Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 22
Filtrar
1.
Am J Cardiol ; 139: 64-70, 2021 01 15.
Artigo em Inglês | MEDLINE | ID: mdl-33115639

RESUMO

Elevated liver fibrosis markers are associated with worse prognosis in acute heart failure (AHF). The aspartate aminotransferase to alanine aminotransferase ratio (AAR) is one such fibrosis marker, and low ALT is a surrogate marker of malnutrition. Here, we evaluated the association between AAR and nutritional status and prognosis in patients with AHF. Consecutive 774 patients who were admitted due to AHF were divided into 3 groups according to AAR at discharge: first tertile, AAR<1.16 (n = 262); second tertile, 1.16≤AAR<1.70 (n = 257); and third tertile, AAR≥1.70 (n = 255). Nutritional indices and a composite of all-cause death or HF rehospitalization were compared in the 3 tertiles. Patients in the third AAR tertile were older and had lower body mass index than patients in other AAR tertiles. A higher AAR was associated with worse nutritional indices (i.e., controlling nutritional status score, geriatric nutritional risk index, and prognostic nutritional index). Clinical outcome rates significantly increased along AAR tertiles (first tertile, 28%; second tertile, 43%; third tertile, 58%, p < 0.001). Cox proportional hazards models including potential prognostic factors revealed high AAR was an independent prognostic factor of AHF. In conclusion, AAR at discharge may be associated with nutritional status and worse clinical outcomes in patients with AHF.


Assuntos
Alanina Transaminase/sangue , Aspartato Aminotransferases/sangue , Insuficiência Cardíaca/enzimologia , Desnutrição/embriologia , Estado Nutricional , Doença Aguda , Idoso , Idoso de 80 Anos ou mais , Biomarcadores/sangue , Índice de Massa Corporal , Causas de Morte/tendências , Feminino , Seguimentos , Insuficiência Cardíaca/complicações , Insuficiência Cardíaca/mortalidade , Humanos , Japão/epidemiologia , Masculino , Desnutrição/etiologia , Pessoa de Meia-Idade , Prognóstico , Estudos Retrospectivos , Fatores de Risco , Taxa de Sobrevida/tendências
2.
Int J Epidemiol ; 48(Suppl 1): i80-i88, 2019 Apr 01.
Artigo em Inglês | MEDLINE | ID: mdl-30883656

RESUMO

BACKGROUND: Levels of child undernutrition have declined in many middle-income countries, whereas overweight and obesity have increased. We describe time trends in nutritional indicators at age 1 year in the 1982, 1993, 2004 and 2015 Pelotas (Brazil) Birth Cohorts. METHODS: Each study included all children born in the urban area of the city, with over 4 200 births in each cohort. Children were measured at approximately 12 months of age. Anthropometric indicators were calculated according to World Health Organization Growth Standards. Stunting and wasting were defined as <-2 Z scores for length for age and weight for length, and overweight as >2 Z scores for weight for length. Prevalence was stratified by sex, maternal skin colour and family income. RESULTS: The prevalence of stunting declined by 53% (from 8.3% to 3.9%) from 1982 to 2015. Wasting prevalence remained stable at low levels (1.8% in 1982 and 1.7% in 2015), whereas overweight increased by 88% (6.5% to 12.2%). Undernutrition was more common among boys, those born to mothers with brown or black skin colour and in the poorest quintile of families. Socioeconomic inequalities in undernutrition decreased markedly over time. Overweight was markedly more common among the rich in 1982, but fast increase among the poor eliminated socioeconomic differences by 2015, when all groups showed similar prevalence. CONCLUSIONS: Our results confirm the rapid nutrition transition in Brazil, with marked reduction in levels and inequalities in undernutrition in parallel with a rapid increase in overweight, which became the main nutritional problem for children.


Assuntos
Antropometria , Transtornos do Crescimento/epidemiologia , Desnutrição/embriologia , Sobrepeso/epidemiologia , Síndrome de Emaciação/epidemiologia , Estatura , Brasil/epidemiologia , Feminino , Humanos , Renda , Lactente , Fenômenos Fisiológicos da Nutrição do Lactente , Masculino , Prevalência , Padrões de Referência , Organização Mundial da Saúde
3.
Int J Dev Neurosci ; 44: 1-5, 2015 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-25882586

RESUMO

It has been reported that prenatal undernutrition affects the development of the peripheral immune system. In this study, the effects of prenatal undernutrition on the febrile response and hypothalamic innate immune system were evaluated in male rats. Pregnant rats were divided into normally nourished (NN) and undernourished groups (UN). The febrile and anorectic responses to lipopolysaccharides (LPS) were evaluated in the offspring of NN and UN dams. The hypothalamic expression levels of pro-inflammatory cytokines, toll-like receptor 4 (TLR4), and neuropeptide Y (NPY) were also evaluated. The UN rats exhibited significantly lighter body weights than the NN rats at birth; however, their mean body weight was the same as that of the NN rats by postnatal day 10. In adulthood, the UN rats exhibited significantly stronger febrile responses than the NN rats, and the anorectic responses of the UN rats also tended to be stronger than those of the NN rats. On the other hand, no differences in hypothalamic interleukin (IL)-1ß, IL-6, tumor necrosis factor-α, TLR4, or NPY mRNA expression were detected between the NN and UN rats. These results suggest that prenatal undernutrition has long-lasting effects on the febrile response to LPS. However, the precise mechanism underlying these effects and their pathophysiological significance remain unclear.


Assuntos
Lipopolissacarídeos/toxicidade , Desnutrição/embriologia , Desnutrição/fisiopatologia , Efeitos Tardios da Exposição Pré-Natal/fisiopatologia , Convulsões Febris/induzido quimicamente , Análise de Variância , Animais , Temperatura Corporal/efeitos dos fármacos , Peso Corporal/efeitos dos fármacos , Citocinas/genética , Citocinas/metabolismo , Ingestão de Alimentos/efeitos dos fármacos , Feminino , Gliceraldeído-3-Fosfato Desidrogenases/genética , Gliceraldeído-3-Fosfato Desidrogenases/metabolismo , Hipotálamo/efeitos dos fármacos , Hipotálamo/metabolismo , Masculino , Desnutrição/metabolismo , Neuropeptídeo Y/genética , Neuropeptídeo Y/metabolismo , Fragmentos de Peptídeos/genética , Fragmentos de Peptídeos/metabolismo , Gravidez , RNA Mensageiro/metabolismo , Ratos , Ratos Sprague-Dawley , Convulsões Febris/patologia , Fatores de Tempo , Receptor 4 Toll-Like/genética , Receptor 4 Toll-Like/metabolismo
4.
Acta Physiol (Oxf) ; 210(1): 84-98, 2014 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-23452307

RESUMO

AIM: Exposure to adverse intra-uterine conditions can predispose for metabolic disorders later in life. By using a sheep model, we studied (i) how programming of glucose-insulin homoeostasis during late gestation is manifested later in life depending on the early post-natal dietary exposure and (ii) whether dietary alteration in obese individuals can prevent adverse outcomes of early life programming. METHODS: During late gestation, twin-pregnant sheep were fed 100% (NORM) or 50% (LOW) of energy and protein requirements. After birth, offspring were exposed to a moderate (CONV) or high-carbohydrate-high-fat (HCHF) diet until around puberty. Offspring remaining thereafter (exclusively females) were fed a moderate diet until young adulthood. RESULTS: LOW lambs had increased insulin secretory responses during intravenous glucose tolerance tests indicative of reduced insulin sensitivity. HCHF lambs were hypertriglyceridaemic, 75% had mild pancreatic collagen infiltration, and their acute insulin secretory response and insulin clearance during intravenous glucose and insulin tolerance tests, respectively, were reduced. However, NORM-HCHF in contrast to LOW-HCHF lambs had normal glucose tolerance, indicating that later health outcomes are highly influenced by pre-natal nutrition. Dietary alteration normalized glucose-insulin homoeostasis in adult HCHF females, whereas late-gestation undernutrition (LOW) permanently depressed insulin sensitivity. CONCLUSION: Maintenance of glucose tolerance in sheep exposed to pre-natal undernutrition relied on pancreatic hypersecretion of insulin to compensate for reduced insulin sensitivity. A mismatching high-fat diet in early post-natal life interfered with this pancreatic hypersecretion resulting in reduced glucose tolerance. Early post-natal, but not late pre-natal, impacts on glucose-insulin homoeostasis could be reversed by dietary correction later in life.


Assuntos
Envelhecimento/metabolismo , Glicemia/metabolismo , Insulina/sangue , Desnutrição/metabolismo , Efeitos Tardios da Exposição Pré-Natal/metabolismo , Animais , Medicina Baseada em Evidências , Feminino , Homeostase , Resistência à Insulina , Desnutrição/embriologia , Modelos Animais , Gravidez , Ovinos
5.
Biol Reprod ; 89(6): 144, 2013 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-24227755

RESUMO

Environmental conditions during perinatal development such as maternal undernutrition, maternal glucocorticoids, placental insufficiency, and maternal sodium overload can program changes in renal Na(+) excretion leading to hypertension. Experimental studies indicate that fetal exposure to an adverse maternal environment may reduce glomerular filtration rate by decreasing the surface area of the glomerular capillaries. Moreover, fetal responses to environmental insults during early life that contribute to the development of hypertension may include increased expression of tubular apical or basolateral membrane Na(+) transporters and increased production of renal superoxide leading to enhanced Na(+) reabsorption. This review will address the role of these potential renal mechanisms in the fetal programming of hypertension in experimental models induced by maternal undernutrition, fetal exposure to glucocorticoids, placental insufficiency, and maternal sodium overload in the rat.


Assuntos
Hipertensão/embriologia , Rim/fisiopatologia , Efeitos Tardios da Exposição Pré-Natal/fisiopatologia , Animais , Feminino , Taxa de Filtração Glomerular , Glucocorticoides/farmacologia , Humanos , Hipertensão/fisiopatologia , Rim/embriologia , Desnutrição/complicações , Desnutrição/embriologia , Desnutrição/fisiopatologia , Fenômenos Fisiológicos da Nutrição Materna , Insuficiência Placentária/fisiopatologia , Gravidez , Ratos , Sódio na Dieta
6.
Tohoku J Exp Med ; 224(3): 163-71, 2011 07.
Artigo em Inglês | MEDLINE | ID: mdl-21666350

RESUMO

Maternal undernutrition during pregnancy is a risk factor that impairs fetal growth and causes cardiovascular diseases. However, the underlying mechanism is still unknown. In this study, we evaluated the effect of maternal undernutrition on the expression levels of transcription factors in the fetal heart. Female mice were given low protein or regular food from 2 weeks before mating and during their pregnancy. The fetal hearts were collected on day 17.5 of gestation, about 1-2 days before birth. Maternal undernutrition resulted in a significant increase in the relative heart weight (heart weight/body weight) in female fetuses, but not in male fetuses. Microarray analysis revealed that expression levels of mRNAs for 133 transcription factors were changed in the fetal heart under maternal undernutrition. Among them, we focused on hypoxia-inducible factor 1 alpha (HIF1α) that is involved in the pathogenesis of cardiovascular diseases on adulthood. Quantitative real-time PCR analysis showed that the expression level of HIF1α mRNA was increased about 1.3-fold in male fetal heart under maternal undernutrition, but remained unchanged in female heart. Moreover, maternal undernutrition increased the mRNA level of prolyl hydroxylase 1 (PHD1), which contributes to degradation of HIF1α, in male heart but not in female heart. Immunohistochemical analysis showed the accumulation of HIF1α protein in the fetal heart of both sexes under maternal undernutrition, without the induction of HIF1α mRNA expression in female heart. These results suggest that maternal undernutrition may induce HIF1α expression in the fetal heart through the distinct mechanisms depending on the sex.


Assuntos
Coração Fetal/metabolismo , Subunidade alfa do Fator 1 Induzível por Hipóxia/genética , Desnutrição/embriologia , Desnutrição/genética , Animais , Regulação para Baixo/genética , Feminino , Coração Fetal/patologia , Regulação da Expressão Gênica no Desenvolvimento , Subunidade alfa do Fator 1 Induzível por Hipóxia/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Tamanho do Órgão/genética , Gravidez , Pró-Colágeno-Prolina Dioxigenase/genética , Pró-Colágeno-Prolina Dioxigenase/metabolismo , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Regulação para Cima/genética , Aumento de Peso/genética
7.
Reprod Sci ; 18(4): 398-405, 2011 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-21079239

RESUMO

Hepatic ß-adrenergic receptors (ß-ARs) play a pivotal role in mobilization of reserves via gluconeogenesis and glycogenolysis to supply the animal with its energy needs during decreased nutrient availability. Using a unique nutrient-deprived baboon model, we have demonstrated for the first time that immunoreactive hepatic ß(1)- and ß(2)-AR subtypes are regionally distributed and localized on cells around the central lobular vein in 0.5 and 0.9 gestation (G) fetuses of ad libitum fed control (CTR) and maternal nutrient restricted (MNR) mothers. Furthermore, MNR decreased fetal liver immunoreactive ß(1)-AR and increased immunoreactive ß(2)-AR at 0.5G. However, at 0.9G, immunohistochemistry and Western blot analysis revealed a decrease in ß(1)-AR and no change in ß(2)-AR levels. Thus, MNR in a nonhuman primate species has effects on hepatic ß(1)- and ß(2)-ARs that are receptor- and gestation stage-specific and may represent compensatory systems whose effects would increase glucose availability in the presence of nutrient deprivation.


Assuntos
Fenômenos Fisiológicos da Nutrição Animal , Fígado/metabolismo , Desnutrição/metabolismo , Fenômenos Fisiológicos da Nutrição Materna , Receptores Adrenérgicos beta 1/metabolismo , Receptores Adrenérgicos beta 2/metabolismo , Animais , Western Blotting , Feminino , Idade Gestacional , Imuno-Histoquímica , Fígado/embriologia , Fígado/patologia , Desnutrição/embriologia , Desnutrição/genética , Desnutrição/patologia , Papio , Gravidez , RNA Mensageiro/metabolismo , Receptores Adrenérgicos beta 1/genética , Receptores Adrenérgicos beta 2/genética , Reação em Cadeia da Polimerase Via Transcriptase Reversa
8.
Reproduction ; 141(1): 119-26, 2011 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-21045167

RESUMO

Maternal nutrition during the period of early organ development can modulate the offspring's ability to metabolise excess fat as young adults when exposed to an obesogenic environment. This study examined the hypothesis that exposing offspring to nutrient restriction coincident with early hepatogenesis would result in endocrine and metabolic adaptations that subsequently lead to increased ectopic lipid accumulation within the liver. Pregnant sheep were fed either 50 or 100% of total metabolisable energy requirements from 30 to 80 days gestation and 100% thereafter. At weaning, offspring were made obese, and at ~1 year of age livers were sampled. Lipid infiltration and molecular indices of gluconeogenesis, lipid metabolism and mitochondrial function were measured. Although hepatic triglyceride accumulation was not affected by obesity per se, it was nearly doubled in obese offspring born to nutrient-restricted mothers. This adaptation was accompanied by elevated gene expression for peroxisome proliferator-activated receptor γ (PPARG) and its co-activator PGC1α, which may be indicative of changes in the rate of hepatic fatty acid oxidation. In contrast, maternal diet had no influence on the stimulatory effect of obesity on gene expression for a range of proteins involved in glucose metabolism and energy balance including glucokinase, glucocorticoid receptors and uncoupling protein 2. Similarly, although gene expressions for the insulin and IGF1 receptors were suppressed by obesity they were not influenced by the prenatal nutritional environment. In conclusion, excess hepatic lipid accumulation with juvenile obesity is promoted by suboptimal nutrition coincident with early development of the fetal liver.


Assuntos
Fenômenos Fisiológicos da Nutrição Animal , Fígado Gorduroso/metabolismo , Fígado/metabolismo , Desnutrição/metabolismo , Fenômenos Fisiológicos da Nutrição Materna , Obesidade/metabolismo , Efeitos Tardios da Exposição Pré-Natal , Fatores Etários , Animais , Modelos Animais de Doenças , Fígado Gorduroso/embriologia , Fígado Gorduroso/genética , Fígado Gorduroso/patologia , Fígado Gorduroso/fisiopatologia , Feminino , Regulação da Expressão Gênica no Desenvolvimento , Idade Gestacional , Gluconeogênese/genética , Metabolismo dos Lipídeos/genética , Fígado/embriologia , Fígado/patologia , Fígado/fisiopatologia , Desnutrição/embriologia , Desnutrição/genética , Desnutrição/fisiopatologia , Mitocôndrias Hepáticas/metabolismo , Obesidade/embriologia , Obesidade/genética , Obesidade/patologia , Obesidade/fisiopatologia , PPAR gama/genética , Gravidez , Ovinos , Triglicerídeos/metabolismo
9.
Theriogenology ; 74(9): 1661-9, 2010 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-20708787

RESUMO

Expression of aquaporin water channels (AQPs) in the male excurrent ducts, is of major importance for local water movements. To study the influence of pre- and postnatal undernutrition on AQP-expression in the adult male genital tract, 4 pregnant female rats were fed ad libitum (control group) and 4 with 33.5% of gestational feed requirements (underfed group). Feeding restriction of underfed group pups continued up to weaning (25 days of age), then all pups were fed ad libitum until slaughtered at 100 days of age. Epididymides were sampled and processed for aquaporin immunohistochemistry. Expression of AQP1 was similar either in the control and underfed groups of rats, strongly evidenced at the apical and lateral plasma membrane of the efferent ducts non-ciliated cells, in the smooth muscle cells surrounding epididymal duct and in blood vessel endothelium throughout the epididymis. AQP2-immunoreactivity was present in the corpus and cauda regions, strongly expressed in the principal cells of both groups of rats. In contrast, AQP9 expression was modified by early life undernourishment, as it was weakly evidenced at the microvilli in the principal cells and strongly diminished or completely lacked in the clear cells of the cauda, in underfed group epididymides. Since it is known that clear cells are involved in luminal fluid acidification, this function might be altered in adult animals, which were underfed during early life.


Assuntos
Aquaporina 1/metabolismo , Aquaporina 2/metabolismo , Aquaporinas/metabolismo , Epididimo/metabolismo , Desnutrição/metabolismo , Animais , Feminino , Imuno-Histoquímica , Rim/metabolismo , Masculino , Desnutrição/embriologia , Gravidez , Ratos , Ratos Sprague-Dawley , Maturidade Sexual , Fatores de Tempo
10.
Recenti Prog Med ; 101(2): 57-60, 2010 Feb.
Artigo em Italiano | MEDLINE | ID: mdl-20433001

RESUMO

In the last century, life expectancy in the economically developed countries has significantly increased. In parallel there has been an increase in the prevalence of obesity and of chronic kidney disease. Abdominal obesity is a relevant risk factor for death and cardiovascular complications in chronic kidney disease and in dialysis patients as well.


Assuntos
Falência Renal Crônica/etiologia , Obesidade/complicações , Índice de Massa Corporal , Doenças Cardiovasculares/mortalidade , Feminino , Humanos , Gordura Intra-Abdominal/fisiopatologia , Falência Renal Crônica/epidemiologia , Desnutrição/complicações , Desnutrição/embriologia , Síndrome Metabólica/epidemiologia , Modelos Biológicos , Obesidade/embriologia , Obesidade/epidemiologia , Gravidez , Efeitos Tardios da Exposição Pré-Natal , Diálise Renal , Fatores de Risco , Relação Cintura-Quadril
11.
J Physiol ; 588(Pt 5): 821-9, 2010 Mar 01.
Artigo em Inglês | MEDLINE | ID: mdl-20083512

RESUMO

Recent studies have suggested that intrauterine undernutrition is closely associated with the pathogenesis of diseases after birth. Perinatal undernutrition is known to disturb the development of reproductive function and delay the onset of puberty in some species. Using a rat model, we determined the effects of prenatal undernutrition on the development of the hypothalamic kisspeptin system and evaluated whether the alteration of the kisspeptin system contributes to the delayed onset of puberty induced by prenatal undernutrition. We also evaluated the effects of prenatal undernutrition on the developmental changes in serum leptin levels because leptin was a putative positive regulator of the hypothalamic kisspeptin system. We compared the timing of vaginal opening (VO) and the developmental changes in body weight, hypothalamic Kiss1 mRNA levels, and serum leptin concentrations between offspring with prenatal undernutrition (UN offspring) and normal nutrition (NN offspring). After birth, the UN offspring showed rapid growth and had caught up to body weight of the NN offspring by postnatal day 12. After postnatal day 16, the UN offspring showed significantly lower Kiss1 mRNA levels than the NN offspring, despite their significantly higher serum leptin levels (at days 20 and 28). The timing of VO in the UN offspring was delayed compared with that in the NN offspring, and chronic central injection of kisspeptin normalized the timing of VO in the UN offspring. These results suggest that decreased hypothalamic kisspeptin action contributes to the delayed onset of puberty in prenatally undernourished female rats. Increased leptin resistance in the kisspeptin system might be involved in these alterations.


Assuntos
Hipotálamo/embriologia , Hipotálamo/metabolismo , Desnutrição/embriologia , Desnutrição/metabolismo , Proteínas/metabolismo , Animais , Feminino , Hipotálamo/crescimento & desenvolvimento , Kisspeptinas , Ratos , Ratos Sprague-Dawley
12.
Rev Invest Clin ; 61(1): 41-52, 2009.
Artigo em Espanhol | MEDLINE | ID: mdl-19507474

RESUMO

Human epidemiological and experimental animal studies have shown that suboptimal environments in the womb and during early neonatal life alter growth and may program offspring susceptibility to lifelong health problems. One of the most interesting and significant feature of developmental programming is the evidence that adverse consequences of altered intrauterine environments can be passed from first generation to second generation offspring. To obtain the transgenerational phenotype, a negative environment is required during fetal or early neonatal life, the physiologic phenotype or disease can be transmitted through the germ line and the subsequent generations are not directly exposed to the environmental factor. The hypothesis has become well accepted by compelling animal studies that define the outcome of specific challenges such as: 1) nutrient restriction or overfeeding during pregnancy and lactation; 2) uterine blood flow restriction; 3) fetal exposure to inappropriately high levels of glucocorticoids, and 4) experimental maternal diabetes. Maternal protein restriction in the rat adversely affects glucose metabolism of male and female second generation offspring in a gender and developmental time window-specific manner. Other studies have proved transgenerational passage of effects resulting from treatment of pregnant rats with dexamethasone by either maternal or paternal lines. First generation female diabetic offspring of F0 rats treated with streptozotocin during pregnancy had F2 offspring with altered glucose and carbohydrate metabolism. The studies suggest that the mechanisms involved in developmental programming are likely epigenetic rather than due to DNA sequence mutations. Many individuals all over the world experience undernutrition, stress, hyperglycemia and other negative environmental factors during pregnancy and/or lactation. Insult during this critical period of development may induce malprogramming and adversely alter not only the F1 generation but also future generations. Preventing or treating these conditions will help to minimize the risk of transmission of metabolic diseases to future generations.


Assuntos
Doenças Metabólicas/etiologia , Efeitos Tardios da Exposição Pré-Natal , Animais , Peso ao Nascer , Diabetes Mellitus Experimental/embriologia , Diabetes Gestacional/metabolismo , Dieta , Suscetibilidade a Doenças , Epigênese Genética , Feminino , Retardo do Crescimento Fetal/etiologia , Macrossomia Fetal/etiologia , Glucocorticoides/efeitos adversos , Humanos , Hiperinsulinismo/embriologia , Hiperinsulinismo/metabolismo , Recém-Nascido , Lactação , Masculino , Desnutrição/complicações , Desnutrição/embriologia , Doenças Metabólicas/embriologia , Doenças Metabólicas/genética , Síndrome Metabólica/embriologia , Síndrome Metabólica/genética , Síndrome Metabólica/metabolismo , Modelos Biológicos , Oxazóis/toxicidade , Gravidez , Complicações na Gravidez/metabolismo , Ratos , Ratos Wistar
13.
Nestle Nutr Workshop Ser Pediatr Program ; 63: 59-73; discussion 74-7, 259-68, 2009.
Artigo em Inglês | MEDLINE | ID: mdl-19346768

RESUMO

Undernutrition continues to be high in many regions of the developing world. Birthweight, a common proxy measure of intrauterine growth, is influenced by nutritional, environmental and lifestyle factors during pregnancy and, in turn, affects immediate survival and function, and is a determinant of later life risk of chronic diseases. Maternal pre-pregnancy weight and height are independently associated with birthweight and also modify the effects of pregnancy weight gain and interventions during pregnancy on birthweight and perinatal mortality. Other prenatal factors commonly known to impact birthweight include maternal age, parity, sex, and birth interval, whereas lifestyle factors such as physical activity and maternal stress, as well as environmental toxicants have variable influences. Tobacco and other substance use and infections, specifically ascending reproductive tract infections, malaria, and HIV, can cause intrauterine growth restriction (IUGR). Few studies have examined the contribution of prenatal factors including low birthweight to childhood wasting and stunting. Studies that have examined this, with adequate adjustment for confounders, have generally found odds ratios associated with low birthweight ranging between 2 and 5. Even fewer studies have examined birth length or maternal nutritional status as risk factors. More research is needed to determine the proportion of childhood under-nutrition attributable to IUGR so that interventions can be targeted to the appropriate life stages.


Assuntos
Doenças Fetais/epidemiologia , Desnutrição/embriologia , Desnutrição/epidemiologia , África/epidemiologia , Ásia/epidemiologia , Peso ao Nascer , Estatura , Peso Corporal , Criança , Pré-Escolar , Países em Desenvolvimento/estatística & dados numéricos , Feminino , Transtornos do Crescimento/epidemiologia , Humanos , Lactente , América Latina/epidemiologia , Estilo de Vida , Desnutrição/etiologia , Gravidez , Prevalência , Fatores de Risco
14.
Diabetes ; 58(6): 1440-4, 2009 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-19258437

RESUMO

OBJECTIVE: Fetal malnutrition may predispose to type 2 diabetes through gene programming and developmental changes. Previous studies showed that these effects may be modulated by genetic variation. Genome-wide association studies discovered and replicated a number of type 2 diabetes-associated genes. We investigated the effects of such well-studied polymorphisms and their interactions with fetal malnutrition on type 2 diabetes risk and related phenotypes in the Dutch Famine Birth Cohort. RESEARCH DESIGN AND METHODS: The rs7754840 (CDKAL1), rs10811661 (CDKN2AB), rs1111875 (HHEX), rs4402960 (IGF2BP2), rs5219 (KCNJ11), rs13266634 (SLC30A8), and rs7903146 (TCF7L2) polymorphisms were genotyped in 772 participants of the Dutch Famine Birth Cohort Study (n = 328 exposed, n = 444 unexposed). Logistic and linear regression models served to analyze their interactions with prenatal exposure to famine on type 2 diabetes, impaired glucose tolerance (IGT), and area under the curves (AUCs) for glucose and insulin during oral glucose tolerance testing (OGTT). RESULTS: In the total population, the TCF7L2 and IGF2BP2 variants most strongly associated with increased risk for type 2 diabetes/IGT and increased AUC for glucose, while the CDKAL1 polymorphism associated with decreased AUC for insulin. The IGF2BP2 polymorphism showed an interaction with prenatal exposure to famine on AUC for glucose (beta = -9.2 [95% CI -16.2 to -2.1], P = 0.009). CONCLUSIONS: The IGF2BP2 variant showed a nominal interaction with exposure to famine in utero, decreasing OGTT AUCs for glucose. This may provide a clue that modulation of the consequences of fetal environment depends on an individual's genetic background.


Assuntos
Glicemia/metabolismo , Diabetes Mellitus Tipo 2/genética , Doenças Fetais/genética , Variação Genética , Desnutrição/genética , Proteínas de Ligação a RNA/genética , Inanição/genética , Diabetes Mellitus Tipo 2/sangue , Diabetes Mellitus Tipo 2/embriologia , Feminino , Perfilação da Expressão Gênica , Teste de Tolerância a Glucose , Humanos , Insulina/metabolismo , Secreção de Insulina , Desnutrição/embriologia , Pessoa de Meia-Idade , Países Baixos , Fenótipo , Gravidez , Inanição/embriologia , Guerra
15.
Br J Nutr ; 101(6): 902-8, 2009 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-18761782

RESUMO

Maternal nutrition during pregnancy has a significant influence in establishing patterns of metabolism and postnatal behaviours in offspring, and therefore shapes their risk of developing disorders in later life. Although it is well established that a mismatch between food consumption and energy expenditure leads to obesity and metabolic dysregulation, little research has investigated the biological origin of such behaviour. We conducted the present experiments to investigate effects of early-life nutrition on preference between wheel running and lever pressing for food during adult life. To address this issue we employed a well-established experimental approach in the rat which has shown that offspring of mothers undernourished during pregnancy develop obesity and metabolic disorders when kept under standard laboratory conditions. Using this experimental approach, two studies were conducted where offspring of ad libitum-fed dams and dams undernourished throughout pregnancy were given the choice between wheel running and pressing a response lever for food. Across subsequent conditions, the rate at which the response lever provided food was varied from 0.22 to 6.0 (study 1) and 0.19 to 3.0 (study 2) pellets per min. Compared with the control group, offspring from dams undernourished during pregnancy showed a consistently greater preference for running over lever pressing for food throughout both experiments of the study. The results of the present study provide experimental evidence that a mother's nutrition during pregnancy can result in a long-term shift in her offspring's lifestyle choices that are relevant to obesity prevention. Such a shift, if endorsed, will have substantial and wide-ranging health consequences throughout the lifespan.


Assuntos
Comportamento de Escolha , Ingestão de Alimentos , Desnutrição/embriologia , Desnutrição/psicologia , Efeitos Tardios da Exposição Pré-Natal , Corrida , Animais , Condicionamento Operante , Metabolismo Energético , Feminino , Modelos Animais , Gravidez , Distribuição Aleatória , Ratos , Ratos Wistar
16.
BJOG ; 115(2): 261-8, 2008 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-18081604

RESUMO

OBJECTIVE: To determine the role of carotid sinus innervation on differential fetal organ growth during maternal nutrient restriction in late pregnancy. DESIGN: Randomised controlled study. SETTING: University research facility. SAMPLE: Thirty-nine Merino ewes. METHODS: At 113 days gestational age (dGA), fetuses were bilaterally carotid sinus denervated or sham denervated. From 118 dGA, the surgery groups were subdivided into two dietary groups, and their ewes were fed 100% of nutrient requirements or 50% until tissue collection at 140 dGA. This provided four groups (sham/control diet, sham/restricted diet, denervated/control diet and denervated/restricted diet). MAIN OUTCOME MEASURES: Fetal organ weights and hormone levels and maternal weight change during the dietary restriction. RESULTS: Adrenal glands were larger in sham/restricted diet fetuses than in sham/control diet or denervated/restricted diet fetuses (P < 0.05). Fetal adrenal weight and brain-to-liver weight ratio were positively related to maternal weight change during the nutritional challenge in sham fetuses only (P < 0.05). Fetal liver weight was negatively related to maternal weight change during nutritional challenge in sham fetuses only (P < 0.05). CONCLUSIONS: We have shown a reduction in liver growth but sparing of adrenal growth in response to moderate maternal undernutrition, which is dependent on intact carotid body innervation. This suggests a new role for the carotid bodies in the control of differential organ growth during such undernutrition.


Assuntos
Seio Carotídeo/inervação , Dieta Redutora/efeitos adversos , Desenvolvimento Fetal/fisiologia , Desnutrição/embriologia , Complicações na Gravidez/etiologia , Glândulas Suprarrenais/embriologia , Hormônio Adrenocorticotrópico/metabolismo , Animais , Peso Corporal/fisiologia , Encéfalo/embriologia , Seio Carotídeo/embriologia , Seio Carotídeo/cirurgia , Eletrólitos/sangue , Feminino , Sangue Fetal/química , Hidrocortisona/metabolismo , Insulina/metabolismo , Fígado/embriologia , Pulmão/embriologia , Tamanho do Órgão/fisiologia , Gravidez , Distribuição Aleatória , Ovinos
17.
Schizophr Res ; 99(1-3): 48-55, 2008 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-18065207

RESUMO

Exposure to prenatal undernutrition or malnutrition increases the risk of schizophrenia, although little is known about the mechanism. Pro-inflammatory factors are critical in brain development, and are believed to play an important role in neurodevelopmental disorders associated with prenatal exposure to infection, including schizophrenia. However it is not known whether pro-inflammatory factors also mediate the effects on the fetus of prenatal malnutrition or undernutrition. In this study, we established a new prenatal undernourished rat model induced by maternal exposure to a diet restricted to 50% of the low (6%) protein diet (RLP50). We observed the disappearance of maternal nest-building behavior in the RLP50 dams, increased levels of TNFA and IL6 in the placentas (P<0.001; P=0.879, respectively) and fetal livers (P<0.001; P<0.05, respectively), and a decrease in the fetal brains (P<0.05; P<0.01, respectively). Our results are similar to previous studies of maternal infection, which implies that a common pathway mediated by pro-inflammatory factors may contribute to the brain development, consequently increasing the risk of schizophrenia and other psychiatric diseases programmed by varied maternal adversities. We also provide a new prenatal undernourished model for researching prenatal problems, which differs from previous malnourished model in terms of the maternal behavior of dams and of observed pro-inflammatory factor levels in fetal tissues.


Assuntos
Citocinas/sangue , Modelos Animais de Doenças , Interleucina-6/sangue , Desnutrição/imunologia , Efeitos Tardios da Exposição Pré-Natal , Desnutrição Proteico-Calórica/imunologia , Esquizofrenia/imunologia , Fator de Necrose Tumoral alfa/sangue , Animais , Encéfalo/imunologia , Ensaio de Imunoadsorção Enzimática , Feminino , Humanos , Fígado/embriologia , Fígado/imunologia , Desnutrição/embriologia , Comportamento Materno/fisiologia , Comportamento de Nidação/fisiologia , Placenta/embriologia , Placenta/imunologia , Gravidez , Desnutrição Proteico-Calórica/embriologia , Ratos , Ratos Sprague-Dawley
18.
Diabetologia ; 50(5): 1099-108, 2007 May.
Artigo em Inglês | MEDLINE | ID: mdl-17370059

RESUMO

AIMS/HYPOTHESIS: Observational studies in humans suggest that low birthweight may decrease the risk of type 1 diabetes, but the mechanism is unknown. We hypothesised that antenatal undernutrition would decrease the incidence of type 1 diabetes in non-obese diabetic (NOD) mice. MATERIALS AND METHODS: A 40% restriction of energy intake was applied to pregnant NOD dams from day 12.5 to day 18.5 of gestation, resulting in intrauterine growth retardation of offspring. All mice were fed a standard diet after weaning. Control and undernourished female offspring were followed to assess diabetes incidence. Male NOD mice were treated with cyclophosphamide to accelerate development of diabetes. Glucose homeostasis, body composition and pancreatic histology were compared in control and undernourished offspring. RESULTS: Mean birthweight was lower in undernourished than in control mice (p = 0.00003). At 24 weeks of age, the cumulative incidence of spontaneous diabetes in female mice was 73% in control and 48% in undernourished mice (p = 0.003). In cyclophosphamide-treated male mice, antenatal undernutrition also tended to reduce the development of diabetes (p = 0.058). Maternal leptin levels were lower in undernourished dams on day 18.5 of pregnancy (p = 0.039), while postnatal leptin levels were significantly higher in undernourished offspring at 4, 20 and 27 weeks of life (p < 0.05). Beta cell mass was similar in both groups (control = 0.4 mg; undernourished = 0.54 mg; p = 0.24). Histological evidence of apoptosis at 20 weeks was greater in control than in undernourished mice (control = 6.3 +/- 1.4%; undernourished = 4.2 +/- 0.3%, p = 0.05). CONCLUSIONS/INTERPRETATION: Antenatal undernutrition reduces the incidence of diabetes in NOD mice, perhaps via alterations in apoptosis.


Assuntos
Diabetes Mellitus Tipo 1/prevenção & controle , Desnutrição/embriologia , Absorciometria de Fóton , Animais , Diabetes Mellitus Tipo 1/epidemiologia , Feminino , Retardo do Crescimento Fetal/fisiopatologia , Marcação In Situ das Extremidades Cortadas , Incidência , Camundongos , Camundongos Endogâmicos NOD , Gravidez
19.
Am J Physiol Endocrinol Metab ; 292(1): E187-95, 2007 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-16912057

RESUMO

We have previously shown that fetuses from undernourished (U) pregnant rats exhibited an increased beta-cell mass probably related to an enhanced IGF-I replicative response. Because IGF-I signaling pathways have been implicated in regulating beta-cell growth, we investigated in this study the IGF-I transduction system in U fetuses. To this end, an in vitro model of primary fetal islets was developed to characterize glucose/IGF-I-mediated signaling that specially influences beta-cell proliferation. We found that U fetal islets showed a greater replicative response to glucose and IGF-I than controls. Furthermore, insulin receptor substrate (IRS)-2 protein and its association with p85 were also increased. In the complete absence of IGF-I or stimulatory glucose, U islets presented an increased basal phosphorylation of downstream signals of the phosphatidylinositol 3-kinase (PI3K) pathway such as PKB, glycogen synthase kinase (GSK)3alpha/beta, PKCzeta, and mammalian target of rapamycin (mTOR). Similarly, phosphorylation of these proteins (except GSK3alpha/beta) by glucose and IGF-I was augmented even though total protein content remained unchanged. Downstream of PKB, direct glucose activation of mTOR was increased as well. In contrast, ERK1/2 phosphorylation was unaffected by undernutrition, but ERK activation seemed to be required to induce a higher proliferative response in U islets. In conclusion, we have demonstrated that fetal U islets show increased IRS-2 content and an enhancement in both basal and glucose/IGF-I activations of the IRS-2/PI3K/PKB pathway. These molecular changes may be responsible for the greater glucose/IGF-I islet replication and contribute to the increased beta-cell mass found in these fetuses.


Assuntos
Feto/citologia , Fator de Crescimento Insulin-Like I/metabolismo , Células Secretoras de Insulina/citologia , Peptídeos e Proteínas de Sinalização Intracelular/metabolismo , Desnutrição/metabolismo , Fosfoproteínas/metabolismo , Prenhez , Animais , Proliferação de Células/efeitos dos fármacos , Células Cultivadas , Feminino , Glucose/farmacologia , Proteínas Substratos do Receptor de Insulina , Fator de Crescimento Insulin-Like I/farmacologia , Células Secretoras de Insulina/metabolismo , Masculino , Desnutrição/embriologia , Fosforilação , Gravidez , Proteínas Tirosina Quinases/metabolismo , Ratos , Ratos Wistar , Receptor IGF Tipo 1/metabolismo , Transdução de Sinais
20.
J Nutr Biochem ; 16(4): 195-204, 2005 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-15808323

RESUMO

Epidemiological studies have revealed strong and reproducible links between indices of poor fetal growth and susceptibility to the development of glucose intolerance and insulin resistance syndrome in adult life. To explain these associations, the thrifty phenotype hypothesis has been proposed. Mitochondrial DNA abnormalities have been known to cause insulin deficiency, insulin resistance and diabetes mellitus. In this review, we propose that mitochondrial dysfunction is a link between malnutrition during early life and disease in adult life. The potential mechanism for mitochondrial dysfunction will be focused on availability of the taurine and nucleotides, and imprinting on the genes.


Assuntos
DNA Mitocondrial/genética , Diabetes Mellitus Tipo 2/genética , Desnutrição/embriologia , Sequência de Bases , Feminino , Humanos , Resistência à Insulina/genética , Dados de Sequência Molecular , Conformação de Ácido Nucleico , Gravidez , Efeitos Tardios da Exposição Pré-Natal , RNA de Transferência de Lisina/genética
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...