Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 242
Filtrar
Mais filtros

Base de dados
País/Região como assunto
Tipo de documento
Intervalo de ano de publicação
1.
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
2.
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
3.
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
4.
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
5.
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
6.
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
7.
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
8.
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
9.
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
10.
Eur J Neurosci ; 29(1): 97-103, 2009 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-19032587

RESUMO

Prenatal stress (PS) produces persistent abnormalities in anxiety-related behaviors, stress responsivity, susceptibility to psychopathology and hippocampal changes in adult offspring. The hippocampus shows a remarkable degree of structural plasticity, notably in response to stress and glucocorticoids. We hypothesized that PS would differentially affect hippocampal neurogenesis in rats selectively bred for genetic differences in anxiety-related behaviors and stress responsivity. Pregnant dams of high anxiety-related behavior (HAB) and low anxiety-related behavior (LAB) strains were stressed between days 5 and 20 of pregnancy. The survival of newly generated hippocampal cells was found to be significantly lower in 43-day-old HAB than in LAB male offspring of unstressed pregnancies. PS further reduced newly generated cell numbers only in HAB rats, and this was paralleled by a reduction in doublecortin-positive cell numbers, indicative of reduced neurogenesis. As maternal plasma corticosterone levels during PS were similar in both strains, we examined placental 11beta-hydroxysteroid dehydrogenase type 2 (11beta-HSD2), which catalyses rapid inactivation of maternal corticosterone to inert 11-dehydrocorticosterone and thus serves as a physiological 'barrier' to maternal glucocorticoids. PS significantly increased placental 11beta-HSD2 activity in LAB, but not HAB, rats. We conclude that PS differentially affects the number of surviving newly generated cells and neurogenesis in HAB and LAB rats. The high sensitivity of hippocampal neurogenesis to PS in HAB rats is paralleled by a failure to increase placental 11beta-HSD2 activity after stress rather than by different maternal corticosterone responses. Hence, stress-induced placental 11beta-HSD2 expression may be critical in protecting the fetal brain from maternal stress-induced effects on adult neurogenesis.


Assuntos
11-beta-Hidroxiesteroide Desidrogenase Tipo 2/metabolismo , Transtornos de Ansiedade/enzimologia , Predisposição Genética para Doença/genética , Neurogênese/genética , Efeitos Tardios da Exposição Pré-Natal/enzimologia , Estresse Psicológico/enzimologia , Animais , Animais Recém-Nascidos , Transtornos de Ansiedade/genética , Transtornos de Ansiedade/fisiopatologia , Contagem de Células , Proliferação de Células , Sobrevivência Celular/genética , Corticosterona/sangue , Corticosterona/metabolismo , Modelos Animais de Doenças , Proteínas do Domínio Duplacortina , Proteína Duplacortina , Feminino , Masculino , Troca Materno-Fetal/fisiologia , Proteínas Associadas aos Microtúbulos/análise , Proteínas Associadas aos Microtúbulos/metabolismo , Neuropeptídeos/análise , Neuropeptídeos/metabolismo , Placenta/enzimologia , Placenta/metabolismo , Gravidez , Efeitos Tardios da Exposição Pré-Natal/genética , Efeitos Tardios da Exposição Pré-Natal/fisiopatologia , Ratos , Ratos Wistar , Estresse Psicológico/genética , Estresse Psicológico/fisiopatologia
11.
Clin Exp Allergy ; 39(5): 731-9, 2009 May.
Artigo em Inglês | MEDLINE | ID: mdl-19178536

RESUMO

BACKGROUND: Dietary fat intake has been associated with obesity and obesity in its turn with attenuated airway function and asthma, but it is unclear whether or how high-fat intake per se alters immune function relevant to development of allergic asthma. OBJECTIVE: To use a non-obese mouse model of mild to moderate allergic asthma to compare effects of high-fat with isocaloric control-diet on allergic immune responses. METHODS: C57BL/6 mice weaned and maintained on control (11% fat calories) or isocaloric high-fat diet (58% fat calories) were systemically sensitized with ovalbumin and challenged in the lungs. Allergic airway inflammation was assessed by measuring lung inflammation; serum antibodies; and, cytokines in serum, bronchoalveolar lavage (BAL) fluid and in supernatants of in vitro stimulated lung draining lymph node and spleen lymphocytes. RESULTS: There was a significant reduction in lung eosinophilia and IL-5 in high-fat fed mice. Lung draining lymph node cells from these mice showed reduced pro-inflammatory cytokine (MCP-1 and TNF-alpha) release after ovalbumin re-stimulation and reduced release of IL-13 after concanavalin-A stimulation, indicating a general rather than just an antigen-specific change. There was no difference in IFN-gamma release. In contrast, pro-inflammatory cytokine release was increased from splenocytes. Decreased eosinophilia was not due to increased regulatory T cell or IL-10 induction in draining lymph nodes or spleen, nor to changes in antibody response to ovalbumin. However, decreased levels of serum and BAL eotaxin were found in high-fat fed animals. CONCLUSIONS: The data indicate that high-fat dietary content redirects local immune responses to allergen in the lungs and systemic responses in the spleen and serum. These effects are not due to changes in regulatory T cell populations but may reflect a failure to mobilize eosinophils in response to allergic challenge.


Assuntos
Asma/imunologia , Citocinas/imunologia , Gorduras na Dieta/efeitos adversos , Pneumonia/imunologia , Eosinofilia Pulmonar/imunologia , Animais , Asma/etiologia , Líquido da Lavagem Broncoalveolar/imunologia , Células Cultivadas , Concanavalina A/farmacologia , Gorduras na Dieta/imunologia , Modelos Animais de Doenças , Pulmão/efeitos dos fármacos , Pulmão/imunologia , Pulmão/patologia , Linfonodos/efeitos dos fármacos , Linfonodos/imunologia , Linfonodos/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Endogâmicos NOD , Ovalbumina/imunologia , Pneumonia/etiologia , Eosinofilia Pulmonar/etiologia , Linfócitos T Reguladores/efeitos dos fármacos , Linfócitos T Reguladores/imunologia , Linfócitos T Reguladores/metabolismo
12.
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
13.
FASEB J ; 22(11): 3896-907, 2008 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-18697839

RESUMO

Glucocorticoid hormones are critical to respond and adapt to stress. Genetic variations in the glucocorticoid receptor (GR) gene alter hypothalamic-pituitary-adrenal (HPA) axis activity and associate with hypertension and susceptibility to metabolic disease. Here we test the hypothesis that reduced GR density alters blood pressure and glucose and lipid homeostasis and limits adaption to obesogenic diet. Heterozygous GR(betageo/+) mice were generated from embryonic stem (ES) cells with a gene trap integration of a beta-galactosidase-neomycin phosphotransferase (betageo) cassette into the GR gene creating a transcriptionally inactive GR fusion protein. Although GR(betageo/+) mice have 50% less functional GR, they have normal lipid and glucose homeostasis due to compensatory HPA axis activation but are hypertensive due to activation of the renin-angiotensin-aldosterone system (RAAS). When challenged with a high-fat diet, weight gain, adiposity, and glucose intolerance were similarly increased in control and GR(betageo/+) mice, suggesting preserved control of intermediary metabolism and energy balance. However, whereas a high-fat diet caused HPA activation and increased blood pressure in control mice, these adaptions were attenuated or abolished in GR(betageo/+) mice. Thus, reduced GR density balanced by HPA activation leaves glucocorticoid functions unaffected but mineralocorticoid functions increased, causing hypertension. Importantly, reduced GR limits HPA and blood pressure adaptions to obesogenic diet.


Assuntos
Pressão Sanguínea/efeitos dos fármacos , Dieta/efeitos adversos , Gorduras na Dieta/efeitos adversos , Hipertensão/metabolismo , Sistema Hipotálamo-Hipofisário/metabolismo , Sistema Hipófise-Suprarrenal/metabolismo , Receptores de Glucocorticoides/metabolismo , Adiposidade/efeitos dos fármacos , Adiposidade/genética , Aldosterona/metabolismo , Angiotensinas/metabolismo , Animais , Glicemia/metabolismo , Linhagem Celular , Gorduras na Dieta/farmacologia , Intolerância à Glucose/genética , Intolerância à Glucose/metabolismo , Humanos , Hipertensão/induzido quimicamente , Hipertensão/genética , Metabolismo dos Lipídeos/genética , Camundongos , Camundongos Transgênicos , Receptores de Glucocorticoides/genética , Renina/metabolismo , Aumento de Peso/efeitos dos fármacos , Aumento de Peso/genética
14.
Science ; 294(5549): 2166-70, 2001 Dec 07.
Artigo em Inglês | MEDLINE | ID: mdl-11739957

RESUMO

The adverse metabolic consequences of obesity are best predicted by the quantity of visceral fat. Excess glucocorticoids produce visceral obesity and diabetes, but circulating glucocorticoid levels are normal in typical obesity. Glucocorticoids can be produced locally from inactive 11-keto forms through the enzyme 11beta hydroxysteroid dehydrogenase type 1 (11beta HSD-1). We created transgenic mice overexpressing 11beta HSD-1 selectively in adipose tissue to an extent similar to that found in adipose tissue from obese humans. These mice had increased adipose levels of corticosterone and developed visceral obesity that was exaggerated by a high-fat diet. The mice also exhibited pronounced insulin-resistant diabetes, hyperlipidemia, and, surprisingly, hyperphagia despite hyperleptinemia. Increased adipocyte 11beta HSD-1 activity may be a common molecular etiology for visceral obesity and the metabolic syndrome.


Assuntos
Tecido Adiposo/enzimologia , Modelos Animais de Doenças , Hidroxiesteroide Desidrogenases/metabolismo , Síndrome Metabólica , Obesidade/enzimologia , Obesidade/genética , 11-beta-Hidroxiesteroide Desidrogenase Tipo 1 , Abdome , Adipócitos/citologia , Adipócitos/patologia , Tecido Adiposo/metabolismo , Animais , Composição Corporal , Tamanho Celular , Corticosterona/sangue , Corticosterona/metabolismo , Gorduras na Dieta/administração & dosagem , Ingestão de Alimentos , Marcação de Genes , Humanos , Hidroxiesteroide Desidrogenases/genética , Hiperglicemia/metabolismo , Hiperinsulinismo/metabolismo , Resistência à Insulina , Leptina/metabolismo , Metabolismo dos Lipídeos , Lipídeos/sangue , Lipase Lipoproteica/genética , Lipase Lipoproteica/metabolismo , Masculino , Camundongos , Camundongos Transgênicos , Receptores de Glucocorticoides/genética , Receptores de Glucocorticoides/metabolismo , Vísceras , Aumento de Peso
15.
Endocrinology ; 149(4): 1861-8, 2008 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-18174284

RESUMO

Glucocorticoid excess promotes visceral obesity and cardiovascular disease. Similar features are found in the highly prevalent metabolic syndrome in the absence of high levels of systemic cortisol. Although elevated activity of the glucocorticoid-amplifying enzyme 11beta-hydroxysteroid dehydrogenase type 1 (11beta-HSD1) within adipocytes might explain this paradox, the potential role of 11beta-HSD1 in preadipocytes is less clear; human omental adipose stromal vascular (ASV) cells exhibit 11beta-dehydrogenase activity (inactivation of glucocorticoids) probably due to the absence of cofactor provision by hexose-6-phosphate dehydrogenase. To clarify the depot-specific impact of 11beta-HSD1, we assessed whether preadipocytes in ASV from mesenteric (as a representative of visceral adipose tissue) and sc tissue displayed 11beta-HSD1 activity in mice. 11beta-HSD1 was highly expressed in freshly isolated ASV cells, predominantly in preadipocytes. 11beta-HSD1 mRNA and protein levels were comparable between ASV and adipocyte fractions in both depots. 11beta-HSD1 was an 11beta-reductase, thus reactivating glucocorticoids in ASV cells, consistent with hexose-6-phosphate dehydrogenase mRNA expression. Unexpectedly, glucocorticoid reactivation was higher in intact mesenteric ASV cells despite a lower expression of 11beta-HSD1 mRNA and protein (homogenate activity) levels than sc ASV cells. This suggests a novel depot-specific control over 11beta-HSD1 enzyme activity. In vivo, high-fat diet-induced obesity was accompanied by increased visceral fat preadipocyte differentiation in wild-type but not 11beta-HSD1(-/-) mice. The results suggest that 11beta-HSD1 reductase activity is augmented in mouse mesenteric preadipocytes where it promotes preadipocyte differentiation and contributes to visceral fat accumulation in obesity.


Assuntos
11-beta-Hidroxiesteroide Desidrogenase Tipo 1/fisiologia , Adipócitos/enzimologia , Gordura Intra-Abdominal/metabolismo , Obesidade/etiologia , Células-Tronco/enzimologia , 11-beta-Hidroxiesteroide Desidrogenase Tipo 1/análise , 11-beta-Hidroxiesteroide Desidrogenase Tipo 1/genética , Animais , Inibidor de Quinase Dependente de Ciclina p27/análise , Gorduras na Dieta/administração & dosagem , Masculino , Camundongos , Camundongos Endogâmicos C57BL , RNA Mensageiro/análise
16.
J Clin Invest ; 103(5): 683-9, 1999 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-10074485

RESUMO

Deficiency of 11beta-hydroxysteroid dehydrogenase type 2 (11beta-HSD2) in humans leads to the syndrome of apparent mineralocorticoid excess (SAME), in which cortisol illicitly occupies mineralocorticoid receptors, causing sodium retention, hypokalemia, and hypertension. However, the disorder is usually incompletely corrected by suppression of cortisol, suggesting additional and irreversible changes, perhaps in the kidney. To examine this further, we produced mice with targeted disruption of the 11beta-HSD2 gene. Homozygous mutant mice (11beta-HSD2(-/-)) appear normal at birth, but approximately 50% show motor weakness and die within 48 hours. Both male and female survivors are fertile but exhibit hypokalemia, hypotonic polyuria, and apparent mineralocorticoid activity of corticosterone. Young adult 11beta-HSD2(-/-) mice are markedly hypertensive, with a mean arterial blood pressure of 146 +/- 2 mmHg, compared with 121 +/- 2 mmHg in wild-type controls and 114 +/- 4 mmHg in heterozygotes. The epithelium of the distal tubule of the nephron shows striking hypertrophy and hyperplasia. These histological changes do not readily reverse with mineralocorticoid receptor antagonism in adulthood. Thus, 11beta-HSD2(-/-) mice demonstrate the major features of SAME, providing a unique rodent model to study the molecular mechanisms of kidney resetting leading to hypertension.


Assuntos
Hidroxiesteroide Desidrogenases/genética , Hipertensão/genética , Camundongos Knockout/fisiologia , 11-beta-Hidroxiesteroide Desidrogenases , Animais , Corticosterona/metabolismo , Feminino , Hidroxiesteroide Desidrogenases/deficiência , Hipertensão/enzimologia , Masculino , Camundongos
17.
J Clin Invest ; 101(10): 2174-81, 1998 May 15.
Artigo em Inglês | MEDLINE | ID: mdl-9593773

RESUMO

Low birth weight in humans is predictive of insulin resistance and diabetes in adult life. The molecular mechanisms underlying this link are unknown but fetal exposure to excess glucocorticoids has been implicated. The fetus is normally protected from the higher maternal levels of glucocorticoids by feto-placental 11beta-hydroxysteroid dehydrogenase type-2 (11beta-HSD2) which inactivates glucocorticoids. We have shown previously that inhibiting 11beta-HSD2 throughout pregnancy in rats reduces birth weight and causes hyperglycemia in the adult offspring. We now show that dexamethasone (a poor substrate for 11beta-HSD2) administered to pregnant rats selectively in the last week of pregnancy reduces birth weight by 10% (P < 0.05), and produces adult fasting hyperglycemia (treated 5.3+/-0.3; control 4.3+/-0.2 mmol/ liter, P = 0.04), reactive hyperglycemia (treated 8.7+/-0.4; control 7.5+/-0.2 mmol/liter, P = 0.03), and hyperinsulinemia (treated 6.1+/-0.4; control 3.8+/-0.5 ng/ml, P = 0.01) on oral glucose loading. In the adult offspring of rats exposed to dexamethasone in late pregnancy, hepatic expression of glucocorticoid receptor (GR) mRNA and phosphoenolpyruvate carboxykinase (PEPCK) mRNA (and activity) are increased by 25% (P = 0.01) and 60% (P < 0.01), respectively, while other liver enzymes (glucose-6-phosphatase, glucokinase, and 11beta-hydroxysteroid dehydrogenase type-1) are unaltered. In contrast dexamethasone, when given in the first or second week of gestation, has no effect on offspring insulin/glucose responses or hepatic PEPCK and GR expression. The increased hepatic GR expression may be crucial, since rats exposed to dexamethasone in utero showed potentiated glucose responses to exogenous corticosterone. These observations suggest that excessive glucocorticoid exposure late in pregnancy predisposes the offspring to glucose intolerance in adulthood. Programmed hepatic PEPCK overexpression, perhaps mediated by increased GR, may promote this process by increasing gluconeogenesis.


Assuntos
Regulação Enzimológica da Expressão Gênica/genética , Glucocorticoides/farmacologia , Intolerância à Glucose/fisiopatologia , Fígado/enzimologia , Receptores de Glucocorticoides/genética , 11-beta-Hidroxiesteroide Desidrogenases , Animais , Peso ao Nascer/efeitos dos fármacos , Glicemia/metabolismo , Peso Corporal/efeitos dos fármacos , Corticosterona/farmacologia , Dexametasona/farmacologia , Feminino , Hidroxiesteroide Desidrogenases/genética , Insulina/sangue , Fosfoenolpiruvato Carboxiquinase (GTP) , Gravidez , Ligação Proteica/efeitos dos fármacos , RNA Mensageiro/metabolismo , Ratos , Ratos Wistar
18.
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
19.
Neuroscience ; 137(3): 865-73, 2006 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-16289840

RESUMO

11beta-Hydroxysteroid dehydrogenase type 2 is a glucocorticoid metabolizing enzyme that catalyzes rapid inactivation of corticosterone and cortisol to inert 11-keto derivatives. As 11beta-hydroxysteroid dehydrogenase type 2 is highly expressed in the developing brain, but not in the adult CNS, we hypothesized that it may represent a protective barrier to the deleterious actions of corticosteroids on proliferating cells. To test this hypothesis we have investigated the development and growth of the cerebellum in neonatal C57BL/6 mice and mice lacking 11beta-hydroxysteroid dehydrogenase type 2 (-/-). 11beta-Hydroxysteroid dehydrogenase type 2-/- mice had consistently lower body weight throughout the neonatal period, coupled with a smaller brain size although this was normalized when corrected for body weight. The cerebellar size was smaller in 11beta-hydroxysteroid dehydrogenase type 2-/- mice, due to decreases in size of both the molecular and internal granule layers. When exogenous corticosterone was administered to the pups between postnatal days 4 and 13, 11beta-hydroxysteroid dehydrogenase type 2(-/-) mice were more sensitive, showing further inhibition of cerebellar growth while the wildtype mice were not affected. Upon withdrawal of exogenous steroid, there was a rebound growth spurt so that at day 21 postnatally, the cerebellar size in 11beta-hydroxysteroid dehydrogenase type 2-/- mice was similar to untreated mice of the same genotype. Furthermore, 11beta-hydroxysteroid dehydrogenase type 2-/- mice had a delay in the attainment of neurodevelopmental landmarks such as negative geotaxis and eye opening. We therefore suggest that 11beta-hydroxysteroid dehydrogenase type 2 acts as to protect the developing nervous system from the deleterious consequences of glucocorticoid overexposure.


Assuntos
11-beta-Hidroxiesteroide Desidrogenase Tipo 2/fisiologia , Animais Recém-Nascidos/fisiologia , Cerebelo/fisiologia , Glucocorticoides/fisiologia , 11-beta-Hidroxiesteroide Desidrogenase Tipo 2/biossíntese , 11-beta-Hidroxiesteroide Desidrogenase Tipo 2/genética , Animais , Peso Corporal/fisiologia , Encéfalo/enzimologia , Encéfalo/crescimento & desenvolvimento , Proliferação de Células , Cerebelo/crescimento & desenvolvimento , Cerebelo/patologia , Corticosterona/sangue , Feminino , Proteína Glial Fibrilar Ácida/biossíntese , Imuno-Histoquímica , Hibridização In Situ , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Tamanho do Órgão/fisiologia , Equilíbrio Postural/fisiologia , Reflexo/fisiologia
20.
J Endocrinol ; 188(3): 435-42, 2006 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-16522724

RESUMO

Excessive exposure to glucocorticoids during gestation reduces birth weight and induces permanent hypertension in adulthood. The mechanisms underlying this programmed elevation of blood pressure have not been established. We hypothesised that prenatal glucocorticoid exposure may lead to vascular dysfunction in adulthood. Pregnant rats received dexamethasone (Dex) (100 microg/kg, s.c.) or vehicle (control) daily throughout pregnancy. Blood pressure was elevated (students t-test, unpaired; P < 0.05) in adult female offspring (aged 12-16 weeks) of Dex-treated mothers (148.0 +/- 3.6 mmHg, n=10) compared with the control group (138.0 +/- 2.5 mmHg, n=8). Vascular responsiveness in aortae and mesenteric arteries was differentially affected by prenatal Dex: aortae were less responsive to angiotensin II, whereas mesenteric arteries were more responsive to norepinephrine, vasopressin and potassium (mesenteric arteries respond poorly to angiotensin II in vitro). Acetylcholine-mediated, endothelium-dependent relaxation was similar in both groups. Prenatal exposure to Dex had no effect on blood pressure or aldosterone response to acute (15 min, i.v.) infusion of angiotensin II (75 ng/kg per min). In contrast, chronic (2-week, s.c.) infusion of angiotensin II (100 ng/kg per min) produced a greater elevation (P < 0.05) of blood pressure in Dex-treated rats (150.0 +/- 3.6 mmHg) than in controls (135.3 +/- 5.4 mmHg), and aldosterone levels were higher in Dex-treated animals. There was no angiotensin II-induced medial hypertrophy/hyperplasia in mesenteric arteries from Dex-treated rats. These results indicate that vascular function is altered in a region-specific manner in rats with glucocorticoid-programmed hypertension. Despite a striking increase in mesenteric artery contraction in Dex-treated rats, in vivo studies suggest that abnormalities of the renin-angiotensin-aldosterone system, rather than enhanced vascular contractility, may be responsible for the elevation of blood pressure in these animals.


Assuntos
Dexametasona/farmacologia , Glucocorticoides/farmacologia , Hipertensão/embriologia , Efeitos Tardios da Exposição Pré-Natal , Vasoconstrição/efeitos dos fármacos , Acetilcolina/farmacologia , Aldosterona/sangue , Angiotensina II/farmacologia , Animais , Animais Recém-Nascidos , Aorta/efeitos dos fármacos , Corticosterona/sangue , Relação Dose-Resposta a Droga , Feminino , Hipertensão/patologia , Hipertensão/fisiopatologia , Técnicas In Vitro , Bombas de Infusão , Artérias Mesentéricas/efeitos dos fármacos , Artérias Mesentéricas/patologia , Gravidez , Ratos , Ratos Wistar , Renina/sangue
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA