Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 84
Filtrar
Mais filtros

Base de dados
País/Região como assunto
Tipo de documento
Intervalo de ano de publicação
1.
Diabet Med ; 41(2): e15243, 2024 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-37845186

RESUMO

AIMS: The impact of maternal metformin use during pregnancy on fetal, infant, childhood and adolescent growth, development, and health remains unclear. Our objective was to systematically review the available evidence from animal experiments on the effects of intrauterine metformin exposure on offspring's anthropometric, cardiovascular and metabolic outcomes. METHODS: A systematic search was conducted in PUBMED and EMBASE from inception (searched on 12th April 2023). We extracted original, controlled animal studies that investigated the effects of maternal metformin use during pregnancy on offspring anthropometric, cardiovascular and metabolic measurements. Subsequently, risk of bias was assessed and meta-analyses using the standardized mean difference and a random effects model were conducted for all outcomes containing data from 3 or more studies. Subgroup analyses were planned for species, strain, sex and type of model in the case of 10 comparisons or more per subgroup. RESULTS: We included 37 articles (n = 3133 offspring from n = 716 litters, containing n = 51 comparisons) in this review, mostly (95%) on rodent models and 5% pig models. Follow-up of offspring ranged from birth to 2 years of age. Thirty four of the included articles could be included in the meta-analysis. No significant effects in the overall meta-analysis of metformin on any of the anthropometric, cardiovascular and metabolic offspring outcome measures were identified. Between-studies heterogeneity was high, and risk of bias was unclear in most studies as a consequence of poor reporting of essential methodological details. CONCLUSION: This systematic review was unable to establish effects of metformin treatment during pregnancy on anthropometric, cardiovascular and metabolic outcomes in non-human offspring. Heterogeneity between studies was high and reporting of methodological details often limited. This highlights a need for additional high-quality research both in humans and model systems to allow firm conclusions to be established. Future research should include focus on the effects of metformin in older offspring age groups, and on outcomes which have gone uninvestigated to date.


Assuntos
Diabetes Mellitus , Metformina , Gravidez , Animais , Feminino , Humanos , Gravidez/efeitos dos fármacos , Experimentação Animal , Antropometria , Hipoglicemiantes/efeitos adversos , Metformina/efeitos adversos , Cuidado Pré-Natal , Suínos , Camundongos , Ratos , Modelos Animais , Diabetes Mellitus/tratamento farmacológico
2.
Reprod Biomed Online ; 38(5): 779-786, 2019 May.
Artigo em Inglês | MEDLINE | ID: mdl-30885667

RESUMO

RESEARCH QUESTION: The physiological processes of pregnancy and lactation require profound changes in maternal metabolism and energy balance. The timescale of metabolic reversion after pregnancy, in particular post-partum weight loss, is highly variable between individuals. Currently, mechanisms influencing post-partum metabolic recovery are not well understood. The hypothesis tested here is that, in common with other metabolic and obesity-related outcomes, capacity for post-partum weight loss is influenced by developmental programming. DESIGN: Adult female Wistar rats exposed to a maternal low-protein diet in utero then weaned onto a control diet post-natally (recuperated group) were compared with controls. Adult females from both groups underwent pregnancy at 3 months of age. Weight changes and metabolic parameters during pregnancy and lactation were compared between control and recuperated groups, and also with non-pregnant littermates. RESULTS: Pregnancy weight gain was not different between the control and recuperated groups, but post-partum recuperated animals remained significantly heavier than both post-partum control animals (P<0.05) and their non-pregnant recuperated littermates (P<0.05) at the end of lactation. Post-partum recuperated animals had more intra-abdominal fat mass (P<0.05) and higher serum triglyceride concentrations (P<0.01) than controls. Post-partum recuperated animals also had increased expression of IL6, NRF2 and ALOX12 (key regulators of inflammation and lipoxygenase activity) in the intra-abdominal adipose tissue compared with control groups. CONCLUSIONS: Mothers who themselves have been exposed to adverse early life environments are likely to have slower metabolic recovery from pregnancy than controls. Failure to return to pre-pregnancy weight after delivery predisposes to persisting sequential inter-pregnancy weight gain, which can represent a significant metabolic burden across a life course involving several pregnancies.


Assuntos
Dieta com Restrição de Proteínas/efeitos adversos , Período Pós-Parto/metabolismo , Efeitos Tardios da Exposição Pré-Natal , Fenômenos Fisiológicos da Nutrição Pré-Natal , Redução de Peso , Animais , Feminino , Folículo Ovariano , Gravidez , Ratos Wistar
3.
Int J Obes (Lond) ; 40(2): 229-38, 2016 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-26367335

RESUMO

Maternal obesity is associated with an increased risk of developing gestational diabetes mellitus and it also results in an increased risk of giving birth to a large baby with increased fat mass. Furthermore, it is also contributes to an increased risk of obesity and insulin resistance in the offspring in childhood, adolescence and adult life. It has been proposed that exposure to maternal obesity may therefore result in an 'intergenerational cycle' of obesity and insulin resistance. There is significant interest in whether exposure to maternal obesity around the time of conception alone contributes directly to poor metabolic outcomes in the offspring and whether dieting in the obese mother before pregnancy or around the time of conception has metabolic benefits for the offspring. This review focusses on experimental and clinical studies that have investigated the specific impact of exposure to maternal obesity during the periconceptional period alone or extending beyond conception on adipogenesis, lipogenesis and on insulin signalling pathways in the fat, liver and muscle of the offspring. Findings from these studies highlight the need for a better evidence base for the development of dietary interventions in obese women before pregnancy and around the time of conception to maximize the metabolic benefits and minimize the metabolic costs for the next generation.


Assuntos
Diabetes Gestacional/etiologia , Obesidade/complicações , Obesidade Infantil/etiologia , Complicações na Gravidez/metabolismo , Efeitos Tardios da Exposição Pré-Natal/etiologia , Adulto , Feminino , Humanos , Lactente , Resistência à Insulina , Fenômenos Fisiológicos da Nutrição Materna , Mães , Obesidade/metabolismo , Obesidade/fisiopatologia , Obesidade Infantil/prevenção & controle , Gravidez , Efeitos Tardios da Exposição Pré-Natal/prevenção & controle , Fatores de Risco , Transdução de Sinais
4.
Int J Obes (Lond) ; 38(10): 1282-9, 2014 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-24785102

RESUMO

OBJECTIVE: Maternal obesity is associated with increased risk of metabolic dysfunction in the offspring. It is not clear whether it is the metabolic changes or chronic low-grade inflammation in the obese state that causes this metabolic programming. We therefore investigated whether low-grade inflammation was present in obese dams compared with controls dams at gestation day 18 (GD18). METHODS: Female mice were fed either a standard chow diet or a highly palatable obesogenic diet for 6 weeks before conception. Mice were either kileed before mating (n=12 in each group) or on GD18 (n=8 in each group). Blood and tissues were collected for analysis. RESULTS: The obesogenic diet increased body weight and decreased insulin sensitivity before conception, while there was no difference between the groups at GD18. Local inflammation was assayed by macrophage count in adipose tissue (AT) and liver. Macrophage count in the AT was increased significantly by the obesogenic diet, and the hepatic count also showed a tendency to increased macrophage infiltration before gestation. This was further supported by a decreased population of monocytes in the blood of the obese animals, which suggested that monocytes are being recruited from the blood to the liver and AT in the obese animals. Gestation reversed macrophage infiltration, such that obese dams showed a lower AT macrophage count at the end of gestation compared with pre-pregnancy obese mice, and there were no longer a tendency toward increased hepatic macrophage count. Placental macrophage count was also similar in the two groups. CONCLUSION: At GD18, obese dams were found to have similar macrophage infiltration in placenta, AT and liver as lean dams, despite an incipient infiltration before gestation. Thus, the obesity-induced inflammation was reversed during gestation.


Assuntos
Desenvolvimento Fetal , Inflamação/patologia , Fígado/metabolismo , Síndrome Metabólica/patologia , Obesidade/patologia , Efeitos Tardios da Exposição Pré-Natal/patologia , Animais , Modelos Animais de Doenças , Feminino , Desenvolvimento Fetal/imunologia , Citometria de Fluxo , Imuno-Histoquímica , Inflamação/imunologia , Síndrome Metabólica/imunologia , Camundongos , Camundongos Endogâmicos C57BL , Obesidade/imunologia , Gravidez , Efeitos Tardios da Exposição Pré-Natal/imunologia , Aumento de Peso/imunologia
5.
Int J Obes (Lond) ; 37(8): 1051-7, 2013 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-23229735

RESUMO

BACKGROUND: It is now widely accepted that the early-life nutritional environment is important in determining susceptibility to metabolic diseases. In particular, intra-uterine growth restriction followed by accelerated postnatal growth is associated with an increased risk of obesity, type-2 diabetes and other features of the metabolic syndrome. The mechanisms underlying these observations are not fully understood. AIM: Using a well-established maternal protein-restriction rodent model, our aim was to determine if exposure to mismatched nutrition in early-life programmes adipose tissue structure and function, and expression of key components of the insulin-signalling pathway. METHODS: Offspring of dams fed a low-protein (8%) diet during pregnancy were suckled by control (20%)-fed dams to drive catch-up growth. This 'recuperated' group was compared with offspring of dams fed a 20% protein diet during pregnancy and lactation (control group). Epididymal adipose tissue from 22-day and 3-month-old control and recuperated male rats was studied using histological analysis. Expression and phosphorylation of insulin-signalling proteins and gene expression were assessed by western blotting and reverse-transcriptase PCR, respectively. RESULTS: Recuperated offspring at both ages had larger adipocytes (P<0.001). Fasting serum glucose, insulin and leptin levels were comparable between groups but increased with age. Recuperated offspring had reduced expression of IRS-1 (P<0.01) and PI3K p110ß (P<0.001) in adipose tissue. In adult recuperated rats, Akt phosphorylation (P<0.01) and protein levels of Akt-2 (P<0.01) were also reduced. Messenger RNA expression levels of these proteins were not different, indicating a post-transcriptional effect. CONCLUSION: Early-life nutrition programmes alterations in adipocyte cell size and impairs the protein expression of several insulin-signalling proteins through post-transcriptional mechanisms. These indices may represent early markers of insulin resistance and metabolic disease risk.


Assuntos
Diabetes Mellitus Tipo 2/patologia , Retardo do Crescimento Fetal/patologia , Resistência à Insulina , Síndrome Metabólica/patologia , Obesidade/patologia , Proteínas Proto-Oncogênicas c-akt/metabolismo , Adipócitos , Tecido Adiposo/patologia , Animais , Western Blotting , Peso Corporal , Feminino , Desenvolvimento Fetal , Retardo do Crescimento Fetal/metabolismo , Expressão Gênica , Insulina , Masculino , Síndrome Metabólica/metabolismo , Fenótipo , Fosforilação , Gravidez , Efeitos Tardios da Exposição Pré-Natal , Ratos , Ratos Wistar
6.
Int J Obes (Lond) ; 36(8): 1040-6, 2012 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-22124449

RESUMO

BACKGROUND: Pups of normally nourished dams that are cross-fostered after birth to dams fed a low-protein (8% by weight) diet (postnatal low protein (PLP)) grow slower during the suckling period and remain small and lean throughout adulthood. At weaning, they have increased expression in the arcuate nucleus (ARC) of the hypothalamus of the orexigenic neuropeptide Y (NPY) and decreased expression of pro-opiomelanocortin, the precursor of anorexigenic melanocortins. OBJECTIVES AND METHODS: We investigated, using third ventricle administration, whether 3-month-old male PLP rats display altered sensitivity to leptin with respect to food intake, NPY and the melanocortin 3/4-receptor agonist MTII, and using in situ hybridization or laser capture microdissection of the ARC followed by RT-PCR, whether the differences observed were associated with changes in the hypothalamic expression of NPY or the leptin receptor, NPY receptors and melanocortin receptors. RESULTS: PLP rats were smaller and had reduced percentage body fat content and plasma leptin concentration compared with control rats. Leptin (5 µg) reduced food intake over 0-48 h more in PLP than control rats (P<0.05). Submaximal doses of NPY increased the food intake less in PLP rats than in controls, whereas submaximal doses of MTII reduced the food intake more in PLP rats. Maximal responses did not differ between PLP and control rats. Leptin and melanocortin-3 receptor (MC3R) expression were increased in both ARC and ventromedial hypothalamic nuclei in PLP animals compared with the controls. MC4R, NPY Y1R, Y5R and NPY expression were unchanged. CONCLUSION: Postnatal undernourishment results in food intake in adult rats being more sensitive to reduction by leptin and melanocortins, and less sensitive to stimulation by NPY. We propose that this contributes to increased leptin sensitivity and resistance to obesity. Increased expression of ObRb and MC3R may partly explain these findings but other downstream mechanisms must also be involved.


Assuntos
Animais Recém-Nascidos/crescimento & desenvolvimento , Núcleo Arqueado do Hipotálamo/patologia , Leptina/metabolismo , Neuropeptídeo Y/metabolismo , Obesidade/genética , Receptor Tipo 3 de Melanocortina/metabolismo , Magreza/genética , Animais , Núcleo Arqueado do Hipotálamo/fisiologia , Peso Corporal/genética , Suscetibilidade a Doenças , Ingestão de Alimentos , Regulação da Expressão Gênica , Leptina/farmacologia , Masculino , Neuropeptídeo Y/farmacologia , Obesidade/metabolismo , Ratos , Ratos Wistar , Magreza/metabolismo , Fatores de Tempo , Aumento de Peso/genética
7.
FASEB J ; 25(1): 420-7, 2011 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-20923964

RESUMO

Molecular mechanisms predisposing people to insulin resistance are starting to emerge. Altered insulin signaling for hepatic gluconeogenesis and muscle glucose uptake is thought to play a central role. Development under suboptimal conditions is also known to increase the risk of insulin resistance in adulthood. However, the partial contributions of reduced oxygen vs. nutrient delivery to the fetus, two common adverse conditions in utero, to developmental programming of insulin resistance remain unknown. The aim of this study was to determine the effects of developmental hypoxia or undernutrition on the expression of insulin-signaling proteins in liver and skeletal muscle in adult rat offspring. We show that the expression of hepatic phospho-Akt and muscle Akt2 were significantly reduced in offspring of hypoxic, relative to offspring from normoxic or undernourished, pregnancies. Hepatic Akt-1, Akt-2, and PKCζ protein expression was reduced in offspring from both hypoxic and undernourished pregnancies. Muscle GLUT4 expression was decreased in undernourished, and further decreased in hypoxic, offspring. These findings link prenatal hypoxia to down-regulation of components of hepatic and muscle Akt expression in adult offspring. Akt may represent a pharmaceutical target for clinical intervention against the developmental programming of metabolic disease resulting from prenatal hypoxia.


Assuntos
Biomarcadores/metabolismo , Hipóxia/fisiopatologia , Resistência à Insulina/fisiologia , Efeitos Tardios da Exposição Pré-Natal , Animais , Western Blotting , Peso Corporal , Feminino , Transportador de Glucose Tipo 4/metabolismo , Insulina/sangue , Lipídeos/sangue , Tamanho da Ninhada de Vivíparos , Fígado/metabolismo , Masculino , Desnutrição/fisiopatologia , Músculo Esquelético/metabolismo , Gravidez , Proteína Quinase C/metabolismo , Proteínas Proto-Oncogênicas c-akt/metabolismo , Ratos , Ratos Wistar , Fatores de Tempo
8.
Diabetes Obes Metab ; 14 Suppl 3: 29-39, 2012 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-22928562

RESUMO

Type 2 diabetes (T2D), also known as non-insulin dependent diabetes mellitus, arises as a consequence of peripheral insulin resistance in combination with an inability of pancreatic islet ß-cells to secrete adequate amounts of insulin. It is widely recognized that the current environment (e.g. an unhealthy diet and sedentary lifestyle) contributes to this process. In recent years, however, the role of the early environment, particularly nutrition, has emerged as an important factor capable of influencing health and disease risk of an individual, including risk of T2D. The impact of early environment on glucose metabolism has been extensively studied. Compelling evidence from epidemiological studies and animal models suggests that early nutrition can affect insulin action as a mediator of glucose homeostasis in peripheral tissues and as an important regulator of appetite and body weight. The early environment can also affect ß-cell mass and function, and hence insulin secretion. The molecular mechanisms underlying the relationship between a suboptimal early environment and impaired insulin action and secretion is thought to include epigenetic modifications of the foetal genome, oxidative stress and mitochondrial dysfunction.


Assuntos
Glicemia/metabolismo , Diabetes Mellitus Tipo 2/metabolismo , Fenômenos Fisiológicos da Nutrição do Lactente , Células Secretoras de Insulina/metabolismo , Insulina/metabolismo , Fenômenos Fisiológicos da Nutrição Materna , Efeitos Tardios da Exposição Pré-Natal/metabolismo , Animais , Peso ao Nascer , Glicemia/genética , Diabetes Mellitus Tipo 2/genética , Modelos Animais de Doenças , Epigênese Genética , Feminino , Humanos , Recém-Nascido , Resistência à Insulina , Secreção de Insulina , Masculino , Gravidez , Efeitos Tardios da Exposição Pré-Natal/genética
9.
Benef Microbes ; 13(5): 365-381, 2022 Nov 16.
Artigo em Inglês | MEDLINE | ID: mdl-36377578

RESUMO

The intestinal microbiota plays a major role in infant health and development. However, the role of the breastmilk microbiota in infant gut colonisation remains unclear. A systematic review was performed to evaluate the composition of the breastmilk microbiota and evidence for transfer to/colonisation of the infant gut. Searches were performed using PUBMED, OVID, LILACS and PROQUEST from inception until 18th March 2020 with a PUBMED update to December 2021. 88 full texts were evaluated before final critique based on study power, sample contamination avoidance, storage, purification process, DNA extraction/analysis, and consideration of maternal health and other potential confounders. Risk of skin contamination was reduced mainly by breast cleaning and rejecting the first milk drops. Sample storage, DNA extraction and bioinformatics varied. Several studies stored samples under conditions that may selectively impact bacterial DNA preservation, others used preculture reducing reliability. Only 15 studies, with acceptable sample size, handling, extraction, and bacterial analysis, considered transfer of bacteria to the infant. Three reported bacterial transfer from infant to breastmilk. Despite consistent evidence for the breastmilk microbiota, and recent studies using improved methods to investigate factors affecting its composition, few studies adequately considered transfer to the infant gut providing very little evidence for effective impact on gut colonisation.


Assuntos
Microbiota , Probióticos , Lactente , Feminino , Humanos , Leite Humano/microbiologia , Reprodutibilidade dos Testes , Bactérias/genética , DNA Bacteriano/genética
10.
Int J Obes (Lond) ; 35(7): 883-90, 2011 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-21587200

RESUMO

Obesity now represents one of the major health care issues of the 21st century. Its prevalence has increased exponentially in both the developed and developing world during the last couple of decades. Such a rapid rise can therefore not be explained by a change in genotype, but must result from environmental factors and their interaction with our genes. There is clear evidence to show that current environmental factors such as current diet and level of physical activity can influence our risk of obesity. However, there is growing evidence to suggest that factors acting during very early life can influence long-term energy balance. One such factor that is emerging as an important player is maternal obesity and/or over-nutrition during pregnancy and lactation. Early life may therefore represent a critical period during which intervention strategies could be developed to reduce the prevalence of obesity.


Assuntos
Diabetes Mellitus Tipo 2/prevenção & controle , Fenômenos Fisiológicos da Nutrição Materna/fisiologia , Obesidade/prevenção & controle , Fenômenos Fisiológicos da Nutrição Pré-Natal/fisiologia , Animais , Animais Recém-Nascidos , Diabetes Mellitus Tipo 2/etiologia , Metabolismo Energético , Feminino , Humanos , Obesidade/etiologia , Gravidez , Efeitos Tardios da Exposição Pré-Natal , Fatores de Risco
11.
Int J Obes (Lond) ; 35(3): 325-35, 2011 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-20680016

RESUMO

The mechanistic link between obese parents and obese offspring and the relative role of genes, and a shared environment is not completely understood. Animal models help us to differentiate between genetic and environmental factors, and the interaction between the two. However, the willingness of researchers to blend results from multiple models makes it difficult for clear mechanisms to be identified for specific hypothesis-driven research. As such we conducted a systematic review of animal models of maternal high fat feeding in an effort to identify the affect on the offspring glycaemic control. Maternal and offspring outcomes are reported in an effort to identify possible relationships to facilitate and focus on future research. We present here data from 11 studies investigating glycaemic control in offspring exposed to a high fat diet (HFD) during maternal gestation only or gestation and lactation. Studies in this review identify a real risk of type 2 diabetes and obesity in male offspring exposed to a maternal HFD. Poor glycaemic control in the offspring appears to be independent of maternal obesity, birth weight or post-weaning macronutrient intake. Inconsistencies between studies however, limit our capacity to identify mechanisms for the developmental origin of these diseases in animal models of overnutrition.


Assuntos
Glicemia/metabolismo , Peso Corporal/fisiologia , Gorduras na Dieta/administração & dosagem , Modelos Animais , Obesidade/metabolismo , Efeitos Tardios da Exposição Pré-Natal/metabolismo , Animais , Animais Recém-Nascidos , Peso ao Nascer/fisiologia , Feminino , Lactação , Masculino , Fenômenos Fisiológicos da Nutrição Materna , Gravidez , Ratos , Ratos Sprague-Dawley , Desmame
12.
Carcinogenesis ; 31(10): 1873-81, 2010 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-20460357

RESUMO

We previously showed that offspring of rat dams receiving a protein-restricted (low protein) diet throughout pregnancy and lactation develop mammary tumors more quickly. Rapid post-weaning mammary growth and mammary tissue overexpression of insulin receptor, insulin-like growth factor-1 receptor (IGF-1R), estrogen receptor isoform alpha and v-erb-b2 erythroblastic leukemia viral oncogene homolog 2 (ERBB2), correlated with this risk. The objectives of this study were therefore (i) to identify underlying mechanisms of increased risk through candidate and global approaches; (ii) to determine if excessive calorie intake further increased risk and if so, (iii) to identify the molecular mechanisms mediating this. We provide evidence for transcriptional upregulation of IGF-1R by Sp1 in LP mammary tissue (P < 0.01). Cell cycle control and DNA damage repair gene cyclin-dependent kinase inhibitor 1A (CDKN1A) (p21waf1) was also upregulated (P < 0.05) as was transcription factor nuclear factor of kappa light polypeptide gene enhancer in B-cell (P < 0.05) and adhesion gene CDH1 (P < 0.05). Invasion and metastasis markers matrix metalloproteinase 9 and serpin peptidase inhibitor, clade E, member 1 (SERPIN1) were upregulated (both P < 0.05), whereas metastasis suppressor gene NME1 was downregulated (P < 0.01). Feeding a highly palatable diet (HPD) to increase calorie intake from puberty, additively and independently increased early mammary tumor risk, which correlated with increased serum insulin and triglyceride concentrations (P < 0.05). PTEN gene expression was reduced both by early protein restriction (P < 0.05) and HPD (P < 0.01), which may induce Akt in cell survival pathways. Progesterone receptor and ERBB2 (both P < 0.05) expression increased as an effect of an interaction between maternal diet and adult nutrition, with subsequent downstream activation of the mitogen-activated protein kinase pathway. We conclude that poor early growth and excessive calorie intake exert independent and additive effects on mitogenic growth factor signaling to influence mammary tumor susceptibility.


Assuntos
Ingestão de Energia , Transtornos do Crescimento/complicações , Neoplasias Mamárias Animais/etiologia , Transdução de Sinais , Animais , Peso Corporal , Suscetibilidade a Doenças , MAP Quinases Reguladas por Sinal Extracelular/metabolismo , Feminino , Perfilação da Expressão Gênica , Glândulas Mamárias Animais/química , Glândulas Mamárias Animais/metabolismo , Ratos , Ratos Wistar , Receptor ErbB-2/genética , Receptor ErbB-2/fisiologia , Receptor IGF Tipo 1/análise , Receptor IGF Tipo 1/genética , Fator de Transcrição Sp1/análise , Fator de Transcrição Sp1/genética
13.
FASEB J ; 23(5): 1521-8, 2009 May.
Artigo em Inglês | MEDLINE | ID: mdl-19126595

RESUMO

Low birth weight (LBW) followed by accelerated postnatal growth is associated with increased risk of developing age-associated diseases such as type 2 diabetes. Gestational protein restriction in rats causes LBW, beta-cell dysfunction, and reduced longevity. These effects may be mediated by accelerated cellular aging. This study tested the hypothesis that LBW followed by rapid postnatal catch-up growth leads to islet telomere shortening through alterations in antioxidant defense capacity, stress/senescence marker proteins, and DNA repair mechanisms at the gene expression level. We used our rat model of gestational protein restriction (recuperated offspring) and control offspring. Southern blotting revealed shorter (P<0.001) islet telomeres in recuperated animals compared to controls. This was associated with increased expression of peroxiredoxin 1 (P<0.05), peroxiredoxin 3 (P<0.01), and heme oxygenase-1 (HO-1) (P<0.05), which are up-regulated under stress conditions. MnSOD expression was significantly (P<0.05) decreased in recuperated offspring, suggesting partial impairment of mitochondrial antioxidant defenses. Markers of cellular senescence p21 and p16 were also increased (P<0.01 and P<0.05, respectively) in the recuperated group. We conclude that maternal diet influences expression of markers of cellular stress and telomere length in pancreatic islets. This may provide a mechanistic link between early nutrition and growth and type 2 diabetes.


Assuntos
Senescência Celular/fisiologia , Ilhotas Pancreáticas/citologia , Fenômenos Fisiológicos da Nutrição Materna , Deficiência de Proteína/complicações , Telômero/metabolismo , Animais , Reparo do DNA/fisiologia , Feminino , Perfilação da Expressão Gênica , Gravidez , Ratos , Ratos Wistar , Superóxido Dismutase/metabolismo
14.
Mol Metab ; 42: 101079, 2020 12.
Artigo em Inglês | MEDLINE | ID: mdl-32919096

RESUMO

OBJECTIVE: Perinatal exposure to maternal obesity results in predisposition of offspring to develop obesity later in life. Increased weight gain in offspring exposed to maternal obesity is usually associated with hyperphagia, implicating altered central regulation of food intake as a cause. We aimed to define how maternal obesity impacts early development of the hypothalamus to program lasting dysfunction in feeding regulatory pathways. METHODS: Mice offspring of diet-induced obese mothers were compared to the offspring of lean control mothers. We analysed gene expression in the fetal hypothalamus, alongside neurosphere assays to investigate the effects of maternal obesity on neural progenitor cell proliferation in vitro. Western blotting was used to investigate the insulin signalling pathway in the fetal hypothalamus. Characterisation of cell type and neuropeptide profile in adulthood was linked with analyses of feeding behaviour. RESULTS: There was a reduction in the expression of proliferative genes in the fetal hypothalamus of offspring exposed to maternal obesity. This reduction in proliferation was maintained in vitro when hypothalamic neural progenitor cells were grown as neurospheres. Hypothalamic fetal gene expression and neurosphere growth correlated with maternal body weight and insulin levels. Foetuses of obese mothers showed hypothalamic insulin resistance, which may be causative of reduced proliferation. Furthermore, maternal obesity activated the Notch signalling pathway in neonatal offspring hypothalamus, resulting in decreased neurogenesis. Adult offspring of obese mothers displayed an altered ratio of anorexigenic and orexigenic signals in the arcuate nucleus, associated with an inability to maintain energy homeostasis when metabolically challenged. CONCLUSIONS: These findings show that maternal obesity alters the molecular signature in the developing hypothalamus, which is associated with disrupted growth and development of hypothalamic precursor cells and defective feeding regulation in adulthood. This is the first report of fetal hypothalamic insulin resistance in an obese pregnancy and suggests a mechanism by which maternal obesity causes permanent changes to hypothalamic structure and function.


Assuntos
Hipotálamo/embriologia , Resistência à Insulina/fisiologia , Obesidade Materna/fisiopatologia , Animais , Núcleo Arqueado do Hipotálamo/metabolismo , Peso Corporal , Encéfalo/metabolismo , Dieta Hiperlipídica , Comportamento Alimentar , Feminino , Feto/metabolismo , Feto/fisiopatologia , Expressão Gênica/genética , Regulação da Expressão Gênica no Desenvolvimento/genética , Hipotálamo/metabolismo , Insulina/metabolismo , Masculino , Troca Materno-Fetal/fisiologia , Camundongos , Camundongos Endogâmicos C57BL , Células-Tronco Neurais/metabolismo , Neurônios/metabolismo , Obesidade/metabolismo , Obesidade/fisiopatologia , Obesidade Materna/metabolismo , Gravidez , Efeitos Tardios da Exposição Pré-Natal/metabolismo , Aumento de Peso
15.
J Physiol ; 587(Pt 17): 4199-211, 2009 Sep 01.
Artigo em Inglês | MEDLINE | ID: mdl-19622603

RESUMO

A world-wide series of epidemiological and experimental studies have demonstrated that there is an association between being small at birth, accelerated growth in early postnatal life and the emergence of insulin resistance in adult life. The aim of this study was to investigate why accelerated growth occurs in postnatal life after in utero growth restriction. Samples of quadriceps muscle were collected at approximately 140 days gestation (term approximately 150 days gestation) from normally grown fetal lambs (Control, n = 7) and from growth restricted fetal lambs (placentally restricted: PR, n = 8) and from Control (n = 14) and PR (n = 9) lambs at 21 days after birth. The abundance of the insulin and IGF1 receptor protein was higher in the quadriceps muscle of the PR fetus, but there was a lower abundance of the insulin signalling molecule PKC, and GLUT4 protein in the PR group. At 21 days of postnatal age, insulin receptor abundance remained higher in the muscle of the PR lamb, and there was also an up-regulation of the insulin signalling molecules, PI3Kinase p85, Akt1 and Akt2 and of the GLUT4 protein in the PR group. Fetal growth restriction therefore results in an increased abundance of the insulin receptor in skeletal muscle, which persists after birth when it is associated with an upregulation of insulin signalling molecules and the glucose transporter, GLUT4. These data provide evidence that the origins of the accelerated growth experienced by the small baby after birth lie in the adaptive response of the growth restricted fetus to its low placental substrate supply.


Assuntos
Desenvolvimento Fetal , Retardo do Crescimento Fetal/metabolismo , Insulina/metabolismo , Modelos Biológicos , Músculo Esquelético/crescimento & desenvolvimento , Músculo Esquelético/metabolismo , Transdução de Sinais , Animais , Feminino , Gravidez , Ovinos
16.
FASEB J ; 22(6): 2037-44, 2008 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-18230683

RESUMO

Low birth weight is associated with increased cardiovascular disease (CVD) in humans. Detrimental effects of low birth weight are amplified by rapid catch-up growth. Conversely, slow growth during lactation reduces CVD risk. Gestational protein restriction causes low birth weight, vascular dysfunction, and accelerated aging in rats. Atherosclerotic aortic tissue has shortened telomeres, and oxidative stress accelerates telomere shortening through generation of DNA single-strand breaks (ssbs). This study tested the hypothesis that maternal diet influences aortic telomere length through changes in DNA ssbs, antioxidant capacity, and oxidative stress. We used our models of gestational protein restriction followed by rapid catch-up growth (the recuperated group) and protein restriction during lactation (the postnatal low-protein [PLP] group). Southern blotting revealed fewer aortic DNA ssbs and subsequently fewer short telomeres (P<0.05) in the PLP group. This result was associated with reduced (P<0.01) 8-hydroxy-2-deoxyguanosine, a marker of oxidative stress. PLP animals expressed increased (P<0.01) manganese superoxide-dismutase, copper-zinc superoxide-dismutase, catalase, and glutathione-reductase. Age-dependent changes in antioxidant defense enzymes indicated more protection to oxidative stress in the PLP animals; conversely, recuperated animals demonstrated age-associated impairment of antioxidant defenses. We conclude that maternal diet has a major influence on aortic telomere length. This finding may provide a mechanistic link between early growth patterns and CVD.


Assuntos
Antioxidantes , Dano ao DNA , Dieta , Estresse Oxidativo , Telômero/ultraestrutura , Fatores Etários , Animais , Aorta/ultraestrutura , Feminino , Modelos Animais , Mães , Ratos
18.
Obes Rev ; 20(5): 675-685, 2019 05.
Artigo em Inglês | MEDLINE | ID: mdl-30633422

RESUMO

Obesity before and during pregnancy leads to reduced offspring cardiometabolic health. Here, we systematically reviewed animal experimental evidence of maternal obesity before and during pregnancy and offspring anthropometry and cardiometabolic health. We systematically searched Embase and Medline from inception until January 2018. Eligible publications compared offspring of mothers with obesity to mothers with a normal weight. We performed meta-analyses and subgroup analyses. We also examined methodological quality and publication bias. We screened 2543 publications and included 145 publications (N = 21 048 animals, five species). Essential methodological details were not reported in the majority of studies. We found evidence of publication bias for birth weight. Offspring of mothers with obesity had higher body weight (standardized mean difference (SMD) 0.76 [95% CI 0.60;0.93]), fat percentage (0.99 [0.64;1.35]), systolic blood pressure (1.33 [0.75;1.91]), triglycerides (0.64 [0.42;0.86], total cholesterol (0.46 [0.18;0.73]), glucose level (0.43 [0.24;0.63]), and insulin level (0.81 [0.61;1.02]) than offspring of control mothers, but similar birth weight. Sex, age, or species did not influence the effect of maternal obesity on offspring's cardiometabolic health. Obesity before and during pregnancy reduces offspring cardiometabolic health in animals. Future intervention studies should investigate whether reducing obesity prior to conception could prevent these detrimental programming effects and improve cardiometabolic health of future generations.


Assuntos
Índice de Massa Corporal , Obesidade Materna/fisiopatologia , Complicações na Gravidez/fisiopatologia , Efeitos Tardios da Exposição Pré-Natal/fisiopatologia , Animais , Feminino , Obesidade Materna/metabolismo , Gravidez , Complicações na Gravidez/metabolismo , Efeitos Tardios da Exposição Pré-Natal/metabolismo
19.
Sci Rep ; 9(1): 7639, 2019 05 21.
Artigo em Inglês | MEDLINE | ID: mdl-31113969

RESUMO

Telomeres are protein-bound regions of repetitive nucleotide sequences (TTAGGG) at the end of human chromosomes, and their length is a marker of cellular aging. Intrauterine growth restriction is associated with shorter blood cell telomeres at birth and individuals with type 2 diabetes have shorter telomeres. Individuals with a low birth weight (LBW) have an increased risk of metabolic disease and type 2 diabetes. Therefore, we aimed to investigate the relationship between birth weight and telomere length and the association between birth weight, telomere length and cardiometabolic phenotype in adulthood. Young, healthy men with LBW (n = 55) and normal birth weight (NBW) (n = 65) were examined including blood pressure, blood samples and body composition. Leukocyte telomere length was determined using a high-throughput qPCR method. The LBW men were more insulin resistant as determined by the HOMA-IR index. There was no difference in telomere length between LBW and NBW subjects. When adjusting for birth weight and cohort effect, significant negative associations between telomere length and fasting glucose (P = 0.003) and HbA1c (P = 0.0008) were found. In conclusion, no significant difference in telomere length was found between LBW and NBW men. The telomere length was negatively associated with glucose concentrations and HbA1c levels within the normal non-diabetic range independent of birth weight.


Assuntos
Peso ao Nascer , Glicemia/genética , Hemoglobinas Glicadas/genética , Síndrome Metabólica/genética , Homeostase do Telômero , Adulto , Glicemia/metabolismo , Hemoglobinas Glicadas/metabolismo , Humanos , Recém-Nascido de Baixo Peso , Recém-Nascido , Leucócitos/metabolismo , Masculino , Síndrome Metabólica/sangue , Síndrome Metabólica/epidemiologia
20.
Physiol Behav ; 94(1): 17-28, 2008 Apr 22.
Artigo em Inglês | MEDLINE | ID: mdl-18155097

RESUMO

It is becoming increasingly apparent that conditions experienced in early life play an important role in the long-term health of individuals. Alterations in development due to impaired, excessive or imbalanced growth, both in utero and during critical periods of relative plasticity beyond birth, can lead to the permanent programming of physiological systems. The regulation of energy balance is one area that is receiving particular attention, as rates of obesity and associated metabolic and cardiovascular disease continue to rise. Over recent decades, much progress has been made toward understanding the way in which metabolic tissues and physiological systems develop, and the impact of early life events and nutrition on these processes. It is apparent within human populations that some individuals are better able to maintain an appropriate body weight in the face of an obesogenic environment. Animal models have been widely used for the investigation of differential susceptibility to diet-induced obesity (DIO) and impaired energy balance regulation, and are shedding light on key pathways that may be involved. Alterations in pathways mediating energy homeostasis, outlined below, are likely candidates for programming effects following disturbed growth in early life.


Assuntos
Doenças Metabólicas/fisiopatologia , Obesidade/fisiopatologia , Tecido Adiposo/crescimento & desenvolvimento , Tecido Adiposo/fisiologia , Animais , Dieta , Modelos Animais de Doenças , Metabolismo Energético/fisiologia , Feminino , Glucocorticoides/fisiologia , Crescimento/fisiologia , Homeostase/fisiologia , Humanos , Leptina/fisiologia , Hipernutrição/fisiopatologia , Fenótipo , Gravidez
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA