Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 34
Filter
1.
Cardiovasc Res ; 119(8): 1740-1750, 2023 Jul 06.
Article in English | MEDLINE | ID: mdl-36368681

ABSTRACT

AIMS: High salt intake is common and contributes to poor cardiovascular health. Urinary sodium excretion correlates directly with glucocorticoid excretion in humans and experimental animals. We hypothesized that high salt intake activates the hypothalamic-pituitary-adrenal axis activation and leads to sustained glucocorticoid excess. METHODS AND RESULTS: In male C57BL/6 mice, high salt intake for 2-8 weeks caused an increase in diurnal peak levels of plasma corticosterone. After 2 weeks, high salt increased Crh and Pomc mRNA abundance in the hypothalamus and anterior pituitary, consistent with basal hypothalamic-pituitary-adrenal axis activation. Additionally, high salt intake amplified glucocorticoid response to restraint stress, indicative of enhanced axis sensitivity. The binding capacity of Corticosteroid-Binding Globulin was reduced and its encoding mRNA downregulated in the liver. In the hippocampus and anterior pituitary, Fkbp5 mRNA levels were increased, indicating increased glucocorticoid exposure. The mRNA expression of the glucocorticoid-regenerating enzyme, 11ß-hydroxysteroid dehydrogenase Type 1, was increased in these brain areas and in the liver. Sustained high salt intake activated a water conservation response by the kidney, increasing plasma levels of the vasopressin surrogate, copeptin. Increased mRNA abundance of Tonebp and Avpr1b in the anterior pituitary suggested that vasopressin signalling contributes to hypothalamic-pituitary-adrenal axis activation by high salt diet. CONCLUSION: Chronic high salt intake amplifies basal and stress-induced glucocorticoid levels and resets glucocorticoid biology centrally, peripherally and within cells.


Subject(s)
Glucocorticoids , Hypothalamo-Hypophyseal System , Humans , Mice , Animals , Male , Glucocorticoids/metabolism , Hypothalamo-Hypophyseal System/metabolism , Sodium Chloride, Dietary , Pituitary-Adrenal System/metabolism , Mice, Inbred C57BL , Vasopressins/genetics , Vasopressins/metabolism , RNA, Messenger/genetics , RNA, Messenger/metabolism
2.
Front Endocrinol (Lausanne) ; 13: 1027164, 2022.
Article in English | MEDLINE | ID: mdl-36465608

ABSTRACT

Decidualization is the hormone-dependent process of endometrial remodeling that is essential for fertility and reproductive health. It is characterized by dynamic changes in the endometrial stromal compartment including differentiation of fibroblasts, immune cell trafficking and vascular remodeling. Deficits in decidualization are implicated in disorders of pregnancy such as implantation failure, intra-uterine growth restriction, and pre-eclampsia. Androgens are key regulators of decidualization that promote optimal differentiation of stromal fibroblasts and activation of downstream signaling pathways required for endometrial remodeling. We have shown that androgen biosynthesis, via 5α-reductase-dependent production of dihydrotestosterone, is required for optimal decidualization of human stromal fibroblasts in vitro, but whether this is required for decidualization in vivo has not been tested. In the current study we used steroid 5α-reductase type 1 (SRD5A1) deficient mice (Srd5a1-/- mice) and a validated model of induced decidualization to investigate the role of SRD5A1 and intracrine androgen signaling in endometrial decidualization. We measured decidualization response (weight/proportion), transcriptomic changes, and morphological and functional parameters of vascular development. These investigations revealed a striking effect of 5α-reductase deficiency on the decidualization response. Furthermore, vessel permeability and transcriptional regulation of angiogenesis signaling pathways, particularly those that involved vascular endothelial growth factor (VEGF), were disrupted in the absence of 5α-reductase. In Srd5a1-/- mice, injection of dihydrotestosterone co-incident with decidualization restored decidualization responses, vessel permeability, and expression of angiogenesis genes to wild type levels. Androgen availability declines with age which may contribute to age-related risk of pregnancy disorders. These findings show that intracrine androgen signaling is required for optimal decidualization in vivo and confirm a major role for androgens in the development of the vasculature during decidualization through regulation of the VEGF pathway. These findings highlight new opportunities for improving age-related deficits in fertility and pregnancy health by targeting androgen-dependent signaling in the endometrium.


Subject(s)
3-Oxo-5-alpha-Steroid 4-Dehydrogenase , Decidua , Vascular Remodeling , Animals , Female , Mice , Pregnancy , 3-Oxo-5-alpha-Steroid 4-Dehydrogenase/genetics , 3-Oxo-5-alpha-Steroid 4-Dehydrogenase/metabolism , Androgens/pharmacology , Cholestenone 5 alpha-Reductase/genetics , Cholestenone 5 alpha-Reductase/metabolism , Decidua/drug effects , Decidua/metabolism , Dihydrotestosterone/pharmacology , Endometrium/drug effects , Endometrium/metabolism , Vascular Endothelial Growth Factor A/genetics , Vascular Endothelial Growth Factor A/metabolism , Vascular Remodeling/drug effects , Vascular Remodeling/genetics , Vascular Remodeling/physiology
3.
Hypertension ; 75(5): 1213-1222, 2020 05.
Article in English | MEDLINE | ID: mdl-32200679

ABSTRACT

GPR81 (G-protein-coupled receptor 81) is highly expressed in adipocytes, and activation by the endogenous ligand lactate inhibits lipolysis. GPR81 is also expressed in the heart, liver, and kidney, but roles in nonadipose tissues are poorly defined. GPR81 agonists, developed to improve blood lipid profile, might also provide insights into GPR81 physiology. Here, we assessed the blood pressure and renal hemodynamic responses to the GPR81 agonist, AZ'5538. In male wild-type mice, intravenous AZ'5538 infusion caused a rapid and sustained increase in systolic and diastolic blood pressure. Renal artery blood flow, intrarenal tissue perfusion, and glomerular filtration rate were all significantly reduced. AZ'5538 had no effect on blood pressure or renal hemodynamics in Gpr81-/- mice. Gpr81 mRNA was expressed in renal artery vascular smooth muscle, in the afferent arteriole, in glomerular and medullary perivascular cells, and in pericyte-like cells isolated from kidney. Intravenous AZ'5538 increased plasma ET-1 (endothelin 1), and pretreatment with BQ123 (endothelin-A receptor antagonist) prevented the pressor effects of GPR81 activation, whereas BQ788 (endothelin-B receptor antagonist) did not. Renal ischemia-reperfusion injury, which increases renal extracellular lactate, increased the renal expression of genes encoding ET-1, KIM-1 (Kidney Injury Molecule 1), collagen type 1-α1, TNF-α (tumor necrosis factor-α), and F4/80 in wild-type mice but not in Gpr81-/- mice. In summary, activation of GPR81 in vascular smooth muscle and perivascular cells regulates renal hemodynamics, mediated by release of the potent vasoconstrictor ET-1. This suggests that lactate may be a paracrine regulator of renal blood flow, particularly relevant when extracellular lactate is high as occurs during ischemic renal disease.


Subject(s)
Endothelin-1/physiology , Hemodynamics/drug effects , Receptors, G-Protein-Coupled/agonists , Animals , Arteries/drug effects , Blood Pressure/drug effects , Blood Pressure/physiology , Bosentan/pharmacology , Endothelin-1/blood , Glomerular Filtration Rate/drug effects , Heart/drug effects , Hemodynamics/physiology , Infusions, Intravenous , Kidney/blood supply , Kidney/drug effects , Lactates/blood , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Muscle, Smooth, Vascular/drug effects , Muscle, Smooth, Vascular/metabolism , Oligopeptides/pharmacology , Paracrine Communication , Peptides, Cyclic/pharmacology , Pericytes/drug effects , Pericytes/metabolism , Piperidines/pharmacology , RNA, Messenger/biosynthesis , RNA, Messenger/genetics , Receptors, G-Protein-Coupled/deficiency , Receptors, G-Protein-Coupled/genetics , Receptors, G-Protein-Coupled/physiology , Renal Circulation/drug effects , Renal Circulation/physiology , Reperfusion Injury/blood , Reperfusion Injury/drug therapy , Reperfusion Injury/genetics , Reperfusion Injury/physiopathology
4.
Endocrinology ; 160(9): 2061-2073, 2019 09 01.
Article in English | MEDLINE | ID: mdl-31199473

ABSTRACT

Inhibition of 5α-reductases impairs androgen and glucocorticoid metabolism and induces insulin resistance in humans and rodents. The contribution of hepatic glucocorticoids to these adverse metabolic changes was assessed using a liver-selective glucocorticoid receptor (GR) antagonist, A-348441. Mice lacking 5α-reductase 1 (5αR1-KO) and their littermate controls were studied during consumption of a high-fat diet, with or without A-348441(120 mg/kg/d). Male C57BL/6 mice (age, 12 weeks) receiving dutasteride (1.8 mg/kg/d)) or vehicle with consumption of a high-fat diet, with or without A-348441, were also studied. In the 5αR1-KO mice, hepatic GR antagonism improved diet-induced insulin resistance but not more than that of the controls. Liver steatosis was not affected by hepatic GR antagonism in either 5αR1KO mice or littermate controls. In a second model of 5α-reductase inhibition using dutasteride and hepatic GR antagonism with A-348441 attenuated the excess weight gain resulting from dutasteride (mean ± SEM, 7.03 ± 0.5 vs 2.13 ± 0.4 g; dutasteride vs dutasteride plus A-348441; P < 0.05) and normalized the associated hyperinsulinemia after glucose challenge (area under the curve, 235.9 ± 17 vs 329.3 ± 16 vs 198.4 ± 25 ng/mL/min; high fat vs high fat plus dutasteride vs high fat plus dutasteride plus A-348441, respectively; P < 0.05). However, A-348441 again did not reverse dutasteride-induced liver steatosis. Thus, overall hepatic GR antagonism improved the insulin resistance but not the steatosis induced by a high-fat diet. Moreover, it attenuated the excessive insulin resistance caused by pharmacological inhibition of 5α-reductases but not genetic disruption of 5αR1. The use of dutasteride might increase the risk of type 2 diabetes mellitus and reduced exposure to glucocorticoids might be beneficial.


Subject(s)
Cholestenone 5 alpha-Reductase/deficiency , Liver/physiology , Receptors, Glucocorticoid/physiology , Animals , Cholestenone 5 alpha-Reductase/physiology , Cholic Acids/pharmacology , Diet, High-Fat , Dutasteride/pharmacology , Estrone/analogs & derivatives , Estrone/pharmacology , Gluconeogenesis , Insulin Resistance , Lipid Metabolism , Male , Mice , Mice, Inbred C57BL
5.
Endocrinology ; 158(11): 4047-4063, 2017 11 01.
Article in English | MEDLINE | ID: mdl-28938454

ABSTRACT

The enzyme 11ß-hydroxysteroid dehydrogenase type 2 (11ß-HSD2) has an essential role in aldosterone target tissues, conferring aldosterone selectivity for the mineralocorticoid receptor (MR) by converting 11ß-hydroxyglucocorticoids to inactive 11-ketosteroids. Congenital deficiency of 11ß-HSD2 causes a form of salt-sensitive hypertension known as the syndrome of apparent mineralocorticoid excess. The disease phenotype, which ranges from mild to severe, correlates well with reduction in enzyme activity. Furthermore, polymorphisms in the 11ß-HSD2 coding gene (HSD11B2) have been linked to high blood pressure and salt sensitivity, major cardiovascular risk factors. 11ß-HSD2 expression is controlled by different factors such as cytokines, sex steroids, or vasopressin, but posttranslational modulation of its activity has not been explored. Analysis of 11ß-HSD2 sequence revealed a consensus site for conjugation of small ubiquitin-related modifier (SUMO) peptide, a major posttranslational regulatory event in several cellular processes. Our results demonstrate that 11ß-HSD2 is SUMOylated at lysine 266. Non-SUMOylatable mutant K266R showed slightly higher substrate affinity and decreased Vmax, but no effects on protein stability or subcellular localization. Despite mild changes in enzyme activity, mutant K266R was unable to prevent cortisol-dependent MR nuclear translocation. The same effect was achieved by coexpression of wild-type 11ß-HSD2 with sentrin-specific protease 1, a protease that catalyzes SUMO deconjugation. In the presence of 11ß-HSD2-K266R, increased nuclear MR localization did not correlate with increased response to cortisol or increased recruitment of transcriptional coregulators. Taken together, our data suggests that SUMOylation of 11ß-HSD2 at residue K266 modulates cortisol-mediated MR nuclear translocation independently of effects on transactivation.


Subject(s)
11-beta-Hydroxysteroid Dehydrogenase Type 2/metabolism , Cell Nucleus/drug effects , Cell Nucleus/metabolism , Hydrocortisone/pharmacology , Receptors, Mineralocorticoid/metabolism , Sumoylation , 11-beta-Hydroxysteroid Dehydrogenase Type 2/chemistry , 11-beta-Hydroxysteroid Dehydrogenase Type 2/genetics , Active Transport, Cell Nucleus/drug effects , Active Transport, Cell Nucleus/genetics , Amino Acid Sequence , Animals , COS Cells , Chlorocebus aethiops , Humans , Models, Molecular , Protein Interaction Domains and Motifs/genetics , Protein Transport/drug effects , Protein Transport/genetics , Receptors, Mineralocorticoid/chemistry , Transcriptional Activation/drug effects
6.
Biochem Pharmacol ; 129: 73-84, 2017 04 01.
Article in English | MEDLINE | ID: mdl-28131845

ABSTRACT

Use of topical glucocorticoid for inflammatory skin conditions is limited by systemic and local side-effects. This investigation addressed the hypothesis that topical 5α-tetrahydrocorticosterone (5αTHB, a corticosterone metabolite) inhibits dermal inflammation without affecting processes responsible for skin thinning and impaired wound healing. The topical anti-inflammatory properties of 5αTHB were compared with those of corticosterone in C57Bl/6 male mice with irritant dermatitis induced by croton oil, whereas its effects on angiogenesis, inflammation, and collagen deposition were investigated by subcutaneous sponge implantation. 5αTHB decreased dermal swelling and total cell infiltration associated with dermatitis similarly to corticosterone after 24h, although at a five fold higher dose, but in contrast did not have any effects after 6h. Pre-treatment with the glucocorticoid receptor antagonist RU486 attenuated the effect of corticosterone on swelling at 24h, but not that of 5αTHB. After 24h 5αTHB reduced myeloperoxidase activity (representative of neutrophil infiltration) to a greater extent than corticosterone. At equipotent anti-inflammatory doses 5αTHB suppressed angiogenesis to a limited extent, unlike corticosterone which substantially decreased angiogenesis compared to vehicle. Furthermore, 5αTHB reduced only endothelial cell recruitment in sponges whereas corticosterone also inhibited smooth muscle cell recruitment and decreased transcripts of angiogenic and inflammatory genes. Strikingly, corticosterone, but not 5αTHB, reduced collagen deposition. However, both 5αTHB and corticosterone attenuated macrophage infiltration into sponges. In conclusion, 5αTHB displays the profile of a safer topical anti-inflammatory compound. With limited effects on angiogenesis and extracellular matrix, it is less likely to impair wound healing or cause skin thinning.


Subject(s)
Corticosterone/analogs & derivatives , Disease Models, Animal , Inflammation/drug therapy , Administration, Topical , Animals , Corticosterone/administration & dosage , Corticosterone/therapeutic use , Male , Mice , Mice, Inbred C57BL
7.
J Endocrinol ; 232(1): 29-36, 2017 01.
Article in English | MEDLINE | ID: mdl-27647861

ABSTRACT

5α-Reductases irreversibly catalyse A-ring reduction of pregnene steroids, including glucocorticoids and androgens. Genetic disruption of 5α-reductase 1 in male mice impairs glucocorticoid clearance and predisposes to glucose intolerance and hepatic steatosis upon metabolic challenge. However, it is unclear whether this is driven by changes in androgen and/or glucocorticoid action. Female mice with transgenic disruption of 5α-reductase 1 (5αR1-KO) were studied, representing a 'low androgen' state. Glucocorticoid clearance and stress responses were studied in mice aged 6 months. Metabolism was assessed in mice on normal chow (aged 6 and 12 m) and also in a separate cohort following 1-month high-fat diet (aged 3 m). Female 5αR1-KO mice had adrenal suppression (44% lower AUC corticosterone after stress), and upon corticosterone infusion, accumulated hepatic glucocorticoids (~27% increased corticosterone). Female 5αR1-KO mice aged 6 m fed normal chow demonstrated insulin resistance (~35% increased area under curve (AUC) for insulin upon glucose tolerance testing) and hepatic steatosis (~33% increased hepatic triglycerides) compared with controls. This progressed to obesity (~12% increased body weight) and sustained insulin resistance (~38% increased AUC insulin) by age 12 m. Hepatic transcript profiles supported impaired lipid ß-oxidation and increased triglyceride storage. Female 5αR1-KO mice were also predisposed to develop high-fat diet-induced insulin resistance. Exaggerated predisposition to metabolic disorders in female mice, compared with that seen in male mice, after disruption of 5αR1 suggests phenotypic changes may be underpinned by altered metabolism of glucocorticoids rather than androgens.


Subject(s)
Cholestenone 5 alpha-Reductase/genetics , Corticosterone/blood , Energy Metabolism/physiology , Hypothalamo-Hypophyseal System/metabolism , Insulin Resistance/physiology , Liver/metabolism , Pituitary-Adrenal System/metabolism , Stress, Physiological/physiology , Animals , Cholestenone 5 alpha-Reductase/metabolism , Corticosterone/pharmacology , Diet, High-Fat , Female , Hypothalamo-Hypophyseal System/drug effects , Hypothalamus/drug effects , Hypothalamus/metabolism , Liver/drug effects , Mice , Mice, Transgenic , Pituitary-Adrenal System/drug effects , Receptors, Glucocorticoid/metabolism
8.
Anal Chem ; 88(21): 10362-10367, 2016 11 01.
Article in English | MEDLINE | ID: mdl-27676129

ABSTRACT

Androgens are essential for male development and reproductive function. They are transported to their site of action as blood-borne endocrine hormones but can also be produced within tissues to act in intracrine and paracrine fashions. Because of this, circulating concentrations may not accurately reflect the androgenic influence within specific tissue microenvironments. Mass spectrometry imaging permits regional analysis of small molecular species directly from tissue surfaces. However, due to poor ionization and localized ion suppression, steroid hormones are difficult to detect. Here, derivatization with Girard T reagent was used to charge-tag testosterone and 5α-dihydrotestosterone allowing direct detection of these steroids in mouse testes, in both basal and maximally stimulated states, and in rat prostate. Limits of detection were ∼0.1 pg for testosterone. Exemplary detection of endogenous steroids was achieved by matrix-assisted laser desorption ionization and either Fourier transform ion cyclotron resonance detection (at 150 µm spatial resolution) or quadrupole-time-of-flight detection (at 50 µm spatial resolution). Structural confirmation was achieved by collision induced fragmentation following liquid extraction surface analysis and electrospray ionization. This application broadens the scope for derivatization strategies on tissue surfaces to elucidate local endocrine signaling in health and disease.

9.
Sci Transl Med ; 8(352): 352ra109, 2016 08 17.
Article in English | MEDLINE | ID: mdl-27535620

ABSTRACT

The aim of treatment in congenital adrenal hyperplasia is to suppress excess adrenal androgens while achieving physiological glucocorticoid replacement. However, current glucocorticoid replacement regimes are inadequate because doses sufficient to suppress excess androgens almost invariably induce adverse metabolic effects. Although both cortisol and corticosterone are glucocorticoids that circulate in human plasma, any physiological role for corticosterone has been neglected. In the brain, the adenosine 5'-triphosphate-binding cassette transporter ABCB1 exports cortisol but not corticosterone. Conversely, ABCC1 exports corticosterone but not cortisol. We show that ABCC1, but not ABCB1, is expressed in human adipose and that ABCC1 inhibition increases intracellular corticosterone, but not cortisol, and induces glucocorticoid-responsive gene transcription in human adipocytes. Both C57Bl/6 mice treated with the ABCC1 inhibitor probenecid and FVB mice with deletion of Abcc1 accumulated more corticosterone than cortisol in adipose after adrenalectomy and corticosteroid infusion. This accumulation was sufficient to increase glucocorticoid-responsive adipose transcript expression. In human adipose tissue, tissue corticosterone concentrations were consistently low, and ABCC1 mRNA was up-regulated in obesity. To test the hypothesis that corticosterone effectively suppresses adrenocorticotropic hormone (ACTH) without the metabolic adverse effects of cortisol, we infused cortisol or corticosterone in patients with Addison's disease. ACTH suppression was similar, but subcutaneous adipose transcripts of glucocorticoid-responsive genes were higher after infusion with cortisol rather than with corticosterone. These data indicate that corticosterone may be a metabolically favorable alternative to cortisol for glucocorticoid replacement therapy when ACTH suppression is desirable, as in congenital adrenal hyperplasia, and justify development of a pharmaceutical preparation.


Subject(s)
Corticosterone/pharmacology , Hydrocortisone/pharmacology , Multidrug Resistance-Associated Proteins/metabolism , Addison Disease/drug therapy , Adipose Tissue/drug effects , Adipose Tissue/metabolism , Adrenal Hyperplasia, Congenital/drug therapy , Adrenal Hyperplasia, Congenital/genetics , Adrenal Hyperplasia, Congenital/metabolism , Adrenocorticotropic Hormone/antagonists & inhibitors , Animals , Biological Transport, Active , Brain/drug effects , Brain/metabolism , Cells, Cultured , Corticosterone/metabolism , Glucocorticoids/metabolism , Humans , Hydrocortisone/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Multidrug Resistance-Associated Proteins/deficiency , Multidrug Resistance-Associated Proteins/genetics , Obesity/metabolism , Organ Specificity , RNA, Messenger/genetics , RNA, Messenger/metabolism , Translational Research, Biomedical
10.
J Clin Endocrinol Metab ; 101(10): 3787-3793, 2016 10.
Article in English | MEDLINE | ID: mdl-27459533

ABSTRACT

CONTEXT: The mechanism of action of metformin remains unclear. Given the regulation of the cortisol-regenerating enzyme 11ßhydroxysteroid dehydrogenase 1 (11ßHSD1) by insulin and the limited efficacy of selective 11ßHSD1 inhibitors to lower blood glucose when co-prescribed with metformin, we hypothesized that metformin reduces 11ßHSD1 activity. OBJECTIVE: To determine whether metformin regulates 11ßHSD1 activity in vivo in obese men with and without type 2 diabetes mellitus. DESIGN: Double-blind, randomized, placebo-controlled, crossover study. SETTING: A hospital clinical research facility. PARTICIPANTS: Eight obese nondiabetic (OND) men and eight obese men with type 2 diabetes (ODM). INTERVENTION: Participants received 28 days of metformin (1 g twice daily), placebo, or (in the ODM group) gliclazide (80 mg twice daily) in random order. A deuterated cortisol infusion at the end of each phase measured cortisol regeneration by 11ßHSD1. Oral cortisone was given to measure hepatic 11ßHSD1 activity in the ODM group. The effect of metformin on 11ßHSD1 was also assessed in human hepatocytes and Simpson-Golabi-Behmel syndrome adipocytes. MAIN OUTCOME MEASURES: The effect of metformin on whole-body and hepatic 11ßHSD1 activity. RESULTS: Whole-body 11ßHSD1 activity was approximately 25% higher in the ODM group than the OND group. Metformin increased whole-body cortisol regeneration by 11ßHSD1 in both groups compared with placebo and gliclazide and tended to increase hepatic 11ßHSD1 activity. In vitro, metformin did not increase 11ßHSD1 activity in hepatocytes or adipocytes. CONCLUSIONS: Metformin increases whole-body cortisol generation by 11ßHSD1 probably through an indirect mechanism, potentially offsetting other metabolic benefits of metformin. Co-prescription with metformin should provide a greater target for selective 11ßHSD1 inhibitors.


Subject(s)
11-beta-Hydroxysteroid Dehydrogenase Type 1/metabolism , Diabetes Mellitus, Type 2/drug therapy , Gliclazide/pharmacology , Hydrocortisone/metabolism , Hypoglycemic Agents/pharmacology , Metformin/pharmacology , Obesity/drug therapy , 11-beta-Hydroxysteroid Dehydrogenase Type 1/drug effects , Comorbidity , Cross-Over Studies , Diabetes Mellitus, Type 2/epidemiology , Double-Blind Method , Gliclazide/administration & dosage , Humans , Hypoglycemic Agents/administration & dosage , Male , Metformin/administration & dosage , Obesity/epidemiology , Treatment Outcome
11.
J Clin Endocrinol Metab ; 101(5): 2040-6, 2016 05.
Article in English | MEDLINE | ID: mdl-26967690

ABSTRACT

CONTEXT: Deficiency of aromatase, the enzyme that catalyzes the conversion of androgens to estrogens, is associated with insulin resistance in humans and mice. OBJECTIVE: We hypothesized that pharmacological aromatase inhibition results in peripheral insulin resistance in humans. DESIGN: This was a double-blind, randomized, controlled, crossover study. SETTING: The study was conducted at a clinical research facility. PARTICIPANTS: Seventeen healthy male volunteers (18-50 y) participated in the study. INTERVENTION: The intervention included oral anastrozole (1 mg daily) and placebo, each for 6 weeks with a 2-week washout period. MAIN OUTCOME MEASURE: Glucose disposal and rates of lipolysis were measured during a stepwise hyperinsulinemic euglycemic clamp. Data are mean (SEM). RESULTS: Anastrozole therapy resulted in significant estradiol suppression (59.9 ± 3.6 vs 102.0 ± 5.7 pmol/L, P = < .001) and a more modest elevation of total T (25.8 ± 1.2 vs 21.4 ± 0.7 nmol/L, P = .003). Glucose infusion rate, during the low-dose insulin infusion, was lower after anastrozole administration (12.16 ± 1.33 vs 14.15 ± 1.55 µmol/kg·min, P = .024). No differences in hepatic glucose production or rate of lipolysis were observed. CONCLUSION: Aromatase inhibition reduces insulin sensitivity, with respect to peripheral glucose disposal, in healthy men. Local generation and action of estradiol, at the level of skeletal muscle, is likely to be an important determinant of insulin sensitivity.


Subject(s)
Aromatase Inhibitors/pharmacology , Glucose/metabolism , Insulin Resistance/physiology , Lipolysis/drug effects , Nitriles/pharmacology , Triazoles/pharmacology , Adolescent , Adult , Anastrozole , Cross-Over Studies , Double-Blind Method , Estradiol/blood , Glucose Clamp Technique , Healthy Volunteers , Humans , Liver/drug effects , Liver/metabolism , Male , Middle Aged , Muscle, Skeletal/drug effects , Muscle, Skeletal/metabolism , Testosterone/blood , Young Adult
12.
Diabetes ; 64(2): 447-58, 2015 Feb.
Article in English | MEDLINE | ID: mdl-25239636

ABSTRACT

5α-Reductase type 1 (5αR1) catalyses A-ring reduction of androgens and glucocorticoids in liver, potentially influencing hepatic manifestations of the metabolic syndrome. Male mice, homozygous for a disrupted 5αR1 allele (5αR1 knockout [KO] mice), were studied after metabolic (high-fat diet) and fibrotic (carbon tetrachloride [CCl4]) challenge. The effect of the 5α-reductase inhibitor finasteride on metabolism was investigated in male obese Zucker rats. While eating a high-fat diet, male 5αR1-KO mice demonstrated greater mean weight gain (21.6 ± 1.4 vs 16.2 ± 2.4 g), hyperinsulinemia (insulin area under the curve during glucose tolerance test 609 ± 103 vs. 313 ± 66 ng ⋅ mL(-1) ⋅ min), and hepatic steatosis (liver triglycerides 136.1 ± 17.0 vs. 89.3 ± 12.1 µmol ⋅ g(-1)). mRNA transcript profiles in liver were consistent with decreased fatty acid ß-oxidation and increased triglyceride storage. 5αR1-KO male mice were more susceptible to fibrosis after CCl4 administration (37% increase in collagen staining). The nonselective 5α-reductase inhibitor finasteride induced hyperinsulinemia and hepatic steatosis (10.6 ± 1.2 vs. 7.0 ± 1.0 µmol ⋅ g(-1)) in obese male Zucker rats, both intact and castrated. 5αR1 deficiency induces insulin resistance and hepatic steatosis, consistent with the intrahepatic accumulation of glucocorticoids, and predisposes to hepatic fibrosis. Hepatic steatosis is independent of androgens in rats. Variations in 5αR1 activity in obesity and with nonselective 5α-reductase inhibition in men with prostate disease may have important consequences for the onset and progression of metabolic liver disease.


Subject(s)
Cholestenone 5 alpha-Reductase/metabolism , Fatty Liver/etiology , Finasteride/pharmacology , Insulin Resistance , Liver Cirrhosis/metabolism , 5-alpha Reductase Inhibitors/pharmacology , Animals , Cholestenone 5 alpha-Reductase/genetics , Dietary Fats/administration & dosage , Dietary Fats/adverse effects , Enzyme Assays , Male , Mice , Mice, Knockout , RNA, Messenger/genetics , RNA, Messenger/metabolism , Rats , Weight Gain
13.
J Clin Endocrinol Metab ; 99(8): E1397-406, 2014 Aug.
Article in English | MEDLINE | ID: mdl-24823464

ABSTRACT

CONTEXT: 5α-Reductase (5αR) types 1 and 2 catalyze the A-ring reduction of steroids, including androgens and glucocorticoids. 5α-R inhibitors lower dihydrotestosterone in benign prostatic hyperplasia; finasteride inhibits 5αR2, and dutasteride inhibits both 5αR2 and 5αR1. In rodents, loss of 5αR1 promotes fatty liver. OBJECTIVE: Our objective was to test the hypothesis that inhibition of 5αR1 causes metabolic dysfunction in humans. DESIGN, SETTING, AND PARTICIPANTS: This double-blind randomized controlled parallel group study at a clinical research facility included 46 men (20-85 years) studied before and after intervention. INTERVENTION: Oral dutasteride (0.5 mg daily; n = 16), finasteride (5 mg daily; n = 16), or control (tamsulosin; 0.4 mg daily; n = 14) was administered for 3 months. MAIN OUTCOME MEASURE: Glucose disposal was measured during a stepwise hyperinsulinemic-euglycemic clamp. Data are mean (SEM). RESULTS: Dutasteride and finasteride had similar effects on steroid profiles, with reduced urinary androgen and glucocorticoid metabolites and reduced circulating DHT but no change in plasma or salivary cortisol. Dutasteride, but not finasteride, reduced stimulation of glucose disposal by high-dose insulin (dutasteride by -5.7 [3.2] µmol/kg fat-free mass/min, versus finasteride +7.2 [3.0], and tamsulosin +7.0 [2.0]). Dutasteride also reduced suppression of nonesterified fatty acids by insulin and increased body fat (by 1.6% [0.6%]). Glucose production and glycerol turnover were unchanged. Consistent with metabolic effects of dutasteride being mediated in peripheral tissues, mRNA for 5αR1 but not 5αR2 was detected in human adipose tissue. CONCLUSION: Dual inhibition of 5αRs, but not inhibition of 5αR2 alone, modulates insulin sensitivity in human peripheral tissues rather than liver. This may have important implications for patients prescribed dutasteride for prostatic disease.


Subject(s)
3-Oxo-5-alpha-Steroid 4-Dehydrogenase/physiology , 5-alpha Reductase Inhibitors/pharmacology , Azasteroids/pharmacology , Finasteride/pharmacology , Insulin Resistance , Adipose Tissue/drug effects , Adipose Tissue/metabolism , Adult , Aged , Aged, 80 and over , Blood Glucose/drug effects , Blood Glucose/metabolism , Body Composition/drug effects , Double-Blind Method , Dutasteride , Humans , Male , Middle Aged , Prostatic Hyperplasia/metabolism , Prostatic Hyperplasia/urine , Young Adult
14.
J Endocrinol ; 222(2): 257-66, 2014 Aug.
Article in English | MEDLINE | ID: mdl-24872577

ABSTRACT

Patients with critical illness or hepatic failure exhibit impaired cortisol responses to ACTH, a phenomenon known as 'relative adrenal insufficiency'. A putative mechanism is that elevated bile acids inhibit inactivation of cortisol in liver by 5α-reductases type 1 and type 2 and 5ß-reductase, resulting in compensatory downregulation of the hypothalamic-pituitary-adrenal axis and adrenocortical atrophy. To test the hypothesis that impaired glucocorticoid clearance can cause relative adrenal insufficiency, we investigated the consequences of 5α-reductase type 1 deficiency in mice. In adrenalectomised male mice with targeted disruption of 5α-reductase type 1, clearance of corticosterone was lower after acute or chronic (eightfold, P<0.05) administration, compared with WT control mice. In intact 5α-reductase-deficient male mice, although resting plasma corticosterone levels were maintained, corticosterone responses were impaired after ACTH administration (26% lower, P<0.05), handling stress (2.5-fold lower, P<0.05) and restraint stress (43% lower, P<0.05) compared with WT mice. mRNA levels of Nr3c1 (glucocorticoid receptor), Crh and Avp in pituitary or hypothalamus were altered, consistent with enhanced negative feedback. These findings confirm that impaired peripheral clearance of glucocorticoids can cause 'relative adrenal insufficiency' in mice, an observation with important implications for patients with critical illness or hepatic failure, and for patients receiving 5α-reductase inhibitors for prostatic disease.


Subject(s)
3-Oxo-5-alpha-Steroid 4-Dehydrogenase/deficiency , Adrenal Insufficiency/etiology , Membrane Proteins/deficiency , 5-alpha Reductase Inhibitors/adverse effects , Adrenocorticotropic Hormone/pharmacology , Animals , Corticosterone/blood , Dexamethasone/pharmacology , Hypothalamo-Hypophyseal System/drug effects , Hypothalamo-Hypophyseal System/physiology , Male , Mice , Mice, Knockout , Pituitary-Adrenal System/drug effects , Pituitary-Adrenal System/physiology , Stress, Psychological/physiopathology
15.
FASEB J ; 27(4): 1519-31, 2013 Apr.
Article in English | MEDLINE | ID: mdl-23303209

ABSTRACT

11ß-Hydroxysteroid dehydrogenase type-1 (11ß-HSD1) converts inert cortisone into active cortisol, amplifying intracellular glucocorticoid action. 11ß-HSD1 deficiency improves cardiovascular risk factors in obesity but exacerbates acute inflammation. To determine the effects of 11ß-HSD1 deficiency on atherosclerosis and its inflammation, atherosclerosis-prone apolipoprotein E-knockout (ApoE-KO) mice were treated with a selective 11ß-HSD1 inhibitor or crossed with 11ß-HSD1-KO mice to generate double knockouts (DKOs) and challenged with an atherogenic Western diet. 11ß-HSD1 inhibition or deficiency attenuated atherosclerosis (74-76%) without deleterious effects on plaque structure. This occurred without affecting plasma lipids or glucose, suggesting independence from classical metabolic risk factors. KO plaques were not more inflamed and indeed had 36% less T-cell infiltration, associated with 38% reduced circulating monocyte chemoattractant protein-1 (MCP-1) and 36% lower lesional vascular cell adhesion molecule-1 (VCAM-1). Bone marrow (BM) cells are key to the atheroprotection, since transplantation of DKO BM to irradiated ApoE-KO mice reduced atherosclerosis by 51%. 11ß-HSD1-null macrophages show 76% enhanced cholesterol ester export. Thus, 11ß-HSD1 deficiency reduces atherosclerosis without exaggerated lesional inflammation independent of metabolic risk factors. Selective 11ß-HSD1 inhibitors promise novel antiatherosclerosis effects over and above their benefits for metabolic risk factors via effects on BM cells, plausibly macrophages.


Subject(s)
11-beta-Hydroxysteroid Dehydrogenase Type 1/deficiency , Atherosclerosis/metabolism , Bone Marrow/metabolism , 11-beta-Hydroxysteroid Dehydrogenase Type 1/antagonists & inhibitors , 11-beta-Hydroxysteroid Dehydrogenase Type 1/metabolism , Adipose Tissue/drug effects , Adipose Tissue/metabolism , Animals , Atherosclerosis/genetics , Bone Marrow/drug effects , Glucocorticoids/metabolism , Mice , Mice, Knockout , Risk Factors , Vascular Cell Adhesion Molecule-1/metabolism
16.
Am J Physiol Renal Physiol ; 303(4): F494-502, 2012 Aug 15.
Article in English | MEDLINE | ID: mdl-22622456

ABSTRACT

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.


Subject(s)
11-beta-Hydroxysteroid Dehydrogenase Type 2/genetics , 11-beta-Hydroxysteroid Dehydrogenase Type 2/metabolism , Diabetes Insipidus/enzymology , Kidney Concentrating Ability/physiology , Kidney/physiology , Aging , Animals , Diabetes Insipidus/genetics , Gene Expression Regulation , Homeostasis , Kidney/anatomy & histology , Kidney Concentrating Ability/genetics , Mice , Mice, Knockout , Osmolar Concentration , Sodium-Potassium-Chloride Symporters/genetics , Sodium-Potassium-Chloride Symporters/metabolism , Water/metabolism , Weight Loss
17.
Diabetes ; 61(4): 790-6, 2012 Apr.
Article in English | MEDLINE | ID: mdl-22357964

ABSTRACT

Recent trials show salicylates improve glycemic control in type 2 diabetes, but the mechanism is poorly understood. Expression of the glucocorticoid-generating enzyme 11ß-hydroxysteroid dehydrogenase type 1 (11ß-HSD1) in adipose tissue is increased in vitro by proinflammatory cytokines and upregulated in obesity. 11ß-HSD1 inhibition enhances insulin sensitivity. We hypothesized that salicylates downregulate 11ß-HSD1 expression, contributing to their metabolic efficacy. We treated diet-induced obese (DIO) 11ß-HSD1-deficient mice and C57Bl/6 mice with sodium salicylate for 4 weeks. Glucose tolerance was assessed in vivo. Tissue transcript levels were assessed by quantitative PCR and enzyme activity by incubation with (3)H-steroid. Two weeks' administration of salsalate was also investigated in a randomized double-blind placebo-controlled crossover study in 16 men, with measurement of liver 11ß-HSD1 activity in vivo and adipose tissue 11ß-HSD1 transcript levels ex vivo. In C57Bl/6 DIO mice, salicylate improved glucose tolerance and downregulated 11ß-HSD1 mRNA and activity selectively in visceral adipose. DIO 11ß-HSD1-deficient mice were resistant to these metabolic effects of salicylate. In men, salsalate reduced 11ß-HSD1 expression in subcutaneous adipose, and in vitro salicylate treatment reduced adipocyte 11ß-HSD1 expression and induced adiponectin expression only in the presence of 11ß-HSD1 substrate. Reduced intra-adipose glucocorticoid regeneration by 11ß-HSD1 is a novel mechanism that contributes to the metabolic efficacy of salicylates.


Subject(s)
11-beta-Hydroxysteroid Dehydrogenase Type 1/metabolism , Adipose Tissue/metabolism , Gene Expression Regulation/drug effects , Insulin Resistance/physiology , Obesity/metabolism , Sodium Salicylate/pharmacology , 11-beta-Hydroxysteroid Dehydrogenase Type 1/genetics , Adipocytes/cytology , Adipocytes/drug effects , Adipocytes/physiology , Adipose Tissue/drug effects , Adult , Animals , Anti-Inflammatory Agents, Non-Steroidal/pharmacology , Body Weight/drug effects , Cell Differentiation , Cell Line , Cross-Over Studies , Glucose Tolerance Test , Humans , Male , Mice , Mice, Knockout , Middle Aged , Salicylates/pharmacology
18.
FASEB J ; 26(5): 1866-74, 2012 May.
Article in English | MEDLINE | ID: mdl-22321728

ABSTRACT

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.


Subject(s)
Fetal Development , Glucocorticoids/physiology , Nutritional Status , 11-beta-Hydroxysteroid Dehydrogenase Type 2/metabolism , Animals , Base Sequence , Corticosterone/blood , DNA Primers , Female , In Situ Hybridization , Maternal Exposure , Mice , Mice, Inbred C57BL , Placenta/enzymology , Pregnancy
19.
J Clin Endocrinol Metab ; 96(12): E2029-33, 2011 Dec.
Article in English | MEDLINE | ID: mdl-21956425

ABSTRACT

CONTEXT: Black South African women are less insulin sensitive than their White counterparts, despite less central and greater peripheral fat deposition. We hypothesized that this paradox may be explained, in part, by differences in the adipogenic capacity of sc adipose tissue (SAT). OBJECTIVE: Our objective was to measure adipogenic and lipogenic gene expression in abdominal and gluteal SAT depots and determine their relationships with insulin sensitivity (S(I)) in South African women. PARTICIPANTS AND DESIGN: Fourteen normal-weight [body mass index (BMI) <25 kg/m(2)] Black, 13 normal-weight White, 14 obese (BMI >30 kg/m(2)) Black, and 13 obese White premenopausal South African women participated in this cross-sectional study. MAIN OUTCOMES: S(I) (frequently sampled i.v. glucose tolerance test) in relation to expression of adipogenic and lipogenic genes in abdominal and gluteal SAT depots. RESULTS: With increasing BMI, Black women had less visceral fat (P = 0.03) and more abdominal (P = 0.017) and gynoid (P = 0.041) SAT but had lower S(I) (P < 0.01) than White women. The expression of adipogenic and lipogenic genes was proportionately lower with obesity in Black but not White women in the gluteal and deep SAT depots (P < 0.05 for ethnicity × BMI effect). In Black women only, the expression of these genes correlated positively with S(I) (all P < 0.05), independently of age and fat mass. CONCLUSIONS: Obese Black women have reduced SAT expression of adipogenic and lipogenic genes compared with White women, which associates with reduced S(I). These findings suggest that obesity in Black women impairs SAT adipogenesis and storage, potentially leading to insulin resistance and increased risk of type 2 diabetes.


Subject(s)
Black People , Insulin Resistance/genetics , Obesity/genetics , Subcutaneous Fat/metabolism , Adolescent , Adult , Body Composition , Female , Gene Expression , Humans , Intra-Abdominal Fat/metabolism , Middle Aged , Obesity/ethnology , Obesity/metabolism , South Africa , White People
20.
Hypertension ; 57(3): 515-520, 2011 Mar.
Article in English | MEDLINE | ID: mdl-21282561

ABSTRACT

Salt sensitivity of blood pressure is an independent risk factor for cardiovascular morbidity. Mechanistically, abnormal mineralocorticoid action and subclinical renal impairment may blunt the natriuretic response to high sodium intake, causing blood pressure to rise. 11ß-Hydroxysteroid dehydrogenase type 2 (11ßHSD2) controls ligand access to the mineralocorticoid receptor, and ablation of the enzyme causes severe hypertension. Polymorphisms in HSD11B2 are associated with salt sensitivity of blood pressure in normotensives. In this study, we used mice heterozygote for a null mutation in Hsd11b2 (Hsd11b2(+/-)) to define the mechanisms linking reduced enzyme activity to salt sensitivity of blood pressure. A high-sodium diet caused a rapid and sustained increase in blood pressure in Hsd11b2(+/-) mice but not in wild-type littermates. During the adaptation to high-sodium diet, heterozygotes displayed impaired sodium excretion, a transient positive sodium balance, and hypokalemia. After 21 days of high-sodium feeding, Hsd11b2(+/-) mice had an increased heart weight. Mineralocorticoid receptor antagonism partially prevented the increase in heart weight but not the increase in blood pressure. Glucocorticoid receptor antagonism prevented the rise in blood pressure. In Hsd11b2(+/-) mice, high-sodium feeding caused suppression of aldosterone and a moderate but sustained increase in corticosterone. This study demonstrates an inverse relationship among 11ßHSD2 activity, heart weight, and blood pressure in a clinically important context. Reduced activity causes salt sensitivity of blood pressure, but this does not reflect illicit activation of mineralocorticoid receptors by glucocorticoids. Instead, we have identified a novel interaction among 11ßHSD2, dietary salt, and circulating glucocorticoids.


Subject(s)
11-beta-Hydroxysteroid Dehydrogenase Type 2/genetics , Blood Pressure/genetics , Haploinsufficiency/genetics , Hypertension/chemically induced , Hypertension/genetics , Sodium Chloride, Dietary/adverse effects , Analysis of Variance , Animals , Blood Pressure/drug effects , Hormone Antagonists/pharmacology , Mice , Mifepristone/pharmacology , Mutation , Polymorphism, Genetic , Receptors, Glucocorticoid/genetics , Receptors, Mineralocorticoid/genetics , Reverse Transcriptase Polymerase Chain Reaction
SELECTION OF CITATIONS
SEARCH DETAIL
...