151
|
Aldosterone-stimulating somatic gene mutations are common in normal adrenal glands. Proc Natl Acad Sci U S A 2015; 112:E4591-9. [PMID: 26240369 DOI: 10.1073/pnas.1505529112] [Citation(s) in RCA: 221] [Impact Index Per Article: 24.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023] Open
Abstract
Primary aldosteronism (PA) represents the most common cause of secondary hypertension, but little is known regarding its adrenal cellular origins. Recently, aldosterone-producing cell clusters (APCCs) with high expression of aldosterone synthase (CYP11B2) were found in both normal and PA adrenal tissue. PA-causing aldosterone-producing adenomas (APAs) harbor mutations in genes encoding ion channels/pumps that alter intracellular calcium homeostasis and cause renin-independent aldosterone production through increased CYP11B2 expression. Herein, we hypothesized that APCCs have APA-related aldosterone-stimulating somatic gene mutations. APCCs were studied in 42 normal adrenals from kidney donors. To clarify APCC molecular characteristics, we used microarrays to compare the APCC transcriptome with conventional adrenocortical zones [zona glomerulosa (ZG), zona fasciculata, and zona reticularis]. The APCC transcriptome was most similar to ZG but with an enhanced capacity to produce aldosterone. To determine if APCCs harbored APA-related mutations, we performed targeted next generation sequencing of DNA from 23 APCCs and adjacent normal adrenal tissue isolated from both formalin-fixed, paraffin-embedded, and frozen tissues. Known aldosterone driver mutations were identified in 8 of 23 (35%) APCCs, including mutations in calcium channel, voltage-dependent, L-type, α1D-subunit (CACNA1D; 6 of 23 APCCs) and ATPase, Na(+)/(K+) transporting, α1-polypeptide (ATP1A1; 2 of 23 APCCs), which were not observed in the adjacent normal adrenal tissue. Overall, we show three major findings: (i) APCCs are common in normal adrenals, (ii) APCCs harbor somatic mutations known to cause excess aldosterone production, and (iii) the mutation spectrum of aldosterone-driving mutations is different in APCCs from that seen in APA. These results provide molecular support for APCC as a precursor of PA.
Collapse
|
152
|
Schiffer L, Anderko S, Hannemann F, Eiden-Plach A, Bernhardt R. The CYP11B subfamily. J Steroid Biochem Mol Biol 2015; 151:38-51. [PMID: 25465475 DOI: 10.1016/j.jsbmb.2014.10.011] [Citation(s) in RCA: 46] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/14/2014] [Revised: 10/10/2014] [Accepted: 10/14/2014] [Indexed: 01/11/2023]
Abstract
The biosynthesis of steroid hormones is dependent on P450-catalyzed reactions. In mammals, cholesterol is the common precursor of all steroid hormones, and its conversion to pregnenolone is the initial and rate-limiting step in hormone biosynthesis in steroidogenic tissues such as gonads and adrenal glands. The production of glucocorticoids and mineralocorticoids takes place in the adrenal gland and the final steps are catalyzed by 2 mitochondrial cytochromes P450, CYP11B1 (11β-hydroxylase or P45011β) and CYP11B2 (aldosterone synthase or P450aldo). The occurrence and development of these 2 enzymes in different species, their contribution to the biosynthesis of steroid hormones as well as their regulation at different levels (gene expression, cellular regulation, regulation on the level of proteins) is the topic of this chapter.
Collapse
Affiliation(s)
- Lina Schiffer
- Institute of Biochemistry, Saarland University, Campus B2.2, 66123 Saarbrücken, Germany
| | - Simone Anderko
- Institute of Biochemistry, Saarland University, Campus B2.2, 66123 Saarbrücken, Germany
| | - Frank Hannemann
- Institute of Biochemistry, Saarland University, Campus B2.2, 66123 Saarbrücken, Germany
| | - Antje Eiden-Plach
- Institute of Biochemistry, Saarland University, Campus B2.2, 66123 Saarbrücken, Germany
| | - Rita Bernhardt
- Institute of Biochemistry, Saarland University, Campus B2.2, 66123 Saarbrücken, Germany.
| |
Collapse
|
153
|
Drelon C, Berthon A, Mathieu M, Martinez A, Val P. Adrenal cortex tissue homeostasis and zonation: A WNT perspective. Mol Cell Endocrinol 2015; 408:156-64. [PMID: 25542843 DOI: 10.1016/j.mce.2014.12.014] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/17/2014] [Revised: 12/18/2014] [Accepted: 12/18/2014] [Indexed: 11/19/2022]
Abstract
The adrenal cortex plays essential roles in the control of sodium and water homeostasis, stress response, inflammation and metabolism, through secretion of glucocorticoids and mineralocorticoids. Coordinated production of these hormones relies on functional zonation of the cortex, characterised by expression of Cyp11b2 under the control of angiotensin II and plasma potassium level in zona glomerulosa (ZG) and Cyp11b1 under the control of ACTH in zona fasciculata (ZF). The mechanisms involved in the establishment of functional zonation and its maintenance during centripetal cortex cell renewal are still poorly understood. Here, we hypothesise that the hormonal and signalling pathways that control adrenal cortex function are also involved in cortical zonation. In particular, we summarise evidence on the role of WNT/β-catenin signalling in ZG differentiation and how tight control of its activity is required to shape the adult cortex. In this context, we discuss the potential role of known WNT regulators and the possibility of a reciprocal cross-talk between PKA and WNT signalling.
Collapse
Affiliation(s)
- Coralie Drelon
- Laboratoire Génétique Reproduction et Développement -GReD- CNRS UMR 6293, Inserm U1103, Clermont Université, 24 Avenue des Landais, Aubière Cedex 63171, France
| | - Annabel Berthon
- Laboratoire Génétique Reproduction et Développement -GReD- CNRS UMR 6293, Inserm U1103, Clermont Université, 24 Avenue des Landais, Aubière Cedex 63171, France; Eunice Kennedy Shriver National Institute of Child Health and Human Development, Bethesda, Maryland 20892-1103, USA
| | - Mickael Mathieu
- Laboratoire Génétique Reproduction et Développement -GReD- CNRS UMR 6293, Inserm U1103, Clermont Université, 24 Avenue des Landais, Aubière Cedex 63171, France
| | - Antoine Martinez
- Laboratoire Génétique Reproduction et Développement -GReD- CNRS UMR 6293, Inserm U1103, Clermont Université, 24 Avenue des Landais, Aubière Cedex 63171, France
| | - Pierre Val
- Laboratoire Génétique Reproduction et Développement -GReD- CNRS UMR 6293, Inserm U1103, Clermont Université, 24 Avenue des Landais, Aubière Cedex 63171, France.
| |
Collapse
|
154
|
Mose FH, Jensen JM, Therwani S, Mortensen J, Hansen AB, Bech JN, Pedersen EB. Effect of nebivolol on renal nitric oxide availability and tubular function in patients with essential hypertension. Br J Clin Pharmacol 2015; 80:425-35. [PMID: 25778445 DOI: 10.1111/bcp.12627] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2014] [Revised: 03/02/2015] [Accepted: 03/04/2015] [Indexed: 12/18/2022] Open
Abstract
AIMS Nebivolol is a selective β1 -receptor antagonist with vasodilating properties. In patients with essential hypertension, we tested the hypothesis that nebivolol increases systemic and renal nitric oxide (NO) availability using L-N(G) -monomethyl arginine (L-NMMA) as an inhibitor of NO production. METHODS In a randomized, placebo-controlled, crossover study, patients with essential hypertension were treated with nebivolol for five days, along with a standardized diet and fluid intake. We examined the acute effects of systemic NO synthase inhibition with L-NMMA on brachial blood pressure (bBP), pulse wave velocity (PWV) and central blood pressure (cBP) estimated by applanation tonometry, glomerular filtration rate (GFR), fractional excretion of sodium (FENa ), urinary excretion of both aquaporin-2 (u-AQP2) and epithelial sodium channels (u-ENaCγ ), and plasma concentrations of nitrate/nitrite (p-NOx ) and vasoactive hormones after five days' treatment with placebo and nebivolol. RESULTS Nebivolol significantly reduced PWV, bBP, cBP and plasma renin, angiotensin II and aldosterone concentrations. The renal parameters, p-NOx and plasma arginine vasopressin concentration were not changed by nebivolol. There was no difference between nebivolol and placebo in the response to L-NMMA, with LMMA inducing a similar increase in PWV, bBP and cBP and a similar decrease in GFR, uAQP2 and u-ENaCγ and FENa [mean change -0.62% (95% confidence interval {CI} -0.40 to -0.84) during placebo vs. -0.57% (95% CI -0.46 to -0.68; P = 0.564) during nebivolol treatment]. Vasoactive hormones were changed to a similar extend by L-NMMA during administration of nebivolol and placebo. CONCLUSIONS Nebivolol did not change p-NOx , and inhibition of NO synthesis induced the same response in blood pressure, GFR, renal tubular function and vasoactive hormones during nebivolol and placebo. Thus, the data did not support the hypothesis that nebivolol changes vascular and renal NO availability in patients with essential hypertension.
Collapse
Affiliation(s)
- Frank H Mose
- University Clinic in Nephrology and Hypertension, Department of Medical Research and University of Aarhus
| | - Janni M Jensen
- University Clinic in Nephrology and Hypertension, Department of Medical Research and University of Aarhus
| | - Safa Therwani
- University Clinic in Nephrology and Hypertension, Department of Medical Research and University of Aarhus
| | | | | | - Jesper N Bech
- University Clinic in Nephrology and Hypertension, Department of Medical Research and University of Aarhus
| | - Erling B Pedersen
- University Clinic in Nephrology and Hypertension, Department of Medical Research and University of Aarhus
| |
Collapse
|
155
|
Seremwe M, Schnellmann RG, Bollag WB. Calpain-10 Activity Underlies Angiotensin II-Induced Aldosterone Production in an Adrenal Glomerulosa Cell Model. Endocrinology 2015; 156:2138-49. [PMID: 25836666 PMCID: PMC4430612 DOI: 10.1210/en.2014-1866] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Aldosterone is a steroid hormone important in the regulation of blood pressure. Aberrant production of aldosterone results in the development and progression of diseases including hypertension and congestive heart failure; therefore, a complete understanding of aldosterone production is important for developing more effective treatments. Angiotensin II (AngII) regulates steroidogenesis, in part through its ability to increase intracellular calcium levels. Calcium can activate calpains, proteases classified as typical or atypical based on the presence or absence of penta-EF-hands, which are involved in various cellular responses. We hypothesized that calpain, in particular calpain-10, is activated by AngII in adrenal glomerulosa cells and underlies aldosterone production. Our studies showed that pan-calpain inhibitors reduced AngII-induced aldosterone production in 2 adrenal glomerulosa cell models, primary bovine zona glomerulosa and human adrenocortical carcinoma (HAC15) cells, as well as CYP11B2 expression in the HAC15 cells. Although AngII induced calpain activation in these cells, typical calpain inhibitors had no effect on AngII-elicited aldosterone production, suggesting a lack of involvement of classical calpains in this process. However, an inhibitor of the atypical calpain, calpain-10, decreased AngII-induced aldosterone production. Consistent with this result, small interfering RNA (siRNA)-mediated knockdown of calpain-10 inhibited aldosterone production and CYP11B2 expression, whereas adenovirus-mediated overexpression of calpain-10 resulted in increased AngII-induced aldosterone production. Our results indicate that AngII-induced activation of calpain-10 in glomerulosa cells underlies aldosterone production and identify calpain-10 or its downstream pathways as potential targets for the development of drug therapies for the treatment of hypertension.
Collapse
Affiliation(s)
- Mutsa Seremwe
- Charlie Norwood Veterans Administration Medical Center (W.B.B.), Augusta, Georgia 30904; Department of Physiology (M.S., W.B.B.) and Section of Dermatology (W.B.B.), Department of Medicine, Georgia Regents University, Augusta, Georgia 30912; and Department of Drug Discovery and Biomedical Sciences (R.G.S.), Medical University of South Carolina, and Ralph H. Johnson VA Medical Center (R.G.S.), Charleston, South Carolina 29425
| | | | | |
Collapse
|
156
|
de Punder K, Pruimboom L. Stress induces endotoxemia and low-grade inflammation by increasing barrier permeability. Front Immunol 2015; 6:223. [PMID: 26029209 PMCID: PMC4432792 DOI: 10.3389/fimmu.2015.00223] [Citation(s) in RCA: 178] [Impact Index Per Article: 19.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2015] [Accepted: 04/24/2015] [Indexed: 12/17/2022] Open
Abstract
Chronic non-communicable diseases (NCDs) are the leading causes of work absence, disability, and mortality worldwide. Most of these diseases are associated with low-grade inflammation. Here, we hypothesize that stresses (defined as homeostatic disturbances) can induce low-grade inflammation by increasing the availability of water, sodium, and energy-rich substances to meet the increased metabolic demand induced by the stressor. One way of triggering low-grade inflammation is by increasing intestinal barrier permeability through activation of various components of the stress system. Although beneficial to meet the demands necessary during stress, increased intestinal barrier permeability also raises the possibility of the translocation of bacteria and their toxins across the intestinal lumen into the blood circulation. In combination with modern life-style factors, the increase in bacteria/bacterial toxin translocation arising from a more permeable intestinal wall causes a low-grade inflammatory state. We support this hypothesis with numerous studies finding associations with NCDs and markers of endotoxemia, suggesting that this process plays a pivotal and perhaps even a causal role in the development of low-grade inflammation and its related diseases.
Collapse
Affiliation(s)
- Karin de Punder
- Institute of Medical Psychology, Charité University Medicine , Berlin , Germany ; Natura Foundation , Numansdorp , Netherlands
| | | |
Collapse
|
157
|
Nanba K, Chen A, Nishimoto K, Rainey WE. Role of Ca(2+)/calmodulin-dependent protein kinase kinase in adrenal aldosterone production. Endocrinology 2015; 156:1750-6. [PMID: 25679868 PMCID: PMC4398758 DOI: 10.1210/en.2014-1782] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
There is considerable evidence supporting the role of calcium signaling in adrenal regulation of both aldosterone synthase (CYP11B2) and aldosterone production. However, there have been no studies that investigated the role played by the Ca(2+)/calmodulin-dependent protein kinase kinase (CaMKK) in adrenal cells. In this study we investigated the role of CaMKK in adrenal cell aldosterone production. To determine the role of CaMKK, we used a selective CaMKK inhibitor (STO-609) in the HAC15 human adrenal cell line. Cells were treated with angiotensin II (Ang II) or K+ and evaluated for the expression of steroidogenic acute regulatory protein and CYP11B2 (mRNA/protein) as well as aldosterone production. We also transduced HAC15 cells with lentiviral short hairpin RNAs of CaMKK1 and CaMKK2 to determine which CaMKK plays a more important role in adrenal cell regulation of the calcium signaling cascade. The CaMKK inhibitor, STO-609, decreased aldosterone production in cells treated with Ang II or K+ in a dose-dependent manner. STO-609 (20 μM) also inhibited steroidogenic acute regulatory protein and CYP11B2 mRNA/protein induction. CaMKK2 knockdown cells showed significant reduction of CYP11B2 mRNA induction and aldosterone production in cells treated with Ang II, although there was no obvious effect in CaMKK1 knockdown cells. In immunohistochemical analysis, CaMKK2 protein was highly expressed in human adrenal zona glomerulosa with lower expression in the zona fasciculata. In conclusion, the present study suggests that CaMKK2 plays a pivotal role in the calcium signaling cascade regulating adrenal aldosterone production.
Collapse
Affiliation(s)
- Kazutaka Nanba
- Department of Molecular and Integrative Physiology, University of Michigan Medical School, Ann Arbor, Michigan 48109
| | | | | | | |
Collapse
|
158
|
Piaditis G, Markou A, Papanastasiou L, Androulakis II, Kaltsas G. Progress in aldosteronism: a review of the prevalence of primary aldosteronism in pre-hypertension and hypertension. Eur J Endocrinol 2015; 172:R191-203. [PMID: 25538205 DOI: 10.1530/eje-14-0537] [Citation(s) in RCA: 76] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
Primary aldosteronism (PA) secondary to excessive and/or autonomous aldosterone secretion from the renin-angiotensin system accounts for ∼10% of cases of hypertension and is primarily caused by bilateral adrenal hyperplasia (BAH) or aldosterone-producing adenomas (APAs). Although the diagnosis has traditionally been supported by low serum potassium levels, normokalaemic and even normotensive forms of PA have been identified expanding further the clinical phenotype. Moreover, recent evidence has shown that serum aldosterone correlates with increased blood pressure (BP) in the general population and even moderately raised aldosterone levels are linked to increased cardiovascular morbidity and mortality. In addition, aldosterone antagonists are effective in BP control even in patients without evidence of dysregulated aldosterone secretion. These findings indicate a higher prevalence of aldosterone excess among hypertensive patients than previously considered that could be attributed to disease heterogeneity, aldosterone level fluctuations related to an ACTH effect or inadequate sensitivity of current diagnostic means to identify apparent aldosterone excess. In addition, functioning aberrant receptors expressed in the adrenal tissue have been found in a subset of PA cases that could also be related to its pathogenesis. Recently a number of specific genetic alterations, mainly involving ion homeostasis across the membrane of zona glomerulosa, have been detected in ∼50% of patients with APAs. Although specific genotype/phenotype correlations have not been clearly identified, differential expression of these genetic alterations could also account for the wide clinical phenotype, variations in disease prevalence and performance of diagnostic tests. In the present review, we critically analyse the current means used to diagnose PA along with the role that ACTH, aberrant receptor expression and genetic alterations may exert, and provide evidence for an increased prevalence of aldosterone dysregulation in patients with essential hypertension and pre-hypertension.
Collapse
Affiliation(s)
- George Piaditis
- Department of Endocrinology and Diabetes CenterG. Gennimatas Hospital, 154 Mesogion Avenue, 11527 Holargos, Athens, Greece andDepartment of PathophysiologyNational University of Athens, Mikras Asias 75, 11527 Athens, Greece
| | - Athina Markou
- Department of Endocrinology and Diabetes CenterG. Gennimatas Hospital, 154 Mesogion Avenue, 11527 Holargos, Athens, Greece andDepartment of PathophysiologyNational University of Athens, Mikras Asias 75, 11527 Athens, Greece
| | - Labrini Papanastasiou
- Department of Endocrinology and Diabetes CenterG. Gennimatas Hospital, 154 Mesogion Avenue, 11527 Holargos, Athens, Greece andDepartment of PathophysiologyNational University of Athens, Mikras Asias 75, 11527 Athens, Greece
| | - Ioannis I Androulakis
- Department of Endocrinology and Diabetes CenterG. Gennimatas Hospital, 154 Mesogion Avenue, 11527 Holargos, Athens, Greece andDepartment of PathophysiologyNational University of Athens, Mikras Asias 75, 11527 Athens, Greece Department of Endocrinology and Diabetes CenterG. Gennimatas Hospital, 154 Mesogion Avenue, 11527 Holargos, Athens, Greece andDepartment of PathophysiologyNational University of Athens, Mikras Asias 75, 11527 Athens, Greece
| | - Gregory Kaltsas
- Department of Endocrinology and Diabetes CenterG. Gennimatas Hospital, 154 Mesogion Avenue, 11527 Holargos, Athens, Greece andDepartment of PathophysiologyNational University of Athens, Mikras Asias 75, 11527 Athens, Greece
| |
Collapse
|
159
|
Bandulik S, Tauber P, Lalli E, Barhanin J, Warth R. Two-pore domain potassium channels in the adrenal cortex. Pflugers Arch 2015; 467:1027-42. [PMID: 25339223 PMCID: PMC4428839 DOI: 10.1007/s00424-014-1628-6] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2014] [Revised: 10/02/2014] [Accepted: 10/03/2014] [Indexed: 12/31/2022]
Abstract
The physiological control of steroid hormone secretion from the adrenal cortex depends on the function of potassium channels. The "two-pore domain K(+) channels" (K2P) TWIK-related acid sensitive K(+) channel 1 (TASK1), TASK3, and TWIK-related K(+) channel 1 (TREK1) are strongly expressed in adrenocortical cells. They confer a background K(+) conductance to these cells which is important for the K(+) sensitivity as well as for angiotensin II and adrenocorticotropic hormone-dependent stimulation of aldosterone and cortisol synthesis. Mice with single deletions of the Task1 or Task3 gene as well as Task1/Task3 double knockout mice display partially autonomous aldosterone synthesis. It appears that TASK1 and TASK3 serve different functions: TASK1 affects cell differentiation and prevents expression of aldosterone synthase in the zona fasciculata, while TASK3 controls aldosterone secretion in glomerulosa cells. TREK1 is involved in the regulation of cortisol secretion in fasciculata cells. These data suggest that a disturbed function of K2P channels could contribute to adrenocortical pathologies in humans.
Collapse
Affiliation(s)
- Sascha Bandulik
- Medical Cell Biology, University of Regensburg, Universitaetsstrasse 31, 93053, Regensburg, Germany,
| | | | | | | | | |
Collapse
|
160
|
Smith JA, Pati D, Wang L, de Kloet AD, Frazier CJ, Krause EG. Hydration and beyond: neuropeptides as mediators of hydromineral balance, anxiety and stress-responsiveness. Front Syst Neurosci 2015; 9:46. [PMID: 25873866 PMCID: PMC4379895 DOI: 10.3389/fnsys.2015.00046] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2014] [Accepted: 03/06/2015] [Indexed: 11/13/2022] Open
Abstract
Challenges to body fluid homeostasis can have a profound impact on hypothalamic regulation of stress responsiveness. Deficiencies in blood volume or sodium concentration leads to the generation of neural and humoral signals relayed through the hindbrain and circumventricular organs that apprise the paraventricular nucleus of the hypothalamus (PVH) of hydromineral imbalance. Collectively, these neural and humoral signals converge onto PVH neurons, including those that express corticotrophin-releasing factor (CRF), oxytocin (OT), and vasopressin, to influence their activity and initiate compensatory responses that alleviate hydromineral imbalance. Interestingly, following exposure to perceived threats to homeostasis, select limbic brain regions mediate behavioral and physiological responses to psychogenic stressors, in part, by influencing activation of the same PVH neurons that are known to maintain body fluid homeostasis. Here, we review past and present research examining interactions between hypothalamic circuits regulating body fluid homeostasis and those mediating behavioral and physiological responses to psychogenic stress.
Collapse
Affiliation(s)
- Justin A. Smith
- Laboratory of Dr. Eric Krause, Department of Pharmacodynamics, College of Pharmacy, University of FloridaGainesville, FL, USA
| | - Dipanwita Pati
- Laboratory of Dr. Charles Frazier, Department of Pharmacodynamics, College of Pharmacy, University of FloridaGainesville, FL, USA
| | - Lei Wang
- Laboratory of Dr. Eric Krause, Department of Pharmacodynamics, College of Pharmacy, University of FloridaGainesville, FL, USA
| | - Annette D. de Kloet
- Laboratory of Dr. Colin Sumners, Department of Physiology and Functional Genomics, College of Medicine, University of FloridaGainesville, FL, USA
| | - Charles J. Frazier
- Laboratory of Dr. Charles Frazier, Department of Pharmacodynamics, College of Pharmacy, University of FloridaGainesville, FL, USA
| | - Eric G. Krause
- Laboratory of Dr. Eric Krause, Department of Pharmacodynamics, College of Pharmacy, University of FloridaGainesville, FL, USA
| |
Collapse
|
161
|
Dembek KA, Hurcombe SD, Stewart AJ, Barr BS, MacGillivray KC, Kinee M, Elam J, Toribio RE. Association of aldosterone and arginine vasopressin concentrations and clinical markers of hypoperfusion in neonatal foals. Equine Vet J 2015; 48:176-81. [PMID: 25421257 DOI: 10.1111/evj.12393] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2014] [Accepted: 11/19/2014] [Indexed: 11/30/2022]
Abstract
REASONS FOR PERFORMING STUDY Critically ill foals often present to veterinary hospitals with impaired organ perfusion which can be demonstrated by increased blood L-lactate concentrations. As a compensatory mechanism to low blood pressure and electrolyte abnormalities, aldosterone and arginine vasopressin (AVP) are released to restore organ perfusion and function. Several studies have investigated the ability of blood L-lactate concentrations to predict severity of disease and outcome in critically ill human patients, adult horses and foals. However, information on the aldosterone and AVP response to hypoperfusion and its association with L-lactate concentrations in neonatal foals is limited. OBJECTIVES To determine the association between clinical hypoperfusion and endocrine markers of reduced tissue perfusion in normo- and hypoperfused foals. STUDY DESIGN Prospective, multicentre, cross-sectional observational study. METHODS Blood samples were collected on admission from 72 clinically hypoperfused, 110 normoperfused (73 hospitalised and 37 healthy) foals of ≤4 days of age. Foals were considered clinically hypoperfused if they had L-lactate concentrations ≥2.5 mmol/l and one of the 3 following findings: heart rate >120 beats/min, packed cell volume (PCV) >0.44 l/l or azotaemia (increased creatinine and blood urea nitrogen [BUN]). Blood concentrations of aldosterone and AVP were determined by radioimmunoassays. RESULTS Aldosterone, AVP, creatinine and BUN concentrations and heart rate, PCV and blood osmolality were higher in clinically hypoperfused compared with normoperfused foals (P<0.05). Risk of hypoperfusion increased with the presence of hypothermic extremities (OR = 5.26) and with each one unit increase in albumin concentrations (OR = 3.5) (P<0.05). The proposed admission L-lactate cut-off value above which nonsurvival could be reliably predicted in hospitalised foals was 10.6 mmol/l with 82% of sensitivity and 74% of specificity. CONCLUSIONS Hyperaldosteronaemia and hypervasopressinaemia as well as hypothermic extremities and increased albumin concentrations are potent predictors of hypoperfusion in hospitalised foals.
Collapse
Affiliation(s)
- K A Dembek
- College of Veterinary Medicine, The Ohio State University, Columbus, USA
| | - S D Hurcombe
- College of Veterinary Medicine, The Ohio State University, Columbus, USA
| | - A J Stewart
- College of Veterinary Medicine, Auburn University, Alabama, USA
| | - B S Barr
- Rood and Riddle Equine Hospital, Lexington, Kentucky, USA
| | | | - M Kinee
- Rood and Riddle Equine Hospital, Lexington, Kentucky, USA
| | - J Elam
- Hagyard Equine Medical Institute, Lexington, Kentucky, USA
| | - R E Toribio
- College of Veterinary Medicine, The Ohio State University, Columbus, USA
| |
Collapse
|
162
|
Butterworth MB. MicroRNAs and the regulation of aldosterone signaling in the kidney. Am J Physiol Cell Physiol 2015; 308:C521-7. [PMID: 25673770 DOI: 10.1152/ajpcell.00026.2015] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2015] [Accepted: 02/11/2015] [Indexed: 01/22/2023]
Abstract
The role of small noncoding RNAs, termed microRNAs (miRs), in development and disease has been recognized for many years. The number of miRs and regulated targets that reinforce a role for miRs in human disease and disease progression is ever-increasing. However, less is known about the involvement of miRs in steady-state, nondisease homeostatic pathways. In the kidney, much of the regulated ion transport is under the control of hormonal signaling. Evidence is emerging that miRs are involved in the hormonal regulation of kidney function and, particularly, in ion transport. In this short review, the production and intra- and extracellular signaling of miRs and the involvement of miRs in kidney disease are discussed. The discussion also focuses on the role of these small biological molecules in the homeostatic control of ion transport in the kidney. MiR regulation of and by corticosteroid hormones, in particular the mineralocorticoid hormone aldosterone, is considered. While information about the role of aldosterone-regulated miRs in the kidney is limited, an increase in the research in this area will undoubtedly highlight the involvement of miRs as central mediators of hormonal signaling in normal physiology.
Collapse
Affiliation(s)
- Michael B Butterworth
- Department of Cell Biology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania
| |
Collapse
|
163
|
Abstract
Aldosterone is a steroid hormone synthesized in and secreted from the outer layer of the adrenal cortex, the zona glomerulosa. Aldosterone is responsible for regulating sodium homeostasis, thereby helping to control blood volume and blood pressure. Insufficient aldosterone secretion can lead to hypotension and circulatory shock, particularly in infancy. On the other hand, excessive aldosterone levels, or those too high for sodium status, can cause hypertension and exacerbate the effects of high blood pressure on multiple organs, contributing to renal disease, stroke, visual loss, and congestive heart failure. Aldosterone is also thought to directly induce end-organ damage, including in the kidneys and heart. Because of the significance of aldosterone to the physiology and pathophysiology of the cardiovascular system, it is important to understand the regulation of its biosynthesis and secretion from the adrenal cortex. Herein, the mechanisms regulating aldosterone production in zona glomerulosa cells are discussed, with a particular emphasis on signaling pathways involved in the secretory response to the main controllers of aldosterone production, the renin-angiotensin II system, serum potassium levels and adrenocorticotrophic hormone. The signaling pathways involved include phospholipase C-mediated phosphoinositide hydrolysis, inositol 1,4,5-trisphosphate, cytosolic calcium levels, calcium influx pathways, calcium/calmodulin-dependent protein kinases, diacylglycerol, protein kinases C and D, 12-hydroxyeicostetraenoic acid, phospholipase D, mitogen-activated protein kinase pathways, tyrosine kinases, adenylate cyclase, and cAMP-dependent protein kinase. A complete understanding of the signaling events regulating aldosterone biosynthesis may allow the identification of novel targets for therapeutic interventions in hypertension, primary aldosteronism, congestive heart failure, renal disease, and other cardiovascular disorders.
Collapse
Affiliation(s)
- Wendy B Bollag
- Charlie Norwood VA Medical Center, Augusta, Georgia; Department of Physiology, Medical College of Georgia at Georgia Regents University, Augusta, Georgia
| |
Collapse
|
164
|
Magill SB. Pathophysiology, diagnosis, and treatment of mineralocorticoid disorders. Compr Physiol 2015; 4:1083-119. [PMID: 24944031 DOI: 10.1002/cphy.c130042] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
The renin-angiotensin-aldosterone system (RAAS) is a major regulator of blood pressure control, fluid, and electrolyte balance in humans. Chronic activation of mineralocorticoid production leads to dysregulation of the cardiovascular system and to hypertension. The key mineralocorticoid is aldosterone. Hyperaldosteronism causes sodium and fluid retention in the kidney. Combined with the actions of angiotensin II, chronic elevation in aldosterone leads to detrimental effects in the vasculature, heart, and brain. The adverse effects of excess aldosterone are heavily dependent on increased dietary salt intake as has been demonstrated in animal models and in humans. Hypertension develops due to complex genetic influences combined with environmental factors. In the last two decades, primary aldosteronism has been found to occur in 5% to 13% of subjects with hypertension. In addition, patients with hyperaldosteronism have more end organ manifestations such as left ventricular hypertrophy and have significant cardiovascular complications including higher rates of heart failure and atrial fibrillation compared to similarly matched patients with essential hypertension. The pathophysiology, diagnosis, and treatment of primary aldosteronism will be extensively reviewed. There are many pitfalls in the diagnosis and confirmation of the disorder that will be discussed. Other rare forms of hyper- and hypo-aldosteronism and unusual disorders of hypertension will also be reviewed in this article.
Collapse
Affiliation(s)
- Steven B Magill
- Division of Endocrinology, Metabolism, and Clinical Nutrition, Department of Medicine, Medical College of Wisconsin, Menomonee Falls, Wisconsin
| |
Collapse
|
165
|
Abstract
The purpose of this article is to review fundamentals in adrenal gland histophysiology. Key findings regarding the important signaling pathways involved in the regulation of steroidogenesis and adrenal growth are summarized. We illustrate how adrenal gland morphology and function are deeply interconnected in which novel signaling pathways (Wnt, Sonic hedgehog, Notch, β-catenin) or ionic channels are required for their integrity. Emphasis is given to exploring the mechanisms and challenges underlying the regulation of proliferation, growth, and functionality. Also addressed is the fact that while it is now well-accepted that steroidogenesis results from an enzymatic shuttle between mitochondria and endoplasmic reticulum, key questions still remain on the various aspects related to cellular uptake and delivery of free cholesterol. The significant progress achieved over the past decade regarding the precise molecular mechanisms by which the two main regulators of adrenal cortex, adrenocorticotropin hormone (ACTH) and angiotensin II act on their receptors is reviewed, including structure-activity relationships and their potential applications. Particular attention has been given to crucial second messengers and how various kinases, phosphatases, and cytoskeleton-associated proteins interact to ensure homeostasis and/or meet physiological demands. References to animal studies are also made in an attempt to unravel associated clinical conditions. Many of the aspects addressed in this article still represent a challenge for future studies, their outcome aimed at providing evidence that the adrenal gland, through its steroid hormones, occupies a central position in many situations where homeostasis is disrupted, thus highlighting the relevance of exploring and understanding how this key organ is regulated. © 2014 American Physiological Society. Compr Physiol 4:889-964, 2014.
Collapse
Affiliation(s)
- Nicole Gallo-Payet
- Division of Endocrinology, Department of Medicine, Faculté de Médecine et des Sciences de la Santé, Université de Sherbrooke, and Centre de Recherche Clinique Étienne-Le Bel of the Centre Hospitalier Universitaire de Sherbrooke (CHUS), Sherbrooke, Quebec, Canada
| | | |
Collapse
|
166
|
Rios FJ, Neves KB, Nguyen Dinh Cat A, Even S, Palacios R, Montezano AC, Touyz RM. Cholesteryl ester-transfer protein inhibitors stimulate aldosterone biosynthesis in adipocytes through Nox-dependent processes. J Pharmacol Exp Ther 2015; 353:27-34. [PMID: 25617244 DOI: 10.1124/jpet.114.221002] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Hyperaldosteronism and hypertension were unexpected side effects observed in trials of torcetrapib, a cholesteryl ester-transfer protein (CETP) inhibitor that increases high-density lipoprotein. Given that CETP inhibitors are lipid soluble, accumulate in adipose tissue, and have binding sites for proteins involved in adipogenesis, and that adipocytes are a source of aldosterone, we questioned whether CETP inhibitors (torcetrapib, dalcetrapib, and anacetrapib) influence aldosterone production by adipocytes. Studies were performed using human adipocytes (SW872), which express CETP, and mouse adipocytes (3T3-L1), which lack the CETP gene. Torcetrapib, dalcetrapib, and anacetrapib increased expression of CYP11B2, CYP11B1, and steroidogenic acute regulatory protein, enzymes involved in mineralocorticoid and glucocorticoid generation. These effects were associated with increased reactive oxygen species formation. Torcetrapib, dalcetrapib, and anacetrapib upregulated signal transducer and activator of transcription 3 (STAT3) and peroxisome proliferation-activated receptor-γ, important in adipogenesis, but only torcetrapib stimulated production of chemerin, a proinflammatory adipokine. To determine mechanisms whereby CETP inhibitors mediate effects, cells were pretreated with inhibitors of Nox1/Nox4 [GKT137831; 2-(2-chlorophenyl)-4-[3-(dimethylamino)phenyl]-5-methyl-1H-pyrazolo[4,3-c]pyridine-3,6(2H,5H)-dione], Nox1 (ML171 [2-acetylphenothiazine]), mitochondria (rotenone), and STAT3 (S3I-201 [2-hydroxy-4-(((4-methylphenyl)sulfonyloxy)acetyl)amino)-benzoic acid]). In torcetrapib-stimulated cells, Nox inhibitors, rotenone, and S3I-201 downregulated CYP11B2 and steroidogenic acute regulatory protein and reduced aldosterone. Dalcetrapib and anacetrapib effects on aldosterone were variably blocked by GKT137831, ML171, rotenone, and S3I-201. In adipocytes, torcetrapib, dalcetrapib, and anacetrapib inhibit enzymatic pathways responsible for aldosterone production through Nox1/Nox4- and mitochondrial-generated reactive oxygen species and STAT3. CETP inhibitors also influence adipokine production. These processes may be CETP independent. Our findings identify novel adipocyte-related mechanisms whereby CETP inhibitors increase aldosterone production. Such phenomena may contribute to hyperaldosteronism observed in CETP inhibitor clinical trials.
Collapse
Affiliation(s)
- Francisco J Rios
- Institute of Cardiovascular and Medical Sciences, British Heart Foundation Glasgow Cardiovascular Research Centre, University of Glasgow, Glasgow, Scotland, United Kingdom (F.J.R., A.N.D.C., S.E., A.C.M., R.M.T.); Faculty of Pharmaceutical Sciences of Ribeirao Preto, University of Sao Paulo, Ribeirao Preto, Brazil (K.B.N.); and Departamento de Bioquímica, Fisiología y Genética Molecular Facultad de CC. de la Salud, Universidad Rey Juan Carlos, Madrid, Spain (R.P.)
| | - Karla B Neves
- Institute of Cardiovascular and Medical Sciences, British Heart Foundation Glasgow Cardiovascular Research Centre, University of Glasgow, Glasgow, Scotland, United Kingdom (F.J.R., A.N.D.C., S.E., A.C.M., R.M.T.); Faculty of Pharmaceutical Sciences of Ribeirao Preto, University of Sao Paulo, Ribeirao Preto, Brazil (K.B.N.); and Departamento de Bioquímica, Fisiología y Genética Molecular Facultad de CC. de la Salud, Universidad Rey Juan Carlos, Madrid, Spain (R.P.)
| | - Aurelie Nguyen Dinh Cat
- Institute of Cardiovascular and Medical Sciences, British Heart Foundation Glasgow Cardiovascular Research Centre, University of Glasgow, Glasgow, Scotland, United Kingdom (F.J.R., A.N.D.C., S.E., A.C.M., R.M.T.); Faculty of Pharmaceutical Sciences of Ribeirao Preto, University of Sao Paulo, Ribeirao Preto, Brazil (K.B.N.); and Departamento de Bioquímica, Fisiología y Genética Molecular Facultad de CC. de la Salud, Universidad Rey Juan Carlos, Madrid, Spain (R.P.)
| | - Sarah Even
- Institute of Cardiovascular and Medical Sciences, British Heart Foundation Glasgow Cardiovascular Research Centre, University of Glasgow, Glasgow, Scotland, United Kingdom (F.J.R., A.N.D.C., S.E., A.C.M., R.M.T.); Faculty of Pharmaceutical Sciences of Ribeirao Preto, University of Sao Paulo, Ribeirao Preto, Brazil (K.B.N.); and Departamento de Bioquímica, Fisiología y Genética Molecular Facultad de CC. de la Salud, Universidad Rey Juan Carlos, Madrid, Spain (R.P.)
| | - Roberto Palacios
- Institute of Cardiovascular and Medical Sciences, British Heart Foundation Glasgow Cardiovascular Research Centre, University of Glasgow, Glasgow, Scotland, United Kingdom (F.J.R., A.N.D.C., S.E., A.C.M., R.M.T.); Faculty of Pharmaceutical Sciences of Ribeirao Preto, University of Sao Paulo, Ribeirao Preto, Brazil (K.B.N.); and Departamento de Bioquímica, Fisiología y Genética Molecular Facultad de CC. de la Salud, Universidad Rey Juan Carlos, Madrid, Spain (R.P.)
| | - Augusto C Montezano
- Institute of Cardiovascular and Medical Sciences, British Heart Foundation Glasgow Cardiovascular Research Centre, University of Glasgow, Glasgow, Scotland, United Kingdom (F.J.R., A.N.D.C., S.E., A.C.M., R.M.T.); Faculty of Pharmaceutical Sciences of Ribeirao Preto, University of Sao Paulo, Ribeirao Preto, Brazil (K.B.N.); and Departamento de Bioquímica, Fisiología y Genética Molecular Facultad de CC. de la Salud, Universidad Rey Juan Carlos, Madrid, Spain (R.P.)
| | - Rhian M Touyz
- Institute of Cardiovascular and Medical Sciences, British Heart Foundation Glasgow Cardiovascular Research Centre, University of Glasgow, Glasgow, Scotland, United Kingdom (F.J.R., A.N.D.C., S.E., A.C.M., R.M.T.); Faculty of Pharmaceutical Sciences of Ribeirao Preto, University of Sao Paulo, Ribeirao Preto, Brazil (K.B.N.); and Departamento de Bioquímica, Fisiología y Genética Molecular Facultad de CC. de la Salud, Universidad Rey Juan Carlos, Madrid, Spain (R.P.)
| |
Collapse
|
167
|
Abstract
The adrenal gland consists of two distinct parts, the cortex and the medulla. Molecular mechanisms controlling differentiation and growth of the adrenal gland have been studied in detail using mouse models. Knowledge also came from investigations of genetic disorders altering adrenal development and/or function. During embryonic development, the adrenal cortex acquires a structural and functional zonation in which the adrenal cortex is divided into three different steroidogenic zones. Significant progress has been made in understanding adrenal zonation. Recent lineage tracing experiments have accumulated evidence for a centripetal differentiation of adrenocortical cells from the subcapsular area to the inner part of the adrenal cortex. Understanding of the mechanism of adrenocortical cancer (ACC) development was stimulated by knowledge of adrenal gland development. ACC is a rare cancer with a very poor overall prognosis. Abnormal activation of the Wnt/β-catenin as well as the IGF2 signaling plays an important role in ACC development. Studies examining rare genetic syndromes responsible for familial ACT have played an important role in identifying genetic alterations in these tumors (like TP53 or CTNNB1 mutations as well as IGF2 overexpression). Recently, genomic analyses of ACT have shown gene expression profiles associated with malignancy as well as chromosomal and methylation alterations in ACT and exome sequencing allowed to describe the mutational landscape of these tumors. This progress leads to a new classification of these tumors, opening new perspectives for the diagnosis and prognostication of ACT. This review summarizes current knowledge of adrenocortical development, growth, and tumorigenesis.
Collapse
Affiliation(s)
- Lucile Lefèvre
- Inserm, U1016, Institut Cochin, Paris, France Cnrs, UMR8104, Paris, France Université Paris Descartes, Sorbonne Paris Cité, France Department of Endocrinology, Referral Center for Rare Adrenal Diseases, Assistance Publique Hôpitaux de Paris, Hôpital Cochin, Paris, France
| | | | | |
Collapse
|
168
|
Monticone S, Else T, Mulatero P, Williams TA, Rainey WE. Understanding primary aldosteronism: impact of next generation sequencing and expression profiling. Mol Cell Endocrinol 2015; 399:311-20. [PMID: 25240470 PMCID: PMC4285708 DOI: 10.1016/j.mce.2014.09.015] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/02/2014] [Revised: 09/11/2014] [Accepted: 09/15/2014] [Indexed: 10/24/2022]
Abstract
Primary aldosteronism (PA) encompasses a broad, heterogeneous group of disorders including both sporadic and familial forms (familial hyperaldosteronism type I, II and III). PA is the most common form of secondary hypertension and associated with a higher rate of cardiovascular complications, compared with essential hypertension. Despite significant progress in the diagnosis and management of PA, until recently the molecular mechanisms leading to inappropriate aldosterone production were largely unknown. The introduction of next-generation sequencing has had a profound impact on the field of human genetics and has given new insight in the molecular determinants that lead to both sporadic and familial forms of PA. Here we review the recent progress toward understanding of the genetic and molecular mechanisms leading to autonomous aldosterone production in PA.
Collapse
Affiliation(s)
- Silvia Monticone
- Department of Medical Sciences, Division of Internal Medicine and Hypertension, University of Torino, Torino, Italy.
| | - Tobias Else
- Department of Internal Medicine, Division of Metabolism, Endocrinology and Diabetes, University of Michigan Medical School, Ann Arbor, MI, USA
| | - Paolo Mulatero
- Department of Medical Sciences, Division of Internal Medicine and Hypertension, University of Torino, Torino, Italy
| | - Tracy A Williams
- Department of Medical Sciences, Division of Internal Medicine and Hypertension, University of Torino, Torino, Italy
| | - William E Rainey
- Department of Molecular and Integrative Physiology, University of Michigan Medical School, Ann Arbor, MI, USA
| |
Collapse
|
169
|
Abstract
Stem cells are endowed with the potential for self-renewal and multipotency. Pluripotent embryonic stem cells have an early role in the formation of the three germ layers (ectoderm, mesoderm and endoderm), whereas adult tissue stem cells and progenitor cells are critical mediators of organ homeostasis. The adrenal cortex is an exceptionally dynamic endocrine organ that is homeostatically maintained by paracrine and endocrine signals throughout postnatal life. In the past decade, much has been learned about the stem and progenitor cells of the adrenal cortex and the multiple roles that these cell populations have in normal development and homeostasis of the adrenal gland and in adrenal diseases. In this Review, we discuss the evidence for the presence of adrenocortical stem cells, as well as the various signalling molecules and transcriptional networks that are critical for the embryological establishment and postnatal maintenance of this vital population of cells. The implications of these pathways and cells in the pathophysiology of disease are also addressed.
Collapse
Affiliation(s)
- Elisabeth M Walczak
- Division of Nephrology, School of Medicine, Stanford University, Stanford, CA 94305, USA
| | - Gary D Hammer
- Center for Organogenesis, Alfred Taubman Biomedical Sciences Research Building, Room 1528, 109 Zina Pitcher Place, Ann Arbor, MI 48109-2200, USA
| |
Collapse
|
170
|
Verwoert GC, Hofland J, Amin N, Mattace-Raso FUS, Sijbrands EJG, Hofman A, van den Meiracker AH, Uitterlinden AG, van Duijn CM, de Jong FH, Danser AHJ. Expression and gene variation studies deny association of human HSD3B1 gene with aldosterone production or blood pressure. Am J Hypertens 2015; 28:113-20. [PMID: 24951726 DOI: 10.1093/ajh/hpu103] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/14/2022] Open
Abstract
BACKGROUND Recent evidence suggests that the type I 3β-hydroxysteroid dehydrogenase, a steroidogenic enzyme encoded by the HSD3B1 gene, could be involved in aldosterone production and that genetic variation in HSD3B1 is associated with blood pressure. These findings challenge the long-standing hypothesis that all adrenocortical steroidogenesis is executed by the type II iso-enzyme, encoded by HSD3B2. METHODS To verify these findings, the adrenal presence of HSD3B1 and its effect on aldosterone synthesis and blood pressure were studied in expression and genetic association analyses, respectively. Expression of HSD3B1 and HSD3B2 was investigated in various adrenocortical tissues (n = 15) and in primary adrenal cell cultures (n = 5) after stimulation with adrenocorticotropin and angiotensin II. Six tagging single nucleotide polymorphisms within the HSD3B1 gene were studied for association with blood pressure and hypertension in a meta-analysis of 4 Dutch cohorts (n = 11,192). RESULTS HSD3B1 expression was minimal or absent in adrenocortical tissues, including 6 aldosterone-producing adenomas. In contrast with the ubiquitously expressed HSD3B2 mRNA, HSD3B1 levels were not stimulated by adrenocorticotropin or angiotensin II. No variants in the HSD3B1 gene were associated with blood pressure or the occurrence of hypertension. CONCLUSIONS We found no evidence to support confirmation that HSD3B1 is involved in aldosterone synthesis in the human adrenal cortex or that genetic variation in HSD3B1 affects blood pressure or hypertension, favoring the hypothesis that all adrenocortical steroidogenesis is primarily dependent on the type II 3β-hydroxysteroid dehydrogenase.
Collapse
Affiliation(s)
- Germaine C Verwoert
- Department of Epidemiology, Erasmus MC, University Medical Center Rotterdam, Rotterdam, The Netherlands; Department of Internal Medicine, Erasmus MC, University Medical Center Rotterdam, Rotterdam, The Netherlands
| | - Johannes Hofland
- Department of Internal Medicine, Erasmus MC, University Medical Center Rotterdam, Rotterdam, The Netherlands
| | - Najaf Amin
- Department of Epidemiology, Erasmus MC, University Medical Center Rotterdam, Rotterdam, The Netherlands
| | - Francesco U S Mattace-Raso
- Department of Epidemiology, Erasmus MC, University Medical Center Rotterdam, Rotterdam, The Netherlands; Department of Internal Medicine, Erasmus MC, University Medical Center Rotterdam, Rotterdam, The Netherlands
| | - Eric J G Sijbrands
- Department of Internal Medicine, Erasmus MC, University Medical Center Rotterdam, Rotterdam, The Netherlands
| | - Albert Hofman
- Department of Epidemiology, Erasmus MC, University Medical Center Rotterdam, Rotterdam, The Netherlands
| | - Anton H van den Meiracker
- Department of Internal Medicine, Erasmus MC, University Medical Center Rotterdam, Rotterdam, The Netherlands
| | - André G Uitterlinden
- Department of Internal Medicine, Erasmus MC, University Medical Center Rotterdam, Rotterdam, The Netherlands
| | - Cornelia M van Duijn
- Department of Epidemiology, Erasmus MC, University Medical Center Rotterdam, Rotterdam, The Netherlands
| | - Frank H de Jong
- Department of Internal Medicine, Erasmus MC, University Medical Center Rotterdam, Rotterdam, The Netherlands
| | - A H Jan Danser
- Department of Internal Medicine, Erasmus MC, University Medical Center Rotterdam, Rotterdam, The Netherlands.
| |
Collapse
|
171
|
Yarimizu D, Doi M, Ota T, Okamura H. Stimulus-selective induction of the orphan nuclear receptor NGFIB underlies different influences of angiotensin II and potassium on the human adrenal gland zona glomerulosa-specific 3β-HSD isoform gene expression in adrenocortical H295R cells. Endocr J 2015; 62:765-76. [PMID: 26096451 DOI: 10.1507/endocrj.ej15-0211] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/23/2022] Open
Abstract
In the adrenal, the type I 3β-hydroxysteroid dehydrogenase (HSD3B1) is expressed exclusively in the zona glomerulosa (ZG), where aldosterone is produced. Angiotensin II (AngII) and potassium (K(+)) are the major physiological regulators of aldosterone synthesis. However, their respective roles in regulation of aldosterone synthesis are not fully defined, particularly in terms of transcriptional regulation of steroidogenic enzyme genes. We previously showed that AngII can stimulate expression of HSD3B1. But, K(+) responsiveness of this gene has remained unexplored. Here, we report that K(+) stimulation lacks the ability to induce HSD3B1 expression in human adrenocortical H295R cells. Both AngII and K(+) were able to enhance transcription of the aldosterone synthase gene (CYP11B2). Promoter analysis revealed that although both AngII and K(+) activate transcription from the Ca(2+)/cAMP-responsive element (CRE) located in the CYP11B2 promoter, the orphan nuclear receptor NGFIB-responsive element (NBRE) located in the HSD3B1 promoter fails to respond to K(+), being only able to enhance transcription after AngII treatment. We found that induction of de novo protein synthesis of NGFIB occurs only after AngII treatment. This sharply contrasts with the phosphorylation that occurs in response to both AngII and K(+) on the CREB/ATF family transcription factor ATF2. Chromatin immunoprecipitation assay confirmed that the NGFIB protein occupies the HSD3B1 promoter only after AngII, while ATF2 binds to the CYP11B2 promoter in response to both AngII and K(+). These data provide evidence that downstream signals from AngII and K(+) can be uncoupled in the regulation of HSD3B1 in the human adrenocortical H295R cells.
Collapse
Affiliation(s)
- Daisuke Yarimizu
- Department of Systems Biology, Graduate School of Pharmaceutical Sciences, Kyoto University, Sakyo-ku, Kyoto 606-8051, Japan
| | | | | | | |
Collapse
|
172
|
Cortés-Puch I, Hicks CW, Sun J, Solomon SB, Eichacker PQ, Sweeney DA, Nieman LK, Whitley EM, Behrend EN, Natanson C, Danner RL. Hypothalamic-pituitary-adrenal axis in lethal canine Staphylococcus aureus pneumonia. Am J Physiol Endocrinol Metab 2014; 307:E994-E1008. [PMID: 25294215 PMCID: PMC4254987 DOI: 10.1152/ajpendo.00345.2014] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
The clinical significance and even existence of critical illness-related corticosteroid insufficiency is controversial. Here, hypothalamic-pituitary-adrenal (HPA) function was characterized in severe canine Staphylococcus aureus pneumonia. Animals received antibiotics and titrated life-supportive measures. Treatment with dexamethasone, a glucocorticoid, but not desoxycorticosterone, a mineralocorticoid, improves outcome in this model. Total and free cortisol, adrenocorticotropic hormone (ACTH). and aldosterone levels, as well as responses to exogenous ACTH were measured serially. At 10 h after the onset of infection, the acute HPA axis stress response, as measured by cortisol levels, exceeded that seen with high-dose ACTH stimulation but was not predictive of outcome. In contrast to cortisol, aldosterone was largely autonomous from HPA axis control, elevated longer, and more closely associated with survival in early septic shock. Importantly, dexamethasone suppressed cortisol and ACTH levels and restored ACTH responsiveness in survivors. Differing strikingly, nonsurvivors, sepsis-induced hypercortisolemia, and high ACTH levels as well as ACTH hyporesponsiveness were not influenced by dexamethasone. During septic shock, only serial measurements and provocative testing over a well-defined timeline were able to demonstrate a strong relationship between HPA axis function and prognosis. HPA axis unresponsiveness and high aldosterone levels identify a septic shock subpopulation with poor outcomes that may have the greatest potential to benefit from new therapies.
Collapse
Affiliation(s)
- Irene Cortés-Puch
- Critical Care Medicine Department, Clinical Center, National Institutes of Health, Bethesda, Maryland;
| | - Caitlin W Hicks
- Critical Care Medicine Department, Clinical Center, National Institutes of Health, Bethesda, Maryland; Department of General Surgery, The Johns Hopkins Hospital, Baltimore, Maryland; National Institutes of Health Research Scholars Program, Howard Hughes Medical Institute, Chevy Chase, Maryland
| | - Junfeng Sun
- Critical Care Medicine Department, Clinical Center, National Institutes of Health, Bethesda, Maryland
| | - Steven B Solomon
- Critical Care Medicine Department, Clinical Center, National Institutes of Health, Bethesda, Maryland
| | - Peter Q Eichacker
- Critical Care Medicine Department, Clinical Center, National Institutes of Health, Bethesda, Maryland
| | - Daniel A Sweeney
- Medical Intensivist Program, Washington Hospital, Fremont, California
| | - Lynnette K Nieman
- Program in Reproductive and Adult Endocrinology, Eunice Kennedy Shriver National Institute of Child Health and Development, National Institutes of Health, Bethesda, Maryland
| | - Elizabeth M Whitley
- Department of Veterinary Pathology, College of Veterinary Medicine, Iowa State University, Ames, Iowa; and
| | - Ellen N Behrend
- Department of Clinical Sciences, College of Veterinary Medicine, Auburn University, Auburn, Alabama
| | - Charles Natanson
- Critical Care Medicine Department, Clinical Center, National Institutes of Health, Bethesda, Maryland
| | - Robert L Danner
- Critical Care Medicine Department, Clinical Center, National Institutes of Health, Bethesda, Maryland
| |
Collapse
|
173
|
Raff H, Gehrand A, Bruder ED, Hoffman MJ, Engeland WC, Moreno C. Renin knockout rat: control of adrenal aldosterone and corticosterone synthesis in vitro and adrenal gene expression. Am J Physiol Regul Integr Comp Physiol 2014; 308:R73-7. [PMID: 25394830 DOI: 10.1152/ajpregu.00440.2014] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
The classic renin-angiotensin system is partly responsible for controlling aldosterone secretion from the adrenal cortex via the peptide angiotensin II (ANG II). In addition, there is a local adrenocortical renin-angiotensin system that may be involved in the control of aldosterone synthesis in the zona glomerulosa (ZG). To characterize the long-term control of adrenal steroidogenesis, we utilized adrenal glands from renin knockout (KO) rats and compared steroidogenesis in vitro and steroidogenic enzyme expression to wild-type (WT) controls (Dahl S rat). Adrenal capsules (ZG; aldosterone production) and subcapsules [zona reticularis/fasciculata (ZFR); corticosterone production] were separately dispersed and studied in vitro. Plasma renin activity and ANG II concentrations were extremely low in the KO rats. Basal and cAMP-stimulated aldosterone production was significantly reduced in renin KO ZG cells, whereas corticosterone production was not different between WT and KO ZFR cells. As expected, adrenal renin mRNA expression was lower in the renin KO compared with the WT rat. Real-time PCR and immunohistochemical analysis showed a significant decrease in P450aldo (Cyp11b2) mRNA and protein expression in the ZG from the renin KO rat. The reduction in aldosterone synthesis in the ZG of the renin KO adrenal seems to be accounted for by a specific decrease in P450aldo and may be due to the absence of chronic stimulation of the ZG by circulating ANG II or to a reduction in locally released ANG II within the adrenal gland.
Collapse
Affiliation(s)
- Hershel Raff
- Endocrine Research Laboratory, Aurora St. Luke's Medical Center, Aurora Research Institute, Milwaukee, Wisconsin; Departments of Medicine, Surgery, and Physiology, Medical College of Wisconsin, Milwaukee, Wisconsin;
| | - Ashley Gehrand
- Endocrine Research Laboratory, Aurora St. Luke's Medical Center, Aurora Research Institute, Milwaukee, Wisconsin
| | - Eric D Bruder
- Endocrine Research Laboratory, Aurora St. Luke's Medical Center, Aurora Research Institute, Milwaukee, Wisconsin
| | - Matthew J Hoffman
- Department of Physiology, Medical College of Wisconsin, Milwaukee, Wisconsin; and
| | - William C Engeland
- Department of Neuroscience, University of Minnesota, Minneapolis, Minnesota
| | - Carol Moreno
- Department of Physiology, Medical College of Wisconsin, Milwaukee, Wisconsin; and
| |
Collapse
|
174
|
Buglioni A, Cannone V, Cataliotti A, Sangaralingham SJ, Heublein DM, Scott CG, Bailey KR, Rodeheffer RJ, Dessì-Fulgheri P, Sarzani R, Burnett JC. Circulating aldosterone and natriuretic peptides in the general community: relationship to cardiorenal and metabolic disease. Hypertension 2014; 65:45-53. [PMID: 25368032 DOI: 10.1161/hypertensionaha.114.03936] [Citation(s) in RCA: 65] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
We sought to investigate the role of aldosterone as a mediator of disease and its relationship with the counter-regulatory natriuretic peptide (NP) system. We measured plasma aldosterone (n=1674; aged≥45 years old) in a random sample of the general population from Olmsted County, MN. In a multivariate logistic regression model, aldosterone analyzed as a continuous variable was associated with hypertension (odds ratio [OR]=1.75; 95% confidence interval [CI]=1.57-1.96; P<0.0001), obesity (OR=1.34; 95% CI=1.21-1.48; P<0.0001), chronic kidney disease (OR=1.39; 95% CI=1.22-1.60; P<0.0001), central obesity (OR=1.47; 95% CI=1.32-1.63; P<0.0001), metabolic syndrome (OR=1.41; 95% CI=1.26-1.58; P<0.0001), high triglycerides (OR=1.23; 95% CI=1.11-1.36; P<0.0001), concentric left ventricular hypertrophy (OR=1.22; 95% CI=1.09-1.38; P=0.0007), and atrial fibrillation (OR=1.24; 95% CI=1.01-1.53; P=0.04), after adjusting for age and sex. The associations with hypertension, central obesity, metabolic syndrome, triglycerides, and concentric left ventricular hypertrophy remained significant after further adjustment for body mass index, NPs, and renal function. Furthermore, aldosterone in the highest tertile correlated with lower NP levels and increased mortality. Importantly, most of these associations remained significant even after excluding subjects with aldosterone levels above the normal range. In conclusion, we report that aldosterone is associated with hypertension, chronic kidney disease, obesity, metabolic syndrome, concentric left ventricular hypertrophy, and lower NPs in the general community. Our data suggest that aldosterone, even within the normal range, may be a biomarker of cardiorenal and metabolic disease. Further studies are warranted to evaluate a therapeutic and preventive strategy to delay the onset and progression of disease, using mineralocorticoid antagonists or chronic NP administration in high-risk subjects identified by plasma aldosterone.
Collapse
Affiliation(s)
- Alessia Buglioni
- From the Cardiorenal Research Laboratory, Division of Cardiovascular Diseases, Departments of Medicine and Health Sciences Research, College of Medicine Mayo Clinic, Rochester, MN (A.B., V.C., A.C., S.J.S., D.M.H., C.G.S., K.R.B., R.J.R., J.C.B.); and Internal Medicine and Geriatrics, Department of Clinical and Molecular Science, University "Politecnica delle Marche" and Italian National Research Centre on Aging, IRCCS/INRCA, Ancona, Italy (A.B., P.D.-F., R.S.).
| | - Valentina Cannone
- From the Cardiorenal Research Laboratory, Division of Cardiovascular Diseases, Departments of Medicine and Health Sciences Research, College of Medicine Mayo Clinic, Rochester, MN (A.B., V.C., A.C., S.J.S., D.M.H., C.G.S., K.R.B., R.J.R., J.C.B.); and Internal Medicine and Geriatrics, Department of Clinical and Molecular Science, University "Politecnica delle Marche" and Italian National Research Centre on Aging, IRCCS/INRCA, Ancona, Italy (A.B., P.D.-F., R.S.)
| | - Alessandro Cataliotti
- From the Cardiorenal Research Laboratory, Division of Cardiovascular Diseases, Departments of Medicine and Health Sciences Research, College of Medicine Mayo Clinic, Rochester, MN (A.B., V.C., A.C., S.J.S., D.M.H., C.G.S., K.R.B., R.J.R., J.C.B.); and Internal Medicine and Geriatrics, Department of Clinical and Molecular Science, University "Politecnica delle Marche" and Italian National Research Centre on Aging, IRCCS/INRCA, Ancona, Italy (A.B., P.D.-F., R.S.)
| | - S Jeson Sangaralingham
- From the Cardiorenal Research Laboratory, Division of Cardiovascular Diseases, Departments of Medicine and Health Sciences Research, College of Medicine Mayo Clinic, Rochester, MN (A.B., V.C., A.C., S.J.S., D.M.H., C.G.S., K.R.B., R.J.R., J.C.B.); and Internal Medicine and Geriatrics, Department of Clinical and Molecular Science, University "Politecnica delle Marche" and Italian National Research Centre on Aging, IRCCS/INRCA, Ancona, Italy (A.B., P.D.-F., R.S.)
| | - Denise M Heublein
- From the Cardiorenal Research Laboratory, Division of Cardiovascular Diseases, Departments of Medicine and Health Sciences Research, College of Medicine Mayo Clinic, Rochester, MN (A.B., V.C., A.C., S.J.S., D.M.H., C.G.S., K.R.B., R.J.R., J.C.B.); and Internal Medicine and Geriatrics, Department of Clinical and Molecular Science, University "Politecnica delle Marche" and Italian National Research Centre on Aging, IRCCS/INRCA, Ancona, Italy (A.B., P.D.-F., R.S.)
| | - Christopher G Scott
- From the Cardiorenal Research Laboratory, Division of Cardiovascular Diseases, Departments of Medicine and Health Sciences Research, College of Medicine Mayo Clinic, Rochester, MN (A.B., V.C., A.C., S.J.S., D.M.H., C.G.S., K.R.B., R.J.R., J.C.B.); and Internal Medicine and Geriatrics, Department of Clinical and Molecular Science, University "Politecnica delle Marche" and Italian National Research Centre on Aging, IRCCS/INRCA, Ancona, Italy (A.B., P.D.-F., R.S.)
| | - Kent R Bailey
- From the Cardiorenal Research Laboratory, Division of Cardiovascular Diseases, Departments of Medicine and Health Sciences Research, College of Medicine Mayo Clinic, Rochester, MN (A.B., V.C., A.C., S.J.S., D.M.H., C.G.S., K.R.B., R.J.R., J.C.B.); and Internal Medicine and Geriatrics, Department of Clinical and Molecular Science, University "Politecnica delle Marche" and Italian National Research Centre on Aging, IRCCS/INRCA, Ancona, Italy (A.B., P.D.-F., R.S.)
| | - Richard J Rodeheffer
- From the Cardiorenal Research Laboratory, Division of Cardiovascular Diseases, Departments of Medicine and Health Sciences Research, College of Medicine Mayo Clinic, Rochester, MN (A.B., V.C., A.C., S.J.S., D.M.H., C.G.S., K.R.B., R.J.R., J.C.B.); and Internal Medicine and Geriatrics, Department of Clinical and Molecular Science, University "Politecnica delle Marche" and Italian National Research Centre on Aging, IRCCS/INRCA, Ancona, Italy (A.B., P.D.-F., R.S.)
| | - Paolo Dessì-Fulgheri
- From the Cardiorenal Research Laboratory, Division of Cardiovascular Diseases, Departments of Medicine and Health Sciences Research, College of Medicine Mayo Clinic, Rochester, MN (A.B., V.C., A.C., S.J.S., D.M.H., C.G.S., K.R.B., R.J.R., J.C.B.); and Internal Medicine and Geriatrics, Department of Clinical and Molecular Science, University "Politecnica delle Marche" and Italian National Research Centre on Aging, IRCCS/INRCA, Ancona, Italy (A.B., P.D.-F., R.S.)
| | - Riccardo Sarzani
- From the Cardiorenal Research Laboratory, Division of Cardiovascular Diseases, Departments of Medicine and Health Sciences Research, College of Medicine Mayo Clinic, Rochester, MN (A.B., V.C., A.C., S.J.S., D.M.H., C.G.S., K.R.B., R.J.R., J.C.B.); and Internal Medicine and Geriatrics, Department of Clinical and Molecular Science, University "Politecnica delle Marche" and Italian National Research Centre on Aging, IRCCS/INRCA, Ancona, Italy (A.B., P.D.-F., R.S.)
| | - John C Burnett
- From the Cardiorenal Research Laboratory, Division of Cardiovascular Diseases, Departments of Medicine and Health Sciences Research, College of Medicine Mayo Clinic, Rochester, MN (A.B., V.C., A.C., S.J.S., D.M.H., C.G.S., K.R.B., R.J.R., J.C.B.); and Internal Medicine and Geriatrics, Department of Clinical and Molecular Science, University "Politecnica delle Marche" and Italian National Research Centre on Aging, IRCCS/INRCA, Ancona, Italy (A.B., P.D.-F., R.S.)
| |
Collapse
|
175
|
Fuller PJ, Young MJ. Duelling receptors: estrogen receptor versus mineralocorticoid receptor in the cardiovascular system. Endocrinology 2014; 155:4117-9. [PMID: 25325425 DOI: 10.1210/en.2014-1778] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Affiliation(s)
- Peter J Fuller
- MIMR-PHI Institute (formerly Prince Henry's Institute of Medical Research), Clayton, Victoria 3168, Australia
| | | |
Collapse
|
176
|
Yurek D, Yu L, Schrementi J, Bell MG, McGee J, Kowala M, Kuo MS, Wang J. Development of a high-throughput assay for aldosterone synthase inhibitors using high-performance liquid chromatography–tandem mass spectrometry. Anal Biochem 2014; 462:44-50. [DOI: 10.1016/j.ab.2014.06.011] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2013] [Revised: 06/13/2014] [Accepted: 06/14/2014] [Indexed: 10/25/2022]
|
177
|
Frau F, Zaninello R, Salvi E, Ortu MF, Braga D, Velayutham D, Argiolas G, Fresu G, Troffa C, Bulla E, Bulla P, Pitzoi S, Piras DA, Glorioso V, Chittani M, Bernini G, Bardini M, Fallo F, Malatino L, Stancanelli B, Regolisti G, Ferri C, Desideri G, Scioli GA, Galletti F, Sciacqua A, Perticone F, Degli Esposti E, Sturani A, Semplicini A, Veglio F, Mulatero P, Williams TA, Lanzani C, Hiltunen TP, Kontula K, Boerwinkle E, Turner ST, Manunta P, Barlassina C, Cusi D, Glorioso N. Genome-wide association study identifies CAMKID variants involved in blood pressure response to losartan: the SOPHIA study. Pharmacogenomics 2014; 15:1643-52. [DOI: 10.2217/pgs.14.119] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
Background: Essential hypertension arises from the combined effect of genetic and environmental factors. A pharmacogenomics approach could help to identify additional molecular mechanisms involved in its pathogenesis. Aim: The aim of SOPHIA study was to identify genetic polymorphisms regulating blood pressure response to the angiotensin II receptor blocker, losartan, with a whole-genome approach. Materials & methods: We performed a genome-wide association study on blood pressure response in 372 hypertensives treated with losartan and we looked for replication in two independent samples. Results: We identified a peak of association in CAMK1D gene (rs10752271, effect size -5.5 ± 0.94 mmHg, p = 1.2 × 10-8). CAMK1D encodes a protein that belongs to the regulatory pathway involved in aldosterone synthesis. We tested the specificity of rs10752271 for losartan in hypertensives treated with hydrochlorothiazide and we validated it in silico in the GENRES cohort. Conclusion: Using a genome-wide approach, we identified the CAMK1D gene as a novel locus associated with blood pressure response to losartan. CAMK1D gene characterization may represent a useful tool to personalize the treatment of essential hypertension. Original submitted 7 May 2014; Revision submitted 29 July 2014
Collapse
Affiliation(s)
- Francesca Frau
- Hypertension & Related Disease Centre, AOU-University of Sassari, Sassari, Italy
- Department of Health Sciences, University of Milan at San Paolo Hospital, Milan, Italy
- Filarete Foundation, Genomics & Bioinformatics Unit, Milan, Italy
| | - Roberta Zaninello
- Hypertension & Related Disease Centre, AOU-University of Sassari, Sassari, Italy
| | - Erika Salvi
- Department of Health Sciences, University of Milan at San Paolo Hospital, Milan, Italy
- Filarete Foundation, Genomics & Bioinformatics Unit, Milan, Italy
| | - Maria Francesca Ortu
- Hypertension & Related Disease Centre, AOU-University of Sassari, Sassari, Italy
| | - Daniele Braga
- Department of Health Sciences, University of Milan at San Paolo Hospital, Milan, Italy
- Filarete Foundation, Genomics & Bioinformatics Unit, Milan, Italy
| | - Dinesh Velayutham
- Department of Health Sciences, University of Milan at San Paolo Hospital, Milan, Italy
- Filarete Foundation, Genomics & Bioinformatics Unit, Milan, Italy
| | - Giuseppe Argiolas
- Hypertension & Related Disease Centre, AOU-University of Sassari, Sassari, Italy
| | - Giovanni Fresu
- Hypertension & Related Disease Centre, AOU-University of Sassari, Sassari, Italy
| | - Chiara Troffa
- Hypertension & Related Disease Centre, AOU-University of Sassari, Sassari, Italy
| | | | - Patrizia Bulla
- Hypertension & Related Disease Centre, AOU-University of Sassari, Sassari, Italy
| | - Silvia Pitzoi
- Hypertension & Related Disease Centre, AOU-University of Sassari, Sassari, Italy
| | | | - Valeria Glorioso
- Hypertension & Related Disease Centre, AOU-University of Sassari, Sassari, Italy
| | - Martina Chittani
- Department of Health Sciences, University of Milan at San Paolo Hospital, Milan, Italy
- Filarete Foundation, Genomics & Bioinformatics Unit, Milan, Italy
| | | | - Michele Bardini
- Department of Internal Medicine, University of Pisa, Pisa, Italy
| | | | - Lorenzo Malatino
- Department of Medicine & Hypertension Center, University of Catania at Cannizzaro Hospital, Catania, Italy
| | - Benedetta Stancanelli
- Department of Medicine & Hypertension Center, University of Catania at Cannizzaro Hospital, Catania, Italy
| | | | - Claudio Ferri
- Department of Internal Medicine & Public Health, University of L'Aquila, L'Aquila, Italy
| | | | | | - Ferruccio Galletti
- Department of Clinical Medicine & Surgery, “Federico II University” Medical School, Napoli, Italy
| | - Angela Sciacqua
- Department of Medical & Surgical Sciences, Cardiovascular Disease Unit, University of Catanzaro “Magna Graecia”, Catanzaro, Italy
| | - Francesco Perticone
- Department of Medical & Surgical Sciences, Cardiovascular Disease Unit, University of Catanzaro “Magna Graecia”, Catanzaro, Italy
| | | | | | - Andrea Semplicini
- UOC Internal Medicine 1, SS. Giovanni e Paolo Hospital, Campo SS. Giovanni e Paolo, Venice, Italy
| | - Franco Veglio
- Division of Internal Medicine & Hypertension Center, Department of Medical Sciences, AO Città Salute e Scienza, University of Torino, Torino, Italy
| | - Paolo Mulatero
- Division of Internal Medicine & Hypertension Center, Department of Medical Sciences, AO Città Salute e Scienza, University of Torino, Torino, Italy
| | - Tracy A Williams
- Division of Internal Medicine & Hypertension Center, Department of Medical Sciences, AO Città Salute e Scienza, University of Torino, Torino, Italy
| | - Chiara Lanzani
- Università Vita Salute San Raffaele, Nephrology, Dialysis & Hypertension Unit, San Raffaele Scientific Institute, Milan, Italy
| | - Timo P Hiltunen
- Department of Medicine, University of Helsinki & Helsinki University Central Hospital, Helsinki, Finland
| | - Kimmo Kontula
- Department of Medicine, University of Helsinki & Helsinki University Central Hospital, Helsinki, Finland
| | - Eric Boerwinkle
- Human Genetics & Institute of Molecular Medicine, University of Texas Health Science Center, Houston, TX, USA
| | - Stephen T Turner
- Division of Nephrology & Hypertension, Department of Medicine, Mayo Clinic, Rochester, MN, USA
| | - Paolo Manunta
- Università Vita Salute San Raffaele, Nephrology, Dialysis & Hypertension Unit, San Raffaele Scientific Institute, Milan, Italy
| | - Cristina Barlassina
- Department of Health Sciences, University of Milan at San Paolo Hospital, Milan, Italy
- Filarete Foundation, Genomics & Bioinformatics Unit, Milan, Italy
| | - Daniele Cusi
- Department of Health Sciences, University of Milan at San Paolo Hospital, Milan, Italy
- Filarete Foundation, Genomics & Bioinformatics Unit, Milan, Italy
| | - Nicola Glorioso
- Hypertension & Related Disease Centre, AOU-University of Sassari, Sassari, Italy
| |
Collapse
|
178
|
Angiotensin II triggers expression of the adrenal gland zona glomerulosa-specific 3β-hydroxysteroid dehydrogenase isoenzyme through de novo protein synthesis of the orphan nuclear receptors NGFIB and NURR1. Mol Cell Biol 2014; 34:3880-94. [PMID: 25092869 DOI: 10.1128/mcb.00852-14] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023] Open
Abstract
The 3β-hydroxysteroid dehydrogenase (3β-HSD) is an enzyme crucial for steroid synthesis. Two different 3β-HSD isoforms exist in humans. Classically, HSD3B2 was considered the principal isoform present in the adrenal. However, we recently showed that the alternative isoform, HSD3B1, is expressed specifically within the adrenal zona glomerulosa (ZG), where aldosterone is produced, raising the question of why this isozyme needs to be expressed in this cell type. Here we show that in both human and mouse, expression of the ZG isoform 3β-HSD is rapidly induced upon angiotensin II (AngII) stimulation. AngII is the key peptide hormone regulating the capacity of aldosterone synthesis. Using the human adrenocortical H295R cells as a model system, we show that the ZG isoform HSD3B1 differs from HSD3B2 in the ability to respond to AngII. Mechanistically, the induction of HSD3B1 involves de novo protein synthesis of the nuclear orphan receptors NGFIB and NURR1. The HSD3B1 promoter contains a functional NGFIB/NURR1-responsive element to which these proteins bind in response to AngII. Knockdown of these proteins and overexpression of a dominant negative NGFIB both reduce the AngII responsiveness of HSD3B1. Thus, the AngII-NGFIB/NURR1 pathway controls HSD3B1. Our work reveals HSD3B1 as a new regulatory target of AngII.
Collapse
|
179
|
Papanastasiou L, Markou A, Pappa T, Gouli A, Tsounas P, Fountoulakis S, Kounadi T, Tsiama V, Dasou A, Gryparis A, Samara C, Zografos G, Kaltsas G, Chrousos G, Piaditis G. Primary aldosteronism in hypertensive patients: clinical implications and target therapy. Eur J Clin Invest 2014; 44:697-706. [PMID: 24909545 DOI: 10.1111/eci.12286] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/23/2013] [Accepted: 06/03/2014] [Indexed: 12/27/2022]
Abstract
BACKGROUND The prevalence of primary aldosteronism (PA) in hypertensive patients varies according to diagnostic testing and ascertained normal cut-offs. The aim of this case-control study was to confirm the high prevalence of PA in a large hypertensive population and evaluate the antihypertensive effect of mineralocorticoid receptor antagonists (MRA) treatment. MATERIAL AND METHODS We investigated 327 hypertensive and 90 matched normotensive subjects with normal adrenal imaging. Serum aldosterone (ALD), active renin (REN) levels and aldosterone/active renin (ALD/REN) ratio were measured before and after a combined sodium chloride, fludrocortisone and dexamethasone suppression test (FDST). Post-FDST values were compared to cut-offs obtained from controls (post-FDST ALD 2·96 ng/dL and post-FDST ALD/REN 0·93 ng/dL/μU/mL). PA patients received MRA treatment. RESULTS By applying the combination of post-FDST ALD levels and ALD/REN ratio, 28·7% of the hypertensive patients had PA. There was a positive, albeit weak, correlation between systolic (SBP) and diastolic blood pressure (DBP) and ALD levels and/or ALD/REN ratio after the FDST (P < 0·0001). SBP was associated with a post-FDST ALD of 3·24 ng/dL and ALD/REN ratio of 0·90 ng/dL/μU/mL, whereas post-FDST ALD had an inverse association at serum K+ values of less than 3·9 mEq/L. MRA treatment in 69 PA patients, resulted in a significant reduction in the maximum SBP and DBP values (28 ± 15 and 14 ± 7 mmHg, respectively, P < 0·0001). CONCLUSIONS Using the FDST, an increased prevalence of PA in hypertensives was observed. Α significant blood pressure lowering effect was obtained with MRA treatment, implying that these agents may be beneficial in a significant number of hypertensive patients.
Collapse
Affiliation(s)
- Labrini Papanastasiou
- Department of Endocrinology and Diabetes Center, 'G Gennimatas' General Hospital, Athens, Greece
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
180
|
Drapala A, Sikora M, Ufnal M. Statins, the renin–angiotensin–aldosterone system and hypertension – a tale of another beneficial effect of statins. J Renin Angiotensin Aldosterone Syst 2014; 15:250-8. [DOI: 10.1177/1470320314531058] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023] Open
Affiliation(s)
- Adrian Drapala
- Department of Experimental and Clinical Physiology, Medical University of Warsaw, Poland
| | - Mariusz Sikora
- Department of Experimental and Clinical Physiology, Medical University of Warsaw, Poland
| | - Marcin Ufnal
- Department of Experimental and Clinical Physiology, Medical University of Warsaw, Poland
| |
Collapse
|
181
|
Olala LO, Choudhary V, Johnson MH, Bollag WB. Angiotensin II-induced protein kinase D activates the ATF/CREB family of transcription factors and promotes StAR mRNA expression. Endocrinology 2014; 155:2524-33. [PMID: 24708239 PMCID: PMC4060184 DOI: 10.1210/en.2013-1485] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Aldosterone synthesis is initiated upon the transport of cholesterol from the outer to the inner mitochondrial membrane, where the cholesterol is hydrolyzed to pregnenolone. This process is the rate-limiting step in acute aldosterone production and is mediated by the steroidogenic acute regulatory (StAR) protein. We have previously shown that angiotensin II (AngII) activation of the serine/threonine protein kinase D (PKD) promotes acute aldosterone production in bovine adrenal glomerulosa cells, but the mechanism remains unclear. Thus, the purpose of this study was to determine the downstream signaling effectors of AngII-stimulated PKD activity. Our results demonstrate that overexpression of the constitutively active serine-to-glutamate PKD mutant enhances, whereas the dominant-negative serine-to-alanine PKD mutant inhibits, AngII-induced StAR mRNA expression relative to the vector control. PKD has been shown to phosphorylate members of the activating transcription factor (ATF)/cAMP response element binding protein (CREB) family of leucine zipper transcription factors, which have been shown previously to bind the StAR proximal promoter and induce StAR mRNA expression. In primary glomerulosa cells, AngII induces ATF-2 and CREB phosphorylation in a time-dependent manner. Furthermore, overexpression of the constitutively active PKD mutant enhances the AngII-elicited phosphorylation of ATF-2 and CREB, and the dominant-negative mutant inhibits this response. Furthermore, the constitutively active PKD mutant increases the binding of phosphorylated CREB to the StAR promoter. Thus, these data provide insight into the previously reported role of PKD in AngII-induced acute aldosterone production, providing a mechanism by which PKD may be mediating steroidogenesis in primary bovine adrenal glomerulosa cells.
Collapse
Affiliation(s)
- Lawrence O Olala
- Charlie Norwood Veterans Administration Medical Center (L.O.O., V.C., W.B.B.), Augusta, Georgia 30904; and Departments of Physiology (L.O.O., V.C., W.B.B.), Biostatistics and Epidemiology (M.H.J.), and Cell Biology and Anatomy and Medicine and Orthopaedic Surgery (W.B.B.), Medical College of Georgia at Georgia Regents University, Augusta, Georgia 30912
| | | | | | | |
Collapse
|
182
|
Abstract
The primary adrenal cortical steroid hormones, aldosterone, and the glucocorticoids cortisol and corticosterone, act through the structurally similar mineralocorticoid (MR) and glucocorticoid receptors (GRs). Aldosterone is crucial for fluid, electrolyte, and hemodynamic homeostasis and tissue repair; the significantly more abundant glucocorticoids are indispensable for energy homeostasis, appropriate responses to stress, and limiting inflammation. Steroid receptors initiate gene transcription for proteins that effect their actions as well as rapid non-genomic effects through classical cell signaling pathways. GR and MR are expressed in many tissues types, often in the same cells, where they interact at molecular and functional levels, at times in synergy, others in opposition. Thus the appropriate balance of MR and GR activation is crucial for homeostasis. MR has the same binding affinity for aldosterone, cortisol, and corticosterone. Glucocorticoids activate MR in most tissues at basal levels and GR at stress levels. Inactivation of cortisol and corticosterone by 11β-HSD2 allows aldosterone to activate MR within aldosterone target cells and limits activation of the GR. Under most conditions, 11β-HSD1 acts as a reductase and activates cortisol/corticosterone, amplifying circulating levels. 11β-HSD1 and MR antagonists mitigate inappropriate activation of MR under conditions of oxidative stress that contributes to the pathophysiology of the cardiometabolic syndrome; however, MR antagonists decrease normal MR/GR functional interactions, a particular concern for neurons mediating cognition, memory, and affect.
Collapse
Affiliation(s)
- Elise Gomez-Sanchez
- G.V.(Sonny) Montgomery V.A. Medical Center and Department of Medicine, University of Mississippi Medical Center, Jackson, Mississippi
| | - Celso E. Gomez-Sanchez
- G.V.(Sonny) Montgomery V.A. Medical Center and Department of Medicine, University of Mississippi Medical Center, Jackson, Mississippi
| |
Collapse
|
183
|
Abstract
The prevalence of both obesity and hypertension are increasing worldwide. Hypertension is a common consequence of obesity. Increased central adiposity is associated with increased aldosterone levels and blood pressure in human beings. A number of small studies have shown an association between obesity-mediated hypertension and mechanisms directly linked to increased levels of aldosterone. These studies have shown a trend toward relatively greater blood pressure reduction using aldosterone-receptor blockers compared with other classes of antihypertensive agents. Other than treatment for weight loss, treatment of hypertension with specific antihypertensive medications that block or reduce aldosterone action are appropriate in obese patients. Further research is needed to understand the exact role of the adipocyte in obesity-mediated hypertension.
Collapse
|
184
|
Luo Y, Kumar P, Chen CC, Latham J, Wang L, Tudela C, Alexander JM, Shelton JM, McKown L, Mendelson CR. Estrogen-related receptor γ serves a role in blood pressure homeostasis during pregnancy. Mol Endocrinol 2014; 28:965-75. [PMID: 24725083 DOI: 10.1210/me.2014-1003] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022] Open
Abstract
Persistent hypoxia caused by shallow trophoblast invasion and poor placental perfusion may underlie the pathophysiology of preeclampsia, a leading cause of maternal and neonatal morbidity and mortality. Previously, we found that estrogen-related receptor γ (ERRγ) serves a critical and O2-dependent role in differentiation of human trophoblasts in culture and expression of tissue kallikrein and voltage-gated K(+) channels. In this study, we surprisingly observed that ERRγ expression was significantly increased in placentas from preeclamptic women compared with that in gestation-matched normotensive women. To further investigate a functional role for ERRγ during pregnancy, we analyzed ERRγ-deficient mice. Maternal systolic blood pressure was significantly reduced in pregnant ERRγ(+/-) females bred to ERRγ(+/-) males compared with that in wild-type (WT) mice and was markedly up-regulated by treatment of WT pregnant mice with the ERRγ agonist DY131. Placentas of ERRγ(+/-) mice manifested increased vascular endothelial growth factor A expression compared with that in WT mice. Notably, circulating levels of the antiangiogenic factor, soluble fms-like tyrosine kinase-1, were significantly reduced in ERRγ(+/-) pregnant mice as was serum aldosterone. These effects were associated with a decrease in maternal adrenal Cyp11b1 (steroid 11β-hydroxylase) and Cyp11b2 (aldosterone synthase) expression. In contrast, adrenal Cyp11b1 and Cyp11b2 mRNA were increased in pregnant WT mice treated with DY131. Moreover, chromatin immunoprecipitation and luciferase reporter assays identified Cyp11b2 as a transcriptional target of ERRγ. Collectively, these findings reveal a potential role of ERRγ in maternal blood pressure homeostasis during pregnancy and suggest that aberrant ERRγ expression may contribute to the pathogenesis of preeclampsia.
Collapse
Affiliation(s)
- Yanmin Luo
- Department of Biochemistry (Y.L., P.K., C.-C.C., J.L., L.W., C.R.M.), North Texas March of Dimes Birth Defects Center, University of Texas Southwestern Medical Center, Dallas, Texas 75390-9038; Department of Obstetrics and Gynecology (Y.L.), The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, 510080, China; and Department of Obstetrics and Gynecology (C.T., J.M.A., C.R.M.) and Department of Internal Medicine (J.M.S., L.M.), University of Texas Southwestern Medical Center, Dallas, Texas 75390-9038
| | | | | | | | | | | | | | | | | | | |
Collapse
|
185
|
Al-Salameh A, Cohen R, Desailloud R. Overview of the genetic determinants of primary aldosteronism. APPLICATION OF CLINICAL GENETICS 2014; 7:67-79. [PMID: 24817817 PMCID: PMC4012345 DOI: 10.2147/tacg.s45620] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
Primary aldosteronism is the most common cause of secondary hypertension. The syndrome accounts for 10% of all cases of hypertension and is primarily caused by bilateral adrenal hyperplasia or aldosterone-producing adenoma. Over the last few years, the use of exome sequencing has significantly improved our understanding of this syndrome. Somatic mutations in the KCNJ5, ATP1A1, ATP2B3 or CACNA1D genes are present in more than half of all cases of aldosterone-producing adenoma (~40%, ~6%, ~1% and ~8%, respectively). Germline gain-of-function mutations in KCNJ5 are now known to cause familial hyperaldosteronism type III, and an additional form of genetic hyperaldosteronism has been reported in patients with germline mutations in CACNA1D. These genes code for channels that control ion homeostasis across the plasma membrane of zona glomerulosa cells. Moreover, all these mutations modulate the same pathway, in which elevated intracellular calcium levels lead to aldosterone hyperproduction and (in some cases) adrenal cell proliferation. From a clinical standpoint, the discovery of these mutations has potential implications for patient management. The mutated channels could be targeted by drugs, in order to control hormonal and overgrowth-related manifestations. Furthermore, some of these mutations are associated with high cell turnover and may be amenable to diagnosis via the sequencing of cell-free (circulating) DNA. However, genotype-phenotype correlations in patients harboring these mutations have yet to be characterized. Despite this recent progress, much remains to be done to elucidate the yet unknown mechanisms underlying sporadic bilateral adrenal hyperplasia.
Collapse
Affiliation(s)
- Abdallah Al-Salameh
- Service de Diabétologie, Endocrinologie et Maladies Métaboliques, Centre Hospitalier de Creil, Creil, France
| | - Régis Cohen
- Service d'Endocrinologie, Centre Hospitalier de Saint-Denis, Saint-Denis, France
| | - Rachel Desailloud
- Service d'Endocrinologie, Diabétologie et Nutrition, Centre Hospitalier Universitaire d'Amiens, Amiens, France
| |
Collapse
|
186
|
Nishimoto K, Harris RBS, Rainey WE, Seki T. Sodium deficiency regulates rat adrenal zona glomerulosa gene expression. Endocrinology 2014; 155:1363-72. [PMID: 24422541 PMCID: PMC3959598 DOI: 10.1210/en.2013-1999] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
Aldosterone is the primary adrenocortical hormone regulating sodium retention, and its production is under the control of the renin-angiotensin-aldosterone system (RAAS). In vitro, angiotensin II can induce aldosterone production in adrenocortical cells without causing cell proliferation. In vivo, a low-sodium diet activates the RAAS and aldosterone production, at least in part, through an expansion of the adrenal zona glomerulosa (zG) layer. Although these mechanisms have been investigated, RAAS effects on zG gene expression have not been fully elucidated. In this study, we took an unbiased approach to define the complete list of zG transcripts involved in RAAS activation. Adrenal glands were collected from 11-week old Sprague-Dawley rats fed either sodium-deficient (SDef), normal sodium (NS), or high-sodium (HS) diet for 72 hours, and laser-captured zG RNA was analyzed on microarrays containing 27 342 probe sets. When the SDef transcriptome was compared with NS transcriptome (SDef/NS comparison), only 79 and 10 probe sets were found to be up- and down-regulated more than two-fold in SDef, respectively. In SDef/HS comparison, 201 and 68 probe sets were up- and down-regulated in SDef, respectively. Upon gene ontology (GO) analysis of these gene sets, we identified three groups of functionally related GO terms: cell proliferation-associated (group 1), response to stimulus-associated (group 2), and cholesterol/steroid metabolism-associated (group 3) GO terms. Although genes in group 1 may play a critical role in zG layer expansion, those in groups 2 and 3 may have important functions in aldosterone production, and further investigations on these genes are warranted.
Collapse
Affiliation(s)
- Koshiro Nishimoto
- Department of Molecular and Integrative Physiology (K.N., W.E.R.), University of Michigan, Ann Arbor, Michigan 48109; Department of Physiology (R.B.S.H., T.S.), Georgia Regents University, Augusta, Georgia 30912; and Department of Urology (K.N.), Tachikawa Hospital, Tachikawa, 190-0022 Tokyo, Japan
| | | | | | | |
Collapse
|
187
|
Matsuda K, Uruno A, Kogure N, Sugawara K, Shimada H, Nezu M, Saito-Ito T, Iki Y, Kudo M, Shimizu K, Sato I, Yoshikawa T, Satoh F, Ito R, Yokoyama A, Rainey WE, Saito-Hakoda A, Ito S, Sugawara A. Angiotensin II receptor blockers differentially affect CYP11B2 expression in human adrenal H295R cells. Mol Cell Endocrinol 2014; 383:60-8. [PMID: 24333837 DOI: 10.1016/j.mce.2013.12.004] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/17/2013] [Revised: 11/08/2013] [Accepted: 12/06/2013] [Indexed: 10/25/2022]
Abstract
We generated a stable H295R cell line expressing aldosterone synthase gene (CYP11B2) promoter/luciferase chimeric reporter construct that is highly sensitive to angiotensin II (AII) and potassium, and defined AII receptor blocker (ARB) effects. In the presence of AII, all ARBs suppressed AII-induced CYP11B2 transcription. However, telmisartan alone increased CYP11B2 transcription in the absence of AII. Telmisartan dose-dependently increased CYP11B2 transcription/mRNA expression and aldosterone secretion. Experiments using CYP11B2 promoter mutants indicated that the Ad5 element was responsible. Among transcription factors involved in the element, telmisartan significantly induced NGFIB/NURR1 expression. KN-93, a CaMK inhibitor, abrogated the telmisartan-mediated increase of CYP11B2 transcription/mRNA expression and NURR1 mRNA expression, but not NGFIB mRNA expression. NURR1 over-expression significantly augmented the telmisartan-mediated CYP11B2 transcription, while high-dose olmesartan did not affect it. Taken together, telmisartan may stimulate CYP11B2 transcription via NGFIB and the CaMK-mediated induction of NURR1 that activates the Ad5 element, independent of AII type 1 receptor.
Collapse
Affiliation(s)
- Ken Matsuda
- Division of Nephrology, Endocrinology and Vascular Medicine, Tohoku University Graduate School of Medicine, Sendai 980-8574, Japan
| | - Akira Uruno
- Department of Medical Biochemistry, Tohoku University Graduate School of Medicine, Sendai 980-8575, Japan
| | - Naotaka Kogure
- Department of Molecular Endocrinology, Tohoku University Graduate School of Medicine, Sendai 980-8575, Japan
| | - Kaori Sugawara
- Department of Molecular Endocrinology, Tohoku University Graduate School of Medicine, Sendai 980-8575, Japan
| | - Hiroki Shimada
- Department of Molecular Endocrinology, Tohoku University Graduate School of Medicine, Sendai 980-8575, Japan
| | - Masahiro Nezu
- Division of Nephrology, Endocrinology and Vascular Medicine, Tohoku University Graduate School of Medicine, Sendai 980-8574, Japan
| | - Takako Saito-Ito
- Department of Molecular Endocrinology, Tohoku University Graduate School of Medicine, Sendai 980-8575, Japan
| | - Yuko Iki
- Department of Molecular Endocrinology, Tohoku University Graduate School of Medicine, Sendai 980-8575, Japan
| | - Masataka Kudo
- Division of Nephrology, Endocrinology and Vascular Medicine, Tohoku University Graduate School of Medicine, Sendai 980-8574, Japan
| | - Kyoko Shimizu
- Department of Molecular Endocrinology, Tohoku University Graduate School of Medicine, Sendai 980-8575, Japan
| | - Ikuko Sato
- Department of Molecular Endocrinology, Tohoku University Graduate School of Medicine, Sendai 980-8575, Japan
| | - Takeo Yoshikawa
- Department of Pharmacology, Tohoku University Graduate School of Medicine, Sendai 980-8575, Japan
| | - Fumitoshi Satoh
- Division of Nephrology, Endocrinology and Vascular Medicine, Tohoku University Graduate School of Medicine, Sendai 980-8574, Japan
| | - Ryo Ito
- Department of Molecular Endocrinology, Tohoku University Graduate School of Medicine, Sendai 980-8575, Japan
| | - Atsushi Yokoyama
- Department of Molecular Endocrinology, Tohoku University Graduate School of Medicine, Sendai 980-8575, Japan
| | - William E Rainey
- Department of Molecular & Integrative Physiology, University of Michigan, Ann Arbor, MI 48109-5622, USA
| | - Akiko Saito-Hakoda
- Department of Molecular Endocrinology, Tohoku University Graduate School of Medicine, Sendai 980-8575, Japan
| | - Sadayoshi Ito
- Division of Nephrology, Endocrinology and Vascular Medicine, Tohoku University Graduate School of Medicine, Sendai 980-8574, Japan
| | - Akira Sugawara
- Department of Molecular Endocrinology, Tohoku University Graduate School of Medicine, Sendai 980-8575, Japan.
| |
Collapse
|
188
|
Bentley L, Esapa CT, Nesbit MA, Head RA, Evans H, Lath D, Scudamore CL, Hough TA, Podrini C, Hannan FM, Fraser WD, Croucher PI, Brown MA, Brown SDM, Cox RD, Thakker RV. An N-ethyl-N-nitrosourea induced corticotropin-releasing hormone promoter mutation provides a mouse model for endogenous glucocorticoid excess. Endocrinology 2014; 155:908-22. [PMID: 24302625 PMCID: PMC4192286 DOI: 10.1210/en.2013-1247] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
Abstract
Cushing's syndrome, which is characterized by excessive circulating glucocorticoid concentrations, may be due to ACTH-dependent or -independent causes that include anterior pituitary and adrenal cortical tumors, respectively. ACTH secretion is stimulated by CRH, and we report a mouse model for Cushing's syndrome due to an N-ethyl-N-nitrosourea (ENU) induced Crh mutation at -120 bp of the promoter region, which significantly increased luciferase reporter activity and was thus a gain-of-function mutation. Crh(-120/+) mice, when compared with wild-type littermates, had obesity, muscle wasting, thin skin, hair loss, and elevated plasma and urinary concentrations of corticosterone. In addition, Crh(-120/+) mice had hyperglycemia, hyperfructosaminemia, hyperinsulinemia, hypercholesterolemia, hypertriglyceridemia, and hyperleptinemia but normal adiponectin. Crh(-120/+) mice also had low bone mineral density, hypercalcemia, hypercalciuria, and decreased concentrations of plasma PTH and osteocalcin. Bone histomorphometry revealed Crh(-120/+) mice to have significant reductions in mineralizing surface area, mineral apposition, bone formation rates, osteoblast number, and the percentage of corticoendosteal bone covered by osteoblasts, which was accompanied by an increase in adipocytes in the bone marrow. Thus, a mouse model for Cushing's syndrome has been established, and this will help in further elucidating the pathophysiological effects of glucocorticoid excess and in evaluating treatments for corticosteroid-induced osteoporosis.
Collapse
|
189
|
Hunter RW, Ivy JR, Bailey MA. Glucocorticoids and renal Na+ transport: implications for hypertension and salt sensitivity. J Physiol 2014; 592:1731-44. [PMID: 24535442 PMCID: PMC4001748 DOI: 10.1113/jphysiol.2013.267609] [Citation(s) in RCA: 42] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023] Open
Abstract
The clinical manifestations of glucocorticoid excess include central obesity, hyperglycaemia, dyslipidaemia, electrolyte abnormalities and hypertension. A century on from Cushing's original case study, these cardinal features are prevalent in industrialized nations. Hypertension is the major modifiable risk factor for cardiovascular and renal disease and reflects underlying abnormalities of Na+ homeostasis. Aldosterone is a master regulator of renal Na+ transport but here we argue that glucocorticoids are also influential, particularly during moderate excess. The hypothalamic–pituitary–adrenal axis can affect renal Na+ homeostasis on multiple levels, systemically by increasing mineralocorticoid synthesis and locally by actions on both the mineralocorticoid and glucocorticoid receptors, both of which are expressed in the kidney. The kidney also expresses both of the 11β-hydroxysteroid dehydrogenase (11βHSD) enzymes. The intrarenal generation of active glucocorticoid by 11βHSD1 stimulates Na+ reabsorption; failure to downregulate the enzyme during adaption to high dietary salt causes salt-sensitive hypertension. The deactivation of glucocorticoid by 11βHSD2 underpins the regulatory dominance for Na+ transport of mineralocorticoids and defines the ‘aldosterone-sensitive distal nephron’. In summary, glucocorticoids can stimulate renal transport processes conventionally attributed to the renin–angiotensin–aldosterone system. Importantly, Na+ and volume homeostasis do not exert negative feedback on the hypothalamic–pituitary–adrenal axis. These actions are therefore clinically relevant and may contribute to the pathogenesis of hypertension in conditions associated with elevated glucocorticoid levels, such as the metabolic syndrome and chronic stress.
Collapse
Affiliation(s)
- Robert W Hunter
- University/BHF Centre for Cardiovascular Science, The Queen's Medical Research Institute, 47 Little France Crescent, Edinburgh EH16 4TJ, UK.
| | | | | |
Collapse
|
190
|
Zhang S, Morrison JL, Gill A, Rattanatray L, MacLaughlin SM, Kleemann D, Walker SK, McMillen IC. Dietary restriction in the periconceptional period in normal-weight or obese ewes results in increased abundance of angiotensin-converting enzyme (ACE) and angiotensin type 1 receptor (AT1R) in the absence of changes in ACE or AT1R methylation in the adrenal of the offspring. Reproduction 2013; 146:443-54. [DOI: 10.1530/rep-13-0219] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Abstract
Exposure to dietary restriction during the periconceptional period in either normal or obese ewes results in increased adrenal growth and a greater cortisol response to stress in the offspring, but the mechanisms that programme these changes are not fully understood. Activation of the angiotensin type 1 receptor (AT1R) has been demonstrated to stimulate adrenal growth and steroidogenesis. We have used an embryo transfer model in the sheep to investigate the effects of exposure to dietary restriction in normal or obese mothers from before and 1 week after conception on the methylation status, expression, abundance and localisation of key components of the renin–angiotensin system (RAS) in the adrenal of post-natal lambs. Maternal dietary restriction in normal or obese ewes during the periconceptional period resulted in an increase in angiotensin-converting enzyme (ACE) and AT1R abundance in the absence of changes in the methylation status or mRNA expression ofACEandAT1Rin the adrenal of the offspring. Exposure to maternal obesity alone also resulted in an increase in adrenal AT1R abundance. There was no effect of maternal dietary restriction or obesity on ACE2 and AT2R or on ERK, calcium/calmodulin-dependent kinase II abundance, and their phosphorylated forms in the lamb adrenal. Thus, weight loss around the time of conception, in both normal-weight and obese ewes, results in changes within the intra-adrenal RAS consistent with increased AT1R activation. These changes within the intra-adrenal RAS system may contribute to the greater adrenal stress response following exposure to signals of adversity in the periconceptional period.
Collapse
|
191
|
Harada E, Mizuno Y, Katoh D, Kashiwagi Y, Morita S, Nakayama Y, Yoshimura M, Masuzaki H, Saito Y, Yasue H. Increased urinary aldosterone excretion is associated with subcutaneous not visceral, adipose tissue area in obese individuals: a possible manifestation of dysfunctional subcutaneous adipose tissue. Clin Endocrinol (Oxf) 2013; 79:510-6. [PMID: 23106315 DOI: 10.1111/cen.12083] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/30/2012] [Revised: 10/19/2012] [Accepted: 10/20/2012] [Indexed: 12/31/2022]
Abstract
BACKGROUND Aldosterone is reported to be associated with obesity and is a risk factor for metabolic syndrome. Metabolic abnormalities are more strongly associated with visceral adipose tissue (VAT) than with subcutaneous adipose tissue (SAT). OBJECTIVE We examined whether aldosterone is more closely associated with VAT area than with SAT area in obese individuals. METHODS We enrolled 81 Japanese patients (46 men, mean age 43 ± 13 years and 35 women, mean age 53 ± 10 years) suspected of metabolic disorders and measured plasma and 24-h urinary aldosterone, as well as SAT and VAT areas. SAT and VAT areas were measured at the umbilical level by computed tomography. RESULTS Spearman's rank correlation analysis showed that urinary aldosterone was significantly and positively correlated with body mass index, waist circumference, SAT area, alanine aminotransferase, C-reactive protein, plasma immune-reactive insulin, plasma renin activity and estimated glomerular filtration rate, and negatively correlated with age and blood glucose. Urinary aldosterone was not correlated with VAT area (r = 0·013, P = 0·906). Multivariate regression analyses revealed that log SAT area, age and diastolic blood pressure were significant (P = 0·001, 0·001 and 0·032, respectively) predictors of log urinary aldosterone excretion rate. CONCLUSION Our results indicate that urinary aldosterone excretion is positively associated with SAT but not with VAT area in the middle-aged obese individuals.Urinary aldosterone is also negatively correlated with age.
Collapse
Affiliation(s)
- Eisaku Harada
- Division of Cardiovascular Medicine, Kumamoto Kinoh Hospital, Kumamoto Aging Research Institute, Kumamoto, Japan.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
192
|
Comparison of stress-induced changes in adults and pups: is aldosterone the main adrenocortical stress hormone during the perinatal period in rats? PLoS One 2013; 8:e72313. [PMID: 24039750 PMCID: PMC3763995 DOI: 10.1371/journal.pone.0072313] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2013] [Accepted: 07/10/2013] [Indexed: 02/06/2023] Open
Abstract
Positive developmental impact of low stress-induced glucocorticoid levels in early development has been recognized for a long time, while possible involvement of mineralocorticoids in the stress response during the perinatal period has been neglected. The present study aimed at verifying the hypothesis that balance between stress-induced glucocorticoid and mineralocorticoid levels is changing during postnatal development. Hormone responses to two different stressors (insulin-induced hypoglycaemia and immune challenge induced by bacterial lipopolysaccharid) measured in 10-day-old rats were compared to those in adults. In pups corticosterone responses to both stressors were significantly lower than in adults, which corresponded well with the stress hyporesponsive period. Importantly, stress-induced elevations in aldosterone concentration were significantly higher in pups compared both to corticosterone elevations and to those in adulthood with comparable adrenocorticotropin concentrations in the two age groups. Greater importance of mineralocorticoids compared to glucocorticoids in postnatal period is further supported by changes in gene expression and protein levels of gluco- (GR) and mineralocorticoid receptors (MR) and selected enzymes measured by quantitative PCR and immunohystochemistry in the hypothalamus, hippocampus, prefrontal cortex, liver and kidney. Gene expression of 11beta-hydroxysteroid dehydrogenase 2 (11β-HSD2), an enzyme enabling preferential effects of aldosterone on mineralocorticoid receptors, was higher in 10-day-old pups compared to adult animals. On the contrary, the expression and protein levels of GR, MR and 11β-HSD1 were decreased. Presented results clearly show higher stress-induced release of aldosterone in pups compared to adults and strongly suggest greater importance of mineralocorticoids compared to glucocorticoids in stress during the postnatal period.
Collapse
|
193
|
Martinez-Arguelles DB, Campioli E, Culty M, Zirkin BR, Papadopoulos V. Fetal origin of endocrine dysfunction in the adult: the phthalate model. J Steroid Biochem Mol Biol 2013; 137:5-17. [PMID: 23333934 DOI: 10.1016/j.jsbmb.2013.01.007] [Citation(s) in RCA: 97] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/22/2012] [Revised: 12/27/2012] [Accepted: 01/07/2013] [Indexed: 11/16/2022]
Abstract
Di-(2-ethylhexyl) phthalate (DEHP) is a plasticizer with endocrine disrupting properties that is found ubiquitously in the environment as well as in human amniotic fluid, umbilical cord blood, human milk, semen, and saliva. It is used in the industry to add flexibility to polyvinyl chloride-derived plastics and its wide spread use and presence has resulted in constant human exposure through fetal development and postnatal life. Epidemiological studies have suggested an association between phthalate exposures and human reproductive effects in infant and adult populations. The effects of fetal exposure to phthalates on the male reproductive system were unequivocally shown on animal models, principally rodents, in which short term deleterious reproductive effects are well established. By contrast, information on the long term effects of DEHP in utero exposure on gonadal function are scarce, while its potential effects on other organs are just starting to emerge. The present review focuses on these novel findings, which suggest that DEHP exerts more complex and broader disruptive effects on the endocrine system and metabolism than previously thought. This article is part of a Special Issue entitled "CSR 2013".
Collapse
Affiliation(s)
- D B Martinez-Arguelles
- The Research Institute of the McGill University Health Centre and Department of Medicine, McGill University, Montreal, Quebec, Canada
| | | | | | | | | |
Collapse
|
194
|
Suppression of primary aldosteronism and resistant hypertension by the peroxisome proliferator-activated receptor gamma agonist pioglitazone. Am J Med Sci 2013; 345:497-500. [PMID: 23313950 DOI: 10.1097/maj.0b013e31827ad893] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
The peroxisome proliferator-activated receptor gamma (PPAR γ) agonists have been reported to have antiproliferative and tumor-suppressive effects. We report a case of 55-year-old man with primary aldosteronism (PA) whose hyperaldosteronism was suppressed with the PPAR γ agonist pioglitazone. He had drug-resistant hypertension, hypokalemia, and diabetes mellitus. The diagnosis of PA was confirmed by the oral sodium loading test (20.5 μg/d of urinary aldosterone) and Captopril challenge test (19.5 ng/dL of plasma aldosterone). Computed tomography imaging revealed no apparent adrenal mass. The result of the posture stimulation test was consistent with the diagnosis of idiopathic adrenal hyperplasia. On administration of pioglitazone (30 mg/d) and nifedipine (40 mg/d), hypertension and hypokalemia improved and plasma aldosterone decreased for more than 6 months. The sodium loading test done after 6 months of the administration revealed the near normal results (11.2 ng/dL of plasma aldosterone and 13.1 μg/d of urinary aldosterone). The findings indicated that pioglitazone suppressed PA.
Collapse
|
195
|
Abstract
Aldosterone, the key hormone in the mineralocorticoid pathway, plays a fundamental role in salt and water homeostasis, blood pressure regulation, and cardiovascular remodeling. Both genomic and nongenomic mechanisms influence aldosterone-induced renal sodium reabsorption. Furthermore, the mineralocorticoid receptors in nonepithelial tissues, including the heart and vascular smooth muscle cells, have recently been discovered. Thus, aldosterone likely has pleiotropic effects that contribute to the modulation of blood pressure. Among patients with hypertension in general, and among those with more severe or resistant hypertension in particular, a higher-than-expected prevalence of primary hyperaldosteronism is noted. Among individuals with resistant hypertension, aldosterone antagonists have also been shown to be effective in lowering blood pressure. Most significantly, recent community-based studies among non-hypertensive individuals in the general population have demonstrated that both higher serum aldosterone concentrations and a higher aldosterone to renin ratio portend a greater risk of developing hypertension. The combination of the aforementioned observations underscores the importance of the mineralocorticoid pathway in the pathogenesis of hypertension.
Collapse
Affiliation(s)
- Vanessa Xanthakis
- Section of Preventive Medicine and Epidemiology, Boston University School of Medicine, 801 Massachusetts Avenue, Suite 470, Boston, MA 02118, USA.
| | | |
Collapse
|
196
|
Nishimoto K, Rainey WE, Bollag WB, Seki T. Lessons from the gene expression pattern of the rat zona glomerulosa. Mol Cell Endocrinol 2013; 371:107-13. [PMID: 23287491 PMCID: PMC3625490 DOI: 10.1016/j.mce.2012.12.023] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/04/2012] [Revised: 12/20/2012] [Accepted: 12/20/2012] [Indexed: 12/24/2022]
Abstract
We recently identified hundreds of transcripts with differential expression in rat zona glomerulosa (zG) and zona fasciculata. Although the genes up-regulated in the zG may be playing important roles in aldosterone production, the relationship between most of these genes and aldosterone production has not been uncovered. Because aldosterone, in the presence of a high sodium diet, is now considered a significant cardiovascular risk factor, in this review we performed gene ontology and pathway analyses on the same microarray data to better define the genes that may influence zG function. Overall, we identified a number of genes that may be involved in aldosterone production through transforming growth factor β (TGF-β), WNT, calcium, potassium, and ACTH signaling pathways. The list of genes we present in the current report may become an important tool for researchers working on primary aldosteronism and aldosterone-related cardiovascular diseases.
Collapse
Affiliation(s)
- Koshiro Nishimoto
- Department of Physiology, Medical College of Georgia, Georgia Health Sciences University, Augusta, Georgia 30912
- Department of Urology, Tachikawa Hospital, Tokyo 190-8531, Japan
| | - William E. Rainey
- Department of Physiology, Medical College of Georgia, Georgia Health Sciences University, Augusta, Georgia 30912
| | - Wendy B. Bollag
- Department of Physiology, Medical College of Georgia, Georgia Health Sciences University, Augusta, Georgia 30912
- Charlie Norwood VA Medical Center, Augusta, GA 30904
| | - Tsugio Seki
- Department of Physiology, Medical College of Georgia, Georgia Health Sciences University, Augusta, Georgia 30912
- Corresponding author: Tsugio Seki, Department of Physiology, Medical College of Georgia, Georgia Health Sciences University, 1120 15th Street, CA3064, Augusta, GA 30912; Tel., +1-706-721-1321; Fax., +1-706-721-7299
| |
Collapse
|
197
|
Yagi S, Akaike M, Aihara KI, Fukuda D, Ishida M, Ise T, Niki T, Sumitomo-Ueda Y, Yamaguchi K, Iwase T, Taketani Y, Yamada H, Soeki T, Wakatsuki T, Shimabukuro M, Sata M. Pharmacology of Aldosterone and the Effects of Mineralocorticoid Receptor Blockade on Cardiovascular Systems. ACTA CARDIOLOGICA SINICA 2013; 29:201-207. [PMID: 27122708 PMCID: PMC4804831] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Received: 03/11/2013] [Accepted: 04/14/2013] [Indexed: 06/05/2023]
Abstract
UNLABELLED It is well-known that aldosterone plays an important role in reabsorption of sodium and fluid, and in potassium excretion in kidneys via epithelial mineralocorticoid receptor (MR) activation. Recent studies have shown that aldosterone causes cardiovascular remodeling not only in a blood pressure-dependent manner, but also in a blood pressure-independent manner by decreasing nitric oxide bioavailability and modulating oxidative stress, leading to vascular inflammation. In addition, MR blockade does provide beneficial effects associated with cardiovascular protection, resulting in a reduction of cardiovascular morbidity and mortality. A growing body of evidence suggests that MR blockade is a promising therapeutic target to help prevent cardiovascular events. KEY WORDS Aldosterone; Mineralocorticoid receptor; Nitrix oxide; Renin-angiotensin-aldosterone system.
Collapse
Affiliation(s)
- Shusuke Yagi
- Department of Cardiovascular Medicine, University of Tokushima Graduate School of Health Biosciences, Japan
| | - Masashi Akaike
- Department of Medical Education, University of Tokushima Graduate School of Health Biosciences, Japan
| | - Ken-ichi Aihara
- Department of Medicine and Bioregulatory Sciences, University of Tokushima Graduate School of Health Biosciences, Japan
| | - Daiju Fukuda
- Department of Cardio-Diabetes Medicine, University of Tokushima Graduate School of Health Biosciences, Japan
| | - Masayoshi Ishida
- Department of Cardiovascular Medicine, University of Tokushima Graduate School of Health Biosciences, Japan
| | - Takayuki Ise
- Department of Cardiovascular Medicine, University of Tokushima Graduate School of Health Biosciences, Japan
| | - Toshiyuki Niki
- Department of Cardiovascular Medicine, University of Tokushima Graduate School of Health Biosciences, Japan
| | - Yuka Sumitomo-Ueda
- Department of Cardiovascular Medicine, University of Tokushima Graduate School of Health Biosciences, Japan
| | - Koji Yamaguchi
- Department of Cardiovascular Medicine, University of Tokushima Graduate School of Health Biosciences, Japan
| | - Takashi Iwase
- Department of Cardiovascular Medicine, University of Tokushima Graduate School of Health Biosciences, Japan
| | - Yoshio Taketani
- Department of Cardiovascular Medicine, University of Tokushima Graduate School of Health Biosciences, Japan
| | - Hirotsugu Yamada
- Department of Cardiovascular Medicine, University of Tokushima Graduate School of Health Biosciences, Japan
| | - Takeshi Soeki
- Department of Cardiovascular Medicine, University of Tokushima Graduate School of Health Biosciences, Japan
| | - Tetsuzo Wakatsuki
- Department of Cardiovascular Medicine, University of Tokushima Graduate School of Health Biosciences, Japan
| | - Michio Shimabukuro
- Department of Cardio-Diabetes Medicine, University of Tokushima Graduate School of Health Biosciences, Japan
| | - Masataka Sata
- Department of Cardiovascular Medicine, University of Tokushima Graduate School of Health Biosciences, Japan
| |
Collapse
|
198
|
Olala LO, Seremwe M, Tsai YY, Bollag WB. A role for phospholipase D in angiotensin II-induced protein kinase D activation in adrenal glomerulosa cell models. Mol Cell Endocrinol 2013. [PMID: 23178798 PMCID: PMC3656657 DOI: 10.1016/j.mce.2012.11.008] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
The mineralocorticoid aldosterone plays an important role in regulating blood pressure, with excess causing hypertension and exacerbating cardiovascular disease. Previous studies have indicated a role for both phospholipase D (PLD) and protein kinase D (PKD) in angiotensin II (AngII)-regulated aldosterone production in adrenal glomerulosa cells. Therefore, the relationship between AngII-activated PLD and PKD was determined in two glomerulosa cell models, primary bovine zona glomerulosa (ZG) and HAC15 human adrenocortical carcinoma cells, using two inhibitors, 1-butanol and the reported PLD inhibitor, fluoro-2-indolyl des-chlorohalopemide (FIPI). FIPI was first confirmed to decrease PLD activation in response to AngII in the two glomerulosa cell models. Subsequently, it was shown that both 1-butanol and FIPI inhibited AngII-elicited PKD activation and aldosterone production. These results indicate that PKD is downstream of PLD and suggest that PKD is one of the mechanisms through which PLD promotes aldosterone production in response to AngII in adrenal glomerulosa cells.
Collapse
Affiliation(s)
- Lawrence O. Olala
- Department of Physiology, Georgia Health Sciences University (formerly the Medical College of Georgia), 1120 15th Street, Augusta, GA 30912
| | - Mutsa Seremwe
- Department of Physiology, Georgia Health Sciences University (formerly the Medical College of Georgia), 1120 15th Street, Augusta, GA 30912
| | - Ying-Ying Tsai
- Department of Physiology, Georgia Health Sciences University (formerly the Medical College of Georgia), 1120 15th Street, Augusta, GA 30912
| | - Wendy B. Bollag
- Charlie Norwood VA Medical Center, One Freedom Way, Augusta, GA 30904
- Department of Physiology, Georgia Health Sciences University (formerly the Medical College of Georgia), 1120 15th Street, Augusta, GA 30912
- Departments of Cell Biology and Anatomy, Medicine, Oral Biology and Orthopaedic Surgery, Georgia Health Sciences University, 1120 15th Street, Augusta, GA 30912
- To whom correspondence should be addressed: Wendy Bollag, Georgia Health Sciences University, Department of Physiology, 1120 15th Street, Augusta, GA 30912, TEL: (706) 721-0698, FAX: (706) 721-7299,
| |
Collapse
|
199
|
Mulatero P, Monticone S, Rainey WE, Veglio F, Williams TA. Role of KCNJ5 in familial and sporadic primary aldosteronism. Nat Rev Endocrinol 2013; 9:104-12. [PMID: 23229280 DOI: 10.1038/nrendo.2012.230] [Citation(s) in RCA: 80] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Primary aldosteronism is characterised by the dysregulation of aldosterone production and comprises both sporadic forms, caused by an aldosterone-producing adenoma or bilateral adrenal hyperplasia, and familial forms (familial hyperaldosteronism types I, II and III). The two principal physiological regulators of aldosterone synthesis are angiotensin II and serum K(+), which reverse the high resting K(+) conductance and hyperpolarized membrane potential of adrenal glomerulosa cells. The resulting membrane depolarization causes the opening of voltage-gated Ca(2+) channels and an increase in intracellular Ca(2+) that stimulates aldosterone biosynthesis. Point mutations in the KCNJ5 gene, which encodes the G-protein-activated inward rectifier K(+) channel 4 (GIRK4), have been implicated in the pathogenesis of both sporadic and familial forms of primary aldosteronism. These mutations interfere with the selectivity filter of GIRK4 causing Na(+) entry, cell depolarization and Ca(2+) channel opening, resulting in constitutive aldosterone production. Seven families with familial hyperaldosteronism caused by KCNJ5 germline mutations have so far been described, and multicentre studies have reported KCNJ5 mutations in approximately 40% of sporadic aldosterone-producing adenomas. Herein, we review the role of GIRK4 in adrenal pathophysiology and provide an overview of the clinical and biochemical phenotypes resulting from KCNJ5 mutations in patients with sporadic and familial primary aldosteronism.
Collapse
Affiliation(s)
- Paolo Mulatero
- University of Torino, Department of Medical Sciences, Division of Internal Medicine and Hypertension, Italy. paolo.mulatero@ unito.it
| | | | | | | | | |
Collapse
|
200
|
Strushkevich N, Gilep AA, Shen L, Arrowsmith CH, Edwards AM, Usanov SA, Park HW. Structural insights into aldosterone synthase substrate specificity and targeted inhibition. Mol Endocrinol 2013; 27:315-24. [PMID: 23322723 PMCID: PMC5417327 DOI: 10.1210/me.2012-1287] [Citation(s) in RCA: 96] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023] Open
Abstract
Aldosterone is a major mineralocorticoid hormone that plays a key role in the regulation of electrolyte balance and blood pressure. Excess aldosterone levels can arise from dysregulation of the renin-angiotensin-aldosterone system and are implicated in the pathogenesis of hypertension and heart failure. Aldosterone synthase (cytochrome P450 11B2, CYP11B2) is the sole enzyme responsible for the production of aldosterone in humans. Blocking of aldosterone synthesis by mediating aldosterone synthase activity is thus a recently emerging pharmacological therapy for hypertension, yet a lack of structural information has limited this approach. Here, we present the crystal structures of human aldosterone synthase in complex with a substrate deoxycorticosterone and an inhibitor fadrozole. The structures reveal a hydrophobic cavity with specific features associated with corticosteroid recognition. The substrate binding mode, along with biochemical data, explains the high 11β-hydroxylase activity of aldosterone synthase toward both gluco- and mineralocorticoid formation. The low processivity of aldosterone synthase with a high extent of intermediates release might be one of the mechanisms of controlled aldosterone production from deoxycorticosterone. Although the active site pocket is lined by identical residues between CYP11B isoforms, most of the divergent residues that confer additional 18-oxidase activity of aldosterone synthase are located in the I-helix (vicinity of the O(2) activation path) and loops around the H-helix (affecting an egress channel closure required for retaining intermediates in the active site). This intrinsic flexibility is also reflected in isoform-selective inhibitor binding. Fadrozole binds to aldosterone synthase in the R-configuration, using part of the active site cavity pointing toward the egress channel. The structural organization of aldosterone synthase provides critical insights into the molecular mechanism of catalysis and enables rational design of more specific antihypertensive agents.
Collapse
Affiliation(s)
- Natallia Strushkevich
- Structural Genomics Consortium, University of Toronto, Toronto, Ontario, Canada M5G 1L7.
| | | | | | | | | | | | | |
Collapse
|