Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 54
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
J Biol Chem ; 293(4): 1151-1162, 2018 01 26.
Artículo en Inglés | MEDLINE | ID: mdl-29123029

RESUMEN

Normal renin synthesis and secretion is important for the maintenance of juxtaglomerular apparatus architecture. Mice lacking a functional Ren1d gene are devoid of renal juxtaglomerular cell granules and exhibit an altered macula densa morphology. Due to the species-specificity of renin activity, transgenic mice are ideal models for experimentally investigating and manipulating expression patterns of the human renin gene in a native cellular environment without confounding renin-angiotensin system interactions. A 55-kb transgene encompassing the human renin locus was crossed onto the mouse Ren1d-null background, restoring granulation in juxtaglomerular cells. Correct processing of human renin in dense core granules was confirmed by immunogold labeling. After stimulation of the renin-angiotensin system, juxtaglomerular cells contained rhomboid protogranules with paracrystalline contents, dilated rough endoplasmic reticulum, and electron-lucent granular structures. However, complementation of Ren1d-/- mice with human renin was unable to rescue the abnormality seen in macula densa structure. The juxtaglomerular apparatus was still able to respond to tubuloglomerular feedback in isolated perfused juxtaglomerular apparatus preparations, although minor differences in glomerular tuft contractility and macula densa cell calcium handling were observed. This study reveals that the human renin protein is able to complement the mouse Ren1d-/- non-granulated defect and suggests that granulopoiesis requires a structural motif that is conserved between the mouse Ren1d and human renin proteins. It also suggests that the altered macula densa phenotype is related to the activity of the renin-1d enzyme in a local juxtaglomerular renin-angiotensin system.


Asunto(s)
Prueba de Complementación Genética , Aparato Yuxtaglomerular/enzimología , Renina/biosíntesis , Transgenes , Animales , Humanos , Aparato Yuxtaglomerular/patología , Ratones , Ratones Noqueados , Renina/genética
2.
Circulation ; 133(14): 1360-70, 2016 Apr 05.
Artículo en Inglés | MEDLINE | ID: mdl-26951843

RESUMEN

BACKGROUND: The hypertensive syndrome of Apparent Mineralocorticoid Excess is caused by loss-of-function mutations in the gene encoding 11ß-hydroxysteroid dehydrogenase type 2 (11ßHSD2), allowing inappropriate activation of the mineralocorticoid receptor by endogenous glucocorticoid. Hypertension is attributed to sodium retention in the distal nephron, but 11ßHSD2 is also expressed in the brain. However, the central contribution to Apparent Mineralocorticoid Excess and other hypertensive states is often overlooked and is unresolved. We therefore used a Cre-Lox strategy to generate 11ßHSD2 brain-specific knockout (Hsd11b2.BKO) mice, measuring blood pressure and salt appetite in adults. METHODS AND RESULTS: Basal blood pressure, electrolytes, and circulating corticosteroids were unaffected in Hsd11b2.BKO mice. When offered saline to drink, Hsd11b2.BKO mice consumed 3 times more sodium than controls and became hypertensive. Salt appetite was inhibited by spironolactone. Control mice fed the same daily sodium intake remained normotensive, showing the intrinsic salt resistance of the background strain. Dexamethasone suppressed endogenous glucocorticoid and abolished the salt-induced blood pressure differential between genotypes. Salt sensitivity in Hsd11b2.BKO mice was not caused by impaired renal sodium excretion or volume expansion; pressor responses to phenylephrine were enhanced and baroreflexes impaired in these animals. CONCLUSIONS: Reduced 11ßHSD2 activity in the brain does not intrinsically cause hypertension, but it promotes a hunger for salt and a transition from salt resistance to salt sensitivity. Our data suggest that 11ßHSD2-positive neurons integrate salt appetite and the blood pressure response to dietary sodium through a mineralocorticoid receptor-dependent pathway. Therefore, central mineralocorticoid receptor antagonism could increase compliance to low-sodium regimens and help blood pressure management in cardiovascular disease.


Asunto(s)
11-beta-Hidroxiesteroide Deshidrogenasa de Tipo 2/metabolismo , Ansia/fisiología , Hipertensión/genética , Síndrome de Exceso Aparente de Mineralocorticoides/fisiopatología , Proteínas del Tejido Nervioso/deficiencia , Receptores de Mineralocorticoides/fisiología , Cloruro de Sodio Dietético/toxicidad , Núcleo Solitario/enzimología , 11-beta-Hidroxiesteroide Deshidrogenasa de Tipo 2/genética , 11-beta-Hidroxiesteroide Deshidrogenasa de Tipo 2/fisiología , Animales , Barorreflejo/efectos de los fármacos , Corticosterona/sangre , Dexametasona/farmacología , Conducta de Ingestión de Líquido , Genes Sintéticos , Hipertensión/fisiopatología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Síndrome de Exceso Aparente de Mineralocorticoides/tratamiento farmacológico , Síndrome de Exceso Aparente de Mineralocorticoides/genética , Antagonistas de Receptores de Mineralocorticoides/uso terapéutico , Nefronas/fisiopatología , Proteínas del Tejido Nervioso/genética , Proteínas del Tejido Nervioso/fisiología , Nestina/genética , Neuronas/fisiología , Potasio/orina , ARN Mensajero/biosíntesis , Reflejo Anormal , Núcleo Solitario/fisiopatología , Espironolactona/farmacología
3.
J Am Soc Nephrol ; 26(7): 1537-48, 2015 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-25349206

RESUMEN

Na(+) transport in the renal distal convoluted tubule (DCT) by the thiazide-sensitive NaCl cotransporter (NCC) is a major determinant of total body Na(+) and BP. NCC-mediated transport is stimulated by aldosterone, the dominant regulator of chronic Na(+) homeostasis, but the mechanism is controversial. Transport may also be affected by epithelial remodeling, which occurs in the DCT in response to chronic perturbations in electrolyte homeostasis. Hsd11b2(-/-) mice, which lack the enzyme 11ß-hydroxysteroid dehydrogenase type 2 (11ßHSD2) and thus exhibit the syndrome of apparent mineralocorticoid excess, provided an ideal model in which to investigate the potential for DCT hypertrophy to contribute to Na(+) retention in a hypertensive condition. The DCTs of Hsd11b2(-/-) mice exhibited hypertrophy and hyperplasia and the kidneys expressed higher levels of total and phosphorylated NCC compared with those of wild-type mice. However, the striking structural and molecular phenotypes were not associated with an increase in the natriuretic effect of thiazide. In wild-type mice, Hsd11b2 mRNA was detected in some tubule segments expressing Slc12a3, but 11ßHSD2 and NCC did not colocalize at the protein level. Thus, the phosphorylation status of NCC may not necessarily equate to its activity in vivo, and the structural remodeling of the DCT in the knockout mouse may not be a direct consequence of aberrant corticosteroid signaling in DCT cells. These observations suggest that the conventional concept of mineralocorticoid signaling in the DCT should be revised to recognize the complexity of NCC regulation by corticosteroids.


Asunto(s)
11-beta-Hidroxiesteroide Deshidrogenasa de Tipo 2/farmacología , Túbulos Renales Distales/patología , Fosforilación/efectos de los fármacos , Simportadores del Cloruro de Sodio/metabolismo , 11-beta-Hidroxiesteroide Deshidrogenasa de Tipo 2/metabolismo , Análisis de Varianza , Animales , Células Cultivadas , ADN Complementario/análisis , Modelos Animales de Enfermedad , Células Epiteliales/citología , Células Epiteliales/efectos de los fármacos , Femenino , Hipertrofia/patología , Túbulos Renales Distales/efectos de los fármacos , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Reacción en Cadena de la Polimerasa/métodos , ARN/análisis , Distribución Aleatoria , Transcitosis/fisiología
4.
Hum Mol Genet ; 22(16): 3269-82, 2013 Aug 15.
Artículo en Inglés | MEDLINE | ID: mdl-23595884

RESUMEN

Glucocorticoids are vital for the structural and functional maturation of foetal organs, yet excessive foetal exposure is detrimental to adult cardiovascular health. To elucidate the role of glucocorticoid signalling in late-gestation cardiovascular maturation, we have generated mice with conditional disruption of glucocorticoid receptor (GR) in cardiomyocytes and vascular smooth muscle cells using smooth muscle protein 22-driven Cre recombinase (SMGRKO mice) and compared them with mice with global deficiency in GR (GR(-/-)). Echocardiography shows impaired heart function in both SMGRKO and GR(-/-) mice at embryonic day (E)17.5, associated with generalized oedema. Cardiac ultrastructure is markedly disrupted in both SMGRKO and GR(-/-) mice at E17.5, with short, disorganized myofibrils and cardiomyocytes that fail to align in the compact myocardium. Failure to induce critical genes involved in contractile function, calcium handling and energy metabolism underpins this common phenotype. However, although hearts of GR(-/-) mice are smaller, with 22% reduced ventricular volume at E17.5, SMGRKO hearts are normally sized. Moreover, while levels of mRNA encoding atrial natriuretic peptide are reduced in E17.5 GR(-/-) hearts, they are normal in foetal SMGRKO hearts. These data demonstrate that structural, functional and biochemical maturation of the foetal heart is dependent on glucocorticoid signalling within cardiomyocytes and vascular smooth muscle, though some aspects of heart maturation (size, ANP expression) are independent of GR at these key sites.


Asunto(s)
Corazón Fetal/crecimiento & desarrollo , Glucocorticoides/metabolismo , Receptores de Glucocorticoides/genética , Receptores de Glucocorticoides/metabolismo , Transducción de Señal , Animales , Corticosterona/sangre , Corticosterona/fisiología , Corazón Fetal/fisiología , Corazón/embriología , Corazón/fisiología , Ratones , Ratones Transgénicos , Músculo Liso Vascular/embriología , Músculo Liso Vascular/metabolismo , Contracción Miocárdica , Miocardio/ultraestructura , Miocitos Cardíacos/citología , Miocitos Cardíacos/metabolismo , Miofibrillas/ultraestructura
5.
Eur J Neurosci ; 41(6): 787-92, 2015 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-25614240

RESUMEN

11ß-Hydroxysteroid dehydrogenase type 1 (11ß-HSD1) locally regenerates active glucocorticoids from their inert forms thereby amplifying intracellular levels within target tissues including the brain. We previously showed greater increases in intra-hippocampal corticosterone (CORT) levels upon Y-maze testing in aged wild-type than in 11ß-HSD1(-/-) mice coinciding with impaired and intact spatial memory, respectively. Here we examined whether ageing influences 11ß-HSD1 regulation of CORT in the dorsal hippocampus under basal conditions during the diurnal cycle and following stress. Intra-hippocampal CORT levels measured by in vivo microdialysis in freely behaving wild-type mice displayed a diurnal variation with peak levels in the evening that were significantly elevated with ageing. In contrast, the diurnal rise in intra-hippocampal CORT levels was greatly diminished in 11ß-HSD1(-/-) mice and there was no rise with ageing; basal intra-hippocampal CORT levels were similar to wild-type controls. Furthermore, a short (3 min) swim stress induced a longer lasting increase in intra-hippocampal CORT levels in wild-type mice than in 11ß-HSD1(-/-) mice despite no genotypic differences in elevation of plasma CORT. These data indicate that 11ß-HSD1 activity contributes substantially to diurnal and stress-induced increases in hippocampal CORT levels. This contribution is even greater with ageing. Thus, 11ß-HSD1 inhibition may be an attractive target for treating cognitive impairments associated with stress or ageing.


Asunto(s)
11-beta-Hidroxiesteroide Deshidrogenasa de Tipo 1/fisiología , Envejecimiento , Ritmo Circadiano , Corticosterona/fisiología , Hipocampo/fisiología , Estrés Psicológico , 11-beta-Hidroxiesteroide Deshidrogenasa de Tipo 1/genética , Animales , Corticosterona/análisis , Hipocampo/química , Masculino , Ratones , Ratones Noqueados , Microdiálisis
6.
Exp Physiol ; 100(11): 1362-78, 2015 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-26337786

RESUMEN

NEW FINDINGS: What is the central question of this study? Glucocorticoids act in the kidney to promote salt and water retention. Renal 11ß-hydroxysteroid dehydrogenase type 1 (11ßHSD1), by increasing local concentrations of glucocorticoids, may exert an antinatriuretic effect. We hypothesized that global deletion of 11ßHSD1 in the mouse would give rise to a salt-wasting renal phenotype. What is the main finding and its importance? We subjected a mouse model of global 11ßHSD1 deletion to studies of water and electrolyte balance, renal clearance, urinary steroid excretion, renin-angiotensin system activation and renal sodium transporter expression. We found no significant effects on renal sodium or water excretion. Any effect of renal 11ßHSD1 on sodium homeostasis is subtle. Glucocorticoids act in the kidney to regulate glomerular haemodynamics and tubular sodium transport; the net effect favours sodium retention. 11ß-Hydroxysteroid dehydrogenase type 1 (11ßHSD1) is expressed in the renal tubules and the interstitial cells of the medulla, where it is likely to regenerate active glucocorticoids from inert 11-keto forms. The physiological function of renal 11ßHSD1 is largely unknown. We hypothesized that loss of renal 11ßHSD1 would result in salt wasting and tested this in a knockout mouse model in which 11ßHSD1 was deleted in all body tissues. In balance studies, 11ßHSD1 deletion had no effect on water, sodium or potassium metabolism; transition to a low-sodium diet did not reveal a natriuretic phenotype. Renal clearance studies demonstrated identical haemodynamic parameters (arterial blood pressure, renal blood flow and glomerular filtration rate) in knockout and wild-type mice, but revealed an augmented kaliuretic response to thiazides in 11ßHSD1 knockout animals. There was no effect on the natriuretic response to the amiloride analogue benzamil. Urinary excretion of deoxycorticosterone was higher in 11ßHSD1 knockout mice, and there was hypertrophy of cells in the zona fasciculata of the adrenal cortex. There was no difference in the activity of the renin-angiotensin and nitric oxide systems, no difference in renal histology and no difference in the abundance of key tubular transporter proteins. We conclude that any effect of 11ßHSD1 on renal sodium excretion is subtle.


Asunto(s)
11-beta-Hidroxiesteroide Deshidrogenasa de Tipo 1/genética , Homeostasis , Riñón/fisiología , Sodio/fisiología , Animales , Glucocorticoides/fisiología , Ratones Noqueados , Potasio/metabolismo , Sistema Renina-Angiotensina , Sodio/metabolismo , Equilibrio Hidroelectrolítico
7.
FASEB J ; 26(5): 1866-74, 2012 May.
Artículo en Inglés | MEDLINE | ID: mdl-22321728

RESUMEN

Fetal growth restriction associates with increased risk of adult cardiometabolic and neuropsychiatric disorders. Both maternal malnutrition [notably a low-protein (LP) diet] and stress/glucocorticoid exposure reduce fetal growth and cause persisting abnormalities (programming) in adult offspring. Deficiency of placental 11ß-hydroxysteroid dehydrogenase-2 (11ß-HSD2), which inactivates glucocorticoids, is reduced by an LP diet and has been proposed as a unifying mechanism. Here, we explored the importance of glucocorticoids and placental 11ß-HSD2 in dietary programming. Pregnant mice were fed a control or isocaloric LP diet throughout gestation. The LP diet first elevated fetal glucocorticoid levels, then reduced placental growth, and finally decreased fetal weight near term by 17%. Whereas the LP diet reduced placental 11ß-HSD2 activity near term by ∼25%, consistent with previous reports, activity was increased between 20 and 40% at earlier ages, implying that glucocorticoid overexposure in LP fetuses occurs via 11ß-HSD2-independent mechanisms. Consistent with this, heterozygous 11ß-HSD2(+/-) crosses showed that although both LP and 11ß-HSD2 deficiency reduced fetal growth, LP indeed acted independently of 11ß-HSD2. Instead, the LP diet induced the fetal hypothalamic-pituitary-adrenal axis per se. Thus, maternal malnutrition and placental 11ß-HSD2 deficiency act via distinct processes to retard fetal growth, both involving fetoplacental overexposure to glucocorticoids but from distinct sources.


Asunto(s)
Desarrollo Fetal , Glucocorticoides/fisiología , Estado Nutricional , 11-beta-Hidroxiesteroide Deshidrogenasa de Tipo 2/metabolismo , Animales , Secuencia de Bases , Corticosterona/sangre , Cartilla de ADN , Femenino , Hibridación in Situ , Exposición Materna , Ratones , Ratones Endogámicos C57BL , Placenta/enzimología , Embarazo
8.
Am J Physiol Renal Physiol ; 303(4): F494-502, 2012 Aug 15.
Artículo en Inglés | MEDLINE | ID: mdl-22622456

RESUMEN

In aldosterone target tissues, 11ß-hydroxysteroid dehydrogenase type 2 (11ßHSD2) is coexpressed with mineralocorticoid receptors (MR) and protects the receptor from activation by glucocorticoids. Null mutations in the encoding gene, HSD11B2, cause apparent mineralocorticoid excess, in which hypertension is thought to reflect volume expansion secondary to sodium retention. Hsd11b2(-/-) mice are indeed hypertensive, but impaired natriuretic capacity is associated with significant volume contraction, suggestive of a urine concentrating defect. Water turnover and the urine concentrating response to a 24-h water deprivation challenge were therefore assessed in Hsd11b2(-/-) mice and controls. Hsd11b2(-/-) mice have a severe and progressive polyuric/polydipsic phenotype. In younger mice (∼2 mo of age), polyuria was associated with decreased abundance of aqp2 and aqp3 mRNA. The expression of other genes involved in water transport (aqp4, slc14a2, and slc12a2) was not changed. The kidney was structurally normal, and the concentrating response to water deprivation was intact. In older Hsd11b2(-/-) mice (>6 mo), polyuria was associated with a severe atrophy of the renal medulla and downregulation of aqp2, aqp3, aqp4, slc14a2, and slc12a2. The concentrating response to water deprivation was impaired, and the natriuretic effect of the loop diuretic bumetanide was lost. In older Hsd11b2(-/-) mice, the V2 receptor agonist desmopressin did not restore full urine concentrating capacity. We find that Hsd11b2(-/-) mice develop nephrogenic diabetes insipidus. Gross changes to renal structure are observed, but these were probably secondary to sustained polyuria, rather than of developmental origin.


Asunto(s)
11-beta-Hidroxiesteroide Deshidrogenasa de Tipo 2/genética , 11-beta-Hidroxiesteroide Deshidrogenasa de Tipo 2/metabolismo , Diabetes Insípida/enzimología , Capacidad de Concentración Renal/fisiología , Riñón/fisiología , Envejecimiento , Animales , Diabetes Insípida/genética , Regulación de la Expresión Génica , Homeostasis , Riñón/anatomía & histología , Capacidad de Concentración Renal/genética , Ratones , Ratones Noqueados , Concentración Osmolar , Simportadores de Cloruro de Sodio-Potasio/genética , Simportadores de Cloruro de Sodio-Potasio/metabolismo , Agua/metabolismo , Pérdida de Peso
9.
Am J Physiol Endocrinol Metab ; 300(6): E1076-84, 2011 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-21406612

RESUMEN

Increased dietary fat intake is associated with obesity, insulin resistance, and metabolic disease. In transgenic mice, adipose tissue-specific overexpression of the glucocorticoid-amplifying enzyme 11ß-hydroxysteroid dehydrogenase type 1 (11ß-HSD1) exacerbates high-fat (HF) diet-induced visceral obesity and diabetes, whereas 11ß-HSD1 gene knockout ameliorates this, favoring accumulation of fat in nonvisceral depots. Paradoxically, in normal mice HF diet-induced obesity (DIO) is associated with marked downregulation of adipose tissue 11ß-HSD1 levels. To identify the specific dietary fats that regulate adipose 11ß-HSD1 and thereby impact upon metabolic disease, we either fed mice diets enriched (45% calories as fat) in saturated (stearate), monounsaturated (oleate), or polyunsaturated (safflower oil) fats ad libitum or we pair fed them a low-fat (11%) control diet for 4 wk. Adipose and liver mass and glucocorticoid receptor and 11ß-HSD1 mRNA and activity levels were determined. Stearate caused weight loss and hypoinsulinemia, partly due to malabsorption, and this markedly increased plasma corticosterone levels and adipose 11ß-HSD1 activity. Oleate induced pronounced weight gain and hyperinsulinemia in association with markedly low plasma corticosterone and adipose 11ß-HSD1 activity. Weight gain and hyperinsulinemia was less pronounced with safflower compared with oleate despite comparable suppression of plasma corticosterone and adipose 11ß-HSD1. However, with pair feeding, safflower caused a selective reduction in visceral fat mass and relative insulin sensitization without affecting plasma corticosterone or adipose 11ß-HSD1. The dynamic depot-selective relationship between adipose 11ß-HSD1 and fat mass strongly implicates a dominant physiological role for local tissue glucocorticoid reactivation in fat mobilization.


Asunto(s)
11-beta-Hidroxiesteroide Deshidrogenasa de Tipo 1/biosíntesis , Tejido Adiposo/enzimología , Tejido Adiposo/fisiología , Composición Corporal/fisiología , Dieta , 11-beta-Hidroxiesteroide Deshidrogenasa de Tipo 1/genética , Adiposidad , Animales , Corticosterona/metabolismo , Ingestión de Alimentos/efectos de los fármacos , Ácidos Grasos/farmacología , Ácidos Grasos Monoinsaturados/farmacología , Ácidos Grasos Insaturados/farmacología , Heces/química , Expresión Génica/efectos de los fármacos , Homeostasis/efectos de los fármacos , Homeostasis/fisiología , Resistencia a la Insulina/fisiología , Hígado/efectos de los fármacos , Hígado/enzimología , Masculino , Ratones , Ratones Endogámicos C57BL , ARN/biosíntesis , ARN/genética , Receptores de Glucocorticoides/biosíntesis , Receptores de Glucocorticoides/genética , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Aumento de Peso/efectos de los fármacos
10.
Physiol Genomics ; 40(3): 158-66, 2010 Feb 04.
Artículo en Inglés | MEDLINE | ID: mdl-19920212

RESUMEN

We investigated the effects on urinary steroid and electrolyte excretion and renal gene expression of chronic infusions of ACTH in the mouse. ACTH caused a sustained increase in corticosteroid excretion; aldosterone excretion was only transiently elevated. There was an increase in the excretion of deoxycorticosterone, a weak mineralocorticoid, to levels of physiological significance. Nevertheless, we observed neither antinatriuresis nor kaliuresis in ACTH-treated mice, and plasma renin activity was not suppressed. We identified no changes in expression of mineralocorticoid target genes. Water turnover was increased in chronic ACTH-treated mice, as were hematocrit and hypertonicity: volume contraction is consistent with high levels of glucocorticoid. ACTH-treated mice exhibited other signs of glucocorticoid excess, such as enhanced weight gain and involution of the thymus. We identified novel ACTH-induced changes in 1) genes involved in vitamin D (Cyp27b1, Cyp24a1, Gc) and calcium (Sgk, Calb1, Trpv5) metabolism associated with calciuria and phosphaturia; 2) genes that would be predicted to desensitize the kidney to glucocorticoid action (Nr3c1, Hsd11b1, Fkbp5); and 3) genes encoding transporters of enzyme systems associated with xenobiotic metabolism and oxidative stress. Although there is evidence that ACTH-induced hypertension is a function of physiological cross talk between glucocorticoids and mineralocorticoids, the present study suggests that the major changes in electrolyte and fluid homeostasis and renal function are attributable to glucocorticoids. The calcium and organic anion metabolism pathways that are affected by ACTH may explain some of the known adverse effects associated with glucocorticoid excess.


Asunto(s)
Hormona Adrenocorticotrópica/farmacología , Hormonas/farmacología , Riñón/efectos de los fármacos , Riñón/fisiología , Transcripción Genética , Hormona Adrenocorticotrópica/metabolismo , Animales , Calcio/metabolismo , Expresión Génica , Hormonas/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Potasio/metabolismo , Transcripción Genética/efectos de los fármacos
11.
Nutrients ; 11(2)2019 Feb 09.
Artículo en Inglés | MEDLINE | ID: mdl-30744113

RESUMEN

Mechanisms to explain post-prandial increases in circulating glucocorticoids are not well understood and may involve increased adrenal secretion and/or altered steroid metabolism. We have compared salivary levels of cortisol and cortisone levels in healthy male and female volunteers fed either a low or cholesterol-rich midday meal. Urinary levels of steroids, bile acids and markers of lipid peroxidation were also measured. Males and females showed expected circadian changes in salivary steroids and postprandial peaks within 1h of feeding. After a high-cholesterol meal, postprandial cortisol increases were higher in males whereas post-prandial cortisone levels were higher in females. Urinary cortisol but not cortisone levels were higher on the day when males and females ate a high-cholesterol meal. Urinary bile acid excretion and anti-oxidant markers of lipid peroxidation, thiobarbituric acid reactive substances (TBARS), and total phenol content were not affected by dietary cholesterol but tended to be higher in males. Cross-tabulation of correlation coefficients indicated positive associations between urinary markers of peroxidation, bile acids, and cortisol:cortisone ratios. We conclude that dietary cholesterol (a substrate for steroidogenesis) does not have an acute effect on adrenal glucocorticoid synthesis and that gender but not a high-cholesterol meal may influence the interconversion of cortisol and cortisone. Longer term studies of the effects of dietary cholesterol are needed to analyze the associations between bile acids, steroid metabolism, and secretion and lipid peroxidation.


Asunto(s)
Ácidos y Sales Biliares/metabolismo , Colesterol en la Dieta/metabolismo , Glucocorticoides/análisis , Periodo Posprandial/fisiología , Saliva/química , Adulto , Ácidos y Sales Biliares/orina , Colesterol en la Dieta/administración & dosificación , Estudios Cruzados , Dieta/estadística & datos numéricos , Femenino , Humanos , Masculino , Adulto Joven
12.
Front Physiol ; 9: 848, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-30038578

RESUMEN

Salt-sensitive hypertension is common in glucocorticoid excess. Glucocorticoid resistance also presents with hypercortisolemia and hypertension but the relationship between salt intake and blood pressure (BP) is not well defined. GRßgeo/+ mice have global glucocorticoid receptor (GR) haploinsufficiency and increased BP. Here we examined the effect of high salt diet on BP, salt excretion and renal blood flow in GRßgeo/+mice. Basal BP was ∼10 mmHg higher in male GRßgeo/+ mice than in GR+/+ littermates. This modest increase was amplified by ∼10 mmHg following a high-salt diet in GRßgeo/+ mice. High salt reduced urinary aldosterone excretion but increased renal mineralocorticoid receptor expression in both genotypes. Corticosterone, and to a lesser extent deoxycorticosterone, excretion was increased in GRßgeo/+ mice following a high-salt challenge, consistent with enhanced 24 h production. GR+/+ mice increased fractional sodium excretion and reduced renal vascular resistance during the high salt challenge, retaining neutral sodium balance. In contrast, sodium excretion and renal vascular resistance did not adapt to high salt in GRßgeo/+ mice, resulting in transient sodium retention and sustained hypertension. With high-salt diet, Slc12a3 and Scnn1a mRNAs were higher in GRßgeo/+ than controls, and this was reflected in an exaggerated natriuretic response to thiazide and benzamil, inhibitors of NCC and ENaC, respectively. Reduction in GR expression causes salt-sensitivity and an adaptive failure of the renal vasculature and tubule, most likely reflecting sustained mineralocorticoid receptor activation. This provides a mechanistic basis to understand the hypertension associated with loss-of-function polymorphisms in GR in the context of habitually high salt intake.

13.
Psychoneuroendocrinology ; 89: 13-22, 2018 03.
Artículo en Inglés | MEDLINE | ID: mdl-29306773

RESUMEN

Chronic exposure to stress during midlife associates with subsequent age-related cognitive decline and may increase the vulnerability to develop psychiatric conditions. Increased hypothalamic-pituitary-adrenal (HPA) axis activity has been implicated in pathogenesis though any causative role for glucocorticoids is unestablished. This study investigated the contribution of local glucocorticoid regeneration by the intracellular enzyme 11ß-hydroxysteroid dehydrogenase type 1 (11ß-HSD1), in persisting midlife stress-induced behavioral effects in mice. Middle-aged (10 months old) 11ß-HSD1-deficient mice and wild-type congenic controls were randomly assigned to 28 days of chronic unpredictable stress or left undisturbed (non-stressed). All mice underwent behavioral testing at the end of the stress/non-stress period and again 6-7 months later. Chronic stress impaired spatial memory in middle-aged wild-type mice. The effects, involving a wide spectrum of behavioral modalities, persisted for 6-7 months after cessation of stress into early senescence. Enduring effects after midlife stress included impaired spatial memory, enhanced contextual fear memory, impaired fear extinction, heightened anxiety, depressive-like behavior, as well as reduced hippocampal glucocorticoid receptor mRNA expression. In contrast, 11ß-HSD1 deficient mice resisted both immediate and enduring effects of chronic stress, despite similar stress-induced increases in systemic glucocorticoid activity during midlife stress. In conclusion, chronic stress in midlife exerts persisting effects leading to cognitive and affective dysfunction in old age via mechanisms that depend, at least in part, on brain glucocorticoids generated locally by 11ß-HSD1. This finding supports selective 11ß-HSD1 inhibition as a novel therapeutic target to ameliorate the long-term consequences of stress-related psychiatric disorders in midlife.


Asunto(s)
11-beta-Hidroxiesteroide Deshidrogenasa de Tipo 1/metabolismo , Afecto/fisiología , Estrés Psicológico/fisiopatología , Animales , Corticosterona/metabolismo , Miedo/fisiología , Glucocorticoides/metabolismo , Hipocampo/metabolismo , Sistema Hipotálamo-Hipofisario/metabolismo , Masculino , Memoria/fisiología , Trastornos de la Memoria/metabolismo , Ratones , Ratones Endogámicos C57BL , Sistema Hipófiso-Suprarrenal/metabolismo , Memoria Espacial/fisiología
14.
Nat Commun ; 9(1): 4525, 2018 10 30.
Artículo en Inglés | MEDLINE | ID: mdl-30375380

RESUMEN

Malaria reduces host fitness and survival by pathogen-mediated damage and inflammation. Disease tolerance mechanisms counter these negative effects without decreasing pathogen load. Here, we demonstrate that in four different mouse models of malaria, adrenal hormones confer disease tolerance and protect against early death, independently of parasitemia. Surprisingly, adrenalectomy differentially affects malaria-induced inflammation by increasing circulating cytokines and inflammation in the brain but not in the liver or lung. Furthermore, without affecting the transcription of hepatic gluconeogenic enzymes, adrenalectomy causes exhaustion of hepatic glycogen and insulin-independent lethal hypoglycemia upon infection. This hypoglycemia is not prevented by glucose administration or TNF-α neutralization. In contrast, treatment with a synthetic glucocorticoid (dexamethasone) prevents the hypoglycemia, lowers cerebral cytokine expression and increases survival rates. Overall, we conclude that in malaria, adrenal hormones do not protect against lung and liver inflammation. Instead, they prevent excessive systemic and brain inflammation and severe hypoglycemia, thereby contributing to tolerance.


Asunto(s)
Glándulas Suprarrenales/metabolismo , Encéfalo/inmunología , Citocinas/inmunología , Hormonas/inmunología , Hipoglucemia/inmunología , Hígado/inmunología , Pulmón/inmunología , Malaria/inmunología , Glándulas Suprarrenales/inmunología , Adrenalectomía , Animales , Glucemia/efectos de los fármacos , Encéfalo/efectos de los fármacos , Corticosterona/inmunología , Corticosterona/metabolismo , Citocinas/efectos de los fármacos , Dexametasona/farmacología , Modelos Animales de Enfermedad , Epinefrina/inmunología , Epinefrina/metabolismo , Glucocorticoides/inmunología , Glucocorticoides/farmacología , Glucógeno/metabolismo , Hidrocortisona/inmunología , Hidrocortisona/metabolismo , Inflamación , Hígado/efectos de los fármacos , Pulmón/efectos de los fármacos , Ratones , Mineralocorticoides/inmunología , Mineralocorticoides/metabolismo , Norepinefrina/inmunología , Norepinefrina/metabolismo , Plasmodium berghei , Plasmodium chabaudi , Tasa de Supervivencia
16.
Endocrinology ; 148(3): 961-6, 2007 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-17170103

RESUMEN

11beta-Hydroxysteroid dehydrogenase type 1 (11beta-HSD1) acts as a reductase in vivo, regenerating active glucocorticoids within cells from circulating inert 11-keto forms, thus amplifying local glucocorticoid action. 11beta-HSD1 is predominantly expressed in liver and also adipose tissue and brain. Mice deficient in 11beta-HSD1 (11beta-HSD1(-/-)) exhibit adrenal hyperplasia, raised basal corticosterone levels, and increased hypothalamic-pituitary-adrenal (HPA) axis responses to stress. Whereas reduced peripheral glucocorticoid regeneration may explain adrenal hypertrophy and exaggerated stress responses, elevated basal glucocorticoid levels support a role for 11beta-HSD1 within the brain in amplifying glucocorticoid feedback. To test this hypothesis, apolipoprotein E-HSD1 mice overexpressing 11beta-HSD1 in liver were intercrossed with 11beta-HSD1(-/-) mice to determine whether complementation of hepatic 11beta-HSD1 can restore adrenal and HPA defects. Transgene-mediated delivery of 11beta-HSD1 activity to the liver rescued adrenal hyperplasia and reversed exaggerated HPA stress responses in 11beta-HSD1(-/-) mice. Unexpectedly, elevated nadir plasma corticosterone levels were also restored to control levels. Consistent with this, CYP11B1 mRNA expression in the adrenal cortex of 11beta-HSD1(-/-) mice was increased by 50% but returned to control levels in 11beta-HSD1(-/-) mice bearing the apolipoprotein E-HSD1 transgene. 11beta-HSD1(-/-) mice have lower plasma glucose levels, but the fall in plasma corticosterone with sucrose supplementation was similar in 11beta-HSD1(-/-) and control mice, suggesting glucose deficiency is not the main mechanism whereby basal corticosterone levels are elevated in the null mice. Thus, regeneration of glucocorticoids by 11beta-HSD1 in the liver normalizes all aspects of HPA axis dysregulation in 11beta-HSD1(-/-) mice, without restoration of enzyme activity in key feedback areas of the forebrain. Therefore, hepatic glucocorticoid metabolism influences basal as well as stress-associated functions of the HPA axis.


Asunto(s)
11-beta-Hidroxiesteroide Deshidrogenasa de Tipo 1/genética , Terapia Genética , Sistema Hipotálamo-Hipofisario/fisiopatología , Hígado/metabolismo , Sistema Hipófiso-Suprarrenal/fisiopatología , 11-beta-Hidroxiesteroide Deshidrogenasa de Tipo 1/metabolismo , Glándulas Suprarrenales/metabolismo , Hiperplasia Suprarrenal Congénita/genética , Hiperplasia Suprarrenal Congénita/terapia , Animales , Apolipoproteínas E/genética , Ritmo Circadiano , Corticosterona/sangre , Sistema Hipotálamo-Hipofisario/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Especificidad de Órganos , Sistema Hipófiso-Suprarrenal/metabolismo , Transgenes
17.
J Clin Invest ; 112(1): 83-90, 2003 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-12840062

RESUMEN

Obesity is closely associated with the metabolic syndrome, a combination of disorders including insulin resistance, diabetes, dyslipidemia, and hypertension. A role for local glucocorticoid reamplification in obesity and the metabolic syndrome has been suggested. The enzyme 11beta-hydroxysteroid dehydrogenase type 1 (11beta-HSD1) regenerates active cortisol from inactive 11-keto forms, and aP2-HSD1 mice with relative transgenic overexpression of this enzyme in fat cells develop visceral obesity with insulin resistance and dyslipidemia. Here we report that aP2-HSD1 mice also have high arterial blood pressure (BP). The mice have increased sensitivity to dietary salt and increased plasma levels of angiotensinogen, angiotensin II, and aldosterone. This hypertension is abolished by selective angiotensin II receptor AT-1 antagonist at a low dose that does not affect BP in non-Tg littermates. These findings suggest that activation of the circulating renin-angiotensin system (RAS) develops in aP2-HSD1 mice. The long-term hypertension is further reflected by an appreciable hypertrophy and hyperplasia of the distal tubule epithelium of the nephron, resembling salt-sensitive or angiotensin II-mediated hypertension. Taken together, our findings suggest that overexpression of 11beta-HSD1 in fat is sufficient to cause salt-sensitive hypertension mediated by an activated RAS. The potential role of adipose 11beta-HSD1 in mediating critical features of the metabolic syndrome extends beyond obesity and metabolic complications to include the most central cardiovascular feature of this disorder.


Asunto(s)
Tejido Adiposo/enzimología , Glucocorticoides/fisiología , Hidroxiesteroide Deshidrogenasas/fisiología , Hipertensión/etiología , 11-beta-Hidroxiesteroide Deshidrogenasas , Animales , Riñón/patología , Masculino , Ratones , Ratones Transgénicos , Sistema Renina-Angiotensina/fisiología , Cloruro de Sodio/farmacología
18.
J Endocrinol ; 194(1): 161-70, 2007 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-17592030

RESUMEN

Proopiomelanocortin (POMC) deficiency causes severe obesity through hyperphagia of hypothalamic origin. However, low glucocorticoid levels caused by adrenal insufficiency mitigate against insulin resistance, hyperphagia and fat accretion in Pomc-/- mice. Upon exogenous glucocorticoid replacement, corticosterone-supplemented (CORT) Pomc-/- mice show exaggerated responses, including excessive fat accumulation, hyperleptinaemia and insulin resistance. To investigate the peripheral mechanisms underlying this glucocorticoid hypersensitivity, we examined the expression levels of key determinants and targets of glucocorticoid action in adipose tissue and liver. Despite lower basal expression of 11beta-hydroxysteroid dehydrogenase type 1 (11beta-HSD1), which generates active glucocorticoids within cells, CORT-mediated induction of 11beta-HSD1 mRNA levels was more pronounced in adipose tissues of Pomc-/- mice. Similarly, CORT treatment increased lipoprotein lipase mRNA levels in all fat depots in Pomc-/- mice, consistent with exaggerated fat accumulation. Glucocorticoid receptor (GR) mRNA levels were selectively elevated in liver and retroperitoneal fat of Pomc-/- mice but were corrected by CORT in the latter depot. In liver, CORT increased phosphoenolpyruvate carboxykinase mRNA levels specifically in Pomc-/- mice, consistent with their insulin-resistant phenotype. Furthermore, CORT induced hypertension in Pomc-/- mice, independently of adipose or liver renin-angiotensin system activation. These data suggest that CORT-inducible 11beta-HSD1 expression in fat contributes to the adverse cardiometabolic effects of CORT in POMC deficiency, whereas higher GR levels may be more important in liver.


Asunto(s)
Tejido Adiposo/metabolismo , Corticosterona/uso terapéutico , Glucocorticoides/metabolismo , Hiperfagia/tratamiento farmacológico , Hígado/metabolismo , Proopiomelanocortina/deficiencia , 11-beta-Hidroxiesteroide Deshidrogenasa de Tipo 1/genética , 11-beta-Hidroxiesteroide Deshidrogenasa de Tipo 1/metabolismo , Animales , Corticosterona/metabolismo , Hiperfagia/metabolismo , Hipertensión/etiología , Hipertensión/metabolismo , Resistencia a la Insulina , Lipoproteína Lipasa/genética , Lipoproteína Lipasa/metabolismo , Ratones , Ratones Noqueados , Fosfoenolpiruvato Carboxiquinasa (ATP)/genética , Fosfoenolpiruvato Carboxiquinasa (ATP)/metabolismo , Proopiomelanocortina/genética , ARN Mensajero/análisis , Receptores de Glucocorticoides/genética , Receptores de Glucocorticoides/metabolismo
19.
Endocr Connect ; 6(7): 446-457, 2017 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-28720595

RESUMEN

Chronic ACTH exposure is associated with adrenal hypertrophy and steroidogenesis. The underlying molecular processes in mice have been analysed by microarray, histological and immunohistochemical techniques. Synacthen infused for 2 weeks markedly increased adrenal mass and plasma corticosterone levels. Microarray analysis found greater than 2-fold changes in expression of 928 genes (P < 0.001; 397 up, 531 down). These clustered in pathways involved in signalling, sterol/lipid metabolism, cell proliferation/hypertrophy and apoptosis. Signalling genes included some implicated in adrenal adenomas but also upregulated genes associated with cyclic AMP and downregulated genes associated with aldosterone synthesis. Sterol metabolism genes were those promoting cholesterol supply (Scarb1, Sqle, Apoa1) and disposal (Cyp27a1, Cyp7b1). Oil red O staining showed lipid depletion consistent with reduced expression of genes involved in lipid synthesis. Genes involved in steroidogenesis (Star, Cyp11a1, Cyp11b1) were modestly affected (P < 0.05; <1.3-fold). Increased Ki67, Ccna2, Ccnb2 and Tk1 expression complemented immunohistochemical evidence of a 3-fold change in cell proliferation. Growth arrest genes, Cdkn1a and Cdkn1c, which are known to be active in hypertrophied cells, were increased >4-fold and cross-sectional area of fasciculata cells was 2-fold greater. In contrast, genes associated with apoptosis (eg Casp12, Clu,) were downregulated and apoptotic cells (Tunel staining) were fewer (P < 0.001) and more widely distributed throughout the cortex. In summary, long-term steroidogenesis with ACTH excess is sustained by genes controlling cholesterol supply and adrenal mass. ACTH effects on adrenal morphology and genes controlling cell hypertrophy, proliferation and apoptosis suggest the involvement of different cell types and separate molecular pathways.

20.
J Endocrinol ; 233(3): 315-327, 2017 06.
Artículo en Inglés | MEDLINE | ID: mdl-28522730

RESUMEN

We have previously demonstrated that neutrophil recruitment to the heart following myocardial infarction (MI) is enhanced in mice lacking 11ß-hydroxysteroid dehydrogenase type 1 (11ß-HSD1) that regenerates active glucocorticoid within cells from intrinsically inert metabolites. The present study aimed to identify the mechanism of regulation. In a mouse model of MI, neutrophil mobilization to blood and recruitment to the heart were higher in 11ß-HSD1-deficient (Hsd11b1-/- ) relative to wild-type (WT) mice, despite similar initial injury and circulating glucocorticoid. In bone marrow chimeric mice, neutrophil mobilization was increased when 11ß-HSD1 was absent from host cells, but not when absent from donor bone marrow-derived cells. Consistent with a role for 11ß-HSD1 in 'host' myocardium, gene expression of a subset of neutrophil chemoattractants, including the chemokines Cxcl2 and Cxcl5, was selectively increased in the myocardium of Hsd11b1-/- mice relative to WT. SM22α-Cre directed disruption of Hsd11b1 in smooth muscle and cardiomyocytes had no effect on neutrophil recruitment. Expression of Cxcl2 and Cxcl5 was elevated in fibroblast fractions isolated from hearts of Hsd11b1-/- mice post MI and provision of either corticosterone or of the 11ß-HSD1 substrate, 11-dehydrocorticosterone, to cultured murine cardiac fibroblasts suppressed IL-1α-induced expression of Cxcl2 and Cxcl5 These data identify suppression of CXCL2 and CXCL5 chemoattractant expression by 11ß-HSD1 as a novel mechanism with potential for regulation of neutrophil recruitment to the injured myocardium, and cardiac fibroblasts as a key site for intracellular glucocorticoid regeneration during acute inflammation following myocardial injury.


Asunto(s)
11-beta-Hidroxiesteroide Deshidrogenasa de Tipo 1/metabolismo , Quimiocina CXCL2/metabolismo , Quimiocina CXCL5/metabolismo , Fibroblastos/fisiología , Neutrófilos/fisiología , 11-beta-Hidroxiesteroide Deshidrogenasa de Tipo 1/genética , Animales , Células de la Médula Ósea , Células Cultivadas , Quimiocina CXCL5/genética , Corticosterona/análogos & derivados , Corticosterona/farmacología , Masculino , Ratones , Ratones Noqueados , Proteínas de Microfilamentos/genética , Proteínas de Microfilamentos/metabolismo , Proteínas Musculares/genética , Proteínas Musculares/metabolismo , Infarto del Miocardio
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA