is comprised of around 25 zinc finger transcription factors that recognize CACCC boxes and GC-rich elements. We have investigated basic Krüppel-like factor (Bklf/Klf3) and show that in erythroid tissues its expression is highly dependent on another family member, erythroid Krüppel-like factor (Eklf/Klf1). We observe that Bklf mRNA is significantly reduced in erythroid tissues from Eklf-null murine embryos. We find that Bklf is driven primarily by two promoters, a ubiquitously active GC-rich upstream promoter, 1a, and an erythroid downstream promoter, 1b. Transcripts from the two promoters encode identical proteins. Interestingly, both the ubiquitous and the erythroid promoter are dependent on Eklf in erythroid cells. Eklf also activates both promoters in transient assays. Experiments utilizing an inducible form of Eklf demonstrate activation of the endogenous Bklf gene in the presence of an inhibitor of protein synthesis. The kinetics of activation are also consistent with Bklf being a direct Eklf target. Chromatin immunoprecipitation assays confirm that Eklf associates with both Bklf promoters. Eklf is typically an activator of transcription, whereas Bklf is noted as a repressor. Our results support the hypothesis that feedback cross-regulation occurs within the Sp/Klf family in vivo.


Assuntos
Células Eritroides/metabolismo , Regulação da Expressão Gênica no Desenvolvimento , Fatores de Transcrição Kruppel-Like/fisiologia , Animais , Sequência de Bases , Feto/metabolismo , Fatores de Transcrição Kruppel-Like/genética , Fígado/crescimento & desenvolvimento , Fígado/metabolismo , Camundongos , Camundongos Knockout , Dados de Sequência Molecular , Regiões Promotoras Genéticas
10.
Proc Natl Acad Sci U S A ; 104(49): 19351-6, 2007 Dec 04.
Artigo em Inglês | MEDLINE | ID: mdl-18042717

RESUMO

A complex combination of adult health-related disorders can originate from developmental events that occur in utero. The periconceptional period may also be programmable. We report on the effects of restricting the supply of specific B vitamins (i.e., B(12) and folate) and methionine, within normal physiological ranges, from the periconceptional diet of mature female sheep. We hypothesized this would lead to epigenetic modifications to DNA methylation in the preovulatory oocyte and/or preimplantation embryo, with long-term health implications for offspring. DNA methylation is a key epigenetic contributor to maintenance of gene silencing that relies on a dietary supply of methyl groups. We observed no effects on pregnancy establishment or birth weight, but this modest early dietary intervention led to adult offspring that were both heavier and fatter, elicited altered immune responses to antigenic challenge, were insulin-resistant, and had elevated blood pressure-effects that were most obvious in males. The altered methylation status of 4% of 1,400 CpG islands examined by restriction landmark genome scanning in the fetal liver revealed compelling evidence of a widespread epigenetic mechanism associated with this nutritionally programmed effect. Intriguingly, more than half of the affected loci were specific to males. The data provide the first evidence that clinically relevant reductions in specific dietary inputs to the methionine/folate cycles during the periconceptional period can lead to widespread epigenetic alterations to DNA methylation in offspring, and modify adult health-related phenotypes.


Assuntos
Pressão Sanguínea , Metilação de DNA/efeitos dos fármacos , Epigênese Genética/efeitos dos fármacos , Resistência à Insulina , Metionina/administração & dosagem , Gravidez/metabolismo , Complexo Vitamínico B/administração & dosagem , Animais , Animais Recém-Nascidos/imunologia , Animais Recém-Nascidos/metabolismo , Composição Corporal/efeitos dos fármacos , Dieta , Embrião de Mamíferos/metabolismo , Feminino , Fertilização , Ácido Fólico/administração & dosagem , Glucose/metabolismo , Frequência Cardíaca/efeitos dos fármacos , Imunidade , Gravidez/efeitos dos fármacos , Gravidez/genética , Resultado da Gravidez , Ovinos/embriologia , Ovinos/metabolismo , Vitamina B 12/administração & dosagem
11.
Br J Nutr ; 101(9): 1333-40, 2009 May.
Artigo em Inglês | MEDLINE | ID: mdl-18782463

