Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 25
Filtrar
Mais filtros

Bases de dados
País/Região como assunto
Tipo de documento
Intervalo de ano de publicação
1.
Nature ; 625(7996): 760-767, 2024 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-38092039

RESUMO

GDF15, a hormone acting on the brainstem, has been implicated in the nausea and vomiting of pregnancy, including its most severe form, hyperemesis gravidarum (HG), but a full mechanistic understanding is lacking1-4. Here we report that fetal production of GDF15 and maternal sensitivity to it both contribute substantially to the risk of HG. We confirmed that higher GDF15 levels in maternal blood are associated with vomiting in pregnancy and HG. Using mass spectrometry to detect a naturally labelled GDF15 variant, we demonstrate that the vast majority of GDF15 in the maternal plasma is derived from the feto-placental unit. By studying carriers of rare and common genetic variants, we found that low levels of GDF15 in the non-pregnant state increase the risk of developing HG. Conversely, women with ß-thalassaemia, a condition in which GDF15 levels are chronically high5, report very low levels of nausea and vomiting of pregnancy. In mice, the acute food intake response to a bolus of GDF15 is influenced bi-directionally by prior levels of circulating GDF15 in a manner suggesting that this system is susceptible to desensitization. Our findings support a putative causal role for fetally derived GDF15 in the nausea and vomiting of human pregnancy, with maternal sensitivity, at least partly determined by prepregnancy exposure to the hormone, being a major influence on its severity. They also suggest mechanism-based approaches to the treatment and prevention of HG.


Assuntos
Fator 15 de Diferenciação de Crescimento , Hiperêmese Gravídica , Náusea , Vômito , Animais , Feminino , Humanos , Camundongos , Gravidez , Talassemia beta/sangue , Talassemia beta/metabolismo , Feto/metabolismo , Fator 15 de Diferenciação de Crescimento/sangue , Fator 15 de Diferenciação de Crescimento/metabolismo , Hormônios/sangue , Hormônios/metabolismo , Hiperêmese Gravídica/complicações , Hiperêmese Gravídica/metabolismo , Hiperêmese Gravídica/prevenção & controle , Hiperêmese Gravídica/terapia , Náusea/sangue , Náusea/complicações , Náusea/metabolismo , Placenta/metabolismo , Vômito/sangue , Vômito/complicações , Vômito/metabolismo
2.
Hum Reprod ; 35(4): 913-928, 2020 04 28.
Artigo em Inglês | MEDLINE | ID: mdl-32325494

RESUMO

STUDY QUESTION: Are maternal serum phthalate metabolite, phenol and paraben concentrations measured at 10-17 weeks of gestation associated with male infant genital developmental outcomes, specifically cryptorchidism, anogenital distance (AGD), penile length and testicular descent distance, at birth and postnatally? SUMMARY ANSWER: Maternal serum bisphenol A (BPA) concentration at 10-17 weeks of gestation was positively associated with congenital or postnatally acquired cryptorchidism, and n-propyl paraben (n-PrP) concentration was associated with shorter AGD from birth to 24 months of age. WHAT IS KNOWN ALREADY: Male reproductive disorders are increasing in prevalence, which may reflect environmental influences on foetal testicular development. Animal studies have implicated phthalates, BPA and parabens, to which humans are ubiquitously exposed. However, epidemiological studies have generated conflicting results and have often been limited by small sample size and/or measurement of chemical exposures outside the most relevant developmental window. STUDY DESIGN, SIZE, DURATION: Case-control study of cryptorchidism nested within a prospective cohort study (Cambridge Baby Growth Study), with recruitment of pregnant women at 10-17 postmenstrual weeks of gestation from a single UK maternity unit between 2001 and 2009 and 24 months of infant follow-up. Of 2229 recruited women, 1640 continued with the infancy study after delivery, of whom 330 mothers of 334 male infants (30 with congenital cryptorchidism, 25 with postnatally acquired cryptorchidism and 279 unmatched controls) were included in the present analysis. PARTICIPANTS/MATERIALS, SETTING, METHODS: Maternal blood was collected at enrolment, and serum levels of 16 phthalate metabolites, 9 phenols (including BPA) and 6 parabens were measured using liquid chromatography/tandem mass spectrometry. Logistic regression was used to model the association of cryptorchidism with serum chemical concentrations, adjusting for putative confounders. Additionally, offspring AGD, penile length and testicular descent distance were assessed at 0, 3, 12, 18 and 24 months of age, and age-specific Z scores were calculated. Associations between serum chemical levels and these outcomes were tested using linear mixed models. MAIN RESULTS AND THE ROLE OF CHANCE: Maternal serum BPA concentration was associated with offspring all-type cryptorchidism both when considered as a continuous exposure (adjusted odds ratio per log10 µg/l: 2.90, 95% CI 1.31-6.43, P = 0.009) and as quartiles (phet = 0.002). Detection of n-PrP in maternal serum was associated with shorter AGD (by 0.242 standard deviations, 95% CI 0.051-0.433, P = 0.01) from birth to 24 months of age; this reduction was independent of body size and other putative confounders. We did not find any consistent associations with offspring outcomes for the other phenols, parabens, and phthalate metabolites measured. LIMITATIONS, REASONS FOR CAUTION: We cannot discount confounding by other demographic factors or endocrine-disrupting chemicals. There may have been misclassification of chemical exposure due to use of single serum measurements. The cohort was not fully representative of pregnant women in the UK, particularly in terms of smoking prevalence and maternal ethnicity. WIDER IMPLICATIONS OF THE FINDINGS: Our observational findings support experimental evidence that intrauterine exposure to BPA and n-PrP during early gestation may adversely affect male reproductive development. More evidence is required before specific public health recommendations can be made. STUDY FUNDING/COMPETING INTEREST(S): This work was supported by a European Union Framework V programme, the World Cancer Research Fund International, the Medical Research Council (UK), Newlife the Charity for Disabled Children, the Mothercare Group Foundation, Mead Johnson Nutrition and the National Institute for Health Research Cambridge Comprehensive Biomedical Research Centre. Visiting Fellowship (J.M.): Regional Programme 'Jiménez de la Espada' for Research Mobility, Cooperation and Internationalization, Seneca Foundation-Science and Technology Agency for the Region of Murcia (No. 20136/EE/17). K.O. is supported by the Medical Research Council (UK) (Unit Programme number: MC_UU_12015/2). The authors declare no conflict of interest.


Assuntos
Parabenos , Fenóis , Compostos Benzidrílicos , Estudos de Casos e Controles , Criança , Feminino , Humanos , Lactente , Masculino , Fenóis/toxicidade , Gravidez , Estudos Prospectivos
3.
Ann Hum Biol ; 47(2): 142-149, 2020 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-32429763

RESUMO

Background: Highly consistent positive associations are reported between infancy growth and later obesity risk. However, it is unclear whether infancy growth parameters beyond body weight add to the prediction of later obesity risk.Aim: To assess whether infancy length and skinfold thicknesses add to infancy weight in the prediction of childhood adiposity.Subjects and methods: This analysis included 254 children with available data on infant growth from birth to 24 months and childhood adiposity at age 6-11 years measured by DXA. Multilevel linear regression was used to examine the predictors of childhood percent body fat (%BF), with adjustment for sex and age at follow-up visit.Results: Birth weight and weight gain (modelled as changes in z-score) between 0-3 months and 3-24 months showed independent positive relationships with childhood %BF. The addition of gains in infant length and skinfolds between 0-3 months, but not 3-24 months, improved overall model prediction, from 18.7% to 20.7% of the variance in childhood %BF (likelihood ratio test, p < 0.0001), although their independent effect estimates were small (infant length gain: negative trend, partial R-square 0.6%, p = 0.2; skinfolds: positive trend, 1.3%, p = 0.09).Conclusion: Infancy length and skinfolds contribute significantly, but only modestly, to the prediction of childhood adiposity.


Assuntos
Adiposidade , Desenvolvimento Infantil , Obesidade Infantil/etiologia , Aumento de Peso , Peso ao Nascer , Criança , Pré-Escolar , Feminino , Humanos , Lactente , Recém-Nascido , Masculino
4.
Diabetes Metab ; 43(4): 323-331, 2017 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-28392167

RESUMO

AIM: We hypothesised that some of the genetic risk for gestational diabetes (GDM) is due to the fetal genome affecting maternal glucose concentrations. Previously, we found associations between fetal IGF2 gene variants and maternal glucose concentrations in late pregnancy. METHODS: In the present study, we tested associations between SNP alleles from 15 fetal imprinted genes and maternal glucose concentrations in late pregnancy in the Cambridge Baby Growth and Wellbeing cohorts (1160 DNA trios). RESULTS: Four fetal SNP alleles with the strongest univariate associations: paternally-transmitted IGF2 rs10770125 (P-value=2×10-4) and INS rs2585 (P-value=7×10-4), and maternally-transmitted KCNQ1(OT1) rs231841 (P-value=1×10-3) and KCNQ1(OT1) rs7929804 (P-value=4×10-3), were used to construct a composite fetal imprinted gene allele score which was associated with maternal glucose concentrations (P-value=4.3×10-6, n=981, r2=2.0%) and GDM prevalence (odds ratio per allele 1.44 (1.15, 1.80), P-value=1×10-3, n=89 cases and 899 controls). Meta-analysis of the associations including data from 1367 Hyperglycaemia and Adverse Pregnancy Outcome Study participants confirmed the paternally-transmitted fetal IGF2/INS SNP associations (rs10770125, P-value=3.2×10-8, rs2585, P-value=3.6×10-5) and the composite fetal imprinted gene allele score association (P-value=1.3×10-8), but not the maternally-transmitted fetal KCNQ1(OT1) associations (rs231841, P-value=0.4; rs7929804, P-value=0.2). CONCLUSION: This study suggests that polymorphic variation in fetal imprinted genes, particularly in the IGF2/INS region, contribute a small but significant part to the risk of raised late pregnancy maternal glucose concentrations.


Assuntos
Alelos , Glicemia/genética , Diabetes Gestacional/genética , Impressão Genômica , Polimorfismo de Nucleotídeo Único , Adulto , Diabetes Gestacional/sangue , Feminino , Humanos , Insulina/genética , Fator de Crescimento Insulin-Like II/genética , Canal de Potássio KCNQ1/genética , Gravidez , Resultado da Gravidez
5.
J Endocrinol ; 162(2): 313-9, 1999 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-10425470

RESUMO

Numerous studies have shown a relationship between early growth restriction and Type 2 diabetes. Studies have shown that offspring of rats fed a low protein (LP) diet during pregnancy and lactation have a worse glucose tolerance in late adult life compared with controls. In contrast, in young adult life LP offspring have a better glucose tolerance which is associated with increased insulin-stimulated glucose uptake into skeletal muscle. The aim of the present study was to compare the regulation of glucose uptake and lipolysis in adipocytes by insulin in control and LP offspring. LP adipocytes had increased basal and insulin-stimulated glucose uptake compared with controls. There was no difference in basal rates of lipolysis. Isoproterenol stimulated lipolysis in both groups, but it was more effective on LP adipocytes. Insulin reduced lipolytic rates in controls to basal levels but had a reduced effect in LP adipocytes. Protein kinase B activity matched glucose uptake, with LP adipocytes having elevated activities. These results suggest that early growth retardation has long-term effects on adipocyte metabolism. In addition, they show selective resistance to different metabolic actions of insulin and provide insight into the mechanisms by which insulin regulates glucose uptake and lipolysis.


Assuntos
Adipócitos/metabolismo , Transtornos do Crescimento/metabolismo , Insulina/metabolismo , Animais , Dieta com Restrição de Proteínas , Glucose/metabolismo , Lipólise/fisiologia , Masculino , Proteínas Quinases/metabolismo , Ratos , Ratos Wistar
6.
J Endocrinol ; 177(2): 235-41, 2003 May.
Artigo em Inglês | MEDLINE | ID: mdl-12740011

RESUMO

Epidemiological studies have revealed a relationship between early growth restriction and the subsequent development of type 2 diabetes. A rat model of maternal protein restriction has been used to investigate the mechanistic basis of this relationship. This model causes insulin resistance and diabetes in adult male offspring. The aim of the present study was to determine the effect of early growth restriction on muscle insulin action in late adult life. Rats were fed either a 20% or an isocaloric 8% protein diet during pregnancy and lactation. Offspring were weaned onto a 20% protein diet and studied at 15 Months of age. Soleus muscle from growth restricted offspring (LP) (of dams fed 8% protein diet) had similar basal glucose uptakes compared with the control group (mothers fed 20% protein diet). Insulin stimulated glucose uptake into control muscle but had no effect on LP muscle. This impaired insulin action was not related to changes in expression of either the insulin receptor or glucose transporter 4 (GLUT 4). However, LP muscle expressed significantly less (P<0.001) of the zeta isoform of protein kinase C (PKC zeta) compared with controls. This PKC isoform has been shown to be positively involved in GLUT 4-mediated glucose transport. Expression levels of other isoforms (betaI, betaII, epsilon, theta) of PKC were similar in both groups. These results suggest that maternal protein restriction leads to muscle insulin resistance. Reduced expression of PKC zeta may contribute to the mechanistic basis of this resistance.


Assuntos
Retardo do Crescimento Fetal/metabolismo , Glucose/metabolismo , Transtornos do Crescimento/metabolismo , Insulina/farmacologia , Proteínas Musculares , Músculo Esquelético/metabolismo , Animais , Dieta com Restrição de Proteínas , Regulação para Baixo , Feminino , Transportador de Glucose Tipo 4 , Técnicas In Vitro , Resistência à Insulina , Masculino , Modelos Animais , Proteínas de Transporte de Monossacarídeos/metabolismo , Gravidez , Proteína Quinase C/metabolismo , Ratos , Ratos Wistar , Receptor de Insulina/metabolismo
7.
Mol Cell Endocrinol ; 185(1-2): 81-91, 2001 Dec 20.
Artigo em Inglês | MEDLINE | ID: mdl-11738797

RESUMO

Studies of animal models were carried out to explore mechanisms that might underlie epidemiological findings linking indices of poor early (fetal and early postnatal) growth to an increased risk of developing poor glucose tolerance, including the metabolic syndrome, in adult life. Adult obesity was also seen to play an important role in adding to these risks. We proposed the 'thrifty phenotype' hypothesis to provide a conceptual and mechanistic framework that could be tested by experimentation in animal models. Our main approach has been to feed a reduced protein diet to pregnant and/or lactating rat dams as a means of reducing growth in the fetal and/or preweaning stages of pup growth. Animals were weaned onto either a normal diet or an obesity-inducing highly palatable, cafeteria-style diet. Alterations in intermediary metabolism were noted in the rats with early growth restriction, which provide support for our hypothesis and clues to the mechanism.


Assuntos
Retardo do Crescimento Fetal/metabolismo , Metabolismo/fisiologia , Efeitos Tardios da Exposição Pré-Natal , Animais , Dieta com Restrição de Proteínas/efeitos adversos , Feminino , Humanos , Estado Nutricional/fisiologia , Fenótipo , Gravidez , Ratos
8.
Metabolism ; 47(12): 1450-4, 1998 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-9867072

RESUMO

Plasma beta-hydroxybutyrate concentrations were measured in the offspring of rats that were fed either a control (20% protein) diet or low-protein (8% protein) diet during pregnancy and lactation. Low-protein offspring had significantly lower plasma beta-hydroxybutyrate compared with controls in the fed state (P < .04) and after fasting for 24 hours (P < .001) and 48 hours (P < .04). There were no differences in blood glucose, acetoacetate, plasma glucagon, cholesterol, or glycerol between control and low-protein offspring. However, plasma nonesterified fatty acids (NEFAs) were significantly higher in low-protein offspring in the fed state (P < .05). In contrast, plasma triglycerides and insulin were significantly lower in low-protein offspring compared with controls when fed (P < .001) and after a 24-hour fast (P < .001). These results suggest that poor maternal and early postnatal nutrition can have long-term effects on ketone body metabolism in the offspring during adulthood. This apparent ketosis resistance is similar to that observed in some forms of human diabetes.


Assuntos
Dieta com Restrição de Proteínas , Cetose/metabolismo , Ácido 3-Hidroxibutírico/sangue , Acetoacetatos/sangue , Animais , Glicemia/metabolismo , Colesterol/sangue , Jejum , Ácidos Graxos não Esterificados/sangue , Glucagon/sangue , Glicerol/sangue , Insulina/sangue , Masculino , Ratos , Ratos Wistar , Triglicerídeos/sangue
9.
Metabolism ; 50(5): 562-7, 2001 May.
Artigo em Inglês | MEDLINE | ID: mdl-11319718

RESUMO

Epidemiologic studies have demonstrated associations between low birth weight and increased rates of adult diseases such as hypertension and diabetes. Maternal iron restriction in the rat has been reported to both reduce birth weight and to elevate blood pressure at 40 days of age. The aim of the present study was to extend these findings to investigate the effects of maternal iron restriction on glucose tolerance and serum lipids, 2 important components of the metabolic syndrome, in adult offspring. Blood pressure, glucose tolerance, and serum lipids were measured in the 3-month-old offspring of iron-restricted dams. Rats were placed on control or iron-restricted diets 1 week before mating. At term, dams on the iron-restricted diet were anemic with decreased haemoglobin, red blood cell (RBC) count, hematocrit, and mean RBC volume compared with controls. Neonates from iron-restricted litters were more severely anemic than the dams. At birth, body weight was lower in the offspring of iron-restricted dams than in controls and was still decreased at 3 months of age. At this same age, systolic blood pressure was significantly elevated in the offspring of iron-restricted dams. Glucose tolerance was improved in the maternal iron-restricted group. Fasting serum insulin levels were not different between the control and maternal iron-restricted groups. Fasting serum triglyceride was decreased in the offspring of iron-restricted dams compared with controls. Fasting serum cholesterol and free fatty acid concentrations were similar in both groups. These results suggest that maternal iron restriction has long-term effects on physiology and metabolism in the offspring. Some of these findings are comparable to those reported for the maternal protein-restriction model. It is thus speculated that the long-term effects of maternal dietary restriction may result from common fetal metabolic responses to this restriction.


Assuntos
Pressão Sanguínea , Teste de Tolerância a Glucose , Deficiências de Ferro , Lipídeos/sangue , Efeitos Tardios da Exposição Pré-Natal , Animais , Glicemia/análise , Peso Corporal , Colesterol/sangue , Contagem de Eritrócitos , Ácidos Graxos não Esterificados/sangue , Feminino , Idade Gestacional , Hematócrito , Hemoglobinas/análise , Insulina/sangue , Ferro/administração & dosagem , Gravidez , Ratos , Ratos Wistar , Triglicerídeos/sangue
10.
Metabolism ; 50(9): 1070-7, 2001 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-11555841

RESUMO

An animal model of protein restriction during pregnancy and lactation with subsequent dietary fatty acid manipulation was used to investigate the association between poor early growth, defective unsaturated fatty acid handling, and later disease. Both control and early growth-restricted animals fed a diet rich in saturated fatty acids showed a doubling of the plasma insulin levels as well as a reduced degree of unsaturation in liver and skeletal muscle membrane phospholipids compared with animals fed diets rich in unsaturated fatty acids. The skeletal muscle of early growth-restricted animals weaned onto a saturated fat diet had reduced proportions of 22:6n-3 and increased proportions of 18:1n-9. This reduction in 22:6n-3 is similar to that observed in Pima Indians, a population with a high prevalence of type 2 diabetes.


Assuntos
Membrana Celular/metabolismo , Ácidos Graxos/metabolismo , Retardo do Crescimento Fetal/metabolismo , Resistência à Insulina/fisiologia , Fosfolipídeos/metabolismo , Animais , Glicemia/efeitos dos fármacos , Peso Corporal/efeitos dos fármacos , Membrana Celular/química , Dieta com Restrição de Proteínas/efeitos adversos , Gorduras na Dieta/metabolismo , Gorduras na Dieta/farmacologia , Modelos Animais de Doenças , Ácidos Graxos/análise , Ácidos Graxos Insaturados/análise , Ácidos Graxos Insaturados/metabolismo , Feminino , Retardo do Crescimento Fetal/etiologia , Insulina/sangue , Lactação/metabolismo , Fígado/química , Fígado/metabolismo , Microssomos Hepáticos/química , Microssomos Hepáticos/metabolismo , Músculo Esquelético/química , Músculo Esquelético/metabolismo , Gravidez , Efeitos Tardios da Exposição Pré-Natal , Ratos , Ratos Sprague-Dawley
12.
Horm Res ; 65 Suppl 3: 34-40, 2006.
Artigo em Inglês | MEDLINE | ID: mdl-16612112

RESUMO

Size at birth is said to be a highly heritable trait, with an estimated 30-70% of the variability a result of genetics. Data from family studies may be confounded, however, by potential interactions between fetal genes and the maternal uterine environment. Overall, the maternal environment tends to restrain fetal growth, and this is most evident in first pregnancies. Restraint of fetal growth appears to be inherited through the maternal line. Potential genetic candidates include the mitochondrial DNA 16189 variant, and common variants of exclusively maternally expressed genes, such as H19, which have been associated with size at birth. Maternal blood glucose levels and blood pressure are also correlated with size at birth, but the degree to which these changes relate to genetic variation in the mother is unclear. Elegant studies in mouse knockout models and rare genetic variants in humans have highlighted the importance of insulin-like growth factor I (IGF-I), IGF-II, insulin and their respective receptors in determining fetal growth. However, data linking common variation in the genes that regulate these proteins and receptors with size at birth are few and inconsistent. Interestingly, common variation in the insulin gene (INS) variable number tandem repeats, which regulates the transcription of insulin and IGF-II, has been associated with size at birth, largely in second and subsequent pregnancies, where maternal restraint is least evident. This suggests that fetal genes, and in particular paternally expressed genes, may have significant effects on fetal growth during pregnancies where maternal restraint of fetal growth is less evident.


Assuntos
Peso ao Nascer/genética , Desenvolvimento Fetal/genética , Variação Genética , Gravidez/fisiologia , Animais , Peso ao Nascer/fisiologia , Feminino , Desenvolvimento Fetal/fisiologia , Humanos , Troca Materno-Fetal , Útero/fisiologia
13.
Hum Reprod ; 20(7): 1837-43, 2005 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-15802318

RESUMO

BACKGROUND: Aromatase catalyses the conversion of androgens to estrogens and thus variation in the aromatase gene could contribute to female syndromes of androgen excess, such as precocious pubarche (PP) and polycystic ovarian syndrome (PCOS). METHODS: Two groups, one case-control containing girls from Barcelona, Spain with PP (n = 186) or healthy controls (n = 71), and the other a population study of young women from Oxford, UK, who volunteered for a study of normal women's health (n = 109), were genotyped at four aromatase gene haplotype-tag single nucleotide polymorphisms (SNP). Clinical features and hormone concentrations relevant to hyperandrogenism were compared across haplotypes or genotypes. RESULTS: Distributions of aromatase haplotypes (P < 0.0001) and aromatase SNP_50 genotype (P = 0.001) were significantly different between PP girls and Spanish controls. The AGGG haplotype was associated with an odds ratio (95% confidence interval) of 0.5 (0.3-0.9) (P = 0.005) for the presence of PP compared to GAGG. In 84 post-pubertal PP girls, aromatase haplotype was associated with functional ovarian hyperandrogenism (P < 0.05), independently of insulin sensitivity. In the Oxford population, SNP_50 was associated with variation in PCOS symptom score (P = 0.008) and circulating testosterone concentrations (P = 0.02). CONCLUSIONS: This study suggests that common variation at the aromatase gene (and not just rare loss-of-function mutations) is associated with androgen excess in girls and young women.


Assuntos
Aromatase/genética , Variação Genética , Hiperandrogenismo/enzimologia , Hiperandrogenismo/genética , Adolescente , Sequência de Bases , Estudos de Casos e Controles , Criança , DNA/genética , Feminino , Genética Populacional , Haplótipos , Humanos , Hiperandrogenismo/etiologia , Resistência à Insulina , Síndrome do Ovário Policístico/enzimologia , Síndrome do Ovário Policístico/etiologia , Síndrome do Ovário Policístico/genética , Polimorfismo de Nucleotídeo Único , Puberdade Precoce/enzimologia , Puberdade Precoce/etiologia , Puberdade Precoce/genética , Espanha , Reino Unido
14.
Hum Reprod Update ; 6(6): 578-86, 2000.
Artigo em Inglês | MEDLINE | ID: mdl-11129690

RESUMO

The number of cell divisions during embryonic and fetal life makes the embryo/fetus particularly vulnerable to effects resulting from exposure to an adverse intrauterine environment. Exposure to drugs and irradiation at this stage of development are able to cause congenital malformations and various cancers in later life. In-utero exposure to hyperglycaemia is able to lead to future diabetes that is heritable, but not genetic in origin. Fetal malnutrition causing growth restriction is able to lead to an increased risk of developing type 2 diabetes, hypertension and ischaemic heart disease in later life, especially if the growth restriction is followed by catch-up growth postnatally. This review discusses the various mechanisms by which these effects may occur, and presents the difficulties that will have to be faced if their world-wide health burdens are to be reduced.


Assuntos
Doenças Fetais , Distúrbios Nutricionais/complicações , Efeitos Tardios da Exposição Pré-Natal , Animais , Doenças Cardiovasculares/etiologia , Diabetes Gestacional/etiologia , Desenvolvimento Embrionário e Fetal , Feminino , Feto/efeitos dos fármacos , Feto/efeitos da radiação , Humanos , Doenças Metabólicas/etiologia , Gravidez , Fatores de Risco , Raios X/efeitos adversos
15.
Clin Sci (Lond) ; 93(2): 147-52, 1997 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-9301429

RESUMO

1. Recent studies have revealed a link between fetal and early post-natal, growth retardation and the development of features of the insulin resistance syndrome in later life. Obesity is also a strong risk factor for this syndrome. The aim of this study was to assess whether maternal and early protein restriction, which causes growth retardation, and obesity are risk factors that are independent for the development of certain features of the insulin resistance syndrome, especially hypertension. 2. Pregnant Sprague-Dawley rats were given either 20% or 8% protein isocaloric diets throughout pregnancy and lactation. Female offspring were weaned onto the same diets as their mothers and they remained on these diets until 70 days of age. Half the rats were then given standard laboratory chow, whilst the remainder were fed a highly palatable cafeteria-style diet. Rats were maintained on these diets for the remainder of the study. 3. Rats given the 8% protein diet remained physically lighter than comparable animals fed the 20% protein diet throughout the study. In contrast, cafeteria-fed rats showed excessive weight gain. At 1 year of age the rats had their systolic blood pressures and fasting lipids measured, as well as undergoing an intraperitoneal glucose-tolerance test. 4. Cafeteria-fed rats had worse glucose tolerances than controls and hypertriacylglycerolaemia. The early 8% protein rats had significantly increased blood pressures, as did the cafeteria-fed rats. These increases were additive, suggesting that early protein restriction, and later obesity, are indeed independent risk factors for the development of hypertension.


Assuntos
Dieta com Restrição de Proteínas , Hipertensão/etiologia , Obesidade/complicações , Análise de Variância , Animais , Peso Corporal , Feminino , Teste de Tolerância a Glucose , Hipertensão/sangue , Tamanho da Ninhada de Vivíparos , Obesidade/sangue , Tamanho do Órgão , Gravidez , Ratos , Ratos Sprague-Dawley , Triglicerídeos/sangue
16.
Horm Metab Res ; 32(2): 71-5, 2000 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-10741689

RESUMO

Male offspring of rats protein restricted during pregnancy and lactation are growth restricted and have changes in insulin action on epididymal adipocytes. Adipocytes from different anatomical depots are thought to have distinct metabolic functions. The aim of the present study was to determine whether the alterations in metabolism of adipocytes from early growth restricted rats is depot-specific or more generalised. Epididymal, intra-abdominal and subcutaneous adipocytes were studied from three-month-old male offspring of control and protein restricted dams. Basal glucose uptakes were higher in low protein adipocytes (p<0.01) compared to controls. However, insulin stimulation was less in the low protein group in all depots compared to controls (p<0.05). Isoproterenol-stimulated lipolysis was greater in low protein adipocytes (p<0.0001), but the magnitude was greater in epididymal (p<0.0001) and intra-abdominal (p<0.0001) adipocytes than in subcutaneous adipocytes. Epididymal and intra-abdominal adipocytes from low protein offspring were also resistant to the anti-lipolytic action of insulin. These results suggest that certain changes associated with early growth retardation are depot-specific, being enhanced in the more metabolically active intra-abdominal and epididymal tissues.


Assuntos
Adipócitos/efeitos dos fármacos , Adipócitos/metabolismo , Retardo do Crescimento Fetal/metabolismo , Hipoglicemiantes/farmacologia , Insulina/farmacologia , Proteínas Musculares , Tecido Adiposo/química , Tecido Adiposo/citologia , Tecido Adiposo/metabolismo , Animais , Western Blotting , Deficiências Nutricionais/metabolismo , Dieta com Restrição de Proteínas , Proteínas Alimentares/administração & dosagem , Epididimo/citologia , Feminino , Glucose/farmacocinética , Transportador de Glucose Tipo 4 , Lipólise/efeitos dos fármacos , Masculino , Proteínas de Transporte de Monossacarídeos/análise , Gravidez , Ratos , Ratos Wistar
17.
Horm Metab Res ; 32(6): 233-9, 2000 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-10898553

RESUMO

Rats were fed a diet containing either 20% ("control") or 8% ("reduced-protein") protein throughout pregnancy and lactation. Their female offspring were weaned onto the same respective diets. At 63 days of age one set of control and reduced-protein rats (n = 16 per group) underwent intraperitoneal glucose tolerance tests and one week later were killed and their pancreatic hormones extracted and measured. The reduced protein rats had better glucose tolerance (p < 0.001) and lower pancreatic insulin (p < 0.01) and amylin (p < 0.01) contents. Further sets of control and reduced-protein rats were then fed either chow or a cafeteria-style diet (n = 16 in each of the four groups). These rats underwent intraperitoneal glucose tolerance tests at 133 days of age, which showed the cafeteria-fed animals to have a worse glucose tolerance than the chow-fed animals irrespective of previous diet exposure (p < 0.0001). One week later reduced-protein rats still had lower pancreatic insulin contents (p < 0.05) (and a trend for lower amylin contents), but also had increased pancreatic glucagon contents (p < 0.05). There were no detectable differences in pancreatic somatostatin-like immunoreactivity or pancreatic polypeptide contents. These results are consistent with pancreatic beta- and alpha-cells being selectively susceptible to effects associated with early dietary protein restriction.


Assuntos
Glicemia/análise , Proteínas Alimentares/administração & dosagem , Obesidade/metabolismo , Hormônios Pancreáticos/análise , Amiloide/análise , Animais , Peso Corporal , Feminino , Glucagon/análise , Insulina/análise , Insulina/sangue , Polipeptídeo Amiloide das Ilhotas Pancreáticas , Tamanho do Órgão , Pâncreas/patologia , Ratos , Ratos Sprague-Dawley
18.
Diabet Med ; 17(12): 848-53, 2000 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-11168327

RESUMO

AIMS: Low birthweight in humans has been shown to lead to increased resting pulse rate in adult life, suggesting possible increased sympathoadrenal activity. The hypothesis that early growth restriction is associated with permanent alterations in catecholamine metabolism was tested. METHODS: Circulating catecholamine concentrations (by radioimmunoassay) and adipocyte adrenoceptor expression from different fat depots (by Western blot) were estimated in 12-week-old male offspring of rat dams fed a reduced protein diet during pregnancy and lactation. RESULTS: In the fed state, median (interquartile range) plasma adrenaline concentrations for male control and low protein offspring rats were: 0.65 (0.48-0.86) vs. 1.42 (0.89-1.87) nmol/l (P < 0.005), respectively. Equivalent noradrenaline concentrations were: 2.71 (2.16-3.46) vs. 3.45 (3.00-4.28) nmol/l (P < 0.05). After 24 h starvation, plasma adrenaline concentrations of controls rose to become similar to those of low protein offspring: 1.03 (0.95-1.31) vs. 1.41 (0.69-1.62) nmol/l (P = 0.3), respectively. Noradrenaline concentrations rose in both groups to become similar: 3.84 (3.33-4.54) vs. 4.32 (3.70-6.54) nmol/l (P = 0.3). In epididymal adipocytes adrenoceptor expression (relative to that of controls) was: alpha2A 0.79 (0.66-0.94) (P = 0.08), beta1 2.60 (2.27-3.07) (P = 0.04), beta3 1.37 (1.27-1.46) (P = 0.02). Similar-pattern differences in adrenoceptor expression were observed in subcutaneous and intra-abdominal adipocytes. CONCLUSIONS: These results are consistent with the suggestion that long-term alterations in catecholamine metabolism are present in adult offspring of rats fed a reduced protein diet during pregnancy and lactation.


Assuntos
Dieta com Restrição de Proteínas , Epinefrina/sangue , Norepinefrina/sangue , Efeitos Tardios da Exposição Pré-Natal , Deficiência de Proteína/complicações , Receptores Adrenérgicos/análise , Adipócitos/química , Animais , Epididimo , Feminino , Lactação , Masculino , Gravidez , Ratos , Ratos Wistar
19.
Am J Physiol ; 272(5 Pt 1): G1083-90, 1997 May.
Artigo em Inglês | MEDLINE | ID: mdl-9176217

RESUMO

Hepatic enzymes associated with glucose hemostasis were studied in offspring of dams fed either a 20% protein (control) or an isocaloric 8% protein (low-protein) diet during pregnancy and lactation. Additionally, offspring were exposed to maternal 8% protein diet only during gestation (recuperated) or lactation (postnatal low-protein). Glucokinase activity decreased (approximately 50%), whereas phosphoenolpyruvate carboxykinase (PEPCK) activity increased (approximately 100%), in the low-protein and recuperated offspring compared with controls (P < 0.001) at 21 days of age. However, the postnatal low-protein offspring had enzyme activities comparable with those of controls. These changes were still evident in 11-mo-old offspring weaned onto a normal laboratory chow. Parallel changes were apparent in mRNA levels of glucokinase and PEPCK in the low-protein male offspring. Thus the effect of programming metabolism extends not only to protein biochemistry but possibly also to the regulation of gene expression. Furthermore, these changes could not be attributed to glucagon or insulin, because ratios of these hormones were comparable between the control and low-protein groups.


Assuntos
Animais Recém-Nascidos/metabolismo , Proteínas Alimentares/administração & dosagem , Insulina/farmacologia , Fígado/enzimologia , Prenhez/fisiologia , Animais , Animais Recém-Nascidos/crescimento & desenvolvimento , Proteínas Alimentares/farmacologia , Feminino , Expressão Gênica , Glucagon/farmacologia , Glucoquinase/genética , Glucoquinase/metabolismo , Masculino , Fosfoenolpiruvato Carboxiquinase (GTP)/genética , Fosfoenolpiruvato Carboxiquinase (GTP)/metabolismo , Gravidez , Ratos , Ratos Wistar
20.
Diabetologia ; 41(11): 1337-42, 1998 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-9833942

RESUMO

Numerous studies show an association between poor fetal growth and adult insulin resistance. Recent studies have shown relation between the long chain polyunsaturated fatty acid composition of skeletal muscle membranes and insulin sensitivity. More detailed analysis has indicated that the activity of delta5 desaturase is inversely correlated to insulin resistance. The amount of docosahexaenoic acid (C22:6n3) is also thought to play a part in determining insulin sensitivity. The purpose of this study was to test the hypothesis that early growth retardation in the rat, as a result of maternal protein restriction, would lead to alterations in desaturase activities similar to those observed in human insulin resistance. There were no differences in phospholipid fatty acid composition in liver or muscle from control and low protein rats. In both muscle and liver the ratio of docosahexaenoic acid to docosapentaenoic acid was, however, reduced in low protein offspring. Direct measurement of delta5 desaturase activity in hepatic microsomes showed a reduction (p < 0.03) in the low protein offspring which was negatively correlated (r = -0.855) with fasting plasma insulin. No correlation was observed in controls. These results show that it is possible to programme the activity of key enzymes involved in the desaturation of long chain polyunsaturated fatty acids. This is possibly a mechanism linking fetal growth retardation to insulin resistance.


Assuntos
Dieta com Restrição de Proteínas , Gorduras na Dieta , Ácidos Graxos Dessaturases/metabolismo , Microssomos Hepáticos/enzimologia , Músculo Esquelético/enzimologia , Efeitos Tardios da Exposição Pré-Natal , Adulto , Animais , Peso Corporal , Membrana Celular/enzimologia , Dessaturase de Ácido Graxo Delta-5 , Ácidos Graxos/análise , Feminino , Humanos , Insulina/sangue , Fosfolipídeos/química , Fosfolipídeos/metabolismo , Gravidez , Ratos , Ratos Wistar
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA