Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 84
Filtrar
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.
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
3.
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
4.
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
5.
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
6.
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
7.
J Dev Orig Health Dis ; 8(5): 513-519, 2017 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-28889823

RESUMO

Developmental origins of health and disease (DOHaD) is the study of how the early life environment can impact the risk of chronic diseases from childhood to adulthood and the mechanisms involved. Epigenetic modifications such as DNA methylation, histone modifications and non-coding RNAs are involved in mediating how early life environment impacts later health. This review is a summary of the Epigenetics and DOHaD workshop held at the 2016 DOHaD Society of Australia and New Zealand Conference. Our extensive knowledge of how the early life environment impacts later risk for chronic disease would not have been possible without animal models. In this review we highlight some animal model examples that demonstrate how an adverse early life exposure results in epigenetic and gene expression changes that may contribute to increased risk of chronic disease later in life. Type 2 diabetes and cardiovascular disease are chronic diseases with an increasing incidence due to the increased number of children and adults that are obese. Epigenetic changes such as DNA methylation have been shown to be associated with metabolic health measures and potentially predict future metabolic health status. Although more difficult to elucidate in humans, recent studies suggest that DNA methylation may be one of the epigenetic mechanisms that mediates the effects of early life exposures on later life risk of obesity and obesity related diseases. Finally, we discuss the role of the microbiome and how it is a new player in developmental programming and mediating early life exposures on later risk of chronic disease.


Assuntos
Exposição Ambiental/efeitos adversos , Epigênese Genética/fisiologia , Microbioma Gastrointestinal/fisiologia , Nível de Saúde , Efeitos Tardios da Exposição Pré-Natal/genética , Animais , Metilação de DNA/fisiologia , Comportamento Alimentar/fisiologia , Feminino , Humanos , Gravidez , Efeitos Tardios da Exposição Pré-Natal/metabolismo
8.
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
9.
Sci Rep ; 5: 16175, 2015 Nov 03.
Artigo em Inglês | MEDLINE | ID: mdl-26525600

RESUMO

Exposure to an adverse early-life environment leads to long-term health problems, many of which are recapitulated in subsequent generations. The female reproductive tract is particularly sensitive to early-life influences, and plays a pivotal role in programming the conceptus. We examine the influence of suboptimal grandmaternal diet on reproductive potential of granddaughters in the absence of any further dietary manipulations in the daughters in a rat low-protein diet model. Exposure to low-protein grand-maternal diet leads to decreased ovarian reserve and increased intra-abdominal fat mass in granddaughters, accompanied by accelerated accumulation of oxidative stress and mtDNA copy number instability in the ovaries. Ovarian telomere length declines more rapidly in the exposed granddaughters, indicating accelerated ageing in the reproductive tract. Thus, we demonstrate that suboptimal grandmaternal diet during pregnancy accelerates reproductive ageing across subsequent generations. These findings have important implications for understanding both individual rates of decline in fertility with age, and the clinical impact of current global trends towards delayed childbearing.


Assuntos
Dieta com Restrição de Proteínas , Reserva Ovariana/fisiologia , Envelhecimento , Aldeídos/análise , Animais , Hormônio Antimülleriano/sangue , Antioxidantes/metabolismo , Glicemia/análise , Peso Corporal , DNA Mitocondrial/química , DNA Mitocondrial/metabolismo , Ensaio de Imunoadsorção Enzimática , Feminino , Dosagem de Genes , Gordura Intra-Abdominal/fisiologia , Ovário/química , Ovário/metabolismo , Estresse Oxidativo , Gravidez , Ratos , Telômero/metabolismo
10.
Acta Physiol (Oxf) ; 214(4): 440-9, 2015 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-26011013

RESUMO

Maternal obesity during pregnancy increases the child's risk of developing obesity and obesity-related diseases later in life. Key components in foetal programming of metabolic risk remain to be identified; however, chronic low-grade inflammation associated with obesity might be responsible for metabolic imprinting in the offspring. We have therefore surveyed the literature to evaluate the role of maternal obesity-induced inflammation in foetal programming of obesity and related diseases. The literature on this topic is limited, so this review also includes animal models where maternal inflammation is mimicked by single injections with lipopolysaccharide (LPS). An LPS challenge results in an immunological response that resembles the obesity-induced immune profile, although LPS injections provoke a stronger response than the subclinical obesity-associated response. Maternal LPS or cytokine exposures result in increased adiposity and impaired metabolic homeostasis in the offspring, similar to the phenotype observed after exposure to maternal obesity. The cytokine levels might be specifically important for the metabolic imprinting, as cytokines are both transferable from maternal to foetal circulation and have the capability to modulate placental nutrient transfer. However, the immune response associated with obesity is moderate and therefore potentially weakened by the pregnancy-driven immune modulation, dominated by anti-inflammatory Treg and Th2 cells. We know from other low-grade inflammatory diseases, such as rheumatoid arthritis, that pregnancy can improve disease state. If pregnancy is also capable of suppressing the obesity-associated inflammation, the immunological markers might be less likely to affect metabolic programming in the developing foetus than otherwise implied.


Assuntos
Inflamação/complicações , Doenças Metabólicas/etiologia , Obesidade/complicações , Complicações na Gravidez , Efeitos Tardios da Exposição Pré-Natal , Animais , Feminino , Desenvolvimento Fetal , Humanos , Gravidez
11.
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
13.
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
14.
J Dev Orig Health Dis ; 4(5): 338-47, 2013 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-24970727

RESUMO

It is widely recognized that environmental insults during adulthood including smoking, lack of exercise and a poor diet increases an individual's risk of cardiovascular disease (CVD). However, research initiated over the last two decades has highlighted that our risk of CVD can be programmed following adverse exposures during early development. Such adverse exposures may include, undernutrition, placental insufficiency, hypoxia, overnutrition and obesity. This review aims to address the current Western obesity crisis by addressing the long-term impact of maternal overnutrition and obesity on the offspring's future risk of CVD. Although current human studies have observed the presence of adverse CVD markers in children born to obese mothers, animal models have proved vital in understanding the underlying mechanisms involved. Mechanisms suggested to be involved in the programming of CVD in the offspring include increased oxidative stress, inflammation, lipotoxicity and epigenetics. CVD remains the greatest cause of death worldwide, therefore further understanding of the mechanisms mediating these effects is important in the development of intervention strategies.

15.
J Dev Orig Health Dis ; 4(5): 421-9, 2013 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-24970733

RESUMO

Intrauterine growth restriction (IUGR) followed by accelerated growth after birth is associated with an increased risk of abdominal (visceral) obesity and insulin resistance in adult life. The aim of the present study was to determine the impact of IUGR on mRNA expression and protein abundance of insulin signaling molecules in one of the major visceral fat depots, the omental adipose depot. IUGR was induced by placental restriction, and samples of omental adipose tissue were collected from IUGR (n = 9, 5 males, 4 females) and Control (n = 14, 8 males, 6 females) neonatal lambs at 21 days of age. The mRNA expression of the insulin signaling molecules, AMP-kinase (AMPK) and adipogenic/lipogenic genes was determined by qRT-PCR, and protein abundance by Western Blotting. AMPKα2 mRNA expression was increased in male IUGR lambs (0.015 ± 0.002 v. 0.0075 ± 0.0009, P < 0.001). The proportion of the AMPK pool that was phosphorylated (%P-AMPK) was lower in IUGR lambs compared with Controls independent of sex (39 ± 9% v. 100 ± 18%, P < 0.001). The mRNA expression and protein abundance of insulin signaling proteins and adipogenic/lipogenic genes was not different between groups. Thus, IUGR is associated with sex-specific alterations in the mRNA expression of AMPKα2 and a reduction in the percentage of the total AMPK pool that is phosphorylated in the omental adipose tissue of neonatal lambs, before the onset of visceral obesity. These molecular changes would be expected to promote lipid accumulation in the omental adipose depot and may therefore contribute to the onset of visceral adiposity in IUGR animals later in life.

16.
Best Pract Res Clin Endocrinol Metab ; 26(5): 569-80, 2012 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-22980041

RESUMO

The global prevalence of type-2 diabetes (T2D) has more than doubled in the last 30 years and is predicted to continue to rise at an alarming rate. The associated health and financial burdens are considerable. The aetiology of common forms of T2D is multifactorial and involves a complex interplay between genetic, epigenetic and environmental factors. The influential role of the environment, in particular our diet and sedentary lifestyles, in diabetes risk is well established. Of major concern is the increasing prevalence of early onset T2D or pre-diabetic characteristics in children. In recent years, the role of the early life environment in programming diabetes risk has been the focus of numerous human and animal studies. Historical studies highlighted an association between low birthweight, a proxy for suboptimal in utero growth, and diabetes risk in adulthood. Over more recent years it has become apparent that a variety of expositions, including maternal obesity and/or maternal diabetes, can have a significant effect on offspring health outcomes. Further complicating matters, paternal and transgenerational transmission of T2D can occur thus mediating a perpetuating cycle of disease risk between generations. It is imperative for the underlying mechanisms to be elucidated so that interventions can be introduced. In doing so, it may be possible to prevent, delay or reverse a pre-programmed risk for T2D induced by pre- and/or postnatal environmental factors to improve health outcomes and curb premature metabolic decline. This review presents evidence for how the early life environment may programme T2D risk and suggests some mechanisms by which this may occur.


Assuntos
Diabetes Mellitus Tipo 2/etiologia , Efeitos Tardios da Exposição Pré-Natal , Adolescente , Adulto , Animais , Criança , Pré-Escolar , Diabetes Mellitus Tipo 2/epidemiologia , Diabetes Gestacional/etiologia , Dieta , Epigênese Genética , Feminino , Retardo do Crescimento Fetal/fisiopatologia , Humanos , Recém-Nascido de Baixo Peso/crescimento & desenvolvimento , Recém-Nascido , Recém-Nascido Pequeno para a Idade Gestacional/crescimento & desenvolvimento , Células Secretoras de Insulina/fisiologia , MicroRNAs/fisiologia , Modelos Animais , Obesidade/complicações , Estresse Oxidativo , Gravidez , Complicações na Gravidez , Fatores de Risco
17.
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
18.
Cell Death Differ ; 19(6): 1003-12, 2012 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-22223106

RESUMO

Nutrition during early mammalian development permanently influences health of the adult, including increasing the risk of type 2 diabetes and coronary heart disease. However, the molecular mechanisms underlying such programming are poorly defined. Here we demonstrate that programmed changes in miRNA expression link early-life nutrition to long-term health. Specifically, we show that miR-483-3p is upregulated in adipose tissue from low-birth-weight adult humans and prediabetic adult rats exposed to suboptimal nutrition in early life. We demonstrate that manipulation of miR-483-3p levels in vitro substantially modulates the capacity of adipocytes to differentiate and store lipids. We show that some of these effects are mediated by translational repression of growth/differentiation factor-3, a target of miR-483-3p. We propose that increased miR-483-3p expression in vivo, programmed by early-life nutrition, limits storage of lipids in adipose tissue, causing lipotoxicity and insulin resistance and thus increasing susceptibility to metabolic disease.


Assuntos
Tecido Adiposo/metabolismo , Diabetes Mellitus Tipo 2/metabolismo , Dieta , Fator 3 de Diferenciação de Crescimento/metabolismo , MicroRNAs/metabolismo , Regiões 3' não Traduzidas , Adulto , Animais , Animais Recém-Nascidos , Sequência de Bases , Diferenciação Celular , Diabetes Mellitus Tipo 2/patologia , Modelos Animais de Doenças , Regulação para Baixo , Feminino , Fator 3 de Diferenciação de Crescimento/antagonistas & inibidores , Fator 3 de Diferenciação de Crescimento/genética , Células HEK293 , Humanos , Metabolismo dos Lipídeos , Masculino , Interferência de RNA , RNA Interferente Pequeno/metabolismo , Ratos , Ratos Wistar
19.
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
20.
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
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...