RESUMO

In humans poor maternal folate status is associated with a decrease in infant birth weight. As low birth weight increases the risk of cardiovascular and metabolic disease in adults, an inadequate supply of folic acid in the mother's diet may increase the susceptibility of the offspring to disease. We have fed laboratory rats diets deficient in folic acid and the related methyl donors methionine and choline to examine the effects on growth, blood pressure and insulin action in the offspring. Poor folate status transiently increased fetal growth but did not produce a long-term change in body weight. There were, however, small changes in the hearts of the female offspring. When folate deficiency was combined with low intakes of methionine and choline, the kidneys of the male offspring were proportionately smaller, probably because of the limited availability of methionine. There was no effect on the blood pressure of either the male or female offspring. The pancreatic insulin content of fetuses from animals fed the folate-deficient diets were higher than those of the controls. Following an oral glucose challenge, there was a weak trend for glucose-stimulated insulin release to be increased in the offspring of dams fed the folate-deficient diet. The changes in insulin concentrations were, however, much smaller than the corresponding changes observed in the offspring of animals fed protein-deficient diets. These results suggest that folate deficiency during gestation causes modest changes to the insulin axis of the fetus.


Assuntos
Pressão Sanguínea/fisiologia , Deficiência de Colina/fisiopatologia , Deficiência de Ácido Fólico/fisiopatologia , Metionina/deficiência , Fenômenos Fisiológicos da Nutrição Pré-Natal/fisiologia , Acetil-CoA Carboxilase/metabolismo , Animais , Carnitina O-Palmitoiltransferase/metabolismo , Deficiência de Colina/metabolismo , Ingestão de Alimentos/fisiologia , Feminino , Desenvolvimento Fetal/fisiologia , Peso Fetal/fisiologia , Deficiência de Ácido Fólico/metabolismo , Teste de Tolerância a Glucose , Crescimento/fisiologia , Insulina/metabolismo , Masculino , Tamanho do Órgão/fisiologia , Pâncreas/embriologia , Pâncreas/metabolismo , Gravidez , Complicações na Gravidez/fisiopatologia , Ratos
12.
Br J Nutr ; 101(12): 1878-84, 2009 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-19055852

RESUMO

The insulin-like growth factor (IGF) system is altered with intra-uterine growth retardation and in adult metabolic disease. The aim of the present study was to observe effects of continued protein restriction on the IGF-I system and body composition in offspring of mothers fed a low-protein (LP) diet. Offspring from Wistar dams fed either a 20 % (CON) or 8 % (LP) protein diet during gestation and lactation were studied at birth, 10 d, weaning and at 12 weeks after maintenance on either the 8 % (lp) or 20 % (con) protein diet from weaning. LP offspring had reduced weaning weights (P < 0.05) and reduced serum insulin (P < 0.005). Serum IGF-I (P < 0.001) and acid-labile subunit (ALS) (P < 0.0001) were reduced at 10 and 21 d. Hepatic expression of IGF-I (P < 0.05) and ALS (P < 0.005) were reduced at 10 and 21 d. IGF binding protein (IGFBP)-1 hepatic expression was elevated at 10 d (P < 0.001) but not at 21 d. Adult LP-con offspring had reduced body weight (P < 0.05), lean (P < 0.0001) and bone (P < 0.0001) but not fat (P = 0.6) mass with no persistent effects on IGF-I, ALS and IGFBP-1.Postnatal lp feeding reduced lean mass (P < 0.0001) and bone mass (P < 0.0001) in CON and LP animals. Percentage fat (LP P = 0.04; CON P = 0.6) and IGFBP-1 (LP P = 0.01; CON P = 0.2) were increased in LP-lp but not CON-lp offspring. This suggests that postnatal nutrition is important in the effects of maternal protein restriction on adult body composition and that IGFBP-1 may be involved.


Assuntos
Fenômenos Fisiológicos da Nutrição Animal , Animais Recém-Nascidos/crescimento & desenvolvimento , Dieta com Restrição de Proteínas , Fenômenos Fisiológicos da Nutrição Materna , Efeitos Tardios da Exposição Pré-Natal , Ração Animal , Animais , Animais Recém-Nascidos/metabolismo , Composição Corporal , Peso Corporal , Proteínas de Transporte/sangue , Feminino , Glicoproteínas/sangue , Insulina/sangue , Proteína 1 de Ligação a Fator de Crescimento Semelhante à Insulina/análise , Fator de Crescimento Insulin-Like I/análise , Lactação/fisiologia , Fígado/química , Gravidez , Ratos , Ratos Wistar , Fatores de Tempo , Desmame
13.
Sci Rep ; 7: 45753, 2017 03 31.
Artigo em Inglês | MEDLINE | ID: mdl-28362001

RESUMO

Unhealthy diets, and ensuing weight gain, predispose individuals to the development of esophageal adenocarcinoma. We examined the effect of chronic high fat diet (HFD) on the esophageal microbiota of Sprague Dawley rats using Illumina MiSeq amplicon sequencing (V4, 515 F/806 R) and on esophageal expression of IL18, PTGS2, PPARA, FFAR3, and CRAT. The relationships among metabolic parameters, esophageal microbiota, and host gene expression were determined. We observed a significant difference between the upper and lower esophageal microbiota in control fed rats, emphasized by enrichment of Lactobacillus species in the lower esophagus. Rats on HFD gained significantly more fat and had reduced insulin sensitivity. Diet type significantly affected the esophageal microbiota, with Clostridium sensu stricto being enriched in both upper and lower segments of HFD fed rats. Of interest, bacterial pathways related to carotenoid biosynthesis were significantly decreased in the lower esophagus of HFD fed rats. We observed strong correlations between metabolic parameters, the esophageal microbial profiles, and host esophageal gene expression. In particular, Fusobacterium, Rothia, and Granulicatella showed consistent correlations across a range of metabolic and gene markers. Our data indicates that unhealthy diets can significantly alter the esophageal microbiota, and enrich for bacterial species previously associated with chronic gastrointestinal diseases.


Assuntos
Dieta Hiperlipídica , Esôfago/metabolismo , Esôfago/microbiologia , Microbioma Gastrointestinal , Expressão Gênica , Animais , Resistência à Insulina , Masculino , Ratos Sprague-Dawley
14.
Nutrients ; 8(9)2016 Aug 23.
Artigo em Inglês | MEDLINE | ID: mdl-27563922

RESUMO

Along with diabetes and obesity, chronic kidney disease (CKD) is increasing across the globe. Although some data support an effect of maternal obesity on offspring kidney, the impact of paternal obesity is unknown; thus, we have studied the effect of paternal obesity prior to conception. Male Sprague Dawley rats were fed chow diet or high fat diet (HFD) for 13-14 weeks before mating with chow-fed females. Male offspring were weaned onto chow and killed at 27 weeks for renal gene expression and histology. Fathers on HFD were 30% heavier than Controls at mating. At 27 weeks of age offspring of obese fathers weighed 10% less; kidney triglyceride content was significantly increased (5.35 ± 0.84 vs. 2.99 ± 0.47 µg/mg, p < 0.05, n = 8 litters per group. Histological analysis of the kidney demonstrated signs of tubule damage, with significantly greater loss of brush border, and increased cell sloughing in offspring of obese compared to Control fathers. Acat1, involved in entry of fatty acid for beta-oxidation, was significantly upregulated, possibly to counteract increased triglyceride storage. However other genes involved in lipid metabolism, inflammation and kidney injury showed no changes. Paternal obesity was associated with renal triglyceride accumulation and histological changes in tubules, suggesting a mild renal insult in offspring, who may be at risk of developing CKD.


Assuntos
Gorduras na Dieta/administração & dosagem , Túbulos Renais/efeitos dos fármacos , Metabolismo dos Lipídeos , Herança Paterna/genética , Insuficiência Renal Crônica/genética , Animais , Creatinina/sangue , Eletrólitos/sangue , Ácidos Graxos não Esterificados/sangue , Feminino , Túbulos Renais/patologia , Metabolismo dos Lipídeos/genética , Lipídeos/administração & dosagem , Lipídeos/sangue , Masculino , Obesidade/genética , Ratos , Ratos Sprague-Dawley , Albumina Sérica
15.
Asian J Androl ; 18(6): 930-936, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-26608942

RESUMO

There is now strong evidence that the paternal contribution to offspring phenotype at fertilisation is more than just DNA. However, the identity and mechanisms of this nongenetic inheritance are poorly understood. One of the more important questions in this research area is: do changes in sperm DNA methylation have phenotypic consequences for offspring? We have previously reported that offspring of obese male rats have altered glucose metabolism compared with controls and that this effect was inherited through nongenetic means. Here, we describe investigations into sperm DNA methylation in a new cohort using the same protocol. Male rats on a high-fat diet were 30% heavier than control-fed males at the time of mating (16-19 weeks old, n = 14/14). A small (0.25%) increase in total 5-methyl-2Ͳ-deoxycytidine was detected in obese rat spermatozoa by liquid chromatography tandem mass spectrometry. Examination of the repetitive fraction of the genome with methyl-CpG binding domain protein-enriched genome sequencing (MBD-Seq) and pyrosequencing revealed that retrotransposon DNA methylation states in spermatozoa were not affected by obesity, but methylation at satellite repeats throughout the genome was increased. However, examination of muscle, liver, and spermatozoa from male 27-week-old offspring from obese and control fathers (both groups from n = 8 fathers) revealed that normal DNA methylation levels were restored during offspring development. Furthermore, no changes were found in three genomic imprints in obese rat spermatozoa. Our findings have implications for transgenerational epigenetic reprogramming. They suggest that postfertilization mechanisms exist for normalising some environmentally-induced DNA methylation changes in sperm cells.


Assuntos
Metilação de DNA/genética , Epigênese Genética , Obesidade/genética , Espermatozoides/metabolismo , Animais , Feminino , Genoma , Masculino , Obesidade/metabolismo , Ratos
16.
PLoS One ; 10(5): e0126931, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-25992554

RESUMO

The gut microbiota is emerging as a new factor in the development of obesity. Many studies have described changes in microbiota composition in response to obesity and high fat diet (HFD) at the phylum level. In this study we used 16s RNA high throughput sequencing on faecal samples from rats chronically fed HFD or control chow (n = 10 per group, 16 weeks) to investigate changes in gut microbiota composition at the species level. 53.17% dissimilarity between groups was observed at the species level. Lactobacillus intestinalis dominated the microbiota in rats under the chow diet. However this species was considerably less abundant in rats fed HFD (P<0.0001), this being compensated by an increase in abundance of propionate/acetate producing species. To further understand the influence of these species on the development of the obese phenotype, we correlated their abundance with metabolic parameters associated with obesity. Of the taxa contributing the most to dissimilarity between groups, 10 presented significant correlations with at least one of the tested parameters, three of them correlated positively with all metabolic parameters: Phascolarctobacterium, Proteus mirabilis and Veillonellaceae, all propionate/acetate producers. Lactobacillus intestinalis was the only species whose abundance was negatively correlated with change in body weight and fat mass. This species decreased drastically in response to HFD, favouring propionate/acetate producing bacterial species whose abundance was strongly correlated with adiposity and deterioration of metabolic factors. Our observations suggest that these species may play a key role in the development of obesity in response to a HFD.


Assuntos
Dieta Hiperlipídica , Metabolismo Energético , Microbioma Gastrointestinal , Obesidade/etiologia , Obesidade/metabolismo , Animais , Biodiversidade , Biomarcadores , Peso Corporal , Dieta , Glucose/metabolismo , Insulina/metabolismo , Masculino , Metagenoma , Obesidade/sangue , Ratos
17.
Genes Nutr ; 8(2): 181-90, 2013 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-22907820

RESUMO

A methyl-deficient diet (MD) lacking folic acid and the associated methyl donors choline and methionine, fed to the laboratory rat during the periods of oocyte and embryo development, has been shown to programme glucose metabolism in the offspring. The hepatic proteome of the male offspring of female rats fed MD diets for 3 weeks prior to mating and for the first 5 days of gestation has been examined by 2-dimensional gel electrophoresis. Three groups of differentially abundant proteins associated with energy metabolism, amino acid metabolism and antioxidant defence were identified in the soluble proteins extracted from the liver from the MD offspring at both 6 and 12 months of age. Altered mitochondrial activity in other programming models leads to a similar pattern of differential protein abundance. Two of the differentially abundant proteins were identified as GAPDH and PGK-1 by mass spectrometry. Western blotting showed that there were multiple isoforms of both proteins with similar molecular weights but different isoelectric points. The differentially abundant spots reduced in the MD offspring corresponded to minor isoforms of GAPDH and PGK-1. The levels of PPAR-alpha, SREBP and glucocorticoid receptor mRNAs associated with other models of prenatal programming were unchanged in the MD offspring. The data suggest that a diet deficient in folic acid and associated methyl donors fed during the peri-conception and early preimplantation periods of mammalian development affects mitochondrial function in the offspring and that the posttranslational modification of proteins may be important.

18.
Epigenetics ; 8(6): 602-11, 2013 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-23764993

RESUMO

Intrauterine nutrition can program metabolism, creating stable changes in physiology that may have significant health consequences. The mechanism underlying these changes is widely assumed to involve epigenetic changes to the expression of metabolic genes, but evidence supporting this idea is limited. Here we have performed the first study of the epigenomic consequences of exposure to maternal obesity and diabetes. We used a mouse model of natural-onset obesity that allows comparison of genetically identical mice whose mothers were either obese and diabetic or lean with a normal metabolism. We find that the offspring of obese mothers have a latent metabolic phenotype that is unmasked by exposure to a Western-style diet, resulting in glucose intolerance, insulin resistance and hepatic steatosis. The offspring show changes in hepatic gene expression and widespread but subtle alterations in cytosine methylation. Contrary to expectation, these molecular changes do not point to metabolic pathways but instead reside in broadly developmental ontologies. We propose that, rather than being adaptive, these changes may simply produce an inappropriate response to suboptimal environments; maladaptive phenotypes may be avoidable if postnatal nutrition is carefully controlled.


Assuntos
Diabetes Mellitus Tipo 2/metabolismo , Epigênese Genética , Expressão Gênica , Fígado/metabolismo , Obesidade/metabolismo , Complicações na Gravidez/metabolismo , Animais , Diabetes Mellitus Tipo 2/genética , Dieta , Feminino , Desenvolvimento Fetal , Fígado/patologia , Masculino , Camundongos , Mitocôndrias Hepáticas/genética , Mitocôndrias Hepáticas/metabolismo , Gravidez , Gravidez em Diabéticas/metabolismo
19.
Obesity (Silver Spring) ; 18(8): 1593-600, 2010 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-20019681

RESUMO

This study tests the hypothesis that a high-fat postnatal diet increases fat mass and reduces improved insulin sensitivity (IS) found in the low-protein model of maternal undernutrition. Offspring from Wistar dams fed either a 20% (control (CON)) or 8% (low protein (LP)) protein diet during gestation and lactation were randomly assigned to a control (con) or cafeteria (caf) diet at weaning (21 days) until 3 months of age at which point IS was measured (hyperinsulinemic-euglycemic clamp). Fat mass, growth, energy intake (EI) and expenditure (EE), fuel utilization, insulin secretion, and leptin and adiponectin levels were measured to identify a possible role in any changes in IS. IS was increased in LP-con in comparison to CON-con animals. Cafeteria feeding prevented this increase in LP animals but had no effect in CON animals (insulin-stimulated glucose infusion rates (GIRs; mg/min/kg); CON-con: 13.9 +/- 1.0, CON caf: 12.1 +/- 2.1, LP-con: 25.4 +/- 2.0, LP-caf: 13.7 +/- 3.7, P < 0.05). CON-caf animals had similar percent epididymal white adipose tissue (%EWAT; CON-con: 1.71 +/- 0.09 vs. CON-caf: 1.66 +/- 0.08) and adiponectin (microg/ml: CON-con: 4.61 +/- 0.34 vs. CON-caf: 3.67 +/- 0.18) except hyperinsulinemia and relative hyperleptinemia in comparison to CON-con. Differently, LP-caf animals had increased %EWAT (LP-con: 1.11 +/- 0.06 vs. LP-caf: 1.44 +/- 0.08, P < 0.05) and adiponectin (microg/ml: LP-con: 5.38 +/- 0.39 vs. LP-caf: 3.75 +/- 0.35, P < 0.05) but did not show cafeteria-induced hyperinsulinemia or relative hyperleptinemia. An increased propensity to store visceral fat in LP animals may prevent the elevated IS in LP offspring.


Assuntos
Glicemia/metabolismo , Dieta com Restrição de Proteínas , Gorduras na Dieta/farmacologia , Resistência à Insulina , Insulina/sangue , Gordura Intra-Abdominal/metabolismo , Obesidade Abdominal , Adiponectina/sangue , Animais , Animais Recém-Nascidos , Feminino , Hiperinsulinismo/sangue , Leptina/sangue , Masculino , Obesidade Abdominal/sangue , Gravidez , Distribuição Aleatória , Ratos , Ratos Wistar
20.
Epigenomics ; 2(4): 539-49, 2010 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-22121973

RESUMO

Within the Western world's aging and increasingly overweight population, we are seeing an increasing prevalence of adult-onset, lifestyle-related disease such as diabetes, hypertension and atherosclerosis. There is significant evidence that suboptimal nutrition in pregnancy can lead to an increased risk of these diseases developing in offspring, and that this increased risk can be heritable. Thus, poor in utero nutrition may be a major contributor to the current cycle of obesity. While the molecular basis of this phenomenon is unknown, available evidence suggests that it can be mediated by epigenetic changes to gene expression. Here, we discuss epigenetics as a mediator of disease risk in response to nutritional cues. The potential for maternal nutrition to heritably alter epigenetic states may have implications for population health and adaptive evolution.


Assuntos
Metilação de DNA/fisiologia , Epigênese Genética/fisiologia , Desenvolvimento Fetal/fisiologia , Regulação da Expressão Gênica no Desenvolvimento/fisiologia , Obesidade/etiologia , Fenômenos Fisiológicos da Nutrição Pré-Natal/fisiologia , Adaptação Fisiológica/genética , Animais , Feminino , Humanos , Camundongos , Camundongos Mutantes , Gravidez
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA