1
|
Marques PL, Galac S, Mateus L, Leal RO. Bridging the gap-Rethinking the role of the adrenal gland in chronic kidney disease from the feline perspective. Domest Anim Endocrinol 2025; 93:106955. [PMID: 40412070 DOI: 10.1016/j.domaniend.2025.106955] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/24/2025] [Revised: 05/16/2025] [Accepted: 05/16/2025] [Indexed: 05/27/2025]
Abstract
Chronic kidney disease (CKD) is the most common metabolic disease in domestic cats. Unlike humans and dogs, CKD in cats seems to have a highly complex and multifactorial etiology. Despite great effort being poured into research trying to elucidate possible pathways for the pathogenesis of CKD, there is still a lack of understanding regarding its initiating and progression factors. There is also a lack of therapeutic options for these patients, with most treatment plans relying on a low-phosphate diet, dietary protein modification and medical management of complications (e.g. hypertension) as they arise. In this review, we propose the hypothalamic-pituitary-adrenal (HPA) axis plays a central role in the development, pathophysiology and progression of feline chronic kidney disease. The adrenal glands and the hormones they secrete, in particular, may act as lynchpins in chronic kidney disease, mediating virtually every aspect of the disease: from the establishment of fibrosis and kidney damage to the development of hypertension and a pro-inflammatory status. By compiling the available research regarding the influence of adrenal hormones and the HPA axis, we hope to highlight possible future areas of scientific interest regarding feline CKD as well as possible aspects in which the cat may act as a model for research in human medicine.
Collapse
Affiliation(s)
- Patricia Lunet Marques
- Centre for Interdisciplinary Research in Animal Health, Faculty of Veterinary Medicine, University of Lisbon, Lisbon, Portugal; Associate Laboratory for Animal and Veterinary Sciences (AL4AnimalS), Lisbon, Portugal.
| | - Sara Galac
- Utrecht University - Faculty of Veterinary Medicine, Utrecht, Netherlands.
| | - Luísa Mateus
- Centre for Interdisciplinary Research in Animal Health, Faculty of Veterinary Medicine, University of Lisbon, Lisbon, Portugal; Associate Laboratory for Animal and Veterinary Sciences (AL4AnimalS), Lisbon, Portugal.
| | - Rodolfo Oliveira Leal
- Centre for Interdisciplinary Research in Animal Health, Faculty of Veterinary Medicine, University of Lisbon, Lisbon, Portugal; Associate Laboratory for Animal and Veterinary Sciences (AL4AnimalS), Lisbon, Portugal.
| |
Collapse
|
2
|
Guo C, Zhang G, Wu C, Lei Y, Wang Y, Yang J. Emerging trends in small molecule inhibitors targeting aldosterone synthase: A new paradigm in cardiovascular disease treatment. Eur J Med Chem 2024; 274:116521. [PMID: 38820853 DOI: 10.1016/j.ejmech.2024.116521] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2024] [Revised: 05/01/2024] [Accepted: 05/19/2024] [Indexed: 06/02/2024]
Abstract
Aldosterone synthase (CYP11B2) is the rate-limiting enzyme in aldosterone production. In recent years, CYP11B2 has become an appealing target for treating conditions associated with excess aldosterone, such as hypertension, heart failure, and cardiometabolic diseases. Several small-molecule inhibitors of CYP11B2 have demonstrated efficacy in both preclinical studies and clinical trials. Among them, the tetrahydroisoquinoline derivative Baxdrostat has entered clinical trial phases and demonstrated efficacy in treating patients with hypertension. However, the high homology (>93 %) between CYP11B2 and steroid-11β-hydroxylase (CYP11B1), which catalyzes cortisol production, implies that insufficient drug specificity can lead to severe side effects. Developing selective inhibitors for CYP11B2 remains a considerable challenge that requires ongoing attention. This review summarizes recent research progress on small-molecule inhibitors targeting CYP11B2, focusing on structure-activity relationships (SAR) and structural optimization. It discusses strategies for enhancing the specificity and inhibitory activity of inhibitors, while also exploring potential applications and future prospects for CYP11B2 inhibitors, providing a theoretical foundation for developing the new generation of CYP11B2-targeted medications.
Collapse
Affiliation(s)
- Cuiyu Guo
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, West China Medical School, Sichuan University, Chengdu, 610041, Sichuan, China; Department of Pulmonary and Critical Care Medicine, Targeted Tracer Research and Development Laboratory, Institute of Respiratory Health, Frontiers Science Center for Disease-related Molecular Network, Precision Medicine Key Laboratory of Sichuan Province & Precision Medicine Research Center, West China Hospital, Sichuan University, Chengdu, 610041, Sichuan, China
| | - Guangbing Zhang
- Frontier Innovation Center for Dental Medicine Plus, West China Hospital of Stomatology, Sichuan University, Chengdu, 610041, Sichuan, China
| | - Chengyong Wu
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, West China Medical School, Sichuan University, Chengdu, 610041, Sichuan, China; Department of Pulmonary and Critical Care Medicine, Targeted Tracer Research and Development Laboratory, Institute of Respiratory Health, Frontiers Science Center for Disease-related Molecular Network, Precision Medicine Key Laboratory of Sichuan Province & Precision Medicine Research Center, West China Hospital, Sichuan University, Chengdu, 610041, Sichuan, China
| | - Yi Lei
- General Practice Ward/International Medical Center Ward, General Practice Medical Center, General Practice Research Institute, West China Hospital, Sichuan University, China.
| | - Yuxi Wang
- Department of Pulmonary and Critical Care Medicine, Targeted Tracer Research and Development Laboratory, Institute of Respiratory Health, Frontiers Science Center for Disease-related Molecular Network, Precision Medicine Key Laboratory of Sichuan Province & Precision Medicine Research Center, West China Hospital, Sichuan University, Chengdu, 610041, Sichuan, China.
| | - Jinliang Yang
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, West China Medical School, Sichuan University, Chengdu, 610041, Sichuan, China; Research Unit of Gene and Immunotherapy, Chinese Academy of Medical Sciences, Chengdu, 610041, Sichuan, China.
| |
Collapse
|
3
|
Gironacci MM, Bruna-Haupt E. Unraveling the crosstalk between renin-angiotensin system receptors. Acta Physiol (Oxf) 2024; 240:e14134. [PMID: 38488216 DOI: 10.1111/apha.14134] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2024] [Revised: 02/23/2024] [Accepted: 03/05/2024] [Indexed: 04/24/2024]
Abstract
The renin-angiotensin system (RAS) plays a key role in blood pressure regulation. The RAS is a complex interconnected system composed of two axes with opposite effects. The pressor arm, represented by angiotensin (Ang) II and the AT1 receptor (AT1R), mediates the vasoconstrictor, proliferative, hypertensive, oxidative, and pro-inflammatory effects of the RAS, while the depressor/protective arm, represented by Ang-(1-7), its Mas receptor (MasR) and the AT2 receptor (AT2R), opposes the actions elicited by the pressor arm. The AT1R, AT2R, and MasR belong to the G-protein-coupled receptor (GPCR) family. GPCRs operate not only as monomers, but they can also function in dimeric (homo and hetero) or higher-order oligomeric states. Due to the interaction with other receptors, GPCR properties may change: receptor affinity, trafficking, signaling, and its biological function may be altered. Thus, heteromerization provides a newly recognized means of modulation of receptor function, as well as crosstalk between GPCRs. This review is focused on angiotensin receptors, and how their properties are influenced by crosstalk with other receptors, adding more complexity to an already complex system and potentially opening up new therapeutic approaches.
Collapse
Affiliation(s)
- Mariela M Gironacci
- Facultad de Farmacia y Bioquímica, IQUIFIB (UBA-CONICET), Universidad de Buenos Aires, Buenos Aires, Argentina
| | - Ezequiel Bruna-Haupt
- INTEQUI (CONICET), Departamento de Química, Facultad de Química, Bioquímica y Farmacia, Universidad Nacional de San Luis, San Luis, Argentina
| |
Collapse
|
4
|
Aldosterone synthase inhibitors for cardiovascular diseases: A comprehensive review of preclinical, clinical and in silico data. Pharmacol Res 2020; 163:105332. [PMID: 33271294 DOI: 10.1016/j.phrs.2020.105332] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/27/2020] [Revised: 10/27/2020] [Accepted: 11/23/2020] [Indexed: 01/23/2023]
Abstract
Aldosterone, the main mineralocorticoid hormone, plays a fundamental role in maintaining blood pressure (BP)and volume under hypovolemic conditions. However, in numerous diseases, where it is produced in excess, it plays a detrimental role and contributes to cardiovascular events and ultimately to death in a multitude of patients. The seminal observation that the fungicide-derivative fadrozole blunted steroidogenesis has led to develop several agents to inhibit aldosterone synthase (AS, CYP11B2), the mitochondrial NADH-dependent enzyme that is necessary for aldosterone biosynthesis. Aldosterone synthase inhibitors (ASI) have, thereafter, been conceived and investigated in phase I and phase II studies. We herein reviewed the ASIs available so far considering their chemical structure, the related aldosterone synthase binding and pharmacodynamic properties. We also examined the promising results obtained with ASIs that have already been tested in phase II human studies.
Collapse
|
5
|
Sinphitukkul K, Manotham K, Eiam-Ong S, Eiam-Ong S. Aldosterone nongenomically induces angiotensin II receptor dimerization in rat kidney: role of mineralocorticoid receptor and NADPH oxidase. Arch Med Sci 2019; 15:1589-1598. [PMID: 31749889 PMCID: PMC6855162 DOI: 10.5114/aoms.2019.87135] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/30/2017] [Accepted: 06/01/2017] [Indexed: 12/13/2022] Open
Abstract
INTRODUCTION Previous in vitro studies demonstrated that aldosterone nongenomically induces transglutaminase (TG) and reactive oxygen species (ROS), which enhanced angiotensin II receptor (ATR) dimerization. There are no in vivo data in the kidney. MATERIAL AND METHODS Male Wistar rats were intraperitoneally injected with normal saline solution, or aldosterone (Aldo: 150 μg/kg BW); or received pretreatment with eplerenone (mineralocorticoid receptor (MR) blocker, Ep. + Aldo), or with apocynin (nicotinamide adenine dinucleotide phosphate (NADPH) oxidase inhibitor, Apo. + Aldo) 30 min before aldosterone. Thirty minutes after aldosterone injection, protein abundances of dimeric and monomeric forms of AT1R and AT2R, and protein abundances and localizations of TG2 and p47phox, a cytosolic subunit of NADPH oxidase, were determined by Western blot analysis and immunohistochemistry, respectively. RESULTS Protein abundances of dimeric forms of AT1R and AT2R were enhanced by 170% and 70%, respectively. Apocynin could block dimeric forms of both receptors while eplerenone inhibited only AT2R. Monomeric protein levels of both receptors were maintained. Aldosterone significantly enhanced TG2 and p47phox protein abundances, which were blunted by eplerenone or apocynin. Aldosterone stimulated p47phox protein expression in both the cortex and the medulla while TG2 was induced mostly in the medulla. Eplerenone or apocynin normalized the immunoreactivity of both TG2 and p47phox. CONCLUSIONS This is the first in vivo study demonstrating that aldosterone nongenomically increases renal TG2 and p47phox protein expression and then activates AT1R and AT2R dimerizations. Aldosterone-stimulated AT1R and AT2R dimerizations are mediated through activation of NADPH oxidase. Aldosterone-induced AT1R dimer formation is an MR-independent pathway, whereas the formation of AT2R dimer is modulated in an MR-dependent manner.
Collapse
Affiliation(s)
| | - Krissanapong Manotham
- Molecular and Cell Biology Unit, Department of Medicine, Lerdsin General Hospital, Bangkok, Thailand
| | - Somchai Eiam-Ong
- Department of Medicine, Division of Nephrology, Faculty of Medicine, Chulalongkorn University, Bangkok, Thailand
| | - Somchit Eiam-Ong
- Department of Physiology, Faculty of Medicine, Chulalongkorn University, Bangkok, Thailand
| |
Collapse
|
6
|
Abstract
Aldosterone is a mineralocorticoid hormone, as its main renal effect has been considered as electrolyte and water homeostasis in the distal tubule, thus maintaining blood pressure and extracellular fluid homeostasis through the activation of mineralocorticoid receptor (MR) in epithelial cells. However, over the past decade, numerous studies have documented the significant role of aldosterone in the progression of chronic kidney disease (CKD) which has become a subject of interest. It is being studied that aldosterone can affect cardiovascular and renal system, thereby contributing to tissue inflammation, injury, glomerulosclerosis, and interstitial fibrosis. Aldosterone acts on renal vessels, renal cells (glomerular mesangial cells, podocytes, vascular smooth muscle cells, tubular epithelial cells, and interstitial fibroblasts), and infiltrating inflammatory cells, inducing reactive oxygen species (ROS) production, upregulated epithelial growth factor receptor (EGFR), and type 1 angiotensin (AT1) receptor expressions, and activating nuclear factor kappa B (NF-κB), activator protein-1 (AP-1), and EGFR to further promote cell proliferation, apoptosis, and proliferation. Phenotypic transformation of epithelial cells stimulates the expression of transforming growth factor-β (TGF-β), connective tissue growth factor (CTGF), osteopontin (OPN), and plasminogen activator inhibitor-1 (PAI-1), eventually leading to renal fibrosis. MR antagonisms are related to inhibition of aldosterone-mediated pro-inflammatory and pro-fibrotic effect. In this review, we will summarize the important role of aldosterone in the pathogenesis of renal injury and fibrosis, emphasizing on its multiple underlying mechanisms and advances in aldosterone research along with the potential therapeutics for targeting MR in a renal fibrosis.
Collapse
|
7
|
Chronic depression symptoms desensitize renin activity to protect against volume-loading hypertension in Blacks: The SABPA study. Physiol Behav 2018; 194:474-480. [DOI: 10.1016/j.physbeh.2018.06.038] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2018] [Revised: 06/26/2018] [Accepted: 06/26/2018] [Indexed: 01/24/2023]
|
8
|
Sinphitukkul K, Manotham K, Eiam-Ong S, Eiam-Ong S. Nongenomic action of aldosterone on colocalization of angiotensin II type 1 and type 2 receptors in rat kidney. J Histotechnol 2018. [DOI: 10.1080/01478885.2018.1438756] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/17/2022]
Affiliation(s)
| | | | - Somchai Eiam-Ong
- Faculty of Medicine, Department of Medicine (Division of Nephrology), Chulalongkorn University, Bangkok, Thailand
| | - Somchit Eiam-Ong
- Department of Physiology, Faculty of Medicine, Chulalongkorn University, Bangkok, Thailand
| |
Collapse
|
9
|
Hermidorff MM, de Assis LVM, Isoldi MC. Genomic and rapid effects of aldosterone: what we know and do not know thus far. Heart Fail Rev 2018; 22:65-89. [PMID: 27942913 DOI: 10.1007/s10741-016-9591-2] [Citation(s) in RCA: 44] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
Aldosterone is the most known mineralocorticoid hormone synthesized by the adrenal cortex. The genomic pathway displayed by aldosterone is attributed to the mineralocorticoid receptor (MR) signaling. Even though the rapid effects displayed by aldosterone are long known, our knowledge regarding the receptor responsible for such event is still poor. It is intense that the debate whether the MR or another receptor-the "unknown receptor"-is the receptor responsible for the rapid effects of aldosterone. Recently, G protein-coupled estrogen receptor-1 (GPER-1) was elegantly shown to mediate some aldosterone-induced rapid effects in several tissues, a fact that strongly places GPER-1 as the unknown receptor. It has also been suggested that angiotensin receptor type 1 (AT1) also participates in the aldosterone-induced rapid effects. Despite this open question, the relevance of the beneficial effects of aldosterone is clear in the kidneys, colon, and CNS as aldosterone controls the important water reabsorption process; on the other hand, detrimental effects displayed by aldosterone have been reported in the cardiovascular system and in the kidneys. In this line, the MR antagonists are well-known drugs that display beneficial effects in patients with heart failure and hypertension; it has been proposed that MR antagonists could also play an important role in vascular disease, obesity, obesity-related hypertension, and metabolic syndrome. Taken altogether, our goal here was to (1) bring a historical perspective of both genomic and rapid effects of aldosterone in several tissues, and the receptors and signaling pathways involved in such processes; and (2) critically address the controversial points within the literature as regarding which receptor participates in the rapid pathway display by aldosterone.
Collapse
Affiliation(s)
- Milla Marques Hermidorff
- Laboratory of Hypertension, Research Center in Biological Science, Institute of Exact and Biological Sciences, Federal University of Ouro Preto, Campus Morro do Cruzeiro, Ouro Preto, MG, 35400-000, Brazil
| | - Leonardo Vinícius Monteiro de Assis
- Laboratory of Comparative Physiology of Pigmentation, Department of Physiology, Institute of Biosciences, University of São Paulo, São Paulo, Brazil
| | - Mauro César Isoldi
- Laboratory of Hypertension, Research Center in Biological Science, Institute of Exact and Biological Sciences, Federal University of Ouro Preto, Campus Morro do Cruzeiro, Ouro Preto, MG, 35400-000, Brazil.
| |
Collapse
|
10
|
Ruhs S, Nolze A, Hübschmann R, Grossmann C. 30 YEARS OF THE MINERALOCORTICOID RECEPTOR: Nongenomic effects via the mineralocorticoid receptor. J Endocrinol 2017; 234:T107-T124. [PMID: 28348113 DOI: 10.1530/joe-16-0659] [Citation(s) in RCA: 51] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/21/2017] [Accepted: 03/27/2017] [Indexed: 12/12/2022]
Abstract
The mineralocorticoid receptor (MR) belongs to the steroid hormone receptor family and classically functions as a ligand-dependent transcription factor. It is involved in water-electrolyte homeostasis and blood pressure regulation but independent from these effects also furthers inflammation, fibrosis, hypertrophy and remodeling in cardiovascular tissues. Next to genomic effects, aldosterone elicits very rapid actions within minutes that do not require transcription or translation and that occur not only in classical MR epithelial target organs like kidney and colon but also in nonepithelial tissues like heart, vasculature and adipose tissue. Most of these effects can be mediated by classical MR and its crosstalk with different signaling cascades. Near the plasma membrane, the MR seems to be associated with caveolin and striatin as well as with receptor tyrosine kinases like EGFR, PDGFR and IGF1R and G protein-coupled receptors like AT1 and GPER1, which then mediate nongenomic aldosterone effects. GPER1 has also been named a putative novel MR. There is a close interaction and functional synergism between the genomic and the nongenomic signaling so that nongenomic signaling can lead to long-term effects and support genomic actions. Therefore, understanding nongenomic aldosterone/MR effects is of potential relevance for modulating genomic aldosterone effects and may provide additional targets for intervention.
Collapse
Affiliation(s)
- Stefanie Ruhs
- Julius Bernstein Institute of PhysiologyMartin Luther University Halle-Wittenberg, Halle, Germany
| | - Alexander Nolze
- Julius Bernstein Institute of PhysiologyMartin Luther University Halle-Wittenberg, Halle, Germany
| | - Ralf Hübschmann
- Julius Bernstein Institute of PhysiologyMartin Luther University Halle-Wittenberg, Halle, Germany
| | - Claudia Grossmann
- Julius Bernstein Institute of PhysiologyMartin Luther University Halle-Wittenberg, Halle, Germany
| |
Collapse
|
11
|
Spasov AA, Yakovlev DS, Brigadirova AA. Angiotensin AT1 Receptors and Their Ligands (Review). Pharm Chem J 2017. [DOI: 10.1007/s11094-017-1546-y] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
|
12
|
Ong GSY, Young MJ. Mineralocorticoid regulation of cell function: the role of rapid signalling and gene transcription pathways. J Mol Endocrinol 2017; 58:R33-R57. [PMID: 27821439 DOI: 10.1530/jme-15-0318] [Citation(s) in RCA: 52] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/17/2016] [Accepted: 11/06/2016] [Indexed: 12/22/2022]
Abstract
The mineralocorticoid receptor (MR) and mineralocorticoids regulate epithelial handling of electrolytes, and induces diverse effects on other tissues. Traditionally, the effects of MR were ascribed to ligand-receptor binding and activation of gene transcription. However, the MR also utilises a number of intracellular signalling cascades, often by transactivating unrelated receptors, to change cell function more rapidly. Although aldosterone is the physiological mineralocorticoid, it is not the sole ligand for MR. Tissue-selective and mineralocorticoid-specific effects are conferred through the enzyme 11β-hydroxysteroid dehydrogenase 2, cellular redox status and properties of the MR itself. Furthermore, not all aldosterone effects are mediated via MR, with implication of the involvement of other membrane-bound receptors such as GPER. This review will describe the ligands, receptors and intracellular mechanisms available for mineralocorticoid hormone and receptor signalling and illustrate their complex interactions in physiology and disease.
Collapse
Affiliation(s)
- Gregory S Y Ong
- Cardiovascular Endocrinology LaboratoryCentre for Endocrinology and Metabolism, Hudson Institute of Medical Research, Clayton, Victoria, Australia
- Department of MedicineSchool of Clinical Sciences, Monash University, Clayton, Victoria, Australia
| | - Morag J Young
- Cardiovascular Endocrinology LaboratoryCentre for Endocrinology and Metabolism, Hudson Institute of Medical Research, Clayton, Victoria, Australia
- Department of PhysiologySchool of Biomedical Sciences, Monash University, Clayton, Victoria, Australia
| |
Collapse
|
13
|
Engholm M, Pinilla E, Mogensen S, Matchkov V, Hedegaard ER, Chen H, Mulvany MJ, Simonsen U. Involvement of transglutaminase 2 and voltage-gated potassium channels in cystamine vasodilatation in rat mesenteric small arteries. Br J Pharmacol 2016; 173:839-55. [PMID: 26603619 DOI: 10.1111/bph.13393] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2015] [Revised: 10/13/2015] [Accepted: 11/10/2015] [Indexed: 01/02/2023] Open
Abstract
BACKGROUND AND PURPOSE Vasodilatation may contribute to the neuroprotective and vascular anti-remodelling effect of the tissue transglutaminase 2 (TG2) inhibitor cystamine. Here, we hypothesized that inhibition of TG2 followed by blockade of smooth muscle calcium entry and/or inhibition of Rho kinase underlies cystamine vasodilatation. EXPERIMENTAL APPROACH We used rat mesenteric small arteries and RT-PCR, immunoblotting, and measurements of isometric wall tension, intracellular Ca(2+) ([Ca(2+)]i ), K(+) currents (patch clamp), and phosphorylation of myosin phosphatase targeting subunit 1 (MYPT1) and myosin regulatory light chain, in our experiments. KEY RESULTS RT-PCR and immunoblotting revealed expression of TG2 in mesenteric small arteries. Cystamine concentration-dependently inhibited responses to phenylephrine, 5-HT and U46619 and for extracellular potassium. Selective inhibitors of TG2, LDN 27129 and T101, also inhibited phenylephrine contraction. An inhibitor of PLC suppressed cystamine relaxation. Cystamine relaxed and reduced [Ca(2+)]i in phenylephrine-contracted arteries. In potassium-contracted arteries, cystamine induced less relaxation without changing [Ca(2+)]i , and these relaxations were blocked by mitochondrial complex inhibitors. Blockers of Kv 7 channels, XE991 and linopirdine, inhibited cystamine relaxation and increases in voltage-dependent smooth muscle currents. Cystamine and the Rho kinase inhibitor Y27632 reduced basal MYPT1-Thr(855) phosphorylation, but only Y27632 reduced phenylephrine-induced increases in MYPT1-Thr(855) and myosin regulatory light chain phosphorylation. CONCLUSIONS AND IMPLICATIONS Cystamine induced vasodilatation by inhibition of receptor-coupled TG2, leading to opening of Kv channels and reduction of intracellular calcium, and by activation of a pathway sensitive to inhibitors of the mitochondrial complexes I and III. Both pathways may contribute to the antihypertensive and neuroprotective effect of cystamine.
Collapse
Affiliation(s)
- Morten Engholm
- Department of Biomedicine, Pulmonary and Cardiovascular Pharmacology, Aarhus University, Denmark
| | - Estéfano Pinilla
- Department of Biomedicine, Pulmonary and Cardiovascular Pharmacology, Aarhus University, Denmark
| | - Susie Mogensen
- Department of Biomedicine, Pulmonary and Cardiovascular Pharmacology, Aarhus University, Denmark
| | - Vladimir Matchkov
- Department of Biomedicine, Pulmonary and Cardiovascular Pharmacology, Aarhus University, Denmark
| | - Elise Røge Hedegaard
- Department of Biomedicine, Pulmonary and Cardiovascular Pharmacology, Aarhus University, Denmark
| | - Hua Chen
- Department of Biomedicine, Pulmonary and Cardiovascular Pharmacology, Aarhus University, Denmark
| | - Michael J Mulvany
- Department of Biomedicine, Pulmonary and Cardiovascular Pharmacology, Aarhus University, Denmark
| | - Ulf Simonsen
- Department of Biomedicine, Pulmonary and Cardiovascular Pharmacology, Aarhus University, Denmark
| |
Collapse
|
14
|
Karnik SS, Unal H, Kemp JR, Tirupula KC, Eguchi S, Vanderheyden PML, Thomas WG. International Union of Basic and Clinical Pharmacology. XCIX. Angiotensin Receptors: Interpreters of Pathophysiological Angiotensinergic Stimuli [corrected]. Pharmacol Rev 2015; 67:754-819. [PMID: 26315714 PMCID: PMC4630565 DOI: 10.1124/pr.114.010454] [Citation(s) in RCA: 225] [Impact Index Per Article: 22.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
The renin angiotensin system (RAS) produced hormone peptides regulate many vital body functions. Dysfunctional signaling by receptors for RAS peptides leads to pathologic states. Nearly half of humanity today would likely benefit from modern drugs targeting these receptors. The receptors for RAS peptides consist of three G-protein-coupled receptors—the angiotensin II type 1 receptor (AT1 receptor), the angiotensin II type 2 receptor (AT2 receptor), the MAS receptor—and a type II trans-membrane zinc protein—the candidate angiotensin IV receptor (AngIV binding site). The prorenin receptor is a relatively new contender for consideration, but is not included here because the role of prorenin receptor as an independent endocrine mediator is presently unclear. The full spectrum of biologic characteristics of these receptors is still evolving, but there is evidence establishing unique roles of each receptor in cardiovascular, hemodynamic, neurologic, renal, and endothelial functions, as well as in cell proliferation, survival, matrix-cell interaction, and inflammation. Therapeutic agents targeted to these receptors are either in active use in clinical intervention of major common diseases or under evaluation for repurposing in many other disorders. Broad-spectrum influence these receptors produce in complex pathophysiological context in our body highlights their role as precise interpreters of distinctive angiotensinergic peptide cues. This review article summarizes findings published in the last 15 years on the structure, pharmacology, signaling, physiology, and disease states related to angiotensin receptors. We also discuss the challenges the pharmacologist presently faces in formally accepting newer members as established angiotensin receptors and emphasize necessary future developments.
Collapse
Affiliation(s)
- Sadashiva S Karnik
- Department of Molecular Cardiology, Lerner Research Institute of Cleveland Clinic, Cleveland, Ohio (S.S.K., H.U., J.R.K., K.C.T.); Cardiovascular Research Center, Temple University School of Medicine, Philadelphia, Pennsylvania (S.E.); Faculty of Sciences and Bioengineering Sciences, Vrije Universiteit Brussel, Brussels, Belgium (P.M.L.V.); and Department of General Physiology, School of Biomedical Sciences, The University of Queensland, Brisbane, Queensland, Australia (W.G.T.)
| | - Hamiyet Unal
- Department of Molecular Cardiology, Lerner Research Institute of Cleveland Clinic, Cleveland, Ohio (S.S.K., H.U., J.R.K., K.C.T.); Cardiovascular Research Center, Temple University School of Medicine, Philadelphia, Pennsylvania (S.E.); Faculty of Sciences and Bioengineering Sciences, Vrije Universiteit Brussel, Brussels, Belgium (P.M.L.V.); and Department of General Physiology, School of Biomedical Sciences, The University of Queensland, Brisbane, Queensland, Australia (W.G.T.)
| | - Jacqueline R Kemp
- Department of Molecular Cardiology, Lerner Research Institute of Cleveland Clinic, Cleveland, Ohio (S.S.K., H.U., J.R.K., K.C.T.); Cardiovascular Research Center, Temple University School of Medicine, Philadelphia, Pennsylvania (S.E.); Faculty of Sciences and Bioengineering Sciences, Vrije Universiteit Brussel, Brussels, Belgium (P.M.L.V.); and Department of General Physiology, School of Biomedical Sciences, The University of Queensland, Brisbane, Queensland, Australia (W.G.T.)
| | - Kalyan C Tirupula
- Department of Molecular Cardiology, Lerner Research Institute of Cleveland Clinic, Cleveland, Ohio (S.S.K., H.U., J.R.K., K.C.T.); Cardiovascular Research Center, Temple University School of Medicine, Philadelphia, Pennsylvania (S.E.); Faculty of Sciences and Bioengineering Sciences, Vrije Universiteit Brussel, Brussels, Belgium (P.M.L.V.); and Department of General Physiology, School of Biomedical Sciences, The University of Queensland, Brisbane, Queensland, Australia (W.G.T.)
| | - Satoru Eguchi
- Department of Molecular Cardiology, Lerner Research Institute of Cleveland Clinic, Cleveland, Ohio (S.S.K., H.U., J.R.K., K.C.T.); Cardiovascular Research Center, Temple University School of Medicine, Philadelphia, Pennsylvania (S.E.); Faculty of Sciences and Bioengineering Sciences, Vrije Universiteit Brussel, Brussels, Belgium (P.M.L.V.); and Department of General Physiology, School of Biomedical Sciences, The University of Queensland, Brisbane, Queensland, Australia (W.G.T.)
| | - Patrick M L Vanderheyden
- Department of Molecular Cardiology, Lerner Research Institute of Cleveland Clinic, Cleveland, Ohio (S.S.K., H.U., J.R.K., K.C.T.); Cardiovascular Research Center, Temple University School of Medicine, Philadelphia, Pennsylvania (S.E.); Faculty of Sciences and Bioengineering Sciences, Vrije Universiteit Brussel, Brussels, Belgium (P.M.L.V.); and Department of General Physiology, School of Biomedical Sciences, The University of Queensland, Brisbane, Queensland, Australia (W.G.T.)
| | - Walter G Thomas
- Department of Molecular Cardiology, Lerner Research Institute of Cleveland Clinic, Cleveland, Ohio (S.S.K., H.U., J.R.K., K.C.T.); Cardiovascular Research Center, Temple University School of Medicine, Philadelphia, Pennsylvania (S.E.); Faculty of Sciences and Bioengineering Sciences, Vrije Universiteit Brussel, Brussels, Belgium (P.M.L.V.); and Department of General Physiology, School of Biomedical Sciences, The University of Queensland, Brisbane, Queensland, Australia (W.G.T.)
| |
Collapse
|
15
|
Zhu S, Zhang M, Davis JE, Wu WH, Surrao K, Wang H, Wu G. A single mutation in helix 8 enhances the angiotensin II type 1a receptor transport and signaling. Cell Signal 2015; 27:2371-9. [PMID: 26342563 DOI: 10.1016/j.cellsig.2015.08.020] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2015] [Accepted: 08/30/2015] [Indexed: 01/01/2023]
Abstract
The amphipathic helix 8 in the membrane-proximal C-terminus is a structurally conserved feature of class A seven transmembrane-spanning G protein-coupled receptors (GPCRs). Mutations of this helical motif often cause receptor misfolding, defective cell surface transport and dysfunction. Surprisingly, we demonstrated here that a single point mutation at Lys308 in helix 8 markedly enhanced the steady-state surface density of the angiotensin II type 1a receptor (AT1aR). Consistent with the enhanced cell surface expression, Lys308 mutation significantly augmented AT1aR-mediated mitogen-activated protein kinase ERK1/2 activation, inositol phosphate production, and vascular smooth muscle cell migration. This mutation also increased the overall expression of AT1aR without altering receptor degradation. More interestingly, Lys308 mutation abolished AT1aR interaction with β-COP, a component of COPI transport vesicles, and impaired AT1aR responsiveness to the inhibition of Rab6 GTPase involved in the Golgi-to-ER retrograde pathway. Furthermore, these functions of Lys308 were largely dependent on its positively charged property. These data reveal previously unappreciated functions of helix 8 and novel mechanisms governing the cell surface transport and function of AT1aR.
Collapse
Affiliation(s)
- Shu Zhu
- Department of Pharmacology and Toxicology, Medical College of Georgia, Georgia Regents University, 1459 Laney Walker Blvd., Augusta GA 30912, United States
| | - Maoxiang Zhang
- Department of Pharmacology and Toxicology, Medical College of Georgia, Georgia Regents University, 1459 Laney Walker Blvd., Augusta GA 30912, United States
| | - Jason E Davis
- Department of Pharmacology and Toxicology, Medical College of Georgia, Georgia Regents University, 1459 Laney Walker Blvd., Augusta GA 30912, United States
| | - William H Wu
- Department of Pharmacology and Toxicology, Medical College of Georgia, Georgia Regents University, 1459 Laney Walker Blvd., Augusta GA 30912, United States
| | - Kristen Surrao
- Department of Pharmacology and Toxicology, Medical College of Georgia, Georgia Regents University, 1459 Laney Walker Blvd., Augusta GA 30912, United States
| | - Hong Wang
- School of Life Sciences and Technology, Tongji University, 1239 Siping Road, Shanghai, China
| | - Guangyu Wu
- Department of Pharmacology and Toxicology, Medical College of Georgia, Georgia Regents University, 1459 Laney Walker Blvd., Augusta GA 30912, United States.
| |
Collapse
|
16
|
Pathophysiology and treatment of resistant hypertension: the role of aldosterone and amiloride-sensitive sodium channels. Semin Nephrol 2015; 34:532-9. [PMID: 25416662 DOI: 10.1016/j.semnephrol.2014.08.007] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Abstract
Resistant hypertension is a clinically distinct subgroup of hypertension defined by the failure to achieve blood pressure control on optimal dosing of at least 3 antihypertensive medications of different classes, including a diuretic. The pathophysiology of hypertension can be attributed to aldosterone excess in more than 20% of patients with resistant hypertension. Existing dogma attributes the increase in blood pressure seen with increases in aldosterone to its antinatriuretic effects in the distal nephron. However, emerging research, which has identified and has begun to define the function of amiloride-sensitive sodium channels and mineralocorticoid receptors in the systemic vasculature, challenges impaired natriuresis as the sole cause of aldosterone-mediated resistant hypertension. This review integrates these findings to better define the role of the vasculature and aldosterone in the pathophysiology of resistant hypertension. In addition, a brief guide to the treatment of resistant hypertension is presented.
Collapse
|
17
|
Maiolino G, Azzolini M, Rossi GP. Mineralocorticoid Receptor Antagonists Therapy in Resistant Hypertension: Time to Implement Guidelines! Front Cardiovasc Med 2015; 2:3. [PMID: 26664875 PMCID: PMC4668865 DOI: 10.3389/fcvm.2015.00003] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2014] [Accepted: 01/12/2015] [Indexed: 12/24/2022] Open
Abstract
Despite the availability of anti-hypertensive medications with increasing efficacy up to 50% of hypertensive patients have blood pressure levels (BP) not at the goals set by international societies. Some of these patients are either not optimally treated or are non-adherent to the prescribed drugs. However, a proportion, despite adequate treatment, have resistant hypertension (RH), which represents an important problem in that it is associated to an excess risk of cardiovascular events. Notwithstanding a complex pathogenesis, an abundance of data suggests a key contribution for the mineralocorticoid receptor (MR) in RH, thus fostering a potential role for its antagonists in RH. Based on these premises randomized clinical trials aimed at testing the efficacy of MR antagonists (MRAs) in RH patients have been completed. Overall, they demonstrated the efficacy of MRAs in reducing BP and surrogate markers of target organ damage, such as microalbuminuria, either compared to placebo or to other drugs. In summary, owing to the key role of the MR in the pathogenesis of RH and on the proven efficacy of MRAs we advocate their inclusion as an essential component of therapy in patients with presumed RH. Conversely, we propose that RH should be diagnosed only in patients whose BP values show to be resistant to an up-titrated dose of these drugs.
Collapse
Affiliation(s)
- Giuseppe Maiolino
- Department of Medicine (DIMED), Internal Medicine 4, University of Padova , Padova , Italy
| | - Matteo Azzolini
- Department of Medicine (DIMED), Internal Medicine 4, University of Padova , Padova , Italy
| | - Gian Paolo Rossi
- Department of Medicine (DIMED), Internal Medicine 4, University of Padova , Padova , Italy
| |
Collapse
|
18
|
Ferrario CM, Schiffrin EL. Role of mineralocorticoid receptor antagonists in cardiovascular disease. Circ Res 2015; 116:206-13. [PMID: 25552697 PMCID: PMC4283558 DOI: 10.1161/circresaha.116.302706] [Citation(s) in RCA: 67] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/31/2014] [Accepted: 10/08/2014] [Indexed: 12/23/2022]
Abstract
Aldosterone exerts its best known sodium homeostasis actions by controlling sodium excretion at the level of the distal tubules via activation of the apical epithelial sodium channel and the basolateral Na(+)/K(+)ATPase pump. Recently, this mineralocorticoid hormone has been demonstrated to act on the heart and blood vessels. Excess release of aldosterone in relation to the salt status induces both genomic and nongenomic effects that by promoting endothelial dysfunction, and vascular and cardiorenal adverse remodeling, contribute to the target organ damage found in hypertension, heart failure, myocardial infarction, and chronic renal failure. Mineralocorticoid receptor blockers have been shown to be highly effective in resistant hypertension and to slow down heart failure progression, and in experimental animals, the development of atherosclerosis. Blockade of the action of aldosterone and potentially other mineralocorticoid steroids has been increasingly demonstrated to be an extremely beneficial therapy in different forms of cardiovascular disease. This review provides a summary of the knowledge that exists on aldosterone actions in the cardiovascular system and, in providing the translational impact of this knowledge to the clinical arena, illustrates how much more needs to be achieved in exploring the use of mineralocorticoid receptor blockers in less advanced stages of heart, renal, and vascular disease.
Collapse
Affiliation(s)
- Carlos M Ferrario
- From the Hypertension Translational Research Laboratory, Departments of Surgery, Internal Medicine-Nephrology, and Physiology-Pharmacology, Wake Forest University Health Science Center, Winston Salem, NC (C.M.F.); and Department of Medicine and Lady Davis Institute for Medical Research, SMBD-Jewish General Hospital, McGill University, Montreal, PQ, Canada (E.L.S.).
| | - Ernesto L Schiffrin
- From the Hypertension Translational Research Laboratory, Departments of Surgery, Internal Medicine-Nephrology, and Physiology-Pharmacology, Wake Forest University Health Science Center, Winston Salem, NC (C.M.F.); and Department of Medicine and Lady Davis Institute for Medical Research, SMBD-Jewish General Hospital, McGill University, Montreal, PQ, Canada (E.L.S.)
| |
Collapse
|
19
|
Meinel S, Gekle M, Grossmann C. Mineralocorticoid receptor signaling: crosstalk with membrane receptors and other modulators. Steroids 2014; 91:3-10. [PMID: 24928729 DOI: 10.1016/j.steroids.2014.05.017] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/11/2013] [Revised: 05/16/2014] [Accepted: 05/28/2014] [Indexed: 12/30/2022]
Abstract
The mineralocorticoid receptor (MR) belongs to the steroid receptor superfamily. Classically, it acts as a ligand-bound transcription factor in epithelial tissues, where it regulates water and electrolyte homeostasis and controls blood pressure. Additionally, the MR has been shown to elicit pathophysiological effects including inflammation, fibrosis and remodeling processes in the cardiovascular system and the kidneys and MR antagonists have proven beneficial for patients with certain cardiovascular and renal disease. The underlying molecular mechanisms that mediate MR effects have not been fully elucidated but very likely rely on interactions with other signaling pathways in addition to genomic actions at hormone response elements. In this review we will focus on interactions of MR signaling with different membrane receptors, namely receptor tyrosine kinases and the angiotensin II receptor because of their potential relevance for disease. In addition, GPR30 is discussed as a new aldosterone receptor. To gain insights into the problem why the MR only seems to mediate pathophysiological effects in the presence of additional permissive factors we will also briefly discuss factors that lead to modulation of MR activity as well. Overall, MR signaling is part of an intricate network that still needs to be investigated further.
Collapse
Affiliation(s)
- S Meinel
- Julius Bernstein Institute of Physiology, Martin Luther University Halle-Wittenberg, Germany
| | - M Gekle
- Julius Bernstein Institute of Physiology, Martin Luther University Halle-Wittenberg, Germany
| | - C Grossmann
- Julius Bernstein Institute of Physiology, Martin Luther University Halle-Wittenberg, Germany.
| |
Collapse
|
20
|
Curione M, Petramala L, Savoriti C, Verrenti M, Baiocco E, Salvatore S, Zinnamosca L, Iannucci G, Sciomer S, Letizia C. Electrical and Myocardial Remodeling in Primary Aldosteronism. Front Cardiovasc Med 2014; 1:7. [PMID: 26664858 PMCID: PMC4668839 DOI: 10.3389/fcvm.2014.00007] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2014] [Accepted: 09/28/2014] [Indexed: 11/13/2022] Open
Abstract
OBJECTIVE AND DESIGN Primary aldosteronism (PA) represents the most common cause of secondary hypertension. A higher risk of cardiovascular events has been reported in patients with PA than in otherwise similar patients with essential hypertension (EH). So far, only a few studies investigated the electrocardiographic changes in PA patients compared to EH patients. METHODS To investigate the electrocardiographic changes and heart remodeling in PA, we enrolled 61 consecutive patients, 30 with PA [12 with aldosterone-producing adrenal cortical adenoma (APA) and 18 with bilateral adrenal hyperplasia-idiopathic adrenal hyperplasia] and 30 with EH. In all subjects, electrocardiographic parameters were evaluated from 12-lead electrocardiograms and heart remodeling with echocardiogram. RESULTS No significant differences in age, sex, body mass index, and blood pressure were found in two groups. The P wave and PR interval duration were significantly prolonged in patients with PA respect to EH (p < 0.003 and <0.002, respectively). A first degree atrio-ventricular block was present in 16% of the patients with PA and only in 3.2% of those with EH. In PA patients, the interventricular septum thickness (IVST) correlated with PR duration (r = 0.51; p < 0.03). Left ventricular hypertrophy was present in 53% of the patients with PA and in 26% of the patients with EH (χ(2), p < 0.03). CONCLUSION In this case-control study, patients with PA show more anatomic and electrical heart remodeling than those with EH. We hypothesize that in patients with PA these cardiac changes may play a role for the increased risk of future cardiovascular events.
Collapse
Affiliation(s)
- Mario Curione
- Department of Internal Medicine and Medical Specialties, Sapienza University of Rome , Rome , Italy
| | - Luigi Petramala
- Specialized Center of Secondary Hypertension, Sapienza University of Rome , Rome , Italy
| | - Claudio Savoriti
- Department of Internal Medicine and Medical Specialties, Sapienza University of Rome , Rome , Italy
| | - Marisa Verrenti
- Department of Internal Medicine and Medical Specialties, Sapienza University of Rome , Rome , Italy
| | - Erika Baiocco
- Department of Internal Medicine and Medical Specialties, Sapienza University of Rome , Rome , Italy
| | - Stephanie Salvatore
- Department of Internal Medicine and Medical Specialties, Sapienza University of Rome , Rome , Italy
| | - Laura Zinnamosca
- Specialized Center of Secondary Hypertension, Sapienza University of Rome , Rome , Italy
| | - Gino Iannucci
- Specialized Center of Secondary Hypertension, Sapienza University of Rome , Rome , Italy
| | - Susanna Sciomer
- Specialized Center of Secondary Hypertension, Sapienza University of Rome , Rome , Italy
| | - Claudio Letizia
- Specialized Center of Secondary Hypertension, Sapienza University of Rome , Rome , Italy
| |
Collapse
|
21
|
Rossi GP, Cesari M, Letizia C, Seccia TM, Cicala MV, Zinnamosca L, Kuppusamy M, Mareso S, Sciomer S, Iacobone M, Mantero F, Pessina AC. KCNJ5 gene somatic mutations affect cardiac remodelling but do not preclude cure of high blood pressure and regression of left ventricular hypertrophy in primary aldosteronism. J Hypertens 2014; 32:1514-21; discussion 1522. [DOI: 10.1097/hjh.0000000000000186] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
|
22
|
Effects of hypertension and exercise on cardiac proteome remodelling. BIOMED RESEARCH INTERNATIONAL 2014; 2014:634132. [PMID: 24877123 PMCID: PMC4022191 DOI: 10.1155/2014/634132] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/19/2013] [Accepted: 02/14/2014] [Indexed: 12/29/2022]
Abstract
Left ventricle hypertrophy is a common outcome of pressure overload stimulus closely associated with hypertension. This process is triggered by adverse molecular signalling, gene expression, and proteome alteration. Proteomic research has revealed that several molecular targets are associated with pathologic cardiac hypertrophy, including angiotensin II, endothelin-1 and isoproterenol. Several metabolic, contractile, and stress-related proteins are shown to be altered in cardiac hypertrophy derived by hypertension. On the other hand, exercise is a nonpharmacologic agent used for hypertension treatment, where cardiac hypertrophy induced by exercise training is characterized by improvement in cardiac function and resistance against ischemic insult. Despite the scarcity of proteomic research performed with exercise, healthy and pathologic heart proteomes are shown to be modulated in a completely different way. Hence, the altered proteome induced by exercise is mostly associated with cardioprotective aspects such as contractile and metabolic improvement and physiologic cardiac hypertrophy. The present review, therefore, describes relevant studies involving the molecular characteristics and alterations from hypertensive-induced and exercise-induced hypertrophy, as well as the main proteomic research performed in this field. Furthermore, proteomic research into the effect of hypertension on other target-demerged organs is examined.
Collapse
|
23
|
Briet M. Mineralocorticoid receptor, the main player in aldosterone-induced large artery stiffness. Hypertension 2013; 63:442-3. [PMID: 24296283 DOI: 10.1161/hypertensionaha.113.02581] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Affiliation(s)
- Marie Briet
- Department of Pharmacology, Centre Hospitalier et Universitaire d'Angers, 4 Rue Larrey, 49100 Angers, France.
| |
Collapse
|
24
|
Tsai CF, Yang SF, Chu HJ, Ueng KC. Cross-talk between mineralocorticoid receptor/angiotensin II type 1 receptor and mitogen-activated protein kinase pathways underlies aldosterone-induced atrial fibrotic responses in HL-1 cardiomyocytes. Int J Cardiol 2013; 169:17-28. [PMID: 24120080 DOI: 10.1016/j.ijcard.2013.06.046] [Citation(s) in RCA: 41] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/15/2012] [Revised: 05/08/2013] [Accepted: 06/28/2013] [Indexed: 10/26/2022]
Abstract
BACKGROUND Aldosterone is increasingly recognized for its involvement in atrial structural remodeling. However, the precise molecular mechanisms and signal pathways underlying aldosterone-induced atrial fibrosis are unknown. METHODS Western blotting was used to investigate the effects of aldosterone on the expression of mineralocorticoid receptor (MR), angiotensin II type I receptor (AT1), mitogen-activated protein kinases (MAPKs), and fibrotic marker proteins in cultured HL-1 cardiomyocytes. RESULTS Aldosterone upregulated MR and AT1 expressions in a concentration-dependent and time-dependent manner. Aldosterone (10(-6)M) significantly and time-dependently increased activation of the extracellular signal-regulated protein kinase (ERK), c-Jun N-terminal kinase (JNK), p38MAPK pathways, and the protein expression of collagen 1A and 3A (COL1A and COL3A), transforming growth factor (TGF)-β1, and α-smooth muscle actin (SMA). Pre-treatment with eplerenone (10(-10)M) prevented the increased expression of MR, MAPK signals and the above profibrotic molecules, but amplified the increase in AT1 level stimulated by aldosterone (10(-6)M). Pre-treatment with losartan (10(-10)M) or MAPK pathway inhibitors (U0126 or SP600125) abolished aldosterone-induced MR upregulation and significantly inhibited the expression of the above fibrotic marker proteins, indicating the critical role of MR and the requirement for active AT1 in the development of aldosterone-induced atrial fibrosis. CONCLUSIONS Elevated MR activity plays a central role in aldosterone-mediated activation of the MAPK signaling pathway and subsequent profibrotic effects in HL-1 atrial cells. MR/AT1 and the MAPK signaling pathway interact to trigger the molecular mechanism underlying the aldosterone-induced atrial fibrotic response. Our results support the view that MR blockade in conjunction with AT1 blockade can prevent the occurrence of atrial fibrillation.
Collapse
Affiliation(s)
- Chin-Feng Tsai
- Division of Cardiology, Department of Internal Medicine, Chung Shan Medical University Hospital, School of Medicine, Chung Shan Medical University, Taichung, Taiwan
| | | | | | | |
Collapse
|
25
|
Michel MC, Foster C, Brunner HR, Liu L. A systematic comparison of the properties of clinically used angiotensin II type 1 receptor antagonists. Pharmacol Rev 2013; 65:809-48. [PMID: 23487168 DOI: 10.1124/pr.112.007278] [Citation(s) in RCA: 218] [Impact Index Per Article: 18.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023] Open
Abstract
Angiotensin II type 1 receptor antagonists (ARBs) have become an important drug class in the treatment of hypertension and heart failure and the protection from diabetic nephropathy. Eight ARBs are clinically available [azilsartan, candesartan, eprosartan, irbesartan, losartan, olmesartan, telmisartan, valsartan]. Azilsartan (in some countries), candesartan, and olmesartan are orally administered as prodrugs, whereas the blocking action of some is mediated through active metabolites. On the basis of their chemical structures, ARBs use different binding pockets in the receptor, which are associated with differences in dissociation times and, in most cases, apparently insurmountable antagonism. The physicochemical differences between ARBs also manifest in different tissue penetration, including passage through the blood-brain barrier. Differences in binding mode and tissue penetration are also associated with differences in pharmacokinetic profile, particularly duration of action. Although generally highly specific for angiotensin II type 1 receptors, some ARBs, particularly telmisartan, are partial agonists at peroxisome proliferator-activated receptor-γ. All of these properties are comprehensively reviewed in this article. Although there is general consensus that a continuous receptor blockade over a 24-hour period is desirable, the clinical relevance of other pharmacological differences between individual ARBs remains to be assessed.
Collapse
Affiliation(s)
- Martin C Michel
- Department of Clinical Development & Medical Affairs, Boehringer Ingelheim, 55216 Ingelheim, Germany.
| | | | | | | |
Collapse
|
26
|
Schiffrin EL. Vascular mineralocorticoid receptors regulate blood pressure effects on myogenic tone and role in aging. Circ Res 2013; 112:415-7. [PMID: 23371898 DOI: 10.1161/circresaha.113.300883] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
Affiliation(s)
- Ernesto L Schiffrin
- Department of Medicine, SMBD-Jewish General Hospital, #B-127, 3755 Côte-Ste-Catherine Rd, Montreal, Quebec, Canada H3T 1E2.
| |
Collapse
|
27
|
Rossitto G, Regolisti G, Rossi E, Negro A, Nicoli D, Casali B, Toniato A, Caroccia B, Seccia TM, Walther T, Rossi GP. Elevation of Angiotensin-II Type-1-Receptor Autoantibodies Titer in Primary Aldosteronism as a Result of Aldosterone-Producing Adenoma. Hypertension 2013; 61:526-33. [DOI: 10.1161/hypertensionaha.112.202945] [Citation(s) in RCA: 49] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023]
Affiliation(s)
- Giacomo Rossitto
- From the Department of Medicine—DIMED, Internal Medicine 4, Padua University—School of Medicine, Padova, Italy (G.Ro., B.C., T.M.S., G.P.R.); Department of Internal Medicine, Santa Maria Nuova Hospital, Reggio Emilia, Italy (G.Re., E.R., A.N., D.N., B.C.); Surgical Pathology Unit, Department of Medical and Surgical Sciences, Padua University, Padua, Italy (A.T.); Hull York Medical School, Hull University, Cottingham, UK (T.W.)
| | - Giuseppe Regolisti
- From the Department of Medicine—DIMED, Internal Medicine 4, Padua University—School of Medicine, Padova, Italy (G.Ro., B.C., T.M.S., G.P.R.); Department of Internal Medicine, Santa Maria Nuova Hospital, Reggio Emilia, Italy (G.Re., E.R., A.N., D.N., B.C.); Surgical Pathology Unit, Department of Medical and Surgical Sciences, Padua University, Padua, Italy (A.T.); Hull York Medical School, Hull University, Cottingham, UK (T.W.)
| | - Ermanno Rossi
- From the Department of Medicine—DIMED, Internal Medicine 4, Padua University—School of Medicine, Padova, Italy (G.Ro., B.C., T.M.S., G.P.R.); Department of Internal Medicine, Santa Maria Nuova Hospital, Reggio Emilia, Italy (G.Re., E.R., A.N., D.N., B.C.); Surgical Pathology Unit, Department of Medical and Surgical Sciences, Padua University, Padua, Italy (A.T.); Hull York Medical School, Hull University, Cottingham, UK (T.W.)
| | - Aurelio Negro
- From the Department of Medicine—DIMED, Internal Medicine 4, Padua University—School of Medicine, Padova, Italy (G.Ro., B.C., T.M.S., G.P.R.); Department of Internal Medicine, Santa Maria Nuova Hospital, Reggio Emilia, Italy (G.Re., E.R., A.N., D.N., B.C.); Surgical Pathology Unit, Department of Medical and Surgical Sciences, Padua University, Padua, Italy (A.T.); Hull York Medical School, Hull University, Cottingham, UK (T.W.)
| | - Davide Nicoli
- From the Department of Medicine—DIMED, Internal Medicine 4, Padua University—School of Medicine, Padova, Italy (G.Ro., B.C., T.M.S., G.P.R.); Department of Internal Medicine, Santa Maria Nuova Hospital, Reggio Emilia, Italy (G.Re., E.R., A.N., D.N., B.C.); Surgical Pathology Unit, Department of Medical and Surgical Sciences, Padua University, Padua, Italy (A.T.); Hull York Medical School, Hull University, Cottingham, UK (T.W.)
| | - Bruno Casali
- From the Department of Medicine—DIMED, Internal Medicine 4, Padua University—School of Medicine, Padova, Italy (G.Ro., B.C., T.M.S., G.P.R.); Department of Internal Medicine, Santa Maria Nuova Hospital, Reggio Emilia, Italy (G.Re., E.R., A.N., D.N., B.C.); Surgical Pathology Unit, Department of Medical and Surgical Sciences, Padua University, Padua, Italy (A.T.); Hull York Medical School, Hull University, Cottingham, UK (T.W.)
| | - Antonio Toniato
- From the Department of Medicine—DIMED, Internal Medicine 4, Padua University—School of Medicine, Padova, Italy (G.Ro., B.C., T.M.S., G.P.R.); Department of Internal Medicine, Santa Maria Nuova Hospital, Reggio Emilia, Italy (G.Re., E.R., A.N., D.N., B.C.); Surgical Pathology Unit, Department of Medical and Surgical Sciences, Padua University, Padua, Italy (A.T.); Hull York Medical School, Hull University, Cottingham, UK (T.W.)
| | - Brasilina Caroccia
- From the Department of Medicine—DIMED, Internal Medicine 4, Padua University—School of Medicine, Padova, Italy (G.Ro., B.C., T.M.S., G.P.R.); Department of Internal Medicine, Santa Maria Nuova Hospital, Reggio Emilia, Italy (G.Re., E.R., A.N., D.N., B.C.); Surgical Pathology Unit, Department of Medical and Surgical Sciences, Padua University, Padua, Italy (A.T.); Hull York Medical School, Hull University, Cottingham, UK (T.W.)
| | - Teresa Maria Seccia
- From the Department of Medicine—DIMED, Internal Medicine 4, Padua University—School of Medicine, Padova, Italy (G.Ro., B.C., T.M.S., G.P.R.); Department of Internal Medicine, Santa Maria Nuova Hospital, Reggio Emilia, Italy (G.Re., E.R., A.N., D.N., B.C.); Surgical Pathology Unit, Department of Medical and Surgical Sciences, Padua University, Padua, Italy (A.T.); Hull York Medical School, Hull University, Cottingham, UK (T.W.)
| | - Thomas Walther
- From the Department of Medicine—DIMED, Internal Medicine 4, Padua University—School of Medicine, Padova, Italy (G.Ro., B.C., T.M.S., G.P.R.); Department of Internal Medicine, Santa Maria Nuova Hospital, Reggio Emilia, Italy (G.Re., E.R., A.N., D.N., B.C.); Surgical Pathology Unit, Department of Medical and Surgical Sciences, Padua University, Padua, Italy (A.T.); Hull York Medical School, Hull University, Cottingham, UK (T.W.)
| | - Gian Paolo Rossi
- From the Department of Medicine—DIMED, Internal Medicine 4, Padua University—School of Medicine, Padova, Italy (G.Ro., B.C., T.M.S., G.P.R.); Department of Internal Medicine, Santa Maria Nuova Hospital, Reggio Emilia, Italy (G.Re., E.R., A.N., D.N., B.C.); Surgical Pathology Unit, Department of Medical and Surgical Sciences, Padua University, Padua, Italy (A.T.); Hull York Medical School, Hull University, Cottingham, UK (T.W.)
| |
Collapse
|
28
|
He BJ, Anderson ME. Aldosterone and cardiovascular disease: the heart of the matter. Trends Endocrinol Metab 2013; 24:21-30. [PMID: 23040074 PMCID: PMC3532553 DOI: 10.1016/j.tem.2012.09.004] [Citation(s) in RCA: 48] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/23/2012] [Revised: 09/01/2012] [Accepted: 09/06/2012] [Indexed: 02/07/2023]
Abstract
Aldosterone contributes to the endocrine basis of heart failure, and studies on cardiac aldosterone signaling have reinforced its value as a therapeutic target. Recent focus has shifted to new roles of aldosterone that appear to depend on coexisting pathologic stimuli, cell type, and disease etiology. This review evaluates recent advances in mechanisms underlying aldosterone-induced cardiac disease and highlights the interplay between aldosterone and Ca(2+)/calmodulin dependent protein kinase II, whose hyperactivity during heart failure contributes to disease progression. Increasing evidence implicates aldosterone in diastolic dysfunction, and there is a need to develop more targeted therapeutics such as aldosterone synthase inhibitors and molecularly specific antioxidants. Despite accumulating knowledge, many questions still persist and will likely dictate areas of future research.
Collapse
Affiliation(s)
- B Julie He
- Department of Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA
| | | |
Collapse
|
29
|
Briet M, Schiffrin EL. Vascular actions of aldosterone. J Vasc Res 2012; 50:89-99. [PMID: 23172373 DOI: 10.1159/000345243] [Citation(s) in RCA: 118] [Impact Index Per Article: 9.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2012] [Accepted: 10/18/2012] [Indexed: 02/03/2023] Open
Abstract
Aldosterone exerts direct effects on the vascular system by inducing oxidative stress, inflammation, hypertrophic remodeling, fibrosis, and endothelial dysfunction. Aldosterone exerts its effects through genomic and nongenomic pathways in a mineralocorticoid receptor (MR)-dependent or independent manner. Other aldosterone receptors such as GPR30 have been identified. A tight relation exists between the aldosterone and angiotensin II pathways, as well as with the endothelin-1 system. There is a correlation between plasma levels of aldosterone and cardiovascular risk. Recently, an increasing body of evidence has underlined the importance of aldosterone in cardiovascular complications associated with the metabolic syndrome, such as arterial remodeling and endothelial dysfunction. Blockade of MR is an increasingly used evidence-based therapy for many forms of cardiovascular disease, including hypertension, heart failure, chronic kidney disease, and diabetes mellitus.
Collapse
Affiliation(s)
- Marie Briet
- Université Paris Descartes, Faculté de Medicine, Assistance Publique des Hôpitaux de Paris, Hôpital Européen Georges Pompidou, INSERM CIC 9201, Paris, France
| | | |
Collapse
|
30
|
Abstract
The RAS (renin–angiotensin system) plays a role not only in the cardiovascular system, including blood pressure regulation, but also in the central nervous system. AngII (angiotensin II) binds two major receptors: the AT1 receptor (AngII type 1 receptor) and AT2 receptor (AngII type 2 receptor). It has been recognized that AT2 receptor activation not only opposes AT1 receptor actions, but also has unique effects beyond inhibitory cross-talk with AT1 receptor signalling. Novel pathways beyond the classical actions of RAS, the ACE (angiotensin-converting enzyme)/AngII/AT1 receptor axis, have been highlighted: the ACE2/Ang-(1–7) [angiotensin-(1–7)]/Mas receptor axis as a new opposing axis against the ACE/AngII/AT1 receptor axis, novel AngII-receptor-interacting proteins and various AngII-receptor-activation mechanisms including dimer formation. ATRAP (AT1-receptor-associated protein) and ATIP (AT2-receptor-interacting protein) are well-characterized AngII-receptor-associated proteins. These proteins could regulate the functions of AngII receptors and thereby influence various pathophysiological states. Moreover, the possible cross-talk between PPAR (peroxisome-proliferator-activated receptor)-γ and AngII receptor subtypes is an intriguing issue to be addressed in order to understand the roles of RAS in the metabolic syndrome, and interestingly some ARBs (AT1-receptor blockers) have been reported to have an AT1-receptor-blocking action with a partial PPAR-γ agonistic effect. These emerging concepts concerning the regulation of AngII receptors are discussed in the present review.
Collapse
|
31
|
Berwick ZC, Dick GM, Tune JD. Heart of the matter: coronary dysfunction in metabolic syndrome. J Mol Cell Cardiol 2012; 52:848-56. [PMID: 21767548 PMCID: PMC3206994 DOI: 10.1016/j.yjmcc.2011.06.025] [Citation(s) in RCA: 51] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/30/2011] [Revised: 06/16/2011] [Accepted: 06/30/2011] [Indexed: 12/14/2022]
Abstract
Metabolic syndrome (MetS) is a collection of risk factors including obesity, dyslipidemia, insulin resistance/impaired glucose tolerance, and/or hypertension. The incidence of obesity has reached pandemic levels, as ~20-30% of adults in most developed countries can be classified as having MetS. This increased prevalence of MetS is critical as it is associated with a two-fold elevated risk for cardiovascular disease. Although the pathophysiology underlying this increase in disease has not been clearly defined, recent evidence indicates that alterations in the control of coronary blood flow could play an important role. The purpose of this review is to highlight current understanding of the effects of MetS on regulation of coronary blood flow and to outline the potential mechanisms involved. In particular, the role of neurohumoral modulation via sympathetic α-adrenoceptors and the renin-angiotensin-aldosterone system (RAAS) are explored. Alterations in the contribution of end-effector K(+), Ca(2+), and transient receptor potential (TRP) channels are also addressed. Finally, future perspectives and potential therapeutic targeting of the microcirculation in MetS are discussed. This article is part of a Special Issue entitled "Coronary Blood Flow".
Collapse
Affiliation(s)
- Zachary C. Berwick
- Department of Cellular and Integrative Physiology, Indiana University School of Medicine, Indianapolis, IN 46202
| | - Gregory M. Dick
- Department of Exercise Physiology Center for Cardiovascular and Respiratory Sciences West Virginia University School of Medicine
| | - Johnathan D. Tune
- Department of Cellular and Integrative Physiology, Indiana University School of Medicine, Indianapolis, IN 46202
| |
Collapse
|
32
|
Batenburg WW, Jansen PM, van den Bogaerdt AJ, J Danser AH. Angiotensin II-aldosterone interaction in human coronary microarteries involves GPR30, EGFR, and endothelial NO synthase. Cardiovasc Res 2012; 94:136-43. [PMID: 22260839 DOI: 10.1093/cvr/cvs016] [Citation(s) in RCA: 58] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/29/2022] Open
Abstract
AIMS The aim of this study was to investigate the aldosterone-angiotensin (Ang) II interaction in human coronary microarteries (HCMAs). METHODS AND RESULTS HCMAs, obtained from 75 heart-beating organ donors, were mounted in myographs and exposed to Ang II, either directly or following a 30-min pre-incubation with aldosterone, 17β-oestradiol, hydrocortisone, the p38 mitogen-activated protein kinase (MAPK) inhibitor SB203580, the extracellular regulated kinase 1/2 (ERK1/2) inhibitor PD98059, the GPR30 antagonist G15, or the epidermal growth factor receptor (EGFR) antagonist AG1478. Ang II constricted HCMAs in a concentration-dependent manner. All steroids, at nanomolar levels, potentiated Ang II and G15 prevented this effect. The potentiation disappeared or was reversed into Ang II antagonism at micromolar steroid levels. NO synthase (NOS) inhibition prevented the latter antagonism in the case of 17β-oestradiol, whereas both aldosterone and 17β-oestradiol at micro- (but not nano-) molar levels induced endothelial NOS phosphorylation in human umbilical vein endothelial cells. AG1478, but not SB203580 or PD98059, abolished the Ang II-induced contraction in the presence of aldosterone or 17β-oestradiol, and none of these drugs affected Ang II alone. CONCLUSION Steroids including aldosterone affect Ang II-induced vasoconstriction in a biphasic manner. Potentiation occurs at nanomolar steroid levels and depends on GPR30 and EGFR transactivation. At micromolar steroid levels, this potentiation either disappears (aldosterone and hydrocortisone) or is reversed into an inhibition (17β-oestradiol), and this is due to the endothelial NOS activation that occurs at such concentrations.
Collapse
Affiliation(s)
- Wendy W Batenburg
- Division of Pharmacology and Vascular Medicine, Department of Internal Medicine, Erasmus MC, Dr Molewaterplein 50, room EE1418b, 3015 GE Rotterdam, The Netherlands
| | | | | | | |
Collapse
|
33
|
Zhang ZH, Yu Y, Wei SG, Felder RB. Aldosterone-induced brain MAPK signaling and sympathetic excitation are angiotensin II type-1 receptor dependent. Am J Physiol Heart Circ Physiol 2011; 302:H742-51. [PMID: 22081704 DOI: 10.1152/ajpheart.00856.2011] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Abstract
Angiotensin II (ANG II)-induced mitogen-activated protein kinase (MAPK) signaling upregulates angiotensin II type-1 receptors (AT(1)R) in hypothalamic paraventricular nucleus (PVN) and contributes to AT(1)R-mediated sympathetic excitation in heart failure. Aldosterone has similar effects to increase AT(1)R expression in the PVN and sympathetic drive. The present study was undertaken to determine whether aldosterone also activates the sympathetic nervous system via MAPK signaling and, if so, whether its effect is independent of ANG II and AT(1)R. In anesthetized rats, a 4-h intravenous infusion of aldosterone induced increases (P < 0.05) in phosphorylated (p-) p44/42 MAPK in PVN, PVN neuronal excitation, renal sympathetic nerve activity (RSNA), mean blood pressure (MBP), and heart rate (HR). Intracerebroventricular or bilateral PVN microinjection of the p44/42 MAPK inhibitor PD-98059 reduced the aldosterone-induced RSNA, HR, and MBP responses. Intracerebroventricular pretreatment (5 days earlier) with pooled small interfering RNAs targeting p44/42 MAPK reduced total and p-p44/42 MAPK, aldosterone-induced c-Fos expression in the PVN, and the aldosterone-induced increases in RSNA, HR, and MBP. Intracerebroventricular infusion of either the mineralocorticoid receptor antagonist RU-28318 or the AT(1)R antagonist losartan blocked aldosterone-induced phosphorylation of p44/42 MAPK and prevented the increases in RSNA, HR, and MBP. These data suggest that aldosterone-induced sympathetic excitation depends upon that AT(1)R-induced MAPK signaling in the brain. The short time course of this interaction suggests a nongenomic mechanism, perhaps via an aldosterone-induced transactivation of the AT(1)R as described in peripheral tissues.
Collapse
Affiliation(s)
- Zhi-Hua Zhang
- Department of Internal Medicine, Roy J. and Lucille A. Carver College of Medicine, University of Iowa, owa City, IA52242, USA.
| | | | | | | |
Collapse
|
34
|
Affiliation(s)
- Tomohiro Osanai
- Department of Cardiology Hirosaki University Graduate School of Medicine Hirosaki, Japan (Osanai, Yamada, Okumura)
| | - Masahiro Yamada
- Department of Cardiology Hirosaki University Graduate School of Medicine Hirosaki, Japan (Osanai, Yamada, Okumura)
| | - Ken Okumura
- Department of Cardiology Hirosaki University Graduate School of Medicine Hirosaki, Japan (Osanai, Yamada, Okumura)
| |
Collapse
|
35
|
Rautureau Y, Paradis P, Schiffrin EL. Cross-talk between aldosterone and angiotensin signaling in vascular smooth muscle cells. Steroids 2011; 76:834-9. [PMID: 21371487 DOI: 10.1016/j.steroids.2011.02.015] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/01/2010] [Revised: 01/17/2011] [Accepted: 02/16/2011] [Indexed: 01/13/2023]
Abstract
In hypertension or other forms of cardiovascular disease, the chronic activation of the renin-angiotensin-aldosterone system (RAAS) leads to dysfunction of the vasculature, including, increased vascular tone, inflammation, fibrosis and thrombosis. Cross-talk between the main mediators of the RAAS, aldosterone and angiotensin (Ang) II, participates in the development of this vascular dysfunction. Recent studies have highlighted the molecular mechanisms supporting this cross-talk in vascular smooth muscle cells (VSMCs). Some of the signaling pathways activated by the Ang II type 1 receptor (AT(1)R) are dependent on the mineralocorticoid receptor (MR) and vice versa. VSMC signaling pathways involved in migration and growth are under the control of cross-talk between aldosterone and Ang II. A synergistic mechanism leads to potentiation of signaling pathways activated by each agent. The genomic and non-genomic mechanisms activated by aldosterone cooperate with Ang II to regulate vascular tone and gene expression of pro-inflammatory and pro-fibrotic molecules. This cross-talk is dependent on the non-receptor tyrosine kinase c-Src, and on receptor tyrosine kinases, EGFR and PDGFR, and leads to activation of MAP kinases and growth, migration and inflammatory effects. These new findings will contribute to development of better treatments for conditions in which the RAAS is excessively activated.
Collapse
MESH Headings
- Aldosterone/metabolism
- Aldosterone/physiology
- Angiotensins/metabolism
- Angiotensins/physiology
- Animals
- Gene Expression
- Humans
- Muscle, Smooth, Vascular/cytology
- Muscle, Smooth, Vascular/metabolism
- Muscle, Smooth, Vascular/physiology
- Myocytes, Smooth Muscle/metabolism
- Myocytes, Smooth Muscle/physiology
- Receptors, Angiotensin/metabolism
- Receptors, Angiotensin/physiology
- Receptors, Mineralocorticoid/metabolism
- Receptors, Mineralocorticoid/physiology
- Signal Transduction
- Transcriptional Activation
Collapse
Affiliation(s)
- Yohann Rautureau
- Lady Davis Institute for Medical Research, Sir Mortimer B. Davis-Jewish General Hospital, McGill University, Montreal, QC, Canada
| | | | | |
Collapse
|
36
|
Abstract
PURPOSE OF THIS REVIEW The purpose of this review is to explain the rationale and limitations for use of mineralocorticoid receptor blockers (MRBs) for the treatment of chronic kidney disease (CKD) and its complications. RECENT FINDINGS Recent studies in animal models of CKD demonstrate that blockade of the mineralocorticoid receptor using spironolactone or eplerenone decreases inflammation, oxidative stress, proteinuria and glomerular and tubular injury. Patients with CKD are at very high risk for progression of kidney disease and major cardiovascular events. Recent studies in patients with CKD demonstrate that administration of low doses of MRBs added onto an angiotensin-converting enzyme inhibitor-based regimen reduces proteinuria--a risk marker for both progressive kidney disease and cardiovascular events. However, incident hyperkalemia, an unwanted side effect, dampened enthusiasm for this approach. There are no large-scale, long-term outcome trials examining whether MRB can slow progression of kidney disease or prevent cardiovascular events. SUMMARY At this time it is unknown whether mineralocorticoid receptor blockade can improve outcomes in patients with CKD. To move this field forward and determine whether these agents can improve the lives of patients with kidney disease, novel strategies to prevent or ameliorate hyperkalemia are needed.
Collapse
|
37
|
Affiliation(s)
- A. H. Jan Danser
- Division of Pharmacology and Vascular Medicine, Department of Internal Medicine, Erasmus Medical Center, Rotterdam, The Netherlands
| |
Collapse
|
38
|
Lemarié CA, Simeone SMC, Nikonova A, Ebrahimian T, Deschênes ME, Coffman TM, Paradis P, Schiffrin EL. Aldosterone-induced activation of signaling pathways requires activity of angiotensin type 1a receptors. Circ Res 2009; 105:852-9. [PMID: 19762686 DOI: 10.1161/circresaha.109.196576] [Citation(s) in RCA: 75] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/21/2023]
Abstract
RATIONALE Aldosterone has been shown to induce vascular damage, endothelial dysfunction, and myocardial fibrosis, which depend in part on activation of angiotensin II (Ang II)-mediated pathways. However, mechanisms underlying crosstalk between Ang II type 1 receptor (AT(1)R) and mineralocorticoid receptor (MR) are mostly unknown. OBJECTIVES We tested whether the lack of Ang II type 1a receptor (AT(1a)R) or Ang II type 1b receptor (AT(1b)R) would decrease cellular effects induced by aldosterone. METHODS AND RESULTS We examined the effect of Ang II or aldosterone after transfection of mesenteric vascular smooth muscle cells (VSMCs) from C57Bl/6 mice with small interference RNA for AT(1a)R, AT(1b)R, or MR for 48 hours. Ang II and aldosterone separately induced ERK1/2, c-Jun NH2-terminal protein kinase (JNK), and nuclear factor (NF)-kappaB phosphorylation after a 20-minute stimulation. Small interference RNA for AT(1a)R downregulated phosphorylation of ERK1/2, JNK, and NF-kappaB after aldosterone stimulation compared to controls. Downregulation of AT(1b)R or MR only abolished the activation of NF-kappaB. In VSMCs from C57Bl/6 mice, aldosterone and Ang II induced the activation of the c-fos promoter from 30 minutes to 1 hour. This effect was blocked when using VSMCs from AT(1a)R knockout mice. CONCLUSION We show for the first time that nongenomic and genomic effects of aldosterone are differentially dependent on activity of both AT(1a)R and AT(1b)R. Our data suggest that aldosterone augments AT(1)R-dependent activation of ERK1/2, JNK, and NF-kappaB in VSMCs. We provide mechanistic understanding and experimental in vitro support for the benefit of combination therapy with dual blockade of AT(1)R and MR to treat hypertension and progression of heart failure as reported in clinical studies and animal models.
Collapse
Affiliation(s)
- Catherine A Lemarié
- Lady Davis Institute for Medical Research, Sir Mortimer B. Davis-Jewish General Hospital, McGill University, Montreal, Quebec, Canada
| | | | | | | | | | | | | | | |
Collapse
|
39
|
Jain G, Campbell RC, Warnock DG. Mineralocorticoid receptor blockers and chronic kidney disease. Clin J Am Soc Nephrol 2009; 4:1685-91. [PMID: 19729430 DOI: 10.2215/cjn.01340209] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
The increasing prevalence of chronic kidney disease (CKD) and the public health initiatives for detection and slowing its progression have placed special emphasis on controlling proteinuria and the renin-angiotensin-aldosterone system (RAAS). In addition to the traditional blockers of angiotensin-converting enzyme and angiotensin receptors, mineralocorticoid receptor blockers (MRBs) have come into focus as anti-proteinuric agents with moderate anti-hypertensive effects. The beneficial effects of MRBs on mortality in patients with cardiac disease have been well described. We review the role of aldosterone in end-organ damage, the rationales for using MRBs as adjuncts to angiotensin-converting enzyme inhibitors (ACEIs) and angiotensin receptor blockers (ARBs) in treating CKD, and the adverse effects that may occur when these agents are used in combination. Suggestions are included for avoiding serious adverse events in CKD patients treated with MRBs. There is a clearly defined need for prospective outcome studies focused on cardiovascular mortality as well as progression of CKD in patients treated with MRBS and other inhibitors of the RAAS.
Collapse
Affiliation(s)
- Gaurav Jain
- Department of Medicine, University of Alabama at Birmingham, Birmingham, Alabama, USA
| | | | | |
Collapse
|