1
|
Oliveira KB, de Souza FMA, de Sá LBM, Pacheco ALD, Prado MR, de Sousa Rodrigues CF, Bassi ÊJ, Santana-Melo I, Silva-Júnior A, Sabino-Silva R, Shetty AK, de Castro OW. Potential Mechanisms Underlying COVID-19-Mediated Central and Peripheral Demyelination: Roles of the RAAS and ADAM-17. Mol Neurobiol 2025; 62:1151-1164. [PMID: 38965171 DOI: 10.1007/s12035-024-04329-8] [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: 12/13/2023] [Accepted: 06/21/2024] [Indexed: 07/06/2024]
Abstract
Demyelination is among the most conspicuous neurological sequelae of SARS-CoV-2 infection (COVID-19) in both the central (CNS) and peripheral (PNS) nervous systems. Several hypotheses have been proposed to explain the mechanisms underlying demyelination in COVID-19. However, none have considered the SARS-CoV-2's effects on the renin-angiotensin-aldosterone system (RAAS). Therefore, our objective in this review is to evaluate how RAAS imbalance, caused by direct and indirect effects of SARS-CoV-2 infection, could contribute to myelin loss in the PNS and CNS. In the PNS, we propose that demyelination transpires from two significant changes induced by SARS-CoV-2 infection, which include upregulation of ADAM-17 and induction of lymphopenia. Whereas, in the CNS, demyelination could result from RAAS imbalance triggering two alterations: (1) a decrease in angiotensin type II receptor (AT2R) activity, responsible for restraining defense cells' action on myelin; (2) upregulation of ADAM-17 activity, leading to impaired maturation of oligodendrocytes and myelin formation. Thus, we hypothesize that increased ADAM-17 activity and decreased AT2R activity play roles in SARS-CoV-2 infection-mediated demyelination in the CNS.
Collapse
Affiliation(s)
- Kellysson Bruno Oliveira
- Institute of Biological Sciences and Health, Federal University of Alagoas (UFAL), Av. Lourival de Melo Mota, Km 14, Campus A. C. Simões, Cidade Universitária, Maceió, AL, CEP, 57072-970, Brazil
| | - Fernanda Maria Araujo de Souza
- Institute of Biological Sciences and Health, Federal University of Alagoas (UFAL), Av. Lourival de Melo Mota, Km 14, Campus A. C. Simões, Cidade Universitária, Maceió, AL, CEP, 57072-970, Brazil
| | - Letícia Barros Maurício de Sá
- Institute of Biological Sciences and Health, Federal University of Alagoas (UFAL), Av. Lourival de Melo Mota, Km 14, Campus A. C. Simões, Cidade Universitária, Maceió, AL, CEP, 57072-970, Brazil
| | - Amanda Larissa Dias Pacheco
- Institute of Biological Sciences and Health, Federal University of Alagoas (UFAL), Av. Lourival de Melo Mota, Km 14, Campus A. C. Simões, Cidade Universitária, Maceió, AL, CEP, 57072-970, Brazil
| | - Mariana Reis Prado
- Institute of Biological Sciences and Health, Federal University of Alagoas (UFAL), Av. Lourival de Melo Mota, Km 14, Campus A. C. Simões, Cidade Universitária, Maceió, AL, CEP, 57072-970, Brazil
| | - Célio Fernando de Sousa Rodrigues
- Institute of Biological Sciences and Health, Federal University of Alagoas (UFAL), Av. Lourival de Melo Mota, Km 14, Campus A. C. Simões, Cidade Universitária, Maceió, AL, CEP, 57072-970, Brazil
| | - Ênio José Bassi
- Institute of Biological Sciences and Health, Federal University of Alagoas (UFAL), Av. Lourival de Melo Mota, Km 14, Campus A. C. Simões, Cidade Universitária, Maceió, AL, CEP, 57072-970, Brazil
| | - Igor Santana-Melo
- Institute of Biological Sciences and Health, Federal University of Alagoas (UFAL), Av. Lourival de Melo Mota, Km 14, Campus A. C. Simões, Cidade Universitária, Maceió, AL, CEP, 57072-970, Brazil
| | - Abelardo Silva-Júnior
- Institute of Biological Sciences and Health, Federal University of Alagoas (UFAL), Av. Lourival de Melo Mota, Km 14, Campus A. C. Simões, Cidade Universitária, Maceió, AL, CEP, 57072-970, Brazil
| | - Robinson Sabino-Silva
- Department of Physiology, Institute of Biomedical Sciences, Federal University of Uberlândia (UFU), Uberlândia, MG, Brazil
| | - Ashok K Shetty
- Institute for Regenerative Medicine, Department of Cell Biology and Genetics, Texas A&M University School of Medicine, College Station, TX, USA.
- Institute for Regenerative Medicine, Department of Molecular and Cellular Medicine, Texas A&M Health Science Center College of Medicine, College Station, TX, 77843, USA.
| | - Olagide Wagner de Castro
- Institute of Biological Sciences and Health, Federal University of Alagoas (UFAL), Av. Lourival de Melo Mota, Km 14, Campus A. C. Simões, Cidade Universitária, Maceió, AL, CEP, 57072-970, Brazil.
| |
Collapse
|
2
|
Fujii W, Shibata S. Mineralocorticoid Receptor Antagonists for Preventing Chronic Kidney Disease Progression: Current Evidence and Future Challenges. Int J Mol Sci 2023; 24:ijms24097719. [PMID: 37175424 PMCID: PMC10178637 DOI: 10.3390/ijms24097719] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2023] [Revised: 04/05/2023] [Accepted: 04/20/2023] [Indexed: 05/15/2023] Open
Abstract
Regulation and action of the mineralocorticoid receptor (MR) have been the focus of intensive research over the past 80 years. Genetic and physiological/biochemical analysis revealed how MR and the steroid hormone aldosterone integrate the responses of distinct tubular cells in the face of environmental perturbations and how their dysregulation compromises fluid homeostasis. In addition to these roles, the accumulation of data also provided unequivocal evidence that MR is involved in the pathophysiology of kidney diseases. Experimental studies delineated the diverse pathological consequences of MR overactivity and uncovered the multiple mechanisms that result in enhanced MR signaling. In parallel, clinical studies consistently demonstrated that MR blockade reduces albuminuria in patients with chronic kidney disease. Moreover, recent large-scale clinical studies using finerenone have provided evidence that the non-steroidal MR antagonist can retard the kidney disease progression in diabetic patients. In this article, we review experimental data demonstrating the critical importance of MR in mediating renal injury as well as clinical studies providing evidence on the renoprotective effects of MR blockade. We also discuss areas of future investigation, which include the benefit of non-steroidal MR antagonists in non-diabetic kidney disease patients, the identification of surrogate markers for MR signaling in the kidney, and the search for key downstream mediators whereby MR blockade confers renoprotection. Insights into these questions would help maximize the benefit of MR blockade in subjects with kidney diseases.
Collapse
Affiliation(s)
- Wataru Fujii
- Division of Nephrology, Department of Internal Medicine, Graduate School of Medicine, Teikyo University, Tokyo 173-8605, Japan
| | - Shigeru Shibata
- Division of Nephrology, Department of Internal Medicine, Graduate School of Medicine, Teikyo University, Tokyo 173-8605, Japan
| |
Collapse
|
3
|
Oliveira KB, de Melo IS, da Silva BRM, Oliveira KLDS, Sabino-Silva R, Anhezini L, Katayama PL, Santos VR, Shetty AK, de Castro OW. SARS-CoV-2 and Hypertension: Evidence Supporting Invasion into the Brain Via Baroreflex Circuitry and the Role of Imbalanced Renin-Angiotensin-Aldosterone-System. Neurosci Insights 2023; 18:26331055231151926. [PMID: 36756280 PMCID: PMC9900164 DOI: 10.1177/26331055231151926] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2022] [Accepted: 01/04/2023] [Indexed: 02/05/2023] Open
Abstract
Hypertension is considered one of the most critical risk factors for COVID-19. Evidence suggests that SARS-CoV-2 infection produces intense effects on the cardiovascular system by weakening the wall of large vessels via vasa-vasorum. In this commentary, we propose that SARS-CoV-2 invades carotid and aortic baroreceptors, leading to infection of the nucleus tractus solitari (NTS) and paraventricular hypothalamic nucleus (PVN), and such dysregulation of NTS and PVN following infection causes blood pressure alteration at the central level. We additionally explored the hypothesis that SARS-CoV-2 favors the internalization of membrane ACE2 receptors generating an imbalance of the renin-angiotensin-aldosterone system (RAAS), increasing the activity of angiotensin II (ANG-II), disintegrin, and metalloproteinase 17 domain (ADAM17/TACE), eventually modulating the integration of afferents reaching the NTS from baroreceptors and promoting increased blood pressure. These mechanisms are related to the increased sympathetic activity, which leads to transient or permanent hypertension associated with SARS-CoV-2 invasion, contributing to the high number of deaths by cardiovascular implications.
Collapse
Affiliation(s)
- Kellysson Bruno Oliveira
- Department of Physiology, Institute of
Biological Sciences and Health, Federal University of Alagoas (UFAL), Maceió,
Alagoas, Brazil
| | - Igor Santana de Melo
- Department of Physiology, Institute of
Biological Sciences and Health, Federal University of Alagoas (UFAL), Maceió,
Alagoas, Brazil
| | - Bianca Rodrigues Melo da Silva
- Department of Physiology, Institute of
Biological Sciences and Health, Federal University of Alagoas (UFAL), Maceió,
Alagoas, Brazil
| | - Keylla Lavínia da Silva Oliveira
- Department of Physiology, Institute of
Biological Sciences and Health, Federal University of Alagoas (UFAL), Maceió,
Alagoas, Brazil
| | - Robinson Sabino-Silva
- Department of Physiology, Institute of
Biomedical Sciences, Federal University of Uberlandia (UFU), Uberlândia, Minas
Gerais, Brazil
| | - Lucas Anhezini
- Department of Histology, Institute of
Biological Sciences and Health, Federal University of Alagoas, Maceió, Alagoas,
Brazil
| | - Pedro Lourenco Katayama
- Department of Physiology and Pathology,
Dentistry School of Araraquara, São Paulo State University, Araraquara, São Paulo,
Brazil
| | - Victor Rodrigues Santos
- Department of Morphology, Institute of
Biological Science, Federal University of Minas Gerais (UFMG), Belo Horizonte, Minas
Gerais, Brazil
| | - Ashok K Shetty
- Institute for Regenerative Medicine,
Department of Cell Biology and Genetics, Texas A&M University School of
Medicine, College Station, TX, USA
| | - Olagide Wagner de Castro
- Department of Physiology, Institute of
Biological Sciences and Health, Federal University of Alagoas (UFAL), Maceió,
Alagoas, Brazil,Olagide Wagner de Castro, Institute of
Biological Sciences and Health, Federal University of Alagoas (UFAL), Av.
Lourival de Melo Mota, km 14, Campus A. C. Simões, Cidade Universitária, Maceió,
Alagoas CEP 57072-970, Brazil.
| |
Collapse
|
4
|
Wish JB, Pergola P. Evolution of Mineralocorticoid Receptor Antagonists in the Treatment of Chronic Kidney Disease Associated with Type 2 Diabetes Mellitus. Mayo Clin Proc Innov Qual Outcomes 2022; 6:536-551. [PMID: 36277502 PMCID: PMC9578990 DOI: 10.1016/j.mayocpiqo.2022.09.002] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
Chronic kidney disease (CKD) is one of the most frequent complications associated with type 2 diabetes mellitus (T2DM) and is also an independent risk factor for cardiovascular disease. The mineralocorticoid receptor (MR) is a nuclear receptor expressed in many tissue types, including kidney and heart. Aberrant and long-term activation of MR by aldosterone in patients with T2DM triggers detrimental effects (eg, inflammation and fibrosis) in these tissues. The suppression of aldosterone at the early stage of T2DM has been a therapeutic strategy for patients with T2DM-associated CKD. Although patients have been treated with renin-angiotensin system (RAS) blockers for decades, RAS blockers alone are not sufficient to prevent CKD progression. Steroidal MR antagonists (MRAs) have been used in combination with RAS blockers; however, undesired adverse effects have restricted their usage, prompting the development of nonsteroidal MRAs with better target specificity and safety profiles. Recently conducted studies, Finerenone in Reducing Kidney Failure and Disease Progression in Diabetic Kidney Disease (FIDELIO-DKD) and Finerenone in Reducing Cardiovascular Mortality and Morbidity in Diabetic Kidney Disease (FIGARO-DKD), have reported that finerenone, a nonsteroidal MRA, improves both renal and cardiovascular outcomes compared with placebo. In this article, we review the history of MRA development and discuss the possibility of its combination with other treatment options, such as sodium-glucose cotransporter 2 inhibitors, glucagon-like peptide-1 receptor agonists, and potassium binders for patients with T2DM-associated CKD.
Collapse
Key Words
- ACEi, angiotensin-converting enzyme inhibitor
- ADA, American Diabetes Association
- AR, androgen receptor
- ARB, angiotensin II receptor blocker
- ARTS, minerAlocorticoid Receptor Antagonist Tolerability Study
- BP, blood pressure
- CKD, chronic kidney disease
- CV, cardiovascular
- CVD, cardiovascular disease
- DM, diabetes mellitus
- DN, diabetic nephropathy
- ESKD, end-stage kidney disease
- FIDELIO-DKD, Finerenone in Reducing Kidney Failure and Disease Progression in Diabetic Kidney Disease
- FIGARO-DKD, Finerenone in Reducing Cardiovascular Mortality and Morbidity in Diabetic Kidney Disease
- GLP-1 RA, glucagon-like peptide 1 receptor agonists
- GR, glucocorticoid receptor
- HF, heart failure
- HFrEF, heart failure with reduced ejection fraction
- KDIGO, Kidney Disease Improving Global Outcomes
- MR, mineralocorticoid receptor
- MRA, mineralocorticoid receptor antagonist
- PR, progesterone receptor
- RAAS, renin–angiotensin–aldosterone system
- RAS, renin–angiotensin system
- SGLT-2i, sodium-glucose cotransporter 2 inhibitor
- T2DM, type 2 diabetes mellitus
- UACR, urinary albumin-creatine ratio
- eGFR, estimated glomerular filtration rate
Collapse
Affiliation(s)
- Jay B. Wish
- Department of Medicine, Indiana University School of Medicine and Indiana University Health, Indianapolis
| | | |
Collapse
|
5
|
Epstein M, Kovesdy CP, Clase CM, Sood MM, Pecoits-Filho R. Aldosterone, Mineralocorticoid Receptor Activation, and CKD: A Review of Evolving Treatment Paradigms. Am J Kidney Dis 2022; 80:658-666. [PMID: 36057467 DOI: 10.1053/j.ajkd.2022.04.016] [Citation(s) in RCA: 43] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2022] [Accepted: 04/19/2022] [Indexed: 02/02/2023]
Abstract
Mineralocorticoid receptor (MR) activation is involved in propagating kidney injury, inflammation, and fibrosis and in the progression of chronic kidney disease (CKD). Multiple clinical studies have defined the efficacy of MR antagonism in attenuating progressive kidney disease, and the US Food and Drug Administration recently approved the nonsteroidal mineralocorticoid receptor antagonist (MRA) finerenone for this indication. In this review, we consider the basic science and clinical applicability of MR antagonism. Because hyperkalemia constitutes a constraint to implementing evidence-based MR blockade, we review MRA-associated hyperkalemia in the context of finerenone and discuss evolving mitigation strategies to enhance the safety and efficacy of this treatment. Although the FIDELIO-DKD and FIGARO-DKD clinical trials focused solely on patients with type 2 diabetes mellitus, we propose that MR activation and the resulting inflammation and fibrosis act as a substantive pathogenetic mediator not only in people with diabetic CKD but also in those with CKD without diabetes. We close by briefly discussing both recently initiated and future clinical trials that focus on extending the attributes of MR antagonism to a wider array of nondiabetic kidney disorders, such as patients with nonalbuminuric CKD.
Collapse
Affiliation(s)
- Murray Epstein
- Division of Nephrology and Hypertension, Miller School of Medicine, University of Miami, Miami, Florida.
| | - Csaba P Kovesdy
- University of Tennessee Health Science Center, Memphis, Tennessee; Nephrology, Memphis Veterans Affairs Medical Center, Memphis, Tennessee
| | | | - Manish M Sood
- Department of Medicine and School of Epidemiology and Public Health, University of Ottawa, Ottawa, Ontario, Canada; Ottawa Hospital Research Institute, Ottawa, Ontario, Canada; Nephrology, The Ottawa Hospital, Ottawa, Ontario, Canada
| | - Roberto Pecoits-Filho
- Arbor Research Collaborative for Health, Ann Arbor, Michigan; Escola de Medicina, Pontifical Catholic University of Paraná, Curitiba, Brazil
| |
Collapse
|
6
|
Januszewicz A, Mulatero P, Dobrowolski P, Monticone S, Van der Niepen P, Sarafidis P, Reincke M, Rexhaj E, Eisenhofer G, Januszewicz M, Kasiakogias A, Kreutz R, Lenders JW, Muiesan ML, Persu A, Agabiti-Rosei E, Soria R, Śpiewak M, Prejbisz A, Messerli FH. Cardiac Phenotypes in Secondary Hypertension. J Am Coll Cardiol 2022; 80:1480-1497. [DOI: 10.1016/j.jacc.2022.08.714] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/15/2022] [Revised: 08/03/2022] [Accepted: 08/04/2022] [Indexed: 11/06/2022]
|
7
|
Hirooka K, Kiuchi Y. The Retinal Renin-Angiotensin-Aldosterone System: Implications for Glaucoma. Antioxidants (Basel) 2022; 11:antiox11040610. [PMID: 35453295 PMCID: PMC9029628 DOI: 10.3390/antiox11040610] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2022] [Revised: 03/18/2022] [Accepted: 03/18/2022] [Indexed: 12/26/2022] Open
Abstract
Aldosterone is one of the main effectors of the renin-angiotensin-aldosterone system (RAAS) along with having roles in hypertension, and cardiovascular and renal diseases. Recent evidence has also shown the presence of an active local RAAS within the human eye. It has been shown that at 12 h after a retinal ischemia-reperfusion injury, there is an upregulation of the protein levels of angiotensin II type 1 receptor (AT1-R) in the retina. Furthermore, at 12 h after reperfusion, there is an increase in reactive oxygen species (ROS) production in the retina that is mediated via an NADPH oxidase pathway. This ischemia-reperfusion injury-induced increase of retinal ROS levels and NADPH oxidase expression can be prevented by the administration of an AT1-R antagonist. This suggests that one of the main retinal ischemic injury pathways is via the local RAAS. It has also been reported that progressive retinal ganglion cell loss and glaucomatous optic nerve degeneration without elevated intraocular pressure occur after administration of local or systemic aldosterone. Elucidation of glaucoma pathogenesis, especially normal-tension glaucoma (NTG) subtype by our current animal model can be used for identifying potential therapeutic targets. Based on these results, we are further evaluating NTG prevalence among primary aldosteronism patients.
Collapse
|
8
|
Barrera-Chimal J, Jaisser F, Anders HJ. The mineralocorticoid receptor in chronic kidney disease. Br J Pharmacol 2021; 179:3152-3164. [PMID: 34786690 DOI: 10.1111/bph.15734] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2021] [Revised: 09/11/2021] [Accepted: 10/22/2021] [Indexed: 11/27/2022] Open
Abstract
Chronic kidney disease (CKD) is a major public health concern, affecting approximately 10% of the population worldwide. CKD of glomerular or tubular origin leads to the activation of stress mechanisms, including the renin angiotensin aldosterone system and mineralocorticoid receptor (MR) activation. Over the last two decades, blockade of the MR has arisen as a potential therapeutic approach against various forms of kidney disease. In this review, we summarize the experimental studies that have shown a protective effect of MR antagonists (MRAs) in non-diabetic and diabetic CKD animal models. Moreover, we review the main clinical trials that have shown the clinical application of MRAs to reduce albuminuria and, importantly, to slow CKD progression. Recent evidence from the FIDELIO trial showed that the MRA finerenone can reduce hard kidney outcomes when added to the standard of care in CKD associated with type 2 diabetes. Finally, we discuss the effects of MRAs relative to those of SGLT2 inhibitors, as well as the potential benefit of combination therapy to maximize organ protection.
Collapse
Affiliation(s)
- Jonatan Barrera-Chimal
- Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México, Ciudad Universitaria, Mexico City, Mexico.,Laboratorio de Fisiología Cardiovascular y Trasplante Renal, Unidad de Investigación UNAM-INC, Instituto Nacional de Cardiología Ignacio Chávez, Mexico City, Mexico
| | - Frederic Jaisser
- INSERM, UMRS 1138, Centre de Recherche des Cordeliers, Sorbonne Université, Université de Paris, Paris, France.,Université de Lorraine, INSERM Centre d'Investigations Cliniques-Plurithématique 1433, UMR 1116, CHRU de Nancy, French-Clinical Research Infrastructure Network (F-CRIN) INI-CRCT, Nancy, France
| | - Hans-Joachim Anders
- Medizinische Klinik und Poliklinik IV, LMU Klinikum, Ziemssenstr. 1, D-80336, München
| |
Collapse
|
9
|
Beta-Arrestins in the Treatment of Heart Failure Related to Hypertension: A Comprehensive Review. Pharmaceutics 2021. [DOI: 10.3390/pharmaceutics13060838
expr 929824082 + 956151497] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/16/2023] Open
Abstract
Heart failure (HF) is a complicated clinical syndrome that is considered an increasingly frequent reason for hospitalization, characterized by a complex therapeutic regimen, reduced quality of life, and high morbidity. Long-standing hypertension ultimately paves the way for HF. Recently, there have been improvements in the treatment of hypertension and overall management not limited to only conventional medications, but several novel pathways and their pharmacological alteration are also conducive to the treatment of hypertension. Beta-arrestin (β-arrestin), a protein responsible for beta-adrenergic receptors’ (β-AR) functioning and trafficking, has recently been discovered as a potential regulator in hypertension. β-arrestin isoforms, namely β-arrestin1 and β-arrestin2, mainly regulate cardiac function. However, there have been some controversies regarding the function of the two β-arrestins in hypertension regarding HF. In the present review, we try to figure out the paradox between the roles of two isoforms of β-arrestin in the treatment of HF.
Collapse
|
10
|
Rakib A, Eva TA, Sami SA, Mitra S, Nafiz IH, Das A, Tareq AM, Nainu F, Dhama K, Emran TB, Simal-Gandara J. Beta-Arrestins in the Treatment of Heart Failure Related to Hypertension: A Comprehensive Review. Pharmaceutics 2021; 13:838. [PMID: 34198801 PMCID: PMC8228839 DOI: 10.3390/pharmaceutics13060838] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2021] [Revised: 05/28/2021] [Accepted: 05/31/2021] [Indexed: 12/24/2022] Open
Abstract
Heart failure (HF) is a complicated clinical syndrome that is considered an increasingly frequent reason for hospitalization, characterized by a complex therapeutic regimen, reduced quality of life, and high morbidity. Long-standing hypertension ultimately paves the way for HF. Recently, there have been improvements in the treatment of hypertension and overall management not limited to only conventional medications, but several novel pathways and their pharmacological alteration are also conducive to the treatment of hypertension. Beta-arrestin (β-arrestin), a protein responsible for beta-adrenergic receptors' (β-AR) functioning and trafficking, has recently been discovered as a potential regulator in hypertension. β-arrestin isoforms, namely β-arrestin1 and β-arrestin2, mainly regulate cardiac function. However, there have been some controversies regarding the function of the two β-arrestins in hypertension regarding HF. In the present review, we try to figure out the paradox between the roles of two isoforms of β-arrestin in the treatment of HF.
Collapse
Affiliation(s)
- Ahmed Rakib
- Department of Pharmacy, Faculty of Biological Sciences, University of Chittagong, Chittagong 4331, Bangladesh; (A.R.); (T.A.E.); (S.A.S.)
| | - Taslima Akter Eva
- Department of Pharmacy, Faculty of Biological Sciences, University of Chittagong, Chittagong 4331, Bangladesh; (A.R.); (T.A.E.); (S.A.S.)
| | - Saad Ahmed Sami
- Department of Pharmacy, Faculty of Biological Sciences, University of Chittagong, Chittagong 4331, Bangladesh; (A.R.); (T.A.E.); (S.A.S.)
| | - Saikat Mitra
- Department of Pharmacy, Faculty of Pharmacy, University of Dhaka, Dhaka 1000, Bangladesh;
| | - Iqbal Hossain Nafiz
- Department of Biochemistry and Molecular Biology, Faculty of Biological Sciences, University of Chittagong, Chittagong 4331, Bangladesh; (I.H.N.); (A.D.)
| | - Ayan Das
- Department of Biochemistry and Molecular Biology, Faculty of Biological Sciences, University of Chittagong, Chittagong 4331, Bangladesh; (I.H.N.); (A.D.)
| | - Abu Montakim Tareq
- Department of Pharmacy, International Islamic University Chittagong, Chittagong 4318, Bangladesh;
| | - Firzan Nainu
- Faculty of Pharmacy, Hasanuddin University, Tamalanrea, Kota Makassar, Sulawesi Selatan 90245, Indonesia;
| | - Kuldeep Dhama
- Division of Pathology, ICAR-Indian Veterinary Research Institute, Izatnagar, Bareilly 243122, Uttar Pradesh, India;
| | - Talha Bin Emran
- Department of Pharmacy, BGC Trust University Bangladesh, Chittagong 4381, Bangladesh
| | - Jesus Simal-Gandara
- Nutrition and Bromatology Group, Department of Analytical and Food Chemistry, Faculty of Food Science and Technology, University of Vigo–Ourense Campus, E32004 Ourense, Spain
| |
Collapse
|
11
|
de Melo IS, Sabino-Silva R, Cunha TM, Goulart LR, Reis WL, Jardim ACG, Shetty AK, de Castro OW. Hydroelectrolytic Disorder in COVID-19 patients: Evidence Supporting the Involvement of Subfornical Organ and Paraventricular Nucleus of the Hypothalamus. Neurosci Biobehav Rev 2021; 124:216-223. [PMID: 33577841 PMCID: PMC7872848 DOI: 10.1016/j.neubiorev.2021.02.008] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2020] [Revised: 02/02/2021] [Accepted: 02/03/2021] [Indexed: 02/07/2023]
Abstract
Multiple neurological problems have been reported in coronavirus disease-2019 (COVID-19) patients because severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) likely spreads to the central nervous system (CNS) via olfactory nerves or through the subarachnoid space along olfactory nerves into the brain's cerebrospinal fluid and then into the brain's interstitial space. We hypothesize that SARS-CoV-2 enters the subfornical organ (SFO) through the above routes and the circulating blood since circumventricular organs (CVOs) such as the SFO lack the blood-brain barrier, and infection of the SFO causes dysfunction of the hypothalamic paraventricular nucleus (PVN) and supraoptic nucleus (SON), leading to hydroelectrolytic disorder. SARS-CoV-2 can readily enter SFO-PVN-SON neurons because these neurons express angiotensin-converting enzyme-2 receptors and proteolytic viral activators, which likely leads to neurodegeneration or neuroinflammation in these regions. Considering the pivotal role of SFO-PVN-SON circuitry in modulating hydroelectrolyte balance, SARS-CoV-2 infection in these regions could disrupt the neuroendocrine control of hydromineral homeostasis. This review proposes mechanisms by which SARS-CoV-2 infection of the SFO-PVN-SON pathway leads to hydroelectrolytic disorder in COVID-19 patients.
Collapse
Affiliation(s)
- Igor Santana de Melo
- Department of Physiology, Institute of Biological Sciences and Health, Federal University of Alagoas (UFAL), Maceio, Brazil
| | - Robinson Sabino-Silva
- Department of Physiology, Institute of Biomedical Sciences, Federal University of Uberlandia (UFU), Uberlândia, MG, Brazil.
| | - Thúlio Marquez Cunha
- Department of Pulmonology, School of Medicine, Federal University of Uberlandia, Minas Gerais, Brazil
| | - Luiz Ricardo Goulart
- Institute of Biotechnology, Federal University of Uberlandia, Minas Gerais, Brazil
| | - Wagner Luis Reis
- Department of Physiological, Sciences Biological Sciences Centre Federal University of Santa Catarina (UFSC) Florianopolis, Santa Catarina, Brazil
| | - Ana Carolina Gomes Jardim
- Laboratory of Virology, Institute of Biomedical Sciences, Federal University of Uberlandia, Minas Gerais, Brazil
| | - Ashok K Shetty
- Institute for Regenerative Medicine, Department of Molecular and Cellular Medicine, Texas A&M University College of Medicine, College Station, TX, 77843, USA.
| | - Olagide Wagner de Castro
- Department of Physiology, Institute of Biological Sciences and Health, Federal University of Alagoas (UFAL), Maceio, Brazil.
| |
Collapse
|
12
|
Young MJ, Kanki M, Fuller PJ, Yang J. Identifying new cellular mechanisms of mineralocorticoid receptor activation in the heart. J Hum Hypertens 2021; 35:124-130. [PMID: 32733061 DOI: 10.1038/s41371-020-0386-5] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2020] [Revised: 07/01/2020] [Accepted: 07/16/2020] [Indexed: 01/30/2023]
Abstract
Recent studies have expanded our understanding of the actions of the mineralocorticoid receptor (MR) to a diverse array of tissue types that differ substantially from the epithelial cells of the renal nephron. In these cell types the role of the MR has been largely, but not exclusively, defined in terms of pathogenic signalling pathways leading to tissue injury and remodelling. Macrophages and cardiomyocytes are two cell types in which the MR plays a central role in the cardiac tissue response to injury, renovascular hypertension and oxidative stress for example. Macrophages are critical for resolution of tissue injury and wound healing and their pleiotropic actions are central to the development of many forms of heart, renal and vascular disease. The MR in cardiomyocytes is not only essential for the chronotropic and ionotropic actions of mineralocorticoids in the short and longer term, but also for induction of hypertrophic and proinflammatory signalling programs. The present review discusses recent studies, presented at the Aldosterone and Hypertension Satellite of the 15th Asian-Pacific Congress of Hypertension, investigating new mechanisms for MR signalling in these cells and how their dysfunction contributes to the onset and progression of cardiovascular disease and heart failure.
Collapse
Affiliation(s)
- Morag J Young
- Centre for Endocrinology and Metabolism, Hudson Institute of Medical Research and the Department of Molecular Translational Science, Monash University, Clayton, VIC, Australia. .,Baker Heart and Diabetes Institute, Melborne, VIC, Australia.
| | - Monica Kanki
- Centre for Endocrinology and Metabolism, Hudson Institute of Medical Research and the Department of Molecular Translational Science, Monash University, Clayton, VIC, Australia.,Baker Heart and Diabetes Institute, Melborne, VIC, Australia
| | - Peter J Fuller
- Centre for Endocrinology and Metabolism, Hudson Institute of Medical Research and the Department of Molecular Translational Science, Monash University, Clayton, VIC, Australia
| | - Jun Yang
- Centre for Endocrinology and Metabolism, Hudson Institute of Medical Research and the Department of Molecular Translational Science, Monash University, Clayton, VIC, Australia
| |
Collapse
|
13
|
Abstract
Nuclear receptors have a broad spectrum of biological functions in normal physiology and in the pathology of various diseases, including glomerular disease. The primary therapies for many glomerular diseases are glucocorticoids, which exert their immunosuppressive and direct podocyte protective effects via the glucocorticoid receptor (GR). As glucocorticoids are associated with important adverse effects and a substantial proportion of patients show resistance to these therapies, the beneficial effects of selective GR modulators are now being explored. Peroxisome proliferator-activated receptor-γ (PPARγ) agonism using thiazolidinediones has potent podocyte cytoprotective and nephroprotective effects. Repurposing of thiazolidinediones or identification of novel PPARγ modulators are potential strategies to treat non-diabetic glomerular disease. Retinoic acid receptor-α is the key mediator of the renal protective effects of retinoic acid, and repair of the endogenous retinoic acid pathway offers another potential therapeutic strategy for glomerular disease. Vitamin D receptor, oestrogen receptor and mineralocorticoid receptor modulators regulate podocyte injury in experimental models. Further studies are needed to better understand the mechanisms of these nuclear receptors, evaluate their synergistic pathways and identify their novel modulators. Here, we focus on the role of nuclear receptors in podocyte biology and non-diabetic glomerular disease.
Collapse
|
14
|
Liao HW, Wang SM, Chan CK, Lin YH, Lin PC, Ho CH, Liu YC, Chueh JS, Wu VC. Transtubular potassium gradient predicts kidney function impairment after adrenalectomy in primary aldosteronism. Ther Adv Chronic Dis 2020; 11:2040622320944792. [PMID: 32922714 PMCID: PMC7457632 DOI: 10.1177/2040622320944792] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2020] [Accepted: 06/26/2020] [Indexed: 11/17/2022] Open
Abstract
Background: In primary aldosteronism (PA), kidney function impairment could be concealed
by relative hyperfiltration and emerge after adrenalectomy. We hypothesized
transtubular gradient potassium gradient (TTKG), a kidney aldosterone
bioactivity indicator, could correlate to end organ damage and forecast
kidney function impairment after adrenalectomy. Methods: In the present prospective study, we enrolled lateralized PA patients who
underwent adrenalectomy and were followed up 12 months after operation in
the Taiwan Primary Aldosteronism Investigation (TAIPAI) registry from 2010
to 2018. The clinical outcome was kidney function impairment, defined as
estimated glomerular filtration rate (eGFR) <60 ml/min/1.73 m2
at 12 months after adrenalectomy. End organ damage is determined by
microalbuminuria and left ventricular mass. Results: In total, 323 patients [mean, 50.8 ± 10.9 years old; female 178 (55.1%)] were
enrolled. Comparing pre-operation and post-operation data, systolic blood
pressure, serum aldosterone, urinary albumin to creatinine ratio and eGFR
decreased. TTKG ⩾ 4.9 correlated with pre-operative urinary albumin to
creatinine ratio >50 mg/g [odds ratio (OR) = 2.42;
p = 0.034] and left ventricular mass (B = 20.10;
p = 0.018). Multivariate logistic regression analysis
demonstrated that TTKG ⩾ 4.9 could predict concealed chronic kidney disease
(OR = 5.42; p = 0.011) and clinical success (OR = 2.90,
p = 0.017) at 12 months after adrenalectomy. Conclusions: TTKG could predict concealed kidney function impairment and cure of
hypertension in PA patients after adrenalectomy. TTKG more than 4.9 as an
adverse surrogate of aldosterone and hypokalaemia correlated with
pre-operative end organ damage in terms of high proteinuria and cardiac
hypertrophy.
Collapse
Affiliation(s)
| | - Shuo-Meng Wang
- Department of Urology, National Taiwan University Hospital, Taipei
| | - Chieh-Kai Chan
- Department of Internal Medicine, National Taiwan University Hospital, Hsin-Chu branch, Hsin-Chu
| | - Yen-Hung Lin
- Department of Internal Medicine, National Taiwan University Hospital, Taipei
| | - Po-Chih Lin
- Department of Internal Medicine, National Taiwan University Hospital, Taipei
| | - Chen-Hsun Ho
- Division of Urology, Department of Surgery, Shin Kong Wu Ho-Su Memorial Hospital, Taipei, Taiwan
| | | | - Jeff S Chueh
- Glickman Urological and Kidney Institute, and Cleveland Clinic Lerner College of Medicine, Cleveland Clinic, Cleveland, OH, USA
| | - Vin-Cent Wu
- Department of Internal Medicine, National Taiwan University Hospital, Room 1555, Clinical Research Building, 7 Chung-Shan South Road, Taipei 100
| |
Collapse
|
15
|
Chen D, Li X, Song Q, Hu C, Su F, Dai J, Ye Y, Huang J, Zhang X. Assessment of Hypokalemia and Clinical Characteristics in Patients With Coronavirus Disease 2019 in Wenzhou, China. JAMA Netw Open 2020; 3:e2011122. [PMID: 32525548 PMCID: PMC7290402 DOI: 10.1001/jamanetworkopen.2020.11122] [Citation(s) in RCA: 148] [Impact Index Per Article: 29.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
IMPORTANCE Severe acute respiratory syndrome coronavirus 2 has caused a global outbreak of coronavirus disease 2019 (COVID-19). Severe acute respiratory syndrome coronavirus 2 binds angiotensin-converting enzyme 2 of the rennin-angiotensin system, resulting in hypokalemia. OBJECTIVE To investigate the prevalence, causes, and clinical implications of hypokalemia, including its possible association with treatment outcomes, among patients with COVID-19. DESIGN, SETTING, AND PARTICIPANTS This cohort study was conducted at Wenzhou Central Hospital and Sixth People's Hospital of Wenzhou, Wenzhou, China, from January 11, 2020, to February 15, 2020. Participants included patients who received a diagnosis of COVID-19 according to the criteria issued by the Chinese Health Bureau and were admitted to the hospital. The patients were classified as having severe hypokalemia (plasma potassium <3 mmol/L), hypokalemia (plasma potassium 3-3.5 mmol/L), and normokalemia (plasma potassium >3.5 mmol/L). The clinical features, therapy, and outcomes were compared between the 3 groups. Data analysis was conducted in March 2020. INTERVENTIONS The patients were given general support and antiviral therapy. Their epidemiological and clinical features were collected. MAIN OUTCOMES AND MEASURES The prevalence of hypokalemia and response to treatment with potassium supplements were measured by analyzing plasma and urine potassium levels. RESULTS One hundred seventy-five patients (87 female patients [50%]; mean [SD] age, 45 [14] years) were classified as having severe hypokalemia (31 patients [18%]), hypokalemia (64 patients [37%]), and normokalemia (80 patients [46%]). Patients with severe hypokalemia had statistically significantly higher body temperature (mean [SD], 37.6 °C [0.9 °C]) than the patients with hypokalemia (mean [SD], 37.2 °C [0.7 °C]; difference, 0.4 °C; 95% CI, 0.2-0.6 °C; P = .02) and the patients with normokalemia (mean [SD], 37.1 °C [0.8 °C]; difference, 0.5 °C; 95% CI, 0.3-0.7 °C; P = .005). Patients with higher levels of hypokalemia also had higher creatine kinase levels (severe hypokalemia, mean [SD], 200 [257] U/L [median, 113 U/L; interquartile range {IQR}, 61-242 U/L]; hypokalemia, mean [SD], 97 [85] U/L; and normokalemia, mean [SD], 82 [57] U/L), higher creatine kinase-MB fraction (severe hypokalemia, mean [SD], 32 [39] U/L [median, 14 U/L; IQR, 11-36 U/L]; hypokalemia, mean [SD], 18 [15] U/L; and normokalemia, mean [SD], 15 [8] U/L), higher lactate dehydrogenase levels (mean [SD], severe hypokalemia, 256 [88] U/L; hypokalemia, 212 [59] U/L; and normokalemia, 199 [61] U/L), and higher C-reactive protein levels (severe hypokalemia, mean [SD], 29 [23] mg/L; hypokalemia, mean [SD], 18 [20] mg/L [median, 12, mg/L; IQR, 4-25 mg/L]; and normokalemia, mean [SD], 15 [18] mg/L [median, 6 U/L; IQR, 3-17 U/L]). Of 40 severely and critically ill patients, 34 (85%) had hypokalemia. Patients with severe hypokalemia were given potassium at a dose of 40 mEq per day, for a total mean (SD) of 453 (53) mEq potassium chloride, during the hospital stay. The patients responded well to potassium supplements as they recovered. CONCLUSIONS AND RELEVANCE The correction of hypokalemia is challenging because of continuous renal potassium loss resulting from the degradation of angiotensin-converting enzyme 2. The high prevalence of hypokalemia among patients with COVID-19 suggests the presence of disordered rennin-angiotensin system activity, which increases as a result of reduced counteractivity of angiotensin-converting enzyme 2, which is bound by severe acute respiratory syndrome coronavirus 2.
Collapse
Affiliation(s)
- Dong Chen
- Department of Infectious Diseases, The Ding Li Clinical College of Wenzhou Medical University, Wenzhou, Zhejiang Province, China
- Department of Infectious Diseases, Sixth People’s Hospital of Wenzhou, Wenzhou, Zhejiang Province, China
| | - Xiaokun Li
- Virus Research Institute, Wenzhou Medical University, Wenzhou, Zhejiang Province, China
| | - Qifa Song
- Department of Microbiology, Ningbo Municipal Centre for Disease Control and Prevention, Ningbo, Zhejiang Province, China
| | - Chenchan Hu
- Department of Infectious Diseases, The Ding Li Clinical College of Wenzhou Medical University, Wenzhou, Zhejiang Province, China
- Department of Infectious Diseases, Sixth People’s Hospital of Wenzhou, Wenzhou, Zhejiang Province, China
| | - Feifei Su
- Department of Infectious Diseases, The Ding Li Clinical College of Wenzhou Medical University, Wenzhou, Zhejiang Province, China
- Department of Infectious Diseases, Sixth People’s Hospital of Wenzhou, Wenzhou, Zhejiang Province, China
| | - Jianyi Dai
- Department of Infectious Diseases, The Ding Li Clinical College of Wenzhou Medical University, Wenzhou, Zhejiang Province, China
- Department of Infectious Diseases, Sixth People’s Hospital of Wenzhou, Wenzhou, Zhejiang Province, China
| | - Yinghai Ye
- Department of Infectious Diseases, The Ding Li Clinical College of Wenzhou Medical University, Wenzhou, Zhejiang Province, China
- Department of Infectious Diseases, Sixth People’s Hospital of Wenzhou, Wenzhou, Zhejiang Province, China
| | - Jianping Huang
- Department of Infectious Diseases, The Ding Li Clinical College of Wenzhou Medical University, Wenzhou, Zhejiang Province, China
- Department of Infectious Diseases, Sixth People’s Hospital of Wenzhou, Wenzhou, Zhejiang Province, China
| | - Xiaoming Zhang
- Department of Microbiology, Wenzhou Municipal Centre for Disease Control and Prevention, Wenzhou, Zhejiang Province, China
| |
Collapse
|
16
|
Leader CJ, Kelly DJ, Sammut IA, Wilkins GT, Walker RJ. Spironolactone mitigates, but does not reverse, the progression of renal fibrosis in a transgenic hypertensive rat. Physiol Rep 2020; 8:e14448. [PMID: 32441493 PMCID: PMC7243196 DOI: 10.14814/phy2.14448] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2020] [Revised: 04/15/2020] [Accepted: 04/24/2020] [Indexed: 01/10/2023] Open
Abstract
Hypertension plays an important role in the development and progression of chronic kidney disease. Studies to date, with mineralocorticoid receptor antagonists (MRA), have demonstrated varying degrees of results in modifying the development of renal fibrosis. This study aimed to investigate whether treatment with a MRA commenced following the establishment of hypertension, a situation more accurately representing the clinical setting, modified the progression of renal fibrosis. Using male Cyp1a1Ren2 rats (n = 28), hypertension was established by addition of 0.167% indole-3-carbinol (w/w) to the rat chow, for 2 weeks prior to treatment. Rats were then divided into normotensive, hypertensive (H), or hypertensive with daily oral spironolactone treatment (H + SP) (human equivalent dose 50 mg/day). Physiological data and tissue were collected after 4 and 12 weeks for analysis. After 4 weeks, spironolactone had no demonstrable effect on systolic blood pressure (SBP), proteinuria, or macrophage infiltration in the renal cortex. However, glomerulosclerosis and renal cortical fibrosis were significantly decreased. Following 12 weeks of spironolactone treatment, SBP was lowered (not back to normotensive levels), proteinuria was reduced, and the progression of glomerulosclerosis and renal cortical fibrosis was significantly blunted. This was associated with a significant reduction in macrophage and myofibroblast infiltration, as well as CTGF and pSMAD2 expression. In summary, in a model of established hypertension, spironolactone significantly blunted the progression of renal fibrosis and glomerulosclerosis, and downregulated the renal inflammatory response, which was associated with reduced proteinuria, despite only a partial reduction in systolic blood pressure. This suggests a blood pressure independent effect of MRA on renal fibrosis.
Collapse
Affiliation(s)
| | - Darren J. Kelly
- Department of MedicineUniversity of MelbourneMelbourneVICAustralia
| | - Ivan A. Sammut
- Department of PharmacologyUniversity of OtagoDunedinNew Zealand
| | | | | |
Collapse
|
17
|
Barrera‐Chimal J, Jaisser F. Vascular and inflammatory mineralocorticoid receptors in kidney disease. Acta Physiol (Oxf) 2020; 228:e13390. [PMID: 31529757 DOI: 10.1111/apha.13390] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2019] [Revised: 08/28/2019] [Accepted: 09/12/2019] [Indexed: 12/25/2022]
Abstract
Mineralocorticoid receptor (MR) activation in the kidney can occur outside the aldosterone-sensitive distal nephron in sites including the endothelium, smooth muscle and inflammatory cells. MR activation in these cells has deleterious effects on kidney structure and function by promoting oxidative injury, endothelial dysfunction and stiffness, vascular remodelling and calcification, decreased relaxation and activation of T cells and pro-inflammatory macrophages. Here, we review the data showing the cellular consequences of MR activation in endothelial, smooth muscle and inflammatory cells and how this affects the kidney in pathological situations. The evidence demonstrating a benefit of pharmacological or genetic MR inhibition in various models of kidney disease is also discussed.
Collapse
Affiliation(s)
- Jonatan Barrera‐Chimal
- Laboratorio de Fisiología Cardiovascular y Trasplante Renal Unidad de Investigación en Medicina Traslacional Universidad Nacional Autónoma de México and Instituto Nacional de Cardiología Ignacio Chávez Instituto de Investigaciones Biomédicas Mexico City Mexico
| | - Frederic Jaisser
- INSERM U1116 Clinical Investigation Centre Lorraine University Vandoeuvre‐lès‐Nancy France
- INI‐CRCT (Cardiovascular and Renal Clinical Trialists) F‐CRIN Network Nancy France
- INSERM UMRS 1138 Centre de Recherche des Cordeliers Sorbonne University Paris Descartes University Paris France
| |
Collapse
|
18
|
Barrera-Chimal J, Girerd S, Jaisser F. Mineralocorticoid receptor antagonists and kidney diseases: pathophysiological basis. Kidney Int 2019; 96:302-319. [PMID: 31133455 DOI: 10.1016/j.kint.2019.02.030] [Citation(s) in RCA: 150] [Impact Index Per Article: 25.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2018] [Revised: 02/04/2019] [Accepted: 02/20/2019] [Indexed: 12/13/2022]
Abstract
Chronic kidney disease (CKD) represents a global health concern, and its prevalence is increasing. The ultimate therapeutic option for CKD is kidney transplantation. However, the use of drugs that target specific pathways to delay or halt CKD progression, such as angiotensin-converting enzyme inhibitors, angiotensin receptor blockers, and sodium-glucose co-transporter-2 (SGLT-2) inhibitors is limited in clinical practice. Mineralocorticoid receptor activation in nonclassical tissues, such as the endothelium, smooth muscle cells, inflammatory cells, podocytes, and fibroblasts may have deleterious effects on kidney structure and function. Several preclinical studies have shown that mineralocorticoid receptor antagonists (MRAs) ameliorate or cure kidney injury and dysfunction in different models of kidney disease. In this review, we present the preclinical evidence showing a benefit of MRAs in acute kidney injury, the transition from acute kidney injury to CKD, hypertensive and diabetic nephropathy, glomerulonephritis, and kidney toxicity induced by calcineurin inhibitors. We also discuss the molecular mechanisms responsible for renoprotection related to MRAs that lead to reduced oxidative stress, inflammation, fibrosis, and hemodynamic alterations. The available clinical data support a benefit of MRA in reducing proteinuria in diabetic kidney disease and improving cardiovascular outcomes in CKD patients. Moreover, a benefit of MRAs in kidney transplantation has also been observed. The past and present clinical trials describing the effect of MRAs on kidney injury are presented, and the risk of hyperkalemia and use of other options, such as potassium binding agents or nonsteroidal MRAs, are also addressed. Altogether, the available preclinical and clinical data support a benefit of using MRAs in CKD, an approach that should be further explored in future clinical trials.
Collapse
Affiliation(s)
- Jonatan Barrera-Chimal
- Laboratorio de Fisiología Cardiovascular y Trasplante Renal, Unidad de Medicina Traslacional, Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México and Instituto Nacional de Cardiología Ignacio Chávez, Mexico City, Mexico
| | - Sophie Girerd
- Transplant Unit, Nephrology Department, Nancy University Hospital, Lorraine University, Vandoeuvre-lès-Nancy, France; Institut national de la santé et de la recherche médicale U1116, Clinical Investigation Centre, Lorraine University, Vandoeuvre-lès-Nancy, France; Investigation Network Initiative - Cardiovascular and Renal Clinical Trialists, French-Clinical Research Infrastructure Network, Nancy, France
| | - Frederic Jaisser
- Institut national de la santé et de la recherche médicale U1116, Clinical Investigation Centre, Lorraine University, Vandoeuvre-lès-Nancy, France; Investigation Network Initiative - Cardiovascular and Renal Clinical Trialists, French-Clinical Research Infrastructure Network, Nancy, France; Institut national de la santé et de la recherche médicale, UMRS 1138, Team 1, Centre de Recherche des Cordeliers, Sorbonne University, Paris Descartes University, Paris, France.
| |
Collapse
|
19
|
Wang T, Wang G, Zhang Y, Zhang J, Cao W, Chen X. Effect of lentivirus-mediated overexpression or silencing of MnSOD on apoptosis of resveratrol-treated fibroblast-like synoviocytes in rheumatoid arthritis. Eur J Pharmacol 2019; 844:65-72. [DOI: 10.1016/j.ejphar.2018.12.001] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2018] [Revised: 11/28/2018] [Accepted: 12/03/2018] [Indexed: 12/17/2022]
|
20
|
Abstract
PURPOSE OF REVIEW Type 2 diabetes (T2D) is associated with an increased risk of diabetic kidney disease (DKD), cardiovascular disease, and heart failure, in part through activation of the renin-angiotensin-aldosterone system (RAAS). Although recent cardiovascular outcome trials have identified newer therapeutic agents such as sodium-glucose cotransporter-2 (SGLT-2) inhibitors and glucagon-like peptide-1 (GLP-1)-receptor agonists that reduce the risk of these complications, patients still exhibit residual cardiorenal morbidity and mortality. Accordingly, the identification of pharmacological agents that attenuate micro- and macrovascular complications related to T2D is a major priority. Our aim was to review evidence for the role of novel mineralocorticoid receptor antagonists (MRAs) that are being developed as adjunctive therapies to reduce the risk of DKD and cardiovascular disease in the setting of T2D. RECENT FINDINGS Dual RAAS blockade with angiotensin-converting enzyme (ACE) inhibitor plus angiotensin receptor blockade (ARB) or ARB plus renin inhibition increases serious adverse events such as acute kidney injury and stroke. Due to the potential for these serious side effects, more recent interest has focused on newer, more selective non-steroidal MRAs such as finerenone as cardiorenal protective therapies. Finerenone reduces albuminuria in the setting of DKD in patients with T2D and has a lower risk of hyperkalemia compared to currently available MRAs. Novel MRAs such as finerenone have the potential to reduce the risk of DKD progression in patients with T2D. The impact of finerenone on hard, long-term cardiorenal endpoints is being examined in the FIGARO and FIDELIO trials in patients with DKD.
Collapse
Affiliation(s)
- Yuliya Lytvyn
- Toronto General Hospital Research Institute, UHN, 585 University Ave, 8N-845, Toronto, Ontario, M5G 2N2, Canada.
| | - Lucas C Godoy
- Peter Munk Cardiac Centre, University of Toronto, Toronto, Canada
- Instituto do Coracao (InCor), Faculdade de Medicina FMUSP, Universidade de Sao Paulo, Sao Paulo, SP, Brazil
| | - Rosalie A Scholtes
- Diabetes Center, Department of Internal Medicine, Amsterdam University Medical Center, location VU University Medical Center, Amsterdam, The Netherlands
| | - Daniël H van Raalte
- Diabetes Center, Department of Internal Medicine, Amsterdam University Medical Center, location VU University Medical Center, Amsterdam, The Netherlands
| | - David Z Cherney
- Department of Medicine, Division of Nephrology, University Health Network, University of Toronto, Toronto, Canada
- Banting and Best Diabetes Centre, Toronto, Canada
- Department of Physiology, University of Toronto, Toronto, Canada
| |
Collapse
|
21
|
|
22
|
Mochel JP, Teng CH, Peyrou M, Giraudel J, Danhof M, Rigel DF. Sacubitril/valsartan (LCZ696) significantly reduces aldosterone and increases cGMP circulating levels in a canine model of RAAS activation. Eur J Pharm Sci 2018; 128:103-111. [PMID: 30508581 DOI: 10.1016/j.ejps.2018.11.037] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2018] [Revised: 11/27/2018] [Accepted: 11/29/2018] [Indexed: 12/13/2022]
Abstract
Simultaneous blockade of angiotensin receptors and enhancement of natriuretic peptides (NP) by the first-in-class angiotensin receptor neprilysin (NEP) inhibitor sacubitril/valsartan constitutes an effective approach to treating heart failure. This study examined the effects of sacubitril/valsartan (225 and 675 mg/day) vs. placebo, sacubitril (360 mg/day), valsartan (900 mg/day), and benazepril (5 mg/day) on the dynamics of the renin-angiotensin-aldosterone system (RAAS) and the NP system in dogs. Beagle dogs (n = 18) were fed a low-salt diet (0.05% Na) for 15 days to model RAAS activation observed in clinical heart failure. Drugs were administered once daily during the last 10 days, while the effects on the RAAS and NPs were assessed on Day 1, 5, and 10. Steady-state pharmacokinetics of the test agents were evaluated on Day 5. Compared with placebo, sacubitril/valsartan (675 mg) substantially increased cGMP circulating levels, while benazepril and valsartan showed no effect. Additionally, sacubitril/valsartan (675 mg) and valsartan significantly increased plasma renin activity, angiotensin I and angiotensin II concentrations. Finally, sacubitril/valsartan (both doses), and valsartan significantly decreased plasma aldosterone vs. placebo. Systemic exposure to valsartan following sacubitril/valsartan 675 mg administration was similar to that observed with valsartan 900 mg administration alone. Sacubitril/valsartan favorably modulates the dynamics of the renin and NP cascades through complementary NEP and RAAS inhibition.
Collapse
Affiliation(s)
- Jonathan P Mochel
- Pharmacometrics, Novartis Pharma AG, Werk Saint Johann, 4056 Basel, Switzerland; Department of Biomedical Sciences, Iowa State University College of Veterinary Medicine, 1800 Christensen Drive, 50010 Ames, USA.
| | - Chi Hse Teng
- Biostatistics NIBR, Novartis Institutes for BioMedical Research, 250 Massachusetts Ave., Cambridge, MA 02139, USA
| | - Mathieu Peyrou
- Department of Research & Development, Elanco Animal Health, c/o Novartis Animal Health, 4002 Basel, Switzerland
| | - Jerome Giraudel
- Department of Research & Development, Elanco Animal Health, c/o Novartis Animal Health, 4002 Basel, Switzerland
| | - Meindert Danhof
- Department of Pharmacology, Leiden-Academic Centre for Drug Research, Pharmacology, 2300 Leiden, the Netherlands
| | - Dean F Rigel
- Novartis Institutes for BioMedical Research, East Hanover, NJ, USA
| |
Collapse
|
23
|
Yu BC, Lee MS, Moon JJ, Choi SJ, Kim JK, Hwang SD, Park MY. Efficacy of low-dose spironolactone on top of angiotensin receptor blockade in patients with glomerulonephritis. Kidney Res Clin Pract 2018; 37:257-265. [PMID: 30254850 PMCID: PMC6147182 DOI: 10.23876/j.krcp.2018.37.3.257] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2018] [Revised: 06/27/2018] [Accepted: 07/06/2018] [Indexed: 12/29/2022] Open
Abstract
Background Previous studies have shown that aldosterone antagonists have a proteinuria-lowering effect in patients with proteinuria and progressive proteinuric disease not adequately controlled by the use of angiotensin receptor blockers (ARBs). Aldosterone antagonists, in combination with ARBs, might improve proteinuria in patients with glomerulonephritis (GN). Methods In the present retrospective study, we evaluated the proteinuria-lowering effect and drug safety of low-dose spironolactone (12.5 mg/day) in 42 patients with GN being treated with an ARB. Results Proteinuria decreased from a mean total-protein-to-creatinine (TP/Cr) ratio of 592.3 ± 42.0 mg/g at baseline to 335.6 ± 43.3 mg/g after three months of treatment with spironolactone (P < 0.001). After the initial three months, the mean TP/Cr ratio increased progressively at six, nine, and 12 months; however, it was still less than the baseline value (P = 0.001, < 0.001, and < 0.001, respectively). Although serum Cr levels increased significantly at three and nine months compared with baseline (P = 0.036 and 0.026, respectively), there was no time effect of treatment (P = 0.071). Serum potassium levels tended to increase with time (P = 0.118), whereas systolic and diastolic blood pressures decreased with time (P = 0.122 and 0.044, respectively). Conclusion Low-dose spironolactone in combination with an ARB reduced proteinuria in patients with GN, which could represent a novel treatment option in individuals whose proteinuria is not optimally controlled by the use of ARBs alone.
Collapse
Affiliation(s)
- Byung Chul Yu
- Department of Internal Medicine, Soonchunhyang University Bucheon Hospital, Bucheon, Korea
| | - Min Sung Lee
- Department of Internal Medicine, Soonchunhyang University Bucheon Hospital, Bucheon, Korea
| | - Jong Joo Moon
- Department of Internal Medicine, Soonchunhyang University Bucheon Hospital, Bucheon, Korea
| | - Soo Jeong Choi
- Department of Internal Medicine, Soonchunhyang University Bucheon Hospital, Bucheon, Korea
| | - Jin Kuk Kim
- Department of Internal Medicine, Soonchunhyang University Bucheon Hospital, Bucheon, Korea
| | - Seung Duk Hwang
- Department of Internal Medicine, Soonchunhyang University Bucheon Hospital, Bucheon, Korea
| | - Moo Yong Park
- Department of Internal Medicine, Soonchunhyang University Bucheon Hospital, Bucheon, Korea
| |
Collapse
|
24
|
Aldosterone and Mineralocorticoid Receptor System in Cardiovascular Physiology and Pathophysiology. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2018; 2018:1204598. [PMID: 30327709 PMCID: PMC6169243 DOI: 10.1155/2018/1204598] [Citation(s) in RCA: 48] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/06/2018] [Revised: 08/14/2018] [Accepted: 08/16/2018] [Indexed: 12/22/2022]
Abstract
The mineralocorticoid hormone aldosterone (Aldo) has been intensively studied for its ability to influence both the physiology and pathophysiology of the cardiovascular system. Indeed, although research on Aldo actions for decades has mainly focused on its effects in the kidney, several lines of evidence have now demonstrated that this hormone exerts disparate extrarenal adverse effects, especially in the circulatory system. Accordingly, in the last lusters, a number of studies in preclinical models (in vitro and in vivo) and in humans have established that Aldo, following the interaction with its receptor-the mineralocorticoid receptor (MR)-is able to activate specific intracellular genomic and nongenomic pathways, thus regulating the homeostasis of the cardiovascular system. Importantly, through this mechanism of action, this hormone becomes a crucial regulator of the function and growth of different types of cells, including fibroblasts, cardiomyocytes, and vascular cells. For this main reason, it is plausible that when Aldo is present at high levels in the blood, it profoundly modifies the physiology of these cells, therefore being at the foundation of several cardiovascular disorders, such as heart failure (HF). On these grounds, in this review, we will provide an updated account on the current knowledge concerning Aldo activity in the cardiovascular system and the most recent preclinical studies and clinical trials designed to test better approaches able to counter the hyperactivity of the Aldo/MR signaling pathway in the setting of cardiovascular diseases.
Collapse
|
25
|
Sun WY, Bai B, Luo C, Yang K, Li D, Wu D, Félétou M, Villeneuve N, Zhou Y, Yang J, Xu A, Vanhoutte PM, Wang Y. Lipocalin-2 derived from adipose tissue mediates aldosterone-induced renal injury. JCI Insight 2018; 3:120196. [PMID: 30185654 DOI: 10.1172/jci.insight.120196] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2018] [Accepted: 07/24/2018] [Indexed: 12/14/2022] Open
Abstract
Lipocalin-2 is not only a sensitive biomarker, but it also contributes to the pathogenesis of renal injuries. The present study demonstrates that adipose tissue-derived lipocalin-2 plays a critical role in causing both chronic and acute renal injuries. Four-week treatment with aldosterone and high salt after uninephrectomy (ANS) significantly increased both circulating and urinary lipocalin-2, and it induced glomerular and tubular injuries in kidneys of WT mice. Despite increased renal expression of lcn2 and urinary excretion of lipocalin-2, mice with selective deletion of lcn2 alleles in adipose tissue (Adipo-LKO) are protected from ANS- or aldosterone-induced renal injuries. By contrast, selective deletion of lcn2 alleles in kidney did not prevent aldosterone- or ANS-induced renal injuries. Transplantation of fat pads from WT donors increased the sensitivity of mice with complete deletion of Lcn2 alleles (LKO) to aldosterone-induced renal injuries. Aldosterone promoted the urinary excretion of a human lipocalin-2 variant, R81E, in turn causing renal injuries in LKO mice. Chronic treatment with R81E triggered significant renal injuries in LKO, resembling those observed in WT mice following ANS challenge. Taken in conjunction, the present results demonstrate that lipocalin-2 derived from adipose tissue causes acute and chronic renal injuries, largely independent of local lcn2 expression in kidney.
Collapse
Affiliation(s)
- Wai Yan Sun
- The State Key Laboratory of Pharmaceutical Biotechnology and.,Department of Pharmacology and Pharmacy, The University of Hong Kong, Hong Kong SAR, China
| | - Bo Bai
- The State Key Laboratory of Pharmaceutical Biotechnology and.,Department of Pharmacology and Pharmacy, The University of Hong Kong, Hong Kong SAR, China
| | - Cuiting Luo
- The State Key Laboratory of Pharmaceutical Biotechnology and.,Department of Pharmacology and Pharmacy, The University of Hong Kong, Hong Kong SAR, China
| | - Kangmin Yang
- The State Key Laboratory of Pharmaceutical Biotechnology and.,Department of Pharmacology and Pharmacy, The University of Hong Kong, Hong Kong SAR, China
| | - Dahui Li
- The State Key Laboratory of Pharmaceutical Biotechnology and.,Department of Pharmacology and Pharmacy, The University of Hong Kong, Hong Kong SAR, China
| | - Donghai Wu
- Key Laboratory of Regenerative Biology, Guangzhou Institute of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, China
| | | | | | - Yang Zhou
- Center for Kidney Disease, Second Affiliated Hospital, Nanjing Medical University, China
| | - Junwei Yang
- Center for Kidney Disease, Second Affiliated Hospital, Nanjing Medical University, China
| | - Aimin Xu
- The State Key Laboratory of Pharmaceutical Biotechnology and.,Department of Pharmacology and Pharmacy, The University of Hong Kong, Hong Kong SAR, China
| | - Paul M Vanhoutte
- The State Key Laboratory of Pharmaceutical Biotechnology and.,Department of Pharmacology and Pharmacy, The University of Hong Kong, Hong Kong SAR, China
| | - Yu Wang
- The State Key Laboratory of Pharmaceutical Biotechnology and.,Department of Pharmacology and Pharmacy, The University of Hong Kong, Hong Kong SAR, China
| |
Collapse
|
26
|
Inampudi C, Alvarez P, Asleh R, Briasoulis A. Therapeutic Approach to Patients with Heart Failure with Reduced Ejection Fraction and End-stage Renal Disease. Curr Cardiol Rev 2018; 14:60-66. [PMID: 29366423 PMCID: PMC5872264 DOI: 10.2174/1573403x14666180123164916] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/01/2017] [Revised: 01/11/2018] [Accepted: 01/15/2018] [Indexed: 12/11/2022] Open
Abstract
BACKGROUND Several risk factors including Ischemic heart disease, uncontrolled hypertension, high output Heart Failure (HF) from shunting through vascular hemodialysis access, and anemia, contribute to development of HF in patients with End-Stage Renal Disease (ESRD). Guidelinedirected medical and device therapy for Heart Failure with Reduced Ejection Fraction (HFrEF) has not been extensively studied and may have limited safety and efficacy in patients with ESRD. RESULTS Maintenance of interdialytic and intradialytic euvolemia is a key component of HF management in these patients but often difficult to achieve. Beta-blockers, especially carvedilol which is poorly dialyzed is associated with cardiovascular benefit in this population. Despite paucity of data, Angiotensin-converting Enzyme Inhibitors (ACEI) or Angiotensin II Receptor Blockers (ARBs) when appropriately adjusted by dose and with close monitoring of serum potassium can also be administered to these patients who tolerate beta-blockers. Mineralocorticoid receptors in patients with HFrEF and ESRD have been shown to reduce mortality in a large randomized controlled trial without any significantly increased risk of hyperkalemia. Implantable Cardiac-defibrillators (ICDs) should be considered for primary prevention of sudden cardiac death in patients with HFrEF and ESRD who meet the implant indications. Furthermore in anemic iron-deficient patients, intravenous iron infusion may improve functional status. Finally, mechanical circulatory support with leftventricular assist devices may be related to increased mortality risk and the presence of ESRD poses a relative contraindication to further evaluation of these devices.
Collapse
Affiliation(s)
- Chakradhari Inampudi
- Division of Cardiovascular Diseases, Section of Heart Failure and Transplant, University of Iowa Hospitals and Clinics, Iowa City, IA, United States
| | - Paulino Alvarez
- Division of Cardiovascular Diseases, Section of Heart Failure and Transplant, University of Iowa Hospitals and Clinics, Iowa City, IA, United States
| | - Rabea Asleh
- Department of Cardiovascular Diseases, Mayo Clinic, Rochester MN, United States
| | - Alexandros Briasoulis
- Division of Cardiovascular Diseases, Section of Heart Failure and Transplant, University of Iowa Hospitals and Clinics, Iowa City, IA, United States
| |
Collapse
|
27
|
Cao W, Zhang J, Wang G, Lu J, Wang T, Chen X. Reducing-Autophagy Derived Mitochondrial Dysfunction during Resveratrol Promotes Fibroblast-Like Synovial Cell Apoptosis. Anat Rec (Hoboken) 2018; 301:1179-1188. [PMID: 29461680 DOI: 10.1002/ar.23798] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2017] [Revised: 12/01/2017] [Accepted: 12/06/2017] [Indexed: 12/14/2022]
Abstract
In rheumatoid arthritis patients, the fibroblast-like synovial cells (FLS) growth is not controlled normally, but is similar to the tumor cells proliferation in histology. Our previous studies have shown that resveratrol inhibits the proliferation of FLS and promotes FLS apoptosis. However, the molecular mechanisms involved in resveratrol-induced FLS apoptosis have not been determined yet. Here, we showed that the FLS cell viability (following pretreatment with 5 µM H2 O2 for 24 hr) exhibited better proliferation performance than at other concentrations via the CCK-8 assay. The cell apoptotic rate increased with the increasing concentration of resveratrol (0, 40, 80, 160, 320 μM), as detected by TdT-mediated dUTP nick-end labeling (TUNEL) staining and western blotting. Furthermore, the expression level of autophagy-related proteins (LC3A/B, ATG-5) decreased with the increased concentration of resveratrol, as determined by immunofluorescence and western blot analysis. We also showed that resveratrol induced FLS mitochondrial morphology change. Moreover, mitochondrial function detection showed that the mitochondrial membrane potential was lost with the increased concentration of resveratrol as examined by the JC-1 assay. The production of ATP in cells was positively and negatively correlated with the resveratrol concentration. Simultaneously, the intracellular calcium release and calcium influx decreased gradually with the increase in resveratrol concentration. Therefore, we proposed that resveratrol can reduce the level of autophagy in FLS. The decrease in the autophagy level can lead to the accumulation of reactive oxygen species, which may result in mitochondrial dysfunction and promotion of FLS apoptosis. Anat Rec, 301:1179-1188, 2018. © 2018 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Wei Cao
- Department of Histology and Embryology, Anhui Medical University, Hefei, 230032, China
| | - Junqiang Zhang
- Department of Histology and Embryology, Anhui Medical University, Hefei, 230032, China
| | - Gaoyuan Wang
- Department of Orthopaedic, the First Affiliated Hospital of Anhui Medical University, Hefei, 230031, China
| | - Jinsen Lu
- Department of Histology and Embryology, Anhui Medical University, Hefei, 230032, China
| | - Taorong Wang
- Department of Histology and Embryology, Anhui Medical University, Hefei, 230032, China
| | - Xiaoyu Chen
- Department of Histology and Embryology, Anhui Medical University, Hefei, 230032, China
| |
Collapse
|
28
|
Esposito D, Pasquali D, Johannsson G. Primary Adrenal Insufficiency: Managing Mineralocorticoid Replacement Therapy. J Clin Endocrinol Metab 2018; 103:376-387. [PMID: 29156052 DOI: 10.1210/jc.2017-01928] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/30/2017] [Accepted: 11/10/2017] [Indexed: 12/28/2022]
Abstract
CONTEXT Mineralocorticoid (MC) replacement therapy in patients with primary adrenal insufficiency (PAI) was introduced more than 60 years ago. Still, there are limited data on how MC substitution should be optimized, because MC dosing regimens have only been systematically investigated in a few studies. We review the management of current standard MC replacement therapy in PAI and its plausible impact on outcome. DESIGN Using PubMed, we conducted a systematic review of the literature from 1939 to 2017, with the following keywords: adrenal insufficiency, MC deficiency, aldosterone, cardiovascular disease, hypertension, and heart failure. RESULTS The current standard treatment consists of fludrocortisone (FC) given once daily in the morning, aiming at normotension, normokalemia, and plasma renin activity in the upper normal range. Available data suggest that patients with PAI may be underreplaced with FC as symptoms and signs indicating chronic MC underreplacement, such as salt craving and postural dizziness persist, in many treated patients with PAI. Data acquired from large registry-based studies show that glucocorticoid doses for replacement in PAI are higher than those estimated from endogenous production. Glucocorticoid overreplacement may reduce the need of MC replacement but may also be a consequence of inadequate MC replacement. CONCLUSIONS The commonly used MC replacement in PAI may not be adequate in some patients. Insufficient MC substitution may be responsible for poor cardiometabolic outcome and the failure to restore well-being adequately in patients with PAI. Well-designed studies oriented at optimizing MC replacement therapy are urgently needed.
Collapse
Affiliation(s)
- Daniela Esposito
- Department of Internal Medicine and Clinical Nutrition, Institute of Medicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
- Department of Endocrinology, Sahlgrenska University Hospital, Gothenburg, Sweden
- Department of Medical, Surgical, Neurological, Metabolic Sciences, and Aging, University of Campania "Luigi Vanvitelli," Naples, Italy
| | - Daniela Pasquali
- Department of Medical, Surgical, Neurological, Metabolic Sciences, and Aging, University of Campania "Luigi Vanvitelli," Naples, Italy
| | - Gudmundur Johannsson
- Department of Internal Medicine and Clinical Nutrition, Institute of Medicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
- Department of Endocrinology, Sahlgrenska University Hospital, Gothenburg, Sweden
| |
Collapse
|
29
|
Medeiros M, Velásquez-Jones L, Hernández AM, Ramón-García G, Valverde S, Fuentes Y, Vargas A, Patiño M, Pérez-Villalva R, Ortega-Trejo JA, Barrera-Chimal J, Bobadilla NA. Randomized Controlled Trial of Mineralocorticoid Receptor Blockade in Children with Chronic Kidney Allograft Nephropathy. Clin J Am Soc Nephrol 2017; 12:1291-1300. [PMID: 28536123 PMCID: PMC5544507 DOI: 10.2215/cjn.05300516] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2016] [Accepted: 04/19/2017] [Indexed: 01/05/2023]
Abstract
BACKGROUND AND OBJECTIVES We showed that mineralocorticoid receptor blockade (MRB) prevented acute and chronic cyclosporine nephropathy (CsA-Nx) in the rat. The aim of this translational study was to investigate the effect of long-term eplerenone administration on renal allograft function in children with biopsy-proven chronic allograft nephropathy (CAN). DESIGN, SETTING, PARTICIPANTS, & MEASUREMENTS Renal transplant children <18 years, biopsy-proven CAN, and a GFR>40 ml/min per 1.73 m2 were included. Patients with BK virus active nephritis, recurrence of renal disease, GFR decline in previous 3 months, or treated with calcium antagonists or antifungal drugs were excluded. They were randomized to receive placebo (n=10) or eplerenone 25 mg/d for 24 months (n=13). Visits were scheduled at baseline, 6, 12, and 24 months. At each period, a complete clinical examination was performed and blood and urine samples were taken. Urine creatinine, 8-hydroxylated-guanosine, heat shock protein 72 (HSP72), and kidney injury molecule (KIM-1) levels were also assessed. In kidney biopsy samples, the tubulo-interstitial area affected by fibrosis (TIF) and glomerulosclerosis were measured at baseline and after 24 months. RESULTS The baseline eGFR was 80±6 in the placebo and 86±6 ml/min per 1.73 m2 in the eplerenone group; at 24 months it was 66±8 and 81±7 ml/min per 1.73 m2, respectively (P=0.33; 95% confidence intervals, -18 to 33 at baseline, and -11 to 40 after 24 months). The albumin-to-creatinine ratio was 110±74 in the placebo, and 265±140 mg/g in the eplerenone group; and after 24 months it was 276±140 and 228±88 mg/g, respectively (P=0.15; 95% confidence intervals, -283 to 593, and -485 to 391, respectively). In addition, the placebo exhibited a greater TIF, glomerulosclerosis, and urinary HSP72 compared with the eplerenone group. CONCLUSIONS Although this study was underpowered to provide definitive evidence that long-term eplerenone administration attenuates the progression of CAN in pediatric transplant patients, it encourages testing the potential benefit of MRB in this pediatric population.
Collapse
Affiliation(s)
- Mara Medeiros
- Nephrology and Mineral Metabolism Research Unit
- Department of Nephrology, and
| | | | | | | | | | | | | | | | - Rosalba Pérez-Villalva
- Molecular Physiology Unit, Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México, Mexico City, Mexico; and
- Department of Nephrology, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Mexico City, Mexico
| | - Juan Antonio Ortega-Trejo
- Molecular Physiology Unit, Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México, Mexico City, Mexico; and
- Department of Nephrology, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Mexico City, Mexico
| | - Jonatan Barrera-Chimal
- Molecular Physiology Unit, Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México, Mexico City, Mexico; and
- Department of Nephrology, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Mexico City, Mexico
| | - Norma A. Bobadilla
- Molecular Physiology Unit, Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México, Mexico City, Mexico; and
- Department of Nephrology, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Mexico City, Mexico
| |
Collapse
|
30
|
Salt-dependent Blood Pressure in Human Aldosterone Synthase-Transgenic Mice. Sci Rep 2017; 7:492. [PMID: 28352088 PMCID: PMC5412599 DOI: 10.1038/s41598-017-00461-9] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2016] [Accepted: 02/28/2017] [Indexed: 01/19/2023] Open
Abstract
Hypertension is one of the most important, preventable causes of premature morbidity and mortality in the developed world. Aldosterone is a major mineralocorticoid hormone that plays a key role in the regulation of blood pressure and is implicated in the pathogenesis of hypertension and heart failure. Aldosterone synthase (AS, cytochrome P450 11B2, cyp11B2) is the sole enzyme responsible for the production of aldosterone in humans. To determine the effects of increased expression of human aldosterone synthase (hAS) on blood pressure (BP), we established transgenic mice carrying the hAS gene (cyp11B2). We showed that hAS overexpression increased levels of aldosterone in hAS+/- mice. On high salt diet (HS), BPs of hAS+/- mice were significantly increased compared with WT mice. Fadrozole (an inhibitor of aldosterone synthase) treatment significantly reduced BPs of hAS+/- mice on HS. This is the first time overexpression of AS in a transgenic mouse line has shown an ability to induce HP. Specifically inhibiting AS activity in these mice is a promising therapy for reducing hypertension. This hAS transgenic mouse model is therefore an ideal animal model for hypertension therapy studies.
Collapse
|
31
|
Lattenist L, Lechner SM, Messaoudi S, Le Mercier A, El Moghrabi S, Prince S, Bobadilla NA, Kolkhof P, Jaisser F, Barrera-Chimal J. Nonsteroidal Mineralocorticoid Receptor Antagonist Finerenone Protects Against Acute Kidney Injury-Mediated Chronic Kidney Disease: Role of Oxidative Stress. Hypertension 2017; 69:870-878. [PMID: 28320854 DOI: 10.1161/hypertensionaha.116.08526] [Citation(s) in RCA: 91] [Impact Index Per Article: 11.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2016] [Revised: 10/30/2016] [Accepted: 02/16/2017] [Indexed: 02/06/2023]
Abstract
Acute kidney injury induced by ischemia/reperfusion (IR) is a frequent complication in hospitalized patients. Mineralocorticoid receptor antagonism has shown to be helpful against renal IR consequences; however, the potential benefit of novel nonsteroidal mineralocorticoid receptor antagonists such as finerenone has to be further explored. In this study, we evaluated the efficacy of finerenone to prevent the acute and chronic consequences of ischemic acute kidney injury. For the acute study (24 hours), 18 rats were divided into sham, bilateral renal ischemia of 25 minutes, and rats that received 3 doses of finerenone at 48, 24, and 1 hour before the ischemia. For the chronic study (4 months), 23 rats were divided into sham, rats that underwent 45 minutes of bilateral ischemia, and rats treated with finerenone at days 2 and 1 and 1 hour before IR. We found that after 24 hours of reperfusion, the untreated IR rats presented kidney dysfunction and tubular injury. Kidney injury molecule-1 and neutrophil gelatinase associated to lipolacin mRNA levels were increased. In contrast, the rats treated with finerenone displayed normal kidney function and significantly lesser tubular injury and kidney injury molecule-1 and neutrophil gelatinase associated to lipolacin levels. After 4 months, the IR rats developed chronic kidney disease, evidenced by kidney dysfunction, increased proteinuria and renal vascular resistance, tubular dilation, extensive tubule-interstitial fibrosis, and an increase in kidney transforming growth factor-β and collagen-I mRNA. The transition from acute kidney injury to chronic kidney disease was fully prevented by finerenone. Altogether, our data show that in the rat, finerenone is able to prevent acute kidney injury induced by IR and the chronic and progressive deterioration of kidney function and structure.
Collapse
Affiliation(s)
- Lionel Lattenist
- From the INSERM, UMRS 1138, Team 1, Centre de Recherche des Cordeliers, Pierre et Marie Curie University, Paris Descartes University, France (L.L., S.M.L., S.M., A.L.M., S.E.M., S.P., F.J., J.B.-C.); Molecular Physiology Unit, Instituto de Investigaciones Biomedicas, Universidad Nacional Autónoma de México and Nephrology Department, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Mexico City (N.A.B., J.B.-C.); BAYER AG, Cardiology Research, Wuppertal, Germany (P.K.); and INSERM, CIC1433, CHRU de Nancy, F-CRIN INI-CRCT Network, France (F.J.)
| | - Sebastian M Lechner
- From the INSERM, UMRS 1138, Team 1, Centre de Recherche des Cordeliers, Pierre et Marie Curie University, Paris Descartes University, France (L.L., S.M.L., S.M., A.L.M., S.E.M., S.P., F.J., J.B.-C.); Molecular Physiology Unit, Instituto de Investigaciones Biomedicas, Universidad Nacional Autónoma de México and Nephrology Department, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Mexico City (N.A.B., J.B.-C.); BAYER AG, Cardiology Research, Wuppertal, Germany (P.K.); and INSERM, CIC1433, CHRU de Nancy, F-CRIN INI-CRCT Network, France (F.J.)
| | - Smail Messaoudi
- From the INSERM, UMRS 1138, Team 1, Centre de Recherche des Cordeliers, Pierre et Marie Curie University, Paris Descartes University, France (L.L., S.M.L., S.M., A.L.M., S.E.M., S.P., F.J., J.B.-C.); Molecular Physiology Unit, Instituto de Investigaciones Biomedicas, Universidad Nacional Autónoma de México and Nephrology Department, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Mexico City (N.A.B., J.B.-C.); BAYER AG, Cardiology Research, Wuppertal, Germany (P.K.); and INSERM, CIC1433, CHRU de Nancy, F-CRIN INI-CRCT Network, France (F.J.)
| | - Alan Le Mercier
- From the INSERM, UMRS 1138, Team 1, Centre de Recherche des Cordeliers, Pierre et Marie Curie University, Paris Descartes University, France (L.L., S.M.L., S.M., A.L.M., S.E.M., S.P., F.J., J.B.-C.); Molecular Physiology Unit, Instituto de Investigaciones Biomedicas, Universidad Nacional Autónoma de México and Nephrology Department, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Mexico City (N.A.B., J.B.-C.); BAYER AG, Cardiology Research, Wuppertal, Germany (P.K.); and INSERM, CIC1433, CHRU de Nancy, F-CRIN INI-CRCT Network, France (F.J.)
| | - Soumaya El Moghrabi
- From the INSERM, UMRS 1138, Team 1, Centre de Recherche des Cordeliers, Pierre et Marie Curie University, Paris Descartes University, France (L.L., S.M.L., S.M., A.L.M., S.E.M., S.P., F.J., J.B.-C.); Molecular Physiology Unit, Instituto de Investigaciones Biomedicas, Universidad Nacional Autónoma de México and Nephrology Department, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Mexico City (N.A.B., J.B.-C.); BAYER AG, Cardiology Research, Wuppertal, Germany (P.K.); and INSERM, CIC1433, CHRU de Nancy, F-CRIN INI-CRCT Network, France (F.J.)
| | - Sonia Prince
- From the INSERM, UMRS 1138, Team 1, Centre de Recherche des Cordeliers, Pierre et Marie Curie University, Paris Descartes University, France (L.L., S.M.L., S.M., A.L.M., S.E.M., S.P., F.J., J.B.-C.); Molecular Physiology Unit, Instituto de Investigaciones Biomedicas, Universidad Nacional Autónoma de México and Nephrology Department, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Mexico City (N.A.B., J.B.-C.); BAYER AG, Cardiology Research, Wuppertal, Germany (P.K.); and INSERM, CIC1433, CHRU de Nancy, F-CRIN INI-CRCT Network, France (F.J.)
| | - Norma A Bobadilla
- From the INSERM, UMRS 1138, Team 1, Centre de Recherche des Cordeliers, Pierre et Marie Curie University, Paris Descartes University, France (L.L., S.M.L., S.M., A.L.M., S.E.M., S.P., F.J., J.B.-C.); Molecular Physiology Unit, Instituto de Investigaciones Biomedicas, Universidad Nacional Autónoma de México and Nephrology Department, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Mexico City (N.A.B., J.B.-C.); BAYER AG, Cardiology Research, Wuppertal, Germany (P.K.); and INSERM, CIC1433, CHRU de Nancy, F-CRIN INI-CRCT Network, France (F.J.)
| | - Peter Kolkhof
- From the INSERM, UMRS 1138, Team 1, Centre de Recherche des Cordeliers, Pierre et Marie Curie University, Paris Descartes University, France (L.L., S.M.L., S.M., A.L.M., S.E.M., S.P., F.J., J.B.-C.); Molecular Physiology Unit, Instituto de Investigaciones Biomedicas, Universidad Nacional Autónoma de México and Nephrology Department, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Mexico City (N.A.B., J.B.-C.); BAYER AG, Cardiology Research, Wuppertal, Germany (P.K.); and INSERM, CIC1433, CHRU de Nancy, F-CRIN INI-CRCT Network, France (F.J.)
| | - Frédéric Jaisser
- From the INSERM, UMRS 1138, Team 1, Centre de Recherche des Cordeliers, Pierre et Marie Curie University, Paris Descartes University, France (L.L., S.M.L., S.M., A.L.M., S.E.M., S.P., F.J., J.B.-C.); Molecular Physiology Unit, Instituto de Investigaciones Biomedicas, Universidad Nacional Autónoma de México and Nephrology Department, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Mexico City (N.A.B., J.B.-C.); BAYER AG, Cardiology Research, Wuppertal, Germany (P.K.); and INSERM, CIC1433, CHRU de Nancy, F-CRIN INI-CRCT Network, France (F.J.).
| | - Jonatan Barrera-Chimal
- From the INSERM, UMRS 1138, Team 1, Centre de Recherche des Cordeliers, Pierre et Marie Curie University, Paris Descartes University, France (L.L., S.M.L., S.M., A.L.M., S.E.M., S.P., F.J., J.B.-C.); Molecular Physiology Unit, Instituto de Investigaciones Biomedicas, Universidad Nacional Autónoma de México and Nephrology Department, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Mexico City (N.A.B., J.B.-C.); BAYER AG, Cardiology Research, Wuppertal, Germany (P.K.); and INSERM, CIC1433, CHRU de Nancy, F-CRIN INI-CRCT Network, France (F.J.)
| |
Collapse
|
32
|
Park SM, Jung WJ, Park JM, Rhee H, Kim IY, Seong EY, Lee DW, Lee SB, Kwak IS, Shin N, Song SH. Unmasked chronic renal function deterioration after unilateral adrenalectomy in patients with primary aldosteronism. Kidney Res Clin Pract 2016; 35:255-258. [PMID: 27957422 PMCID: PMC5142303 DOI: 10.1016/j.krcp.2016.05.002] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2016] [Revised: 05/02/2016] [Accepted: 05/10/2016] [Indexed: 12/05/2022] Open
Abstract
We report 2 cases of chronic estimated glomerular filtration rate (eGFR) decline after unilateral adrenalectomy due to primary aldosteronism. The patients were diagnosed with unilateral adrenal cortical adenoma releasing aldosterone. Two patients were examined for hypertension and hypokalemia. Unilateral laparoscopic adrenalectomy was performed in both cases, and pathology confirmed adrenal cortical adenoma. After adrenalectomy, hypertension and hypokalemia improved to within normal range. However, the eGFR decreased postoperatively, and abdominal computed tomography scan showed decreased kidney size compared to previous images. Kidney biopsy was performed to delineate the exact cause of renal function deterioration and revealed hypertensive changes with chronic interstitial changes, indicating that glomerular hyperfiltration with aldosterone excess masked renal function damage. Physicians have to consider the probability of postadrenalectomy eGFR decline related to chronic hypertensive change.
Collapse
Affiliation(s)
- Su Min Park
- Department of Internal Medicine, Pusan National University School of Medicine, Busan, Korea
| | - Woo Jin Jung
- Department of Internal Medicine, Pusan National University School of Medicine, Busan, Korea
| | - Jong Man Park
- Department of Internal Medicine, Pusan National University School of Medicine, Busan, Korea
| | - Harin Rhee
- Department of Internal Medicine, Pusan National University School of Medicine, Busan, Korea
| | - Il Young Kim
- Department of Internal Medicine, Pusan National University School of Medicine, Busan, Korea
| | - Eun Young Seong
- Department of Internal Medicine, Pusan National University School of Medicine, Busan, Korea
| | - Dong Won Lee
- Department of Internal Medicine, Pusan National University School of Medicine, Busan, Korea
| | - Soo Bong Lee
- Department of Internal Medicine, Pusan National University School of Medicine, Busan, Korea
| | - Ihm Soo Kwak
- Department of Internal Medicine, Pusan National University School of Medicine, Busan, Korea
| | - Nari Shin
- Department of Pathology, Pusan National University School of Medicine, Busan, Korea
| | - Sang Heon Song
- Department of Internal Medicine, Pusan National University School of Medicine, Busan, Korea
| |
Collapse
|
33
|
Singh A, Pezeshki A, Zapata RC, Yee NJ, Knight CG, Tuor UI, Chelikani PK. Diets enriched in whey or casein improve energy balance and prevent morbidity and renal damage in salt-loaded and high-fat-fed spontaneously hypertensive stroke-prone rats. J Nutr Biochem 2016; 37:47-59. [DOI: 10.1016/j.jnutbio.2016.07.011] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2016] [Revised: 06/03/2016] [Accepted: 07/15/2016] [Indexed: 01/11/2023]
|
34
|
Pitt B, Stier CT, Rajagopalan S. Mineralocorticoid receptor blockade: new insights into the mechanism of action in patients with cardiovascular disease. J Renin Angiotensin Aldosterone Syst 2016; 4:164-8. [PMID: 14608520 DOI: 10.3317/jraas.2003.025] [Citation(s) in RCA: 42] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
Mineralocorticoid receptor (MR) blockade is effective in reducing total mortality and the incidence of heart failure in patients with systolic left ventricular dysfunction (SLVD) associated with chronic heart failure or post myocardial infarction. Pre-clinical and clinical studies in SLVD have shown that MR blockade reduces sudden cardiac death, left ventricular remodelling, left ventricular hypertrophy, endothelial dysfunction, autonomic imbalance, renal dysfunction and improves fibrinolysis. While MR blockade promotes sodium excretion and the combination of an angiotensin-converting enzyme inhibitor and a MR blocker have been shown to be more effective than either alone in causing natriuresis, it is unlikely that their beneficial effects can be explained solely on this basis. Aldosterone has been shown to have a number of adverse effects, including activation of other neurohumeral mediators, stimulation of active reactive oxygen species (ROS), activation of the NF-κβ and AP-1 signalling pathways, vascular inflammation and fibrosis, myocardial hypertrophy, autonomic imbalance, and a decrease in fibrinolysis. MR blockade is, however, effective both in situations with and without an increase in serum aldosterone level, since the MR can be occupied and activated by cortisol as well as by aldosterone. In view of these mechanisms, MR blockade may play an important role not only on SLVD, but also in essential hypertension with normal systolic function, diastolic heart failure, valvular heart disease, vascular stiffening with ageing, progression of renal disease, and diabetes mellitus. This hypothesis will, however, require further prospective evaluation.
Collapse
Affiliation(s)
- Bertram Pitt
- Division of Cardiology, University of Michigan, USA.
| | | | | |
Collapse
|
35
|
Abstract
OBJECTIVE: To review the pharmacology, pharmacokinetics, clinical efficacy, and safety of eplerenone, a new selective aldosterone blocker. DATA SOURCES: Primary literature and review articles were obtained via MEDLINE search (1966–April 2002). Additional studies and abstracts were identified from the bibliographies of reviewed literature. STUDY SELECTION AND DATA EXTRACTION: Studies and review articles related to eplerenone, aldosterone, aldosterone antagonist, and spironolactone were reviewed. Data pertinent to this article were included. DATA SYNTHESIS: Eplerenone is a selective aldosterone blocker. Recent data have demonstrated the deleterious effects of aldosterone in several chronic disease states including hypertension and heart failure. Animal studies using eplerenone have shown a positive role for aldosterone antagonism in the treatment of hypertension, heart failure, myocardial infarction, renal disease, and atherosclerosis. In humans, eplerenone appears to be effective for the treatment of hypertension. An ongoing study will examine the effect of eplerenone for heart failure. To date, the incidence of adverse effects with eplerenone has been slightly lower than with spironolactone. CONCLUSIONS: Eplerenone appears to be a promising drug in a new class of agents called selective aldosterone blockers. The drug may be approved for treatment of hypertension in 2002. Additional studies are ongoing that may provide information on other clinical uses for this medication.
Collapse
Affiliation(s)
- Alan J Zillich
- Division of Clinical and Administrative Pharmacy and Department of Family Medicine, College of Pharmacy, University of Iowa, Iowa City, IA 52242, USA.
| | | |
Collapse
|
36
|
Local and systemic renin–angiotensin system participates in cardiopulmonary–renal interactions in monocrotaline-induced pulmonary hypertension in the rat. Mol Cell Biochem 2016; 418:147-57. [DOI: 10.1007/s11010-016-2740-z] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2016] [Accepted: 06/15/2016] [Indexed: 10/21/2022]
|
37
|
Shi H, Zhang A, He Y, Yang M, Gan W. Effects of p53 on aldosterone-induced mesangial cell apoptosis in vivo and in vitro. Mol Med Rep 2016; 13:5102-8. [PMID: 27109859 PMCID: PMC4878551 DOI: 10.3892/mmr.2016.5156] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2015] [Accepted: 03/29/2016] [Indexed: 12/26/2022] Open
Abstract
Aldosterone (ALD) is a well‑known hormone, which may initiate renal injury by inducing mesangial cell (MC) injury in chronic kidney disease (CKD); however, the molecular mechanism remains unknown. The aim of the present study was to investigate the effects of p53 on ALD‑induced MC apoptosis and elucidate the underlying molecular mechanism. For the in vivo studies, rats were randomly assigned to receive normal saline or ALD for 4 weeks. The ratio of MC apoptosis was analysed by terminal deoxynucleotidyl transferase dUTP nick end labelling (TUNEL) assay. In addition, the expression level and localisation of p53, a well-known cell apoptosis-associated key protein, were detected by immunofluorescence. For the in vitro studies, rat MCs were incubated in medium containing either buffer (control) or ALD (10‑6 M) for 24 h. The cell apoptosis ratio was assessed by flow cytometry, and the expression level of p53 was assessed by reverse transcription quantitative polymerase chain reaction and western blotting. In order to confirm the role of p53 in ALD‑regulated cell apoptosis, a rescue experiment was performed using targeted small interfering (si)RNA to downregulate the expression of p53. The ALD‑treated rats exhibited greater numbers of TUNEL‑positive MCs and higher expression levels of p53 when compared with the control group. Furthermore, the ratio of MC apoptosis and the p53 expression level were significantly increased following ALD exposure, compared with the control group. Additionally, in the rescue experiment, the effects of ALD on MC were blocked by downregulating the expression level of p53 in MCs. The present study hypothesized that ALD may directly contribute to the occurrence of MC apoptosis via p53, which may participate in ALD-induced renal injury.
Collapse
Affiliation(s)
- Huimin Shi
- Department of Pediatric Nephrology, The Second Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu 210011, P.R. China
| | - Aiqing Zhang
- Department of Pediatric Nephrology, The Second Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu 210011, P.R. China
| | - Yanfang He
- Department of Pediatric Nephrology, The Second Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu 210011, P.R. China
| | - Min Yang
- Department of Nephrology, The Third Affiliated Hospital of Soochow University, Changzhou, Jiangsu 200040, P.R. China
| | - Weihua Gan
- Department of Pediatric Nephrology, The Second Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu 210011, P.R. China
| |
Collapse
|
38
|
Affiliation(s)
- Richard J Auchus
- Division of Metabolism, Diabetes, and Endocrinology, Department of Internal Medicine, Department of Pharmacology, University of Michigan School of Medicine, Ann Arbor, Michigan 48019
| |
Collapse
|
39
|
Sakamoto T, Fujii A, Saito N, Kondo H, Ohuchi A. Alteration of amiloride-sensitive salt taste nerve responses in aldosterone/NaCl-induced hypertensive rats. Neurosci Res 2016; 108:60-6. [PMID: 26828852 DOI: 10.1016/j.neures.2016.01.007] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2015] [Revised: 01/21/2016] [Accepted: 01/21/2016] [Indexed: 01/23/2023]
Abstract
Salt taste sensitivity is related to physiological condition, and declined in hypertensive patients. However, little is known about the mechanism underlying changes in salt taste sensitivity during the development of hypertension. This is largely due to lack of an appropriate animal model which shows the decline of salt taste sensitivity caused by hypertension. Previous studies have suggested that one of main causes of salt-sensitive hypertension is dysfunction of the renin-angiotensin-aldosterone system (RAAS). To examine the involvement of RAAS in modulation of salt taste sensitivity, we utilized aldosterone/NaCl-treated rats as a well-established model of salt-sensitive hypertension caused by RAAS dysfunction. Amount of sodium intake in aldosterone/NaCl-treated rats was higher than that in control rats. In addition to behavioral changes, the amiloride-sensitive salt taste nerve responses in aldosterone/NaCl-treated rats were remarkably lower by approximately 90% than those in the other groups. Moreover, αENaC mRNA expression in the epithelium of circumvallate papillae was significantly low in aldosterone/NaCl-treated rats. Thus, RAAS modulates salt taste system as is case in hypertensive patients. This report is to our knowledge the first to describe an animal model with decline of amiloride-sensitive salt taste nerve responses by RAAS dysfunction-mediated salt-sensitive hypertension.
Collapse
Affiliation(s)
- Takashi Sakamoto
- Kansei Science Laboratories, Kao Corporation, 2606 Akabane, Ichikaimachi, Haga, Tochigi 321-3497, Japan
| | - Akihiko Fujii
- Biological Science Laboratories, Kao Corporation, 2606 Akabane, Ichikaimachi, Haga, Tochigi 321-3497, Japan
| | - Naoko Saito
- Kansei Science Laboratories, Kao Corporation, 2606 Akabane, Ichikaimachi, Haga, Tochigi 321-3497, Japan
| | - Hidehiko Kondo
- Kansei Science Laboratories, Kao Corporation, 2606 Akabane, Ichikaimachi, Haga, Tochigi 321-3497, Japan
| | - Atsushi Ohuchi
- Kansei Science Laboratories, Kao Corporation, 2606 Akabane, Ichikaimachi, Haga, Tochigi 321-3497, Japan.
| |
Collapse
|
40
|
Bai M, Che R, Zhang Y, Yuan Y, Zhu C, Ding G, Jia Z, Huang S, Zhang A. Reactive oxygen species-initiated autophagy opposes aldosterone-induced podocyte injury. Am J Physiol Renal Physiol 2016; 310:F669-F678. [PMID: 26764202 DOI: 10.1152/ajprenal.00409.2015] [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: 09/08/2015] [Accepted: 01/12/2016] [Indexed: 12/28/2022] Open
Abstract
Evidence has demonstrated that aldosterone (Aldo) is involved in the development and progression of chronic kidney diseases. The purpose of the present study was to investigate the role of autophagy in Aldo-induced podocyte damage and the underlying mechanism. Mouse podocytes were treated with Aldo in the presence or absence of 3-methyladenine and N-acetylcysteine. Cell apoptosis was investigated by detecting annexin V conjugates, apoptotic bodies, caspase-3 activity, and alterations of the podocyte protein nephrin. Autophagy was evaluated by measuring the expressions of light chain 3, p62, beclin-1, and autophagy-related gene 5. Aldo (10-7 mol/l) induced podocyte apoptosis, autophagy, and downregulation of nephrin protein in a time-dependent manner. Aldo-induced apoptosis was further promoted by the inhibition of autophagy via 3-methyladenine and autophagy-related gene 5 small interfering RNA pretreatment. Moreover, Aldo time dependently increased ROS generation, and H2O2 (10-4 mol/l) application remarkably elevated podocyte autophagy. After treatment with N-acetylcysteine, the autophagy induced by Aldo or H2O2 was markedly attenuated, suggesting a key role of ROS in mediating autophagy formation in podocytes. Inhibition of ROS could also lessen Aldo-induced podocyte injury. Taken together, our findings suggest that ROS-triggered autophagy played a protective role against Aldo-induced podocyte injury, and targeting autophagy in podocytes may represent a new therapeutic strategy for the treatment of podocytopathy.
Collapse
Affiliation(s)
- Mi Bai
- Department of Nephrology, Nanjing Children's Hospital, Nanjing Medical University, Nanjing, China.,Institute of Pediatrics, Nanjing Medical University, Nanjing, China; and
| | - Ruochen Che
- Department of Nephrology, Nanjing Children's Hospital, Nanjing Medical University, Nanjing, China.,Institute of Pediatrics, Nanjing Medical University, Nanjing, China; and
| | - Yue Zhang
- Department of Nephrology, Nanjing Children's Hospital, Nanjing Medical University, Nanjing, China.,Institute of Pediatrics, Nanjing Medical University, Nanjing, China; and
| | - Yanggang Yuan
- Department of Nephrology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Chunhua Zhu
- Department of Nephrology, Nanjing Children's Hospital, Nanjing Medical University, Nanjing, China.,Institute of Pediatrics, Nanjing Medical University, Nanjing, China; and
| | - Guixia Ding
- Department of Nephrology, Nanjing Children's Hospital, Nanjing Medical University, Nanjing, China.,Institute of Pediatrics, Nanjing Medical University, Nanjing, China; and
| | - Zhanjun Jia
- Department of Nephrology, Nanjing Children's Hospital, Nanjing Medical University, Nanjing, China.,Institute of Pediatrics, Nanjing Medical University, Nanjing, China; and
| | - Songming Huang
- Department of Nephrology, Nanjing Children's Hospital, Nanjing Medical University, Nanjing, China.,Institute of Pediatrics, Nanjing Medical University, Nanjing, China; and
| | - Aihua Zhang
- Department of Nephrology, Nanjing Children's Hospital, Nanjing Medical University, Nanjing, China; .,Institute of Pediatrics, Nanjing Medical University, Nanjing, China; and
| |
Collapse
|
41
|
The renal effects of mineralocorticoid receptor antagonists. Int J Cardiol 2015; 200:20-4. [DOI: 10.1016/j.ijcard.2015.05.125] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/04/2014] [Accepted: 05/17/2015] [Indexed: 01/25/2023]
|
42
|
Ding W, Xu C, Wang B, Zhang M. Rotenone Attenuates Renal Injury in Aldosterone-Infused Rats by Inhibiting Oxidative Stress, Mitochondrial Dysfunction, and Inflammasome Activation. Med Sci Monit 2015; 21:3136-43. [PMID: 26474533 PMCID: PMC4614375 DOI: 10.12659/msm.895945] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022] Open
Abstract
BACKGROUND Reactive oxygen species (ROS) and inflammation both contribute to the progression of aldosterone-induced renal injury. To better understand the underlying mechanisms, we examined mitochondrial dysfunction and NLRP3 inflammasome activation in aldosterone-infused rats, and explored the role of rotenone in attenuating these injuries. MATERIAL AND METHODS Sprague-Dawley rats were divided into 3 groups: vehicle-treated, aldosterone-infused, and aldosterone plus rotenone. Renal damage was evaluated using PAS staining and electron microscopy. Levels of ROS were measured from renal tissue and serum; immunohistochemistry analysis examined the inflammation pathway; Western blot and real-time PCR assessed NLRP3 inflammasome activity. RESULTS Glomerular segmental sclerosis, foot process effacement, and proteinuria were demonstrated in the aldosterone-infused rats. Specifically, the thiobarbituric acid-reactive substances (TBARS) oxidative stress marker, MDA, was significantly increased; ATP content and mtDNA copy number were markedly decreased; inflammatory mediators NF-κB p65 and CTGF were upregulated; and NLRP3 inflammasome and its related target proteins, IL-1β and IL-18, were also increased. Treatment with rotenone, an inhibitor of mitochondrial complex I, significantly attenuated oxidative stress, mitochondrial dysfunction, and inflammasome response in aldosterone-infused rats. CONCLUSIONS Rotenone ameliorated aldosterone-infused renal injury, possibly by inhibiting oxidative stress, mitochondrial dysfunction, and NLRP3 inflammasome activity. These results provide novel evidence for the role of rotenone in aldosterone-induced renal injury or other chronic kidney disease.
Collapse
Affiliation(s)
- Wei Ding
- Division of Nephrology, Shanghai Ninth People's Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai, China (mainland)
| | - Chengyan Xu
- Division of Nephrology, Huashan Hospital and Institute of Nephrology, Fudan University, Shanghai, China (mainland)
| | - Bin Wang
- Division of Nephrology, Huashan Hospital and Institute of Nephrology, Fudan University, Shanghai, China (mainland)
| | - Minmin Zhang
- Division of Nephrology, Huashan Hospital and Institute of Nephrology, Fudan University, Shanghai, China (mainland)
| |
Collapse
|
43
|
Modulation of Immunity and Inflammation by the Mineralocorticoid Receptor and Aldosterone. BIOMED RESEARCH INTERNATIONAL 2015; 2015:652738. [PMID: 26448944 PMCID: PMC4581510 DOI: 10.1155/2015/652738] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/20/2015] [Revised: 05/07/2015] [Accepted: 06/03/2015] [Indexed: 02/07/2023]
Abstract
The mineralocorticoid receptor (MR) is a ligand dependent transcription factor. MR has been traditionally associated with the control of water and electrolyte homeostasis in order to keep blood pressure through aldosterone activation. However, there is growing evidence indicating that MR expression is not restricted to vascular and renal tissues, as it can be also expressed by cells of the immune system, where it responds to stimulation or antagonism, controlling immune cell function. On the other hand, aldosterone also has been associated with proinflammatory immune effects, such as the release of proinflammatory cytokines, generating oxidative stress and inducing fibrosis. The inflammatory participation of MR and aldosterone in the cardiovascular disease suggests an association with alterations in the immune system. Hypertensive patients show higher levels of proinflammatory mediators that can be modulated by MR antagonism. Although these proinflammatory properties have been observed in other autoimmune and chronic inflammatory diseases, the cellular and molecular mechanisms that mediate these effects remain unknown. Here we review and discuss the scientific work aimed at determining the immunological role of MR and aldosterone in humans, as well as animal models.
Collapse
|
44
|
Du Bois P, Pablo Tortola C, Lodka D, Kny M, Schmidt F, Song K, Schmidt S, Bassel-Duby R, Olson EN, Fielitz J. Angiotensin II Induces Skeletal Muscle Atrophy by Activating TFEB-Mediated MuRF1 Expression. Circ Res 2015; 117:424-36. [PMID: 26137861 DOI: 10.1161/circresaha.114.305393] [Citation(s) in RCA: 78] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/05/2014] [Accepted: 07/02/2015] [Indexed: 11/16/2022]
Abstract
RATIONALE Skeletal muscle wasting with accompanying cachexia is a life threatening complication in congestive heart failure. The molecular mechanisms are imperfectly understood, although an activated renin-angiotensin aldosterone system has been implicated. Angiotensin (Ang) II induces skeletