Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 242
Filtrar
1.
J Endocrinol ; 258(3)2023 09 01.
Artigo em Inglês | MEDLINE | ID: mdl-37343234

RESUMO

11ß-Hydroxysteroid dehydrogenase 1 (11ßHSD1) is a drug target to attenuate adverse effects of chronic glucocorticoid excess. It catalyses intracellular regeneration of active glucocorticoids in tissues including brain, liver and adipose tissue (coupled to hexose-6-phosphate dehydrogenase, H6PDH). 11ßHSD1 activity in individual tissues is thought to contribute significantly to glucocorticoid levels at those sites, but its local contribution vs glucocorticoid delivery via the circulation is unknown. Here, we hypothesised that hepatic 11ßHSD1 would contribute significantly to the circulating pool. This was studied in mice with Cre-mediated disruption of Hsd11b1 in liver (Alac-Cre) vs adipose tissue (aP2-Cre) or whole-body disruption of H6pdh. Regeneration of [9,12,12-2H3]-cortisol (d3F) from [9,12,12-2H3]-cortisone (d3E), measuring 11ßHSD1 reductase activity was assessed at steady state following infusion of [9,11,12,12-2H4]-cortisol (d4F) in male mice. Concentrations of steroids in plasma and amounts in liver, adipose tissue and brain were measured using mass spectrometry interfaced with matrix-assisted laser desorption ionisation or liquid chromatography. Amounts of d3F were higher in liver, compared with brain and adipose tissue. Rates of appearance of d3F were ~6-fold slower in H6pdh-/- mice, showing the importance for whole-body 11ßHSD1 reductase activity. Disruption of liver 11ßHSD1 reduced the amounts of d3F in liver (by ~36%), without changes elsewhere. In contrast disruption of 11ßHSD1 in adipose tissue reduced rates of appearance of circulating d3F (by ~67%) and also reduced regenerated of d3F in liver and brain (both by ~30%). Thus, the contribution of hepatic 11ßHSD1 to circulating glucocorticoid levels and amounts in other tissues is less than that of adipose tissue.


Assuntos
Cortisona , Glucocorticoides , Masculino , Camundongos , Animais , Hidrocortisona , Tecido Adiposo , Esteroides , 11-beta-Hidroxiesteroide Desidrogenase Tipo 1/genética
2.
J Neuroendocrinol ; 29(1)2017 01.
Artigo em Inglês | MEDLINE | ID: mdl-27859809

RESUMO

Mice lacking the intracellular glucocorticoid-regenerating enzyme 11ß-hydroxysteroid dehydrogenase type 1 (11ß-HSD1) are protected from age-related spatial memory deficits. 11ß-HSD1 is expressed predominantly in the brain, liver and adipose tissue. Reduced glucocorticoid levels in the brain in the absence of 11ß-HSD1 may underlie the improved memory in aged 11ß-HSD1 deficient mice. However, the improved glucose tolerance, insulin sensitisation and cardioprotective lipid profile associated with reduced peripheral glucocorticoid regeneration may potentially contribute to the cognitive phenotype of aged 11ß-HSD1 deficient mice. In the present study, transgenic mice with forebrain-specific overexpression of 11ß-HSD1 (Tg) were intercrossed with global 11ß-HSD1 knockout mice (HSD1KO) to examine the influence of forebrain and peripheral 11ß-HSD1 activity on spatial memory in aged mice. Transgene-mediated delivery of 11ß-HSD1 to the hippocampus and cortex of aged HSD1KO mice reversed the improved spatial memory retention in the Y-maze but not spatial learning in the watermaze. Brain-derived neurotrophic factor (BDNF) mRNA levels in the hippocampus of aged HSD1KO mice were increased compared to aged wild-type mice. Rescue of forebrain 11ß-HSD1 reduced BDNF mRNA in aged HSD1KO mice to levels comparable to aged wild-type mice. These findings indicate that 11ß-HSD1 regenerated glucocorticoids in the forebrain and decreased levels of BDNF mRNA in the hippocampus play a role in spatial memory deficits in aged wild-type mice, although 11ß-HSD1 activity in peripheral tissues may also contribute to spatial learning impairments in aged mice.


Assuntos
11-beta-Hidroxiesteroide Desidrogenase Tipo 1/fisiologia , Envelhecimento/psicologia , Fator Neurotrófico Derivado do Encéfalo/biossíntese , Terapia Genética , Transtornos da Memória/fisiopatologia , Transtornos da Memória/terapia , Prosencéfalo/fisiologia , 11-beta-Hidroxiesteroide Desidrogenase Tipo 1/deficiência , 11-beta-Hidroxiesteroide Desidrogenase Tipo 1/genética , Envelhecimento/genética , Animais , Corticosterona/sangue , Hipocampo/metabolismo , Masculino , Aprendizagem em Labirinto/fisiologia , Transtornos da Memória/genética , Camundongos , Camundongos Knockout , Camundongos Transgênicos , RNA Mensageiro/genética , RNA Mensageiro/metabolismo
3.
J Neuroendocrinol ; 28(1)2016 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-26563879

RESUMO

Mice deficient in the glucocorticoid-regenerating enzyme 11ß-HSD1 resist age-related spatial memory impairment. To investigate the mechanisms and pathways involved, we used microarrays to identify differentially expressed hippocampal genes that associate with cognitive ageing and 11ß-HSD1. Aged wild-type mice were separated into memory-impaired and unimpaired relative to young controls according to their performance in the Y-maze. All individual aged 11ß-HSD1-deficient mice showed intact spatial memory. The majority of differentially expressed hippocampal genes were increased with ageing (e.g. immune/inflammatory response genes) with no genotype differences. However, the neuronal-specific transcription factor, Npas4, and immediate early gene, Arc, were reduced (relative to young) in the hippocampus of memory-impaired but not unimpaired aged wild-type or aged 11ß-HSD1-deficient mice. A quantitative reverse transcriptase-polymerase chain reaction and in situ hybridisation confirmed reduced Npas4 and Arc mRNA expression in memory-impaired aged wild-type mice. These findings suggest that 11ß-HSD1 may contribute to the decline in Npas4 and Arc mRNA levels associated with memory impairment during ageing, and that decreased activity of synaptic plasticity pathways involving Npas4 and Arc may, in part, underlie the memory deficits seen in cognitively-impaired aged wild-type mice.


Assuntos
11-beta-Hidroxiesteroide Desidrogenase Tipo 1/metabolismo , Envelhecimento/metabolismo , Fatores de Transcrição Hélice-Alça-Hélice Básicos/metabolismo , Proteínas do Citoesqueleto/metabolismo , Hipocampo/metabolismo , Transtornos da Memória/metabolismo , Proteínas do Tecido Nervoso/metabolismo , 11-beta-Hidroxiesteroide Desidrogenase Tipo 1/genética , Envelhecimento/genética , Animais , Fatores de Transcrição Hélice-Alça-Hélice Básicos/genética , Proteínas do Citoesqueleto/genética , Aprendizagem em Labirinto/fisiologia , Memória/fisiologia , Transtornos da Memória/genética , Camundongos , Camundongos Knockout , Proteínas do Tecido Nervoso/genética , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Memória Espacial/fisiologia
4.
Int J Obes (Lond) ; 39(10): 1539-47, 2015 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-26032810

RESUMO

BACKGROUND: Although the prevalence of obesity is higher among women than men, they are somewhat protected from the associated cardiometabolic consequences. The increase in cardiovascular disease risk seen after the menopause suggests a role for estrogens. There is also growing evidence for the importance of estrogen on body fat and metabolism in males. We hypothesized that that estrogen administration would ameliorate the adverse effects of obesity on metabolic parameters in males. METHODS: Male and female C57Bl/6 mice were fed control or obesogenic (DIO) diets from 5 weeks of age until adulthood. Glucose tolerance testing was performed at 13 weeks of age. Mice were killed at 15 weeks of age and liver and adipose tissue were collected for analysis of gene expression. A second cohort of male mice underwent the same experimental design with the addition of estradiol pellet implantation or sham surgery at 6 weeks. RESULTS: DIO males had greater mesenteric adipose deposition and more severe increases in plasma glucose, insulin and lipids than females. Treatment of males with estradiol from 6 weeks of age prevented DIO-induced increases in adipose tissue mass and alterations in glucose-insulin homeostasis. We also identified sex differences in the transcript levels and activity of hepatic and adipose glucocorticoid metabolizing enzymes. Estrogen treatment feminized the pattern of DIO-induced changes in glucocorticoid metabolism, rendering males similar to females. CONCLUSIONS: Thus, DIO induces sex-specific changes in glucose-insulin homeostasis, which are ameliorated in males treated with estrogen, highlighting the importance of sex steroids in metabolism. Given that altered peripheral glucocorticoid metabolism has been observed in rodent and human obesity, our results also suggest that sexually dimorphic expression and activity of glucocorticoid metabolizing enzymes may have a role in the differential metabolic responses to obesity in males and females.


Assuntos
Tecido Adiposo/metabolismo , Estrogênios/farmacologia , Glucocorticoides/metabolismo , Fígado/metabolismo , Obesidade/metabolismo , Receptores de Estrogênio/efeitos dos fármacos , Adiposidade , Animais , Dieta Hiperlipídica , Modelos Animais de Doenças , Feminino , Teste de Tolerância a Glucose , Inflamação/prevenção & controle , Metabolismo dos Lipídeos , Masculino , Camundongos , Camundongos Endogâmicos C57BL
5.
Psychol Med ; 45(15): 3217-26, 2015 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-26096463

RESUMO

BACKGROUND: Glucocorticoids and serotonin may mediate the link between maternal environment, fetal brain development and 'programming' of offspring behaviors. The placenta regulates fetal exposure to maternal hormonal signals in animal studies, but few data address this in humans. We measured prospectively maternal depressive symptoms during pregnancy and mRNAs encoding key gene products determining glucocorticoid and serotonin function in term human placenta and explored associations with infant regulatory behaviors. METHOD: Bi-weekly self-ratings of the Center for Epidemiologic Studies Depression Scale from 12th to 13th gestational week onwards and term placental mRNAs of 11beta-hydroxysteroid dehydrogenase type 2 (HSD2B11), type 1 (HSD1B11), glucocorticoid (NR3C1), mineralocorticoid receptors (NR3C2) and serotonin transporter (SLC6A4) were obtained from 54 healthy mothers aged 32.2 ± 5.3 years with singleton pregnancies and without pregnancy complications. Infant regulatory behaviors (crying, feeding, spitting, elimination, sleeping and predictability) were mother-rated at 15.6 ± 4.2 days. RESULTS: Higher placental mRNA levels of HSD2B11 [0.41 standard deviation (s.d.) unit increase per s.d. unit increase; 95% confidence interval (CI) 0.13-0.69, p = 0.005], HSD1B11 (0.30, 0.03-0.57, p = 0.03), NR3C1 (0.44, 0.19-0.68, p = 0.001) and SLC6A4 (0.26, 0.00-0.53, p = 0.05) were associated with more regulatory behavioral challenges of the infant. Higher placental NR3C1 mRNA partly mediated the association between maternal depressive symptoms during pregnancy and infant regulatory behaviors (p < 0.05). CONCLUSIONS: Higher placental expression of genes regulating feto-placental glucocorticoid and serotonin exposure is characteristic of infants with more regulatory behavioral challenges. Maternal depression acts, at least partly, via altering glucocorticoid action in the placenta to impact on offspring regulatory behaviors.


Assuntos
Depressão/metabolismo , Glucocorticoides/metabolismo , Comportamento do Lactente/fisiologia , Placenta/metabolismo , Complicações na Gravidez/metabolismo , Efeitos Tardios da Exposição Pré-Natal/metabolismo , Comportamento Problema , Serotonina/metabolismo , Adulto , Feminino , Seguimentos , Expressão Gênica , Glucocorticoides/genética , Humanos , Lactente , Masculino , Gravidez , RNA Mensageiro/metabolismo , Serotonina/genética , Proteínas da Membrana Plasmática de Transporte de Serotonina/metabolismo
6.
Psychol Med ; 45(10): 2023-30, 2015 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-25628053

RESUMO

BACKGROUND: Maternal prenatal depression predicts post-partum depression and increases risk of prematurity and low birth weight. These effects may be mediated by altered placental function. We hypothesized that placental function would be influenced by the gestational week of experiencing depressive symptoms and aimed to examine associations between maternal depressive symptoms during pregnancy and placental expression of genes involved in glucocorticoid and serotonin transfer between mother and fetus. METHOD: We studied women participating in a prospective pregnancy cohort: the Prediction and Prevention of Preeclampsia (PREDO) Study, Helsinki, Finland. Maternal depressive symptoms were assessed at 2-week intervals throughout pregnancy in 56 healthy women with singleton, term pregnancies. Messenger ribonucleic acid (mRNA) levels of glucocorticoid (GR) and mineralocorticoid (MR) receptors and serotonin transporter (SLC6A4), 11ß-hydroxysteroid dehydrogenase type 1 (HSD1) and 2 (HSD2) were quantified in placental biopsies. RESULTS: In adjusted analyses women who reported higher depressive symptoms across the whole pregnancy had higher mRNA levels of GR [effect size 0.31 s.d. units, 95% confidence interval (CI) 0.01-0.60, p = 0.042] and MR (effect size 0.34 s.d. units, 95% CI 0.01-0.68, p = 0.047). These effects were significant for symptoms experienced in the third trimester of pregnancy for GR; findings for MR were also significant for symptoms experienced in the second trimester. GR and MR mRNA levels increased linearly by having the trimester-specific depressive symptoms scores 0, 1 or 2-3 times above the clinical cut-off for depression (p = 0.003, p = 0.049, respectively, and p = 0.004, p = 0.15 in adjusted analyses). CONCLUSIONS: Our findings offer potential gestational-age-specific mechanisms linking maternal depressive symptoms during pregnancy via placental biology. Future studies will test whether these also link with adverse offspring outcomes.


Assuntos
Depressão/fisiopatologia , Glucocorticoides/metabolismo , Complicações na Gravidez/fisiopatologia , RNA Mensageiro/metabolismo , 11-beta-Hidroxiesteroide Desidrogenases/análise , Adulto , Feminino , Finlândia , Glucocorticoides/genética , Humanos , Modelos Lineares , Placenta/química , Gravidez , Trimestres da Gravidez , Escalas de Graduação Psiquiátrica , RNA Mensageiro/análise , RNA Mensageiro/genética , Receptores de Mineralocorticoides/análise , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Proteínas da Membrana Plasmática de Transporte de Serotonina/análise , Proteínas da Membrana Plasmática de Transporte de Serotonina/genética , Adulto Jovem
7.
Int J Obes (Lond) ; 39(2): 303-11, 2015 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-24854429

RESUMO

BACKGROUND: Black women have lower visceral adipose tissue (VAT) but are less insulin sensitive than white women; the mechanisms responsible are unknown. OBJECTIVE: The study aimed to test the hypothesis that variation in subcutaneous adipose tissue (SAT) sensitivity to glucocorticoids might underlie these differences. METHODS: Body fatness (dual energy X-ray absorptiometry) and distribution (computerized tomography), insulin sensitivity (SI, intravenous and oral glucose tolerance tests), and expression of 11ß-hydroxysteroid dehydrogenase-1 (11HSD1), hexose-6-phosphate dehydrogenase and glucocorticoid receptor-α (GRα), as well as genes involved in adipogenesis and inflammation were measured in abdominal deep SAT, superficial SAT and gluteal SAT (GLUT) depots of 56 normal-weight or obese black and white premenopausal South African (SA) women. We used a combination of univariate and multivariate statistics to evaluate ethnic-specific patterns in adipose gene expression and related body composition and insulin sensitivity measures. RESULTS: Although 11HSD1 activity and mRNA did not differ by ethnicity, GRα mRNA levels were significantly lower in SAT of black compared with white women, particularly in the GLUT depot (0.52±0.21 vs 0.91±0.26 AU, respectively, P<0.01). In black women, lower SAT GRα mRNA levels were associated with increased inflammatory gene transcript levels and abdominal SAT area, and reduced adipogenic gene transcript levels, VAT/SAT ratio and SI. Abdominal SAT 11HSD1 activity associated with increased VAT area and decreased SI in white, but not in black women. CONCLUSIONS: In black SA women, downregulation of GRα mRNA levels with obesity and reduced insulin sensitivity, possibly via increased SAT inflammation, is associated with reduced VAT accumulation.


Assuntos
11-beta-Hidroxiesteroide Desidrogenases/metabolismo , População Negra , Gordura Intra-Abdominal/metabolismo , Síndrome Metabólica/metabolismo , Receptores de Glucocorticoides/metabolismo , Gordura Subcutânea/metabolismo , População Branca , 11-beta-Hidroxiesteroide Desidrogenases/genética , Absorciometria de Fóton , Adulto , Composição Corporal/genética , Estudos Transversais , Feminino , Teste de Tolerância a Glucose , Humanos , Síndrome Metabólica/etnologia , Síndrome Metabólica/genética , África do Sul/epidemiologia
8.
Transl Psychiatry ; 4: e448, 2014 Sep 23.
Artigo em Inglês | MEDLINE | ID: mdl-25247593

RESUMO

Early-life stress (ELS) is known to be associated with an increased risk of neuropsychiatric and cardiometabolic disease in later life. One of the potential mechanisms underpinning this is through effects on the epigenome, particularly changes in DNA methylation. Using a well-phenotyped cohort of 83 men from the Helsinki Birth Cohort Study, who experienced ELS in the form of separation from their parents during childhood, and a group of 83 matched controls, we performed a genome-wide analysis of DNA methylation in peripheral blood. We found no differences in DNA methylation between men who were separated from their families and non-separated men; however, we did identify differences in DNA methylation in association with the development of at least mild depressive symptoms over the subsequent 5-10 years. Notably, hypomethylation was identified at a number of genes with roles in brain development and/or function in association with depressive symptoms. Pathway analysis revealed an enrichment of DNA methylation changes in pathways associated with development and morphogenesis, DNA and transcription factor binding and programmed cell death. Our results support the concept that DNA methylation differences may be important in the pathogenesis of psychiatric disease.


Assuntos
Metilação de DNA/genética , Epigênese Genética/genética , Homens/psicologia , Transtornos Mentais/genética , Estresse Psicológico/genética , Idoso , Estudos de Coortes , Epigenômica , Feminino , Finlândia , Humanos , Masculino , Pessoa de Meia-Idade
9.
J Dev Orig Health Dis ; 5(5): 351-60, 2014 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-25081820

RESUMO

Rates of obesity are increasing in women of child bearing age with negative impacts on maternal and offspring health. Emerging evidence suggests in utero origins of respiratory health in offspring of obese mothers but mechanisms are unknown. Changes in maternal cortisol levels are one potential factor as cortisol levels are altered in obesity and cortisol is separately implicated in development of offspring wheeze. We aimed to assess whether increased pre-pregnancy maternal body mass index (BMI) was associated with offspring early life wheezing, and whether this was mediated by altered cortisol levels in the mother. In a prospective community-based cohort (Amsterdam Born Children and their Development cohort), women completed questionnaires during pregnancy and at 3-5 months post-delivery regarding self-history of asthma and atopy, and of wheezing of their offspring (n=4860). Pre-pregnancy BMI was recorded and serum total cortisol levels were measured in a subset of women (n=2227) at their first antenatal visit. A total of 20.2% (n=984) women were overweight or obese and 10.3% reported wheezing in their offspring. Maternal BMI was associated with offspring wheezing (1 unit (kg/m2) increase, OR: 1.03; 95% CI: 1.00-1.05), after correction for confounders. Although maternal cortisol levels were lower in overweight mothers and those with a history of asthma, maternal cortisol levels did not mediate the increased offspring wheezing. Pre-pregnancy BMI impacts on baby wheezing, which is not mediated by lower cortisol levels. As the prevalence of obesity in women of child-bearing age is increasing, further studies are needed to investigate modifiable maternal factors to avoid risk of wheezing in young children.


Assuntos
Índice de Massa Corporal , Obesidade/complicações , Obesidade/fisiopatologia , Complicações na Gravidez , Sons Respiratórios/etiologia , Glândulas Suprarrenais/fisiopatologia , Adulto , Asma/complicações , Feminino , Humanos , Hidrocortisona/sangue , Hipersensibilidade/complicações , Sistema Hipotálamo-Hipofisário/fisiopatologia , Lactente , Masculino , Países Baixos , Razão de Chances , Gravidez , Complicações na Gravidez/fisiopatologia , Estudos Prospectivos , Sons Respiratórios/fisiopatologia , Estações do Ano , Fumar/efeitos adversos , Inquéritos e Questionários
10.
Acta Physiol (Oxf) ; 210(2): 288-95, 2014 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-24330050

RESUMO

Foetal growth restriction (FGR), reflective of an adverse intrauterine environment, confers a significantly increased risk of perinatal mortality and morbidity. In addition, low birthweight associates with adult diseases including hypertension, metabolic dysfunction and behavioural disorders. A key mechanism underlying FGR is exposure of the foetus to glucocorticoids which, while critical for foetal development, in excess can reduce foetal growth and permanently alter organ structure and function, predisposing to disease in later life. Foetal glucocorticoid exposure is regulated, at least in part, by the enzyme 11ß-hydroxysteroid dehydrogenase type 2 (11ß-HSD2), which catalyses the intracellular inactivation of glucocorticoids. This enzyme is highly expressed within the placenta at the maternal-foetal interface, limiting the passage of glucocorticoids to the foetus. Expression of 11ß-HSD2 is also high in foetal tissues, particularly within the developing central nervous system. Down-regulation or genetic deficiency of placental 11ß-HSD2 is associated with significant reductions in foetal growth and birth weight, and programmed outcomes in adulthood. To unravel the direct significance of 11ß-HSD2 for developmental programming, placental function, neurodevelopment and adult behaviour have been extensively investigated in a mouse knockout of 11ß-HSD2. This review highlights the evidence obtained from this mouse model for a critical role of feto-placental 11ß-HSD2 in determining the adverse programming outcomes.


Assuntos
11-beta-Hidroxiesteroide Desidrogenase Tipo 2/metabolismo , Desenvolvimento Fetal/fisiologia , Glucocorticoides/metabolismo , Placenta/metabolismo , Animais , Feminino , Retardo do Crescimento Fetal/etiologia , Retardo do Crescimento Fetal/metabolismo , Feto , Glucocorticoides/efeitos adversos , Síndrome Metabólica/embriologia , Síndrome Metabólica/metabolismo , Camundongos , Gravidez
11.
Placenta ; 34(11): 1087-90, 2013 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-24090886

RESUMO

Exposure to overnutrition in utero may increase offspring cardiometabolic disease risk. A mouse model of maternal exposure to an obesogenic diet (DIO) was used to determine effects on fetal and placental weight and gene expression in mid- and late gestation. DIO altered placental gene expression in mid-gestation without differences in fetal or placental weights. Weight gain was attenuated in DIO dams in late gestation and male pup weight was reduced, however there were no persistent changes in placental gene expression. Differences in maternal weight gain and/or specific dietary components may impact on fetal and placental growth and later disease risk.


Assuntos
Dieta Hiperlipídica/efeitos adversos , Sacarose Alimentar/efeitos adversos , Desenvolvimento Fetal , Regulação da Expressão Gênica no Desenvolvimento , Fenômenos Fisiológicos da Nutrição Materna , Placenta/metabolismo , Placentação , Animais , Animais Recém-Nascidos , Peso ao Nascer , Feminino , Peso Fetal , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Obesidade/etiologia , Hipernutrição/metabolismo , Gravidez , Proteínas da Gravidez/biossíntese , Proteínas da Gravidez/genética , Proteínas da Gravidez/metabolismo , RNA Mensageiro/metabolismo , Caracteres Sexuais , Aumento de Peso
12.
Psychoneuroendocrinology ; 38(5): 648-58, 2013 May.
Artigo em Inglês | MEDLINE | ID: mdl-22980941

RESUMO

An imbalance between central glucocorticoid (GR) and mineralocorticoid (MR) receptors is proposed to underlie the HPA axis dysregulation that associates with susceptibility to psychopathology (anxiety, PTSD). To test this 'balance hypothesis' we examined whether the impact of MR levels upon HPA-axis control and behaviour depended on the relative levels of GR and vice versa. Avoiding antenatal maternal 'programming' effects by using littermates, we generated mice with forebrain MR over-expression (MR(hi)) and/or simultaneous global GR under-expression (GR(lo)). We found a significant interaction between MR and GR in control of the HPA-axis under stressed but not basal conditions. With reduced GR levels, HPA-axis activity in response to restraint stress was enhanced, likely due to impaired negative feedback. However, high MR in concert with reduced GR minimised this HPA-axis overshoot in response to stress. MR:GR balance also played a role in determining strategies of spatial memory during a watermaze probe trial: when coupled with GR under-expression, MR(hi) show enhanced perseveration, suggesting enhanced spatial recall or reduced exploratory flexibility. Other alterations in cognitive functions were specific to a single receptor without interaction, with both MR(hi) and GR(lo) manipulations independently impairing reversal learning in spatial and fear memory tasks. Thus, MR and GR interact in specific domains of neuroendocrine and cognitive control, but for other limbic-associated behaviours each receptor mediates its own repertoire of responses. Since modulation of HPA-axis and behavioural dysfunction associated with high levels of MR, selective ligands or transcriptional regulators may afford novel therapeutic approaches to affective psychopathologies.


Assuntos
Comportamento Animal/fisiologia , Sistema Hipotálamo-Hipofisário/metabolismo , Sistema Hipófise-Suprarrenal/metabolismo , Receptores de Glucocorticoides/genética , Receptores de Mineralocorticoides/genética , Animais , Cognição/fisiologia , Regulação da Expressão Gênica/fisiologia , Sistema Hipotálamo-Hipofisário/fisiologia , Sistema Hipotálamo-Hipofisário/fisiopatologia , Masculino , Memória/fisiologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Sistema Hipófise-Suprarrenal/fisiopatologia , Receptores de Glucocorticoides/metabolismo , Receptores de Mineralocorticoides/metabolismo , Restrição Física/fisiologia
13.
Placenta ; 33(3): 220-3, 2012 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-22226642

RESUMO

There is increasing evidence for a role for epigenetic modifications in early life 'programming' effects. Altered placental methyl donor transport may impact on the establishment of epigenetic marks in the fetus. This study investigated the effects of prenatal glucocorticoid overexposure on placental methyl donor transport. Glucocorticoids increased folate but decreased choline transport and reduced fetal plasma methionine levels. There was no change in global DNA methylation in fetal liver. These data suggest prenatal glucocorticoid overexposure causes complex alterations in the placental transport of key methyl donors which may have important implications for maternal diet and nutrient supplementation in pregnancy.


Assuntos
Dexametasona/farmacologia , Desenvolvimento Embrionário/efeitos dos fármacos , Metano/análogos & derivados , Placenta/efeitos dos fármacos , Animais , Transporte Biológico/efeitos dos fármacos , Transporte Biológico/genética , Colina/farmacocinética , Metilação de DNA/efeitos dos fármacos , Metilação de DNA/fisiologia , Dexametasona/efeitos adversos , Desenvolvimento Embrionário/genética , Epistasia Genética/efeitos dos fármacos , Epistasia Genética/genética , Feminino , Ácido Fólico/farmacocinética , Metano/metabolismo , Metionina/farmacocinética , Placenta/metabolismo , Gravidez , Efeitos Tardios da Exposição Pré-Natal/induzido quimicamente , Efeitos Tardios da Exposição Pré-Natal/genética , Efeitos Tardios da Exposição Pré-Natal/metabolismo , Ratos , Ratos Wistar
14.
Stress ; 14(6): 590-603, 2011 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-21875300

RESUMO

A suboptimal prenatal environment may induce permanent changes in cells, organs and physiology that alter social, emotional and cognitive functioning, and increase the risk of cardiometabolic and mental disorders in subsequent life ("developmental programming"). Although animal studies have provided a wealth of data on programming and its mechanisms, including on the role of stress and its glucocorticoid mediators, empirical evidence of these mechanisms in humans is still scanty. We review the existing human evidence on the effects of prenatal maternal stress, anxiety and depression, glucocorticoids and intake of liquorice (which inhibits the placental barrier to maternal glucocorticoids) on offspring developmental outcomes including, for instance, alterations in psychophysiological and neurocognitive functioning and mental health. This work lays the foundations for biomarker discovery and affords opportunities for prevention and interventions to ameliorate adverse outcomes in humans.


Assuntos
Ansiedade/psicologia , Depressão/complicações , Glucocorticoides/efeitos adversos , Glycyrrhiza/efeitos adversos , Complicações na Gravidez/psicologia , Efeitos Tardios da Exposição Pré-Natal , Estresse Psicológico/complicações , Criança , Pré-Escolar , Feminino , Glucocorticoides/fisiologia , Humanos , Sistema Hipotálamo-Hipofisário/efeitos dos fármacos , Sistema Hipotálamo-Hipofisário/fisiologia , Recém-Nascido de Baixo Peso , Recém-Nascido , Sistema Hipófise-Suprarrenal/efeitos dos fármacos , Sistema Hipófise-Suprarrenal/fisiologia , Gravidez
15.
Diabetologia ; 54(9): 2392-403, 2011 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-21667214

RESUMO

AIMS/HYPOTHESIS: Urocortins are the endogenous ligands for the corticotropin-releasing factor receptor type 2 (CRFR2), which is implicated in regulating energy balance and/or glucose metabolism. We determined the effects of chronic CRFR2 activation on metabolism in vivo, by generating and phenotyping transgenic mice overproducing the specific CRFR2 ligand urocortin 3. METHODS: Body composition, glucose metabolism, insulin sensitivity, energy efficiency and expression of key metabolic genes were assessed in adult male urocortin 3 transgenic mice (Ucn3(+)) under control conditions and following an obesogenic high-fat diet (HFD) challenge. RESULTS: Ucn3(+) mice had increased skeletal muscle mass with myocyte hypertrophy. Accelerated peripheral glucose disposal, increased respiratory exchange ratio and hypoglycaemia on fasting demonstrated increased carbohydrate metabolism. Insulin tolerance and indices of insulin-stimulated signalling were unchanged, indicating these effects were not mediated by increased insulin sensitivity. Expression of the transgene in Crfr2 (also known as Crhr2)-null mice negated key aspects of the Ucn3(+) phenotype. Ucn3(+) mice were protected from the HFD-induced hyperglycaemia and increased adiposity seen in control mice despite consuming more energy. Expression of uncoupling proteins 2 and 3 was higher in Ucn3(+) muscle, suggesting increased catabolic processes. IGF-1 abundance was upregulated in Ucn3(+) muscle, providing a potential paracrine mechanism in which urocortin 3 acts upon CRFR2 to link the altered metabolism and muscular hypertrophy observed. CONCLUSIONS/INTERPRETATION: Urocortin 3 acting on CRFR2 in skeletal muscle of Ucn3(+) mice results in a novel metabolically favourable phenotype, with lean body composition and protection against diet-induced obesity and hyperglycaemia. Urocortins and CRFR2 may be of interest as potential therapeutic targets for obesity.


Assuntos
Gorduras na Dieta/efeitos adversos , Hiperglicemia/metabolismo , Hiperglicemia/prevenção & controle , Obesidade/metabolismo , Obesidade/prevenção & controle , Urocortinas/genética , Urocortinas/metabolismo , Animais , Composição Corporal/efeitos dos fármacos , Composição Corporal/fisiologia , Gorduras na Dieta/farmacologia , Modelos Animais de Doenças , Metabolismo Energético/efeitos dos fármacos , Metabolismo Energético/fisiologia , Glucose/metabolismo , Insulina/sangue , Fator de Crescimento Insulin-Like I/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Camundongos Knockout , Camundongos Transgênicos , Músculo Esquelético/metabolismo , Músculo Esquelético/patologia , Fenótipo , Receptores de Hormônio Liberador da Corticotropina/deficiência , Receptores de Hormônio Liberador da Corticotropina/genética , Receptores de Hormônio Liberador da Corticotropina/metabolismo
16.
Horm Metab Res ; 42(11): 821-5, 2010 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-20711951

RESUMO

Peroxisomal proliferator-activated receptors (PPARs) play an important role in the regulation of lipid metabolism. The aim of this study was to investigate the effects of a maternal high-fat (HF) diet on serum lipid concentration and PPAR gene expression in liver and adipose tissue in the early life of the rat offspring. Female Sprague-Dawley rats were fed either an HF or control (CON) diet 6 weeks before mating and throughout gestation and lactation. Blood and tissue samplings of male offspring were carried out at birth or weaning. Birth weights were similar and serum triglyceride (TG) and nonesterified fatty acid (NEFA) levels showed no significant difference between HF and CON newborns, despite greatly increased hepatic PPARα mRNA expression in the HF newborns (p<0.05). Both HF newborns and weanlings revealed significantly decreased hepatic PPARγ expression compared with controls (p<0.0001). Hepatic PPARα expression in the HF weanlings was reduced markedly compared with CON weanlings (p<0.0001) and showed a negative correlation with serum TG levels (r=-0.743, p<0.05). However, epididymal expression of PPARγ in the HF weanlings was upregulated significantly compared with controls (p<0.05) and demonstrated a positive correlation with epididymal fat mass (r=0.733, p<0.05). These were accompanied by obesity as well as a rise in serum TG by 79% (p<0.05) and NEFA concentration by 36% (p<0.05) in these HF weanlings. Our findings suggest that maternal HF diet leads to alterations in PPAR gene expression in the weanling offspring, which is associated with the disturbed lipid homeostasis.


Assuntos
Gorduras na Dieta/farmacologia , Lipídeos/sangue , Receptores Ativados por Proliferador de Peroxissomo/metabolismo , Animais , Animais Recém-Nascidos , Peso Corporal/efeitos dos fármacos , Feminino , Ratos , Ratos Sprague-Dawley
17.
Lab Anim ; 44(3): 199-205, 2010 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-20071412

RESUMO

Low birth weight in humans is predictive of hypertension in adult life, and while the mechanisms underlying this link remain unknown, fetal overexposure to glucocorticoids has been implicated. We have previously shown that prenatal dexamethasone (DEX) exposure in the rat lowers birth weight and programmes adult hypertension. This current study aimed to unravel the molecular nature of this hypertension. However, unknowingly, post hoc investigations revealed that our animals had been subjected to environmental noise stresses from an adjacent construction site, which were sufficient to confound our prenatal DEX-programming experiments. This perinatal stress successfully established low birth weight, hypercorticosteronaemia, insulin resistance, hypertension and hypothalamic-pituitary-adrenal axis dysfunction in vehicle (VEH)-treated offspring, such that the typical distinctions between both treatment groups were ameliorated. The lack of an additional effect on DEX-treated offspring is suggestive of a maximal effect of perinatal stress and glucocorticoids, serving to prevent against the potentially detrimental effects of sustained glucocorticoid hyper-exposure. Finally, this paper serves to inform researchers of the potential detrimental effects of neighbouring construction sites to their experiments.


Assuntos
Dexametasona/toxicidade , Meio Ambiente , Glucocorticoides/toxicidade , Hipertensão/etiologia , Ruído/efeitos adversos , Efeitos Tardios da Exposição Pré-Natal/etiologia , Animais , Animais Recém-Nascidos , Peso ao Nascer/efeitos dos fármacos , Peso ao Nascer/fisiologia , Corticosterona/sangue , Feminino , Teste de Tolerância a Glucose , Sistema Hipotálamo-Hipofisário/efeitos dos fármacos , Sistema Hipotálamo-Hipofisário/fisiopatologia , Resistência à Insulina , Masculino , Exposição Materna , Sistema Hipófise-Suprarrenal/efeitos dos fármacos , Sistema Hipófise-Suprarrenal/fisiopatologia , Gravidez , Efeitos Tardios da Exposição Pré-Natal/fisiopatologia , Ratos , Ratos Wistar , Estresse Fisiológico
18.
Diabetologia ; 53(3): 467-71, 2010 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-20012009

RESUMO

AIMS/HYPOTHESIS: The aim of the study was to identify risk factors for depression and anxiety in a well-characterised cohort of individuals with type 2 diabetes mellitus. METHODS: We used baseline data from participants (n = 1,066, 48.7% women, aged 67.9 +/- 4.2 years) from the Edinburgh Type 2 Diabetes Study. Symptoms of anxiety and depression were assessed using the Hospital Anxiety and Depression Scale (HADS). Obesity was characterised according to both overall (body mass index, fat mass) and abdominal (waist circumference) measurements. Cardiovascular disease was assessed by questionnaire, physical examination and review of medical records. Stepwise multiple linear regression was performed to identify explanatory variables related to either anxiety or depression HADS scores. RESULTS: Abdominal obesity (waist circumference) and cardiovascular disease (ischaemic heart disease and ankle-brachial pressure index) were related to depression but not anxiety. Lifetime history of severe hypoglycaemia was associated with anxiety. Other cardiovascular risk factors or microvascular complications were not related to either anxiety or depressive symptoms. CONCLUSIONS/INTERPRETATION: Depression but not anxiety is associated with abdominal obesity and cardiovascular disease in people with type 2 diabetes mellitus. This knowledge may help to identify depressive symptoms among patients with type 2 diabetes who are at greatest risk.


Assuntos
Ansiedade/complicações , Depressão/complicações , Complicações do Diabetes/diagnóstico , Diabetes Mellitus Tipo 2/patologia , Obesidade Abdominal/complicações , Idoso , Ansiedade/diagnóstico , Índice de Massa Corporal , Depressão/diagnóstico , Feminino , Humanos , Masculino , Pessoa de Meia-Idade , Obesidade Abdominal/diagnóstico , Análise de Regressão , Fatores de Risco , Inquéritos e Questionários
19.
J Neuroendocrinol ; 21(11): 879-87, 2009 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-19602102

RESUMO

Inter-individual differences in hypothalamic-pituitary-adrenal (HPA) axis activity underlie differential vulnerability to neuropsychiatric and metabolic disorders, although the basis of this variation is poorly understood. 11beta-Hydroxysteroid dehydrogenase type 1 (11beta-HSD1) has previously been shown to influence HPA axis activity. 129/MF1 mice null for 11beta-HSD1 (129/MF1 HSD1(-/-)) have greatly increased adrenal gland size and altered HPA activity, consistent with reduced glucocorticoid negative feedback. On this background, concentrations of plasma corticosterone and adrenocorticotrophic hormone (ACTH) were elevated in unstressed mice, and showed a delayed return to baseline after stress in HSD1-null mice with reduced sensitivity to exogenous glucocorticoid feedback compared to same-background genetic controls. In the present study, we report that the genetic background can dramatically alter this pattern. By contrast to HSD1(-/-) mice on a 129/MF1 background, HSD1(-/-) mice congenic on a C57Bl/6J background have normal basal plasma corticosterone and ACTH concentrations and exhibit normal return to baseline of plasma corticosterone and ACTH concentrations after stress. Furthermore, in contrast to 129/MF1 HSD1(-/-) mice, C57Bl/6J HSD1(-/-) mice have increased glucocorticoid receptor expression in areas of the brain involved in glucocorticoid negative feedback (hippocampus and paraventricular nucleus), suggesting this may be a compensatory response to normalise feedback control of the HPA axis. In support of this hypothesis, C57Bl/6J HSD1(-/-) mice show increased sensitivity to dexamethasone-mediated suppression of peak corticosterone. Thus, although 11beta-HSD1 appears to contribute to regulation of the HPA axis, the genetic background is crucial in governing the response to (and hence the consequences of) its loss. Similar variations in plasticity may underpin inter-individual differences in vulnerability to disorders associated with HPA axis dysregulation. They also indicate that 11beta-HSD1 inhibition does not inevitably activate the HPA axis.


Assuntos
11-beta-Hidroxiesteroide Desidrogenase Tipo 1/genética , Sistema Hipotálamo-Hipofisário , Sistema Hipófise-Suprarrenal , 11-beta-Hidroxiesteroide Desidrogenase Tipo 1/metabolismo , Glândulas Suprarrenais/patologia , Hormônio Adrenocorticotrópico/sangue , Animais , Sequência de Bases , Ritmo Circadiano , Corticosterona/sangue , Primers do DNA , Hibridização In Situ , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Tamanho do Órgão , Reação em Cadeia da Polimerase
20.
Neuropathol Appl Neurobiol ; 35(2): 156-64, 2009 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-19284477

RESUMO

BACKGROUND: We have previously reported that neuronal endangerment in vitro and hypothermic transient global ischaemia in vivo each result in increased mineralocorticoid receptor (MR) expression. In both models MR induction is associated with increased neuronal survival, and blocking MR signalling reduces neuronal survival. Furthermore, transgenic overexpression of human MR promotes neuronal survival both in vitro and in vivo. AIMS: Here we have assessed whether brief periods of cerebral ischaemia in human subjects, such as occurs in cardiac arrest from which successful resuscitation is achieved, are associated with a sustained increase in hippocampal MR mRNA expression. METHODS: Human post-mortem brain sections from patients who had died in the weeks following cardiac arrest were analysed for MR mRNA expression by in situ hybridization. RESULTS: Sustained upregulation of MR mRNA expression was observed in the dentate gyrus region of human hippocampus following a brief episode of cerebral ischaemia. CONCLUSIONS: This confirms that MR mRNA expression is regulated following neuronal injury in human brain, and suggests that the benefits of increased MR expression seen in animal models of ischaemia may also be observed in humans.


Assuntos
Isquemia Encefálica/metabolismo , Hipocampo/metabolismo , Receptores de Mineralocorticoides/metabolismo , Adulto , Idoso , Feminino , Parada Cardíaca/mortalidade , Parada Cardíaca/patologia , Humanos , Hibridização In Situ , Masculino , Pessoa de Meia-Idade , Fotomicrografia , Proteínas Proto-Oncogênicas c-bcl-2/metabolismo , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Receptores de Mineralocorticoides/genética , Regulação para Cima , Proteína X Associada a bcl-2/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA