Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 13 de 13
Filtrar
Mais filtros










Base de dados
Tipo de estudo
Intervalo de ano de publicação
1.
Front Pharmacol ; 15: 1341333, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-38595917

RESUMO

Butyrate, a metabolite produced by gut bacteria, has demonstrated beneficial effects in the colon and has been used to treat inflammatory bowel diseases. However, the mechanism by which butyrate operates remains incompletely understood. Given that oral butyrate can exert either a direct impact on the gut mucosa or an indirect influence through its interaction with the gut microbiome, this study aimed to investigate three key aspects: (1) whether oral intake of butyrate modulates the expression of genes encoding short-chain fatty acid (SCFA) transporters (Slc16a1, Slc16a3, Slc16a4, Slc5a8, Abcg2) and receptors (Hcar2, Ffar2, Ffar3, Olfr78, Olfr558) in the colon, (2) the potential involvement of gut microbiota in this modulation, and (3) the impact of oral butyrate on the expression of colonic SCFA transporters and receptors during colonic inflammation. Specific pathogen-free (SPF) and germ-free (GF) mice with or without DSS-induced inflammation were provided with either water or a 0.5% sodium butyrate solution. The findings revealed that butyrate decreased the expression of Slc16a1, Slc5a8, and Hcar2 in SPF but not in GF mice, while it increased the expression of Slc16a3 in GF and the efflux pump Abcg2 in both GF and SPF animals. Moreover, the presence of microbiota was associated with the upregulation of Hcar2, Ffar2, and Ffar3 expression and the downregulation of Slc16a3. Interestingly, the challenge with DSS did not alter the expression of SCFA transporters, regardless of the presence or absence of microbiota, and the effect of butyrate on the transporter expression in SPF mice remained unaffected by DSS. The expression of SCFA receptors was only partially affected by DSS. Our results indicate that (1) consuming a relatively low concentration of butyrate can influence the expression of colonic SCFA transporters and receptors, with their expression being modulated by the gut microbiota, (2) the effect of butyrate does not appear to result from direct substrate-induced regulation but rather reflects an indirect effect associated with the gut microbiome, and (3) acute colon inflammation does not lead to significant changes in the transcriptional regulation of most SCFA transporters and receptors, with the effect of butyrate in the inflamed colon remaining intact.

2.
Front Immunol ; 15: 1330094, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-38361932

RESUMO

Microbiota plays a role in shaping the HPA-axis response to psychological stressors. To examine the role of microbiota in response to acute immune stressor, we stimulated the adaptive immune system by anti-CD3 antibody injection and investigated the expression of adrenal steroidogenic enzymes and profiling of plasma corticosteroids and their metabolites in specific pathogen-free (SPF) and germ-free (GF) mice. Using UHPLC-MS/MS, we showed that 4 hours after immune challenge the plasma levels of pregnenolone, progesterone, 11-deoxycorticosterone, corticosterone (CORT), 11-dehydroCORT and their 3α/ß-, 5α-, and 20α-reduced metabolites were increased in SPF mice, but in their GF counterparts, only CORT was increased. Neither immune stress nor microbiota changed the mRNA and protein levels of enzymes of adrenal steroidogenesis. In contrast, immune stress resulted in downregulated expression of steroidogenic genes (Star, Cyp11a1, Hsd3b1, Hsd3b6) and upregulated expression of genes of the 3α-hydroxysteroid oxidoreductase pathway (Akr1c21, Dhrs9) in the testes of SPF mice. In the liver, immune stress downregulated the expression of genes encoding enzymes with 3ß-hydroxysteroid dehydrogenase (HSD) (Hsd3b2, Hsd3b3, Hsd3b4, Hsd3b5), 3α-HSD (Akr1c14), 20α-HSD (Akr1c6, Hsd17b1, Hsd17b2) and 5α-reductase (Srd5a1) activities, except for Dhrs9, which was upregulated. In the colon, microbiota downregulated Cyp11a1 and modulated the response of Hsd11b1 and Hsd11b2 expression to immune stress. These data underline the role of microbiota in shaping the response to immune stressor. Microbiota modulates the stress-induced increase in C21 steroids, including those that are neuroactive that could play a role in alteration of HPA axis response to stress in GF animals.


Assuntos
Sistema Hipotálamo-Hipofisário , Microbiota , Masculino , Camundongos , Animais , Sistema Hipotálamo-Hipofisário/metabolismo , Enzima de Clivagem da Cadeia Lateral do Colesterol/genética , Enzima de Clivagem da Cadeia Lateral do Colesterol/metabolismo , Espectrometria de Massas em Tandem , Sistema Hipófise-Suprarrenal/metabolismo , Esteroides/metabolismo , Corticosterona/metabolismo
3.
Sci Rep ; 13(1): 7278, 2023 05 04.
Artigo em Inglês | MEDLINE | ID: mdl-37142643

RESUMO

Stress increases plasma concentrations of corticosteroids, however, their tissue levels are unclear. Using a repeated social defeat paradigm, we examined the impact of chronic stress on tissue levels of corticosterone (CORT), progesterone (PROG), 11-deoxycorticosterone (11DOC) and 11-dehydrocorticosterone (11DHC) and on gut microbiota, which may reshape the stress response. Male BALB/c mice, liquid chromatography-tandem mass spectrometry and 16S RNA gene sequencing were used to screen steroid levels and fecal microbiome, respectively. Stress induced greater increase of CORT in the brain, liver, and kidney than in the colon and lymphoid organs, whereas 11DHC was the highest in the colon, liver and kidney and much lower in the brain and lymphoid organs. The CORT/11DHC ratio in plasma was similar to the brain but much lower in other organs. Stress also altered tissue levels of PROG and 11DOC and the PROG/11DOC ratio was much higher in lymphoid organs that in plasma and other organs. Stress impacted the ß- but not the α-diversity of the gut microbiota and LEfSe analysis revealed several biomarkers associated with stress treatment. Our data indicate that social defeat stress modulates gut microbiota diversity and induces tissue-dependent changes in local levels of corticosteroids, which often do not reflect their systemic levels.


Assuntos
Corticosterona , Progesterona , Camundongos , Animais , Masculino , Desoxicorticosterona , Esteroides , Encéfalo , Cromatografia Líquida
4.
Int J Mol Sci ; 22(8)2021 Apr 19.
Artigo em Inglês | MEDLINE | ID: mdl-33921780

RESUMO

Glucocorticoids (GCs) are hormones that are released in response to stressors and exhibit many activities, including immunomodulatory and anti-inflammatory activities. They are primarily synthesized in the adrenal gland but are also produced in peripheral tissues via regeneration of adrenal 11-oxo metabolites or by de novo synthesis from cholesterol. The present study investigated the influence of the microbiota on de novo steroidogenesis and regeneration of corticosterone in the intestine of germ-free (GF) and specific pathogen-free mice challenged with a physical stressor (anti-CD3 antibody i.p. injection). In the small intestine, acute immune stress resulted in increased mRNA levels of the proinflammatory cytokines IL1ß, IL6 and Tnfα and genes involved in de novo steroidogenesis (Stard3 and Cyp11a1), as well as in regeneration of active GCs from their 11-oxo metabolites (Hsd11b1). GF mice showed a generally reduced transcriptional response to immune stress, which was accompanied by decreased intestinal corticosterone production and reduced expression of the GC-sensitive marker Fkbp5. In contrast, the interaction between stress and the microbiota was not detected at the level of plasma corticosterone or the transcriptional response of adrenal steroidogenic enzymes. The results indicate a differential immune stress-induced intestinal response to proinflammatory stimuli and local corticosterone production driven by the gut microbiota.


Assuntos
Corticosterona/metabolismo , Microbioma Gastrointestinal/fisiologia , Intestino Delgado/metabolismo , 11-beta-Hidroxiesteroide Desidrogenases/genética , 11-beta-Hidroxiesteroide Desidrogenases/metabolismo , Animais , Masculino , Camundongos , Reação em Cadeia da Polimerase em Tempo Real , Esteroides/metabolismo , Espectrometria de Massas em Tandem
5.
Mech Ageing Dev ; 190: 111317, 2020 09.
Artigo em Inglês | MEDLINE | ID: mdl-32745473

RESUMO

Aging and tumorigenesis are associated with decline and disruption of circadian rhythms in many tissues and accumulating evidence indicates molecular link between circadian clock and cell cycle. The aim of this study was to investigate the effect of aging and tumorigenesis on coupling between cell cycle and circadian clock oscillators in colon, which undergoes regular rhythmicity of cell cycle and expresses peripheral circadian clock. Using healthy 14-week-old mice and 33-week-old mice with and without colorectal tumors, we showed that the 24-h expression profiles of clock genes and clock-controlled genes were mostly unaffected by aging, whereas the genes of cell cycle and cell proliferation were rhythmic in the young colons but were silenced during aging. On the other hand, tumorigenesis completely silenced or dampened the circadian rhythmicity of the clock genes but only a few genes associated with cell cycle progression and cell proliferation. These results suggest that aging impacts the colonic circadian clock moderately but markedly suppresses the rhythms of cell cycle genes and appears to uncouple the cell cycle machinery from circadian clock control. Conversely, tumorigenesis predominantly affects the rhythms of colonic circadian clocks but is not associated with uncoupling of circadian clock and cell cycle.


Assuntos
Envelhecimento , Carcinogênese , Ciclo Celular/fisiologia , Relógios Circadianos/fisiologia , Ritmo Circadiano/fisiologia , Neoplasias Colorretais , Mucosa Intestinal , Envelhecimento/metabolismo , Envelhecimento/patologia , Animais , Carcinogênese/metabolismo , Carcinogênese/patologia , Proliferação de Células , Transformação Celular Neoplásica , Colo/fisiologia , Neoplasias Colorretais/metabolismo , Neoplasias Colorretais/patologia , Mucosa Intestinal/metabolismo , Mucosa Intestinal/patologia , Camundongos
6.
Front Immunol ; 10: 2655, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-31798585

RESUMO

The gut microbiota play an important role in shaping brain functions and behavior, including the activity of the hypothalamus-pituitary-adrenocortical (HPA) axis. However, little is known about the effect of the microbiota on the distinct structures (hypothalamus, pituitary, and adrenals) of the HPA axis. In the present study, we analyzed the influence of the microbiota on acute restraint stress (ARS) response in the pituitary, adrenal gland, and intestine, an organ of extra-adrenal glucocorticoid synthesis. Using specific pathogen-free (SPF) and germ-free (GF) male BALB/c mice, we showed that the plasma corticosterone response to ARS was higher in GF than in SPF mice. In the pituitary, stress downregulated the expression of the gene encoding CRH receptor type 1 (Crhr1), upregulated the expression of the Fkbp5 gene regulating glucocorticoid receptor sensitivity and did not affect the expression of the proopiomelanocortin (Pomc) and glucocorticoid receptor (Gr) genes. In contrast, the microbiota downregulated the expression of pituitary Pomc and Crhr1 but had no effect on Fkbp5 and Gr. In the adrenals, the steroidogenic pathway was strongly stimulated by ARS at the level of the steroidogenic transcriptional regulator Sf-1, cholesterol transporter Star and Cyp11a1, the first enzyme of steroidogenic pathway. In contrast, the effect of the microbiota was significantly detected at the level of genes encoding steroidogenic enzymes but not at the level of Sf-1 and Star. Unlike adrenal Sf-1, the expression of the gene Lrh-1, which encodes the crucial transcriptional regulator of intestinal steroidogenesis, was modulated by the microbiota and ARS and this effect differed between the ileum and colon. The findings demonstrate that gut microbiota have an impact on the response of the pituitary, adrenals and intestine to ARS and that the interaction between stress and the microbiota during activation of glucocorticoid steroidogenesis differs between organs. The results suggest that downregulated expression of pituitary Pomc and Crhr1 in SPF animals might be an important factor in the exaggerated HPA response of GF mice to stress.


Assuntos
Microbioma Gastrointestinal , Sistema Hipotálamo-Hipofisário , Sistema Hipófise-Suprarrenal , Restrição Física , Estresse Psicológico/microbiologia , Glândulas Suprarrenais/metabolismo , Animais , Enzima de Clivagem da Cadeia Lateral do Colesterol/genética , Colo/metabolismo , Colo/microbiologia , Corticosterona/sangue , Regulação da Expressão Gênica , Íleo/metabolismo , Íleo/microbiologia , Masculino , Camundongos Endogâmicos BALB C , Fosfoproteínas/genética , Hipófise/metabolismo , Pró-Opiomelanocortina/genética , Receptores de Hormônio Liberador da Corticotropina/genética , Fator Esteroidogênico 1/genética , Estresse Psicológico/sangue
7.
Artigo em Inglês | MEDLINE | ID: mdl-31422161

RESUMO

The circadian clock system drives many physiological processes, including plasma concentration of glucocorticoids and epithelial transport of some ions and nutrients. As glucocorticoids entrain the circadian rhythms in various peripheral organs, we examined whether adrenalectomy affects the expression and circadian rhythmicity of intestinal transporters of the solute carrier (SLC) and ATP-binding cassette (ABC) families, which participate in intestinal barriers for absorption of nutrients, nonnutrients and oral drugs. The rat jejunum showed rhythmic circadian profiles of Sglt1, Pept1, Nhe3, Mdr1 and Mrp2 but not Mct1, Oct1, Octn1, Oatp1, Cnt1 and Bcrp. With the exception of Pept1 and Mct1, adrenalectomy decreased the expression of all rhythmic and arrhythmic transporters including the amplitude of Sglt1 and Nhe3 rhythms but minimally affected the phases of rhythmic transporters except of Nhe3. Similarly, adrenalectomy downregulated the expression of rhythmic (Pparα, Hlf, Pgc1α) and arrhythmic (Hnf1ß, Hnf4α) transcription factors, which are known to regulate the expression of transporters. We conclude that endogenous corticosteroids have a profound effect on the expression of intestinal SLC and ABC transporters and their nuclear transcription factors. The circulating corticosteroids are necessary for maintaining upregulated expression of Sglt1, Oct1, Octn1, Oatp1, Cnt1, Nhe3, Mdr1, Bcrp, Mrp2, Pparα, Pgc1α, Hnf1ß, Hnf4α and Hlf and for maintaining the high amplitude of Sglt1, Nhe3, Pparα, Pgc1α and Hlf circadian rhythms. The study demonstrates that signals from the adrenal gland are necessary for maintaining the expression of arrhythmic and rhythmic intestinal transporters and that changes in the secretion of corticosteroids associated with stress might reorganize intestinal transport barriers.


Assuntos
Transportadores de Cassetes de Ligação de ATP/metabolismo , Adrenalectomia/efeitos adversos , Jejuno/metabolismo , Proteínas Carreadoras de Solutos/metabolismo , Animais , Ritmo Circadiano , Masculino , Ratos , Ratos Wistar
8.
Endocr Connect ; 7(12): 1389-1396, 2018 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-30400029

RESUMO

Stress is an important risk factors for human diseases. It activates the hypothalamic-pituitary-adrenal (HPA) axis and increases plasma glucocorticoids, which are powerful regulators of immune system. The response of the target cells to glucocorticoids depends not only on the plasma concentrations of cortisol and corticosterone but also on their local metabolism. This metabolism is catalyzed by 11ß-hydroxysteroid dehydrogenases type 1 and 2, which interconvert glucocorticoid hormones cortisol and corticosterone and their 11-oxo metabolites cortisone and 11-dehydrocorticosterone. The goal of this study was to determine whether stress modulates glucocorticoid metabolism within lymphoid organs - the structures where immune cells undergo development and activation. Using the resident-intruder paradigm, we studied the effect of social stress on glucocorticoid metabolism in primary and secondary lymphoid organs of Fisher 344 (F344) and Lewis (LEW) rats, which exhibit marked differences in their HPA axis response to social stressors and inflammation. We show that repeated social defeat increased the regeneration of corticosterone from 11-dehydrocorticosterone in the thymus, spleen and mesenteric lymphatic nodes (MLN). Compared with the F344 strain, LEW rats showed higher corticosterone regeneration in splenocytes of unstressed rats and in thymic and MLN mobile cells after stress but corticosterone regeneration in the stroma of all lymphoid organs was similar in both strains. Inactivation of corticosterone to 11-dehydrocorticosterone was found only in the stroma of lymphoid organs but not in mobile lymphoid cells and was not upregulated by stress. Together, our findings demonstrate the tissue- and strain-dependent regeneration of glucocorticoids following social stress.

9.
Steroids ; 126: 66-73, 2017 10.
Artigo em Inglês | MEDLINE | ID: mdl-28754259

RESUMO

The bioavailability of glucocorticoids is modulated by enzyme 11ß-hydroxysteroid dehydrogenase type 1 (11HSD1), which catalyzes the conversion of inactive 11-oxo-glucocorticoids to active 11-hydroxy-glucocorticoids cortisol and corticosterone and is regulated by pro-inflammatory cytokines. Our aim was to assess the effect of colitis on the expression of 11HSD1 in specific microanatomical compartments of the mucosal immune system. Using qRT-PCR we quantified the expression of 11HSD1 and cytokines in the colon, mesenteric lymph nodes (MLN) and spleen of mice with colitis. Microsamples of the MLN cortex, paracortex and medulla, colonic crypt epithelium (CCE), lamina propria and isolated intestinal lymphoid follicles (ILF) were harvested by laser microdissection, whereas splenic and MLN lymphocytes by flow cytometry. Colitis increased 11HSD1 in the CCE, ILF, and MLN cortex but not in the lamina propria and the MLN paracortex and medulla. Expression of IL-4, IL-21 and TNFα was increased in both the cortex of MLN and ILF, whereas IL-1ß and IL-10 were only increased in the follicles. No positive effect was observed in the case of IFNγ and TGFß. 11HSD1 was positively correlated with TNFα and less strongly with IL-21, IL-1ß, and IL-4. Colitis also upregulated the 11HSD1 expression of T cells in the spleen and MLN. The study demonstrates the stimulatory effect of inflammation on local glucocorticoid metabolism only in particular compartments of the mucosal immune system. The correlation between cytokines and 11HSD1 in the ILF and MLN cortex indicates that pro-inflammatory cytokines may amplify glucocorticoid signals in inductive compartments of the mucosal immune system.


Assuntos
11-beta-Hidroxiesteroide Desidrogenase Tipo 1/metabolismo , Mucosa Intestinal/imunologia , 11-beta-Hidroxiesteroide Desidrogenase Tipo 1/genética , Animais , Colite/enzimologia , Colite/genética , Colite/imunologia , Citocinas/metabolismo , Regulação Enzimológica da Expressão Gênica , Inflamação/enzimologia , Inflamação/genética , Inflamação/imunologia , Masculino , Camundongos , Camundongos Endogâmicos BALB C
10.
Steroids ; 76(6): 577-81, 2011 May.
Artigo em Inglês | MEDLINE | ID: mdl-21352843

RESUMO

Glucocorticoids are metabolized in vascular tissue by two types of 11ß-hydroxysteroid dehydrogenases (11HSD1, 11HSD2) and thus these enzymes are considered to be important factors that modulate the diverse and complex effects of glucocorticoids on cardiovascular function. The present study evaluated the effect of peroxisome proliferator-activated receptor-γ (PPARγ) agonist pioglitazone on 11HSD1 vascular smooth muscle cells (VSMC) and compared the effect with that of corticosterone. Using primary cultures of VSMC derived from rat aorta, we showed that pioglitazone significantly increases 11HSD1 activity and mRNA expression in a dose-dependent manner with EC(50) 243 nM and that this effect is not blocked by RU 486, an antagonist of the glucocorticoid receptor. In contrast, corticosterone had no effect on 11HSD1. Pioglitazone positively regulated transcription of two CCAAT/enhancer-binding proteins (C/EBPs), specifically C/EBPα a potent activator of 11HSD1 gene transcription in some cells types, and C/EBPζ, whereas C/EBPß and C/EBPδ were not changed. In contrast, corticosterone stimulated the expression of C/EBPß and C/EBPδ, but the levels of C/EBPα and C/EBPζ were not changed. In conclusion, activation of PPARγ in VSMC up-regulates vascular 11HSD1 and thus reactivates 11-oxo metabolites to biologically active glucocorticoids through a mechanism that seems to involve C/EBPα and C/EBPζ. Our data provide one of the possible explanations for PPARγ agonists' effects on the cardiovascular system.


Assuntos
11-beta-Hidroxiesteroide Desidrogenase Tipo 1/metabolismo , Músculo Liso Vascular/citologia , Miócitos de Músculo Liso/enzimologia , PPAR gama/agonistas , 11-beta-Hidroxiesteroide Desidrogenase Tipo 1/genética , Animais , Células Cultivadas , Corticosterona/farmacologia , Ativação Enzimática , Ensaios Enzimáticos , Masculino , Miócitos de Músculo Liso/efeitos dos fármacos , PPAR gama/metabolismo , Pioglitazona , Ratos , Ratos Wistar , Tiazolidinedionas/farmacologia , Transcrição Gênica
11.
Mol Cell Endocrinol ; 323(2): 155-60, 2010 Jul 29.
Artigo em Inglês | MEDLINE | ID: mdl-20226838

RESUMO

11beta-Hydroxysteroid dehydrogenase 1 (11HSD1) regulates local glucocorticoid activity and plays an important role in various diseases. Here, we studied whether arthritis modulates 11HSD1, what is the role of pro-inflammatory cytokines in this process and whether altered local metabolism of glucocorticoids may contribute to the feedback regulation of inflammation. Adjuvant arthritis increased synovial 11HSD1 mRNA and 11-reductase activity but treatments with tumor necrosis factor alpha (TNF-alpha) and interleukin 1beta (IL-1beta) antagonists etanercept and anakinra reduced 11HSD1 upregulation. Treatment with carbenoxolone, an 11HSD inhibitor, increased expression of TNF-alpha, cyclooxygenase 2, and osteopontin mRNA without any changes in the plasma levels of corticosterone. Similar changes were observed when arthritic rats were treated with RU486, an antagonist of GR. This study suggests that arthritis upregulates synovial 11HSD1, this upregulation is controlled by TNF-alpha and IL-1beta and that the increased supply of local corticosterone might contribute to feedback regulation of inflammation.


Assuntos
11-beta-Hidroxiesteroide Desidrogenases/metabolismo , Artrite Experimental/metabolismo , Glucocorticoides/metabolismo , Isoenzimas/metabolismo , 11-beta-Hidroxiesteroide Desidrogenases/genética , Animais , Antiulcerosos/farmacologia , Artrite Experimental/genética , Carbenoxolona/farmacologia , Células Cultivadas , Citocinas/metabolismo , Antagonistas de Hormônios/farmacologia , Humanos , Isoenzimas/genética , Masculino , Mifepristona/farmacologia , RNA Mensageiro/metabolismo , Ratos , Ratos Endogâmicos Lew , Membrana Sinovial/citologia , Membrana Sinovial/efeitos dos fármacos , Membrana Sinovial/metabolismo
12.
Reprod Toxicol ; 28(1): 46-51, 2009 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-19490994

RESUMO

Placental 11 beta-hydroxysteroid dehydrogenase type 2 (11 beta-HSD2) is the key enzyme which protects the fetus from overexposure to glucocorticoids (GCs) by their oxidation into inactive derivates. Several recent studies suggest that 11 beta-HSD2 expression is subjected to regulation by antenatal steroid therapy. In our study we investigated the effect of two commonly used synthetic steroids, dexamethasone (DXM) and betamethasone (BTM), on the expression and function of 11 beta-HSD2 in the rat placenta. Pregnant rats were pretreated with low (0.2mg/kg) or high (5mg/kg and 11.5mg/kg for DXM and BTM, respectively) i.m. doses of GCs. 11 beta-HSD2 expression was investigated using real-time RT-PCR and Western blotting; conversion capacity of 11 beta-HSD2 was assessed by dual perfusion of the rat placenta. Significant increase in placental 11 beta-HSD2 mRNA expression was found in rats treated with DXM, however, this alteration was not observed on protein level. BTM had no effect on either mRNA or protein levels of 11 beta-HSD2. Functional studies revealed that both GCs significantly reduced the metabolism of corticosterone by the placenta. Our data indicate that placental barrier function mediated by 11 beta-HSD2 might be considerably impaired by the antenatal therapy with DXM and BTM. In addition, the discrepancy between expressional and functional studies suggests that sole analysis of expressional changes of 11 beta-HSD2 at mRNA and/or protein levels cannot convincingly predict the role of GC treatment on 11 beta-HSD2 function in the placental barrier.


Assuntos
11-beta-Hidroxiesteroide Desidrogenase Tipo 2/metabolismo , Betametasona/toxicidade , Dexametasona/toxicidade , Glucocorticoides/toxicidade , Placenta/efeitos dos fármacos , 11-beta-Hidroxiesteroide Desidrogenase Tipo 2/genética , Animais , Betametasona/administração & dosagem , Betametasona/metabolismo , Peso ao Nascer/efeitos dos fármacos , Western Blotting , Dexametasona/administração & dosagem , Dexametasona/metabolismo , Relação Dose-Resposta a Droga , Feminino , Regulação da Expressão Gênica/efeitos dos fármacos , Glucocorticoides/administração & dosagem , Glucocorticoides/metabolismo , Injeções Intramusculares , Perfusão , Placenta/enzimologia , Gravidez , RNA Mensageiro/metabolismo , Ratos , Ratos Wistar , Reação em Cadeia da Polimerase Via Transcriptase Reversa
13.
Endocrinology ; 150(9): 4270-7, 2009 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-19470702

RESUMO

Corticosteroids are known to not only regulate electrolyte homeostasis but also play a role in the cardiovascular system, including myocardial remodeling. Because transgenic mice that overexpress 11beta-hydroxysteroid dehydrogenase (11HSD) type 2 in cardiomyocytes have been shown to spontaneously develop cardiac hypertrophy and fibrosis, we investigated whether changes in the cardiac metabolism of glucocorticoids accompany remodeling of the heart under physiological conditions. In the present study, glucocorticoid metabolism and 11HSD2 were explored in the hearts of rats exposed to chronic intermittent hypobaric hypoxia (CIH), which induces hypertrophy and fibrosis of the right and less of the left ventricle. We first demonstrated that adaptation to CIH led to a significant increase in 11HSD2 transcript levels and activity in the myocardium. In contrast, neither 11HSD1 activity and mRNA level nor the abundance of mineralocorticoid and glucocorticoid receptor mRNA were up-regulated. The adaptation to CIH also led to an increase of 11HSD2 mRNA in isolated cardiomyocytes, whereas 11HSD1, glucocorticoid receptor, and mineralocorticoid receptor mRNA levels were not changed in comparison with the cardiomyocytes of control normoxic rats. The changes in cardiac metabolism of glucocorticoids were accompanied by inflammatory responses. The expression levels of the proinflammatory markers cyclooxygenase-2 and osteopontin were significantly increased in both the myocardium and the cardiomyocytes isolated from rats exposed to CIH. These findings suggest that myocardial remodeling induced by CIH is associated with the up-regulation of cardiac 11HSD2. Consequently, local metabolism of glucocorticoids could indeed play a role in cardiac hypertrophy and fibrosis.


Assuntos
11-beta-Hidroxiesteroide Desidrogenase Tipo 2/biossíntese , Hipóxia/metabolismo , Miocárdio/enzimologia , 11-beta-Hidroxiesteroide Desidrogenase Tipo 1/biossíntese , Aldosterona/sangue , Animais , Cardiomegalia , Corticosterona/sangue , Fibrose , Masculino , Miocárdio/patologia , Ratos , Ratos Wistar , Receptores de Glucocorticoides/biossíntese , Receptores de Mineralocorticoides/biossíntese , Regulação para Cima
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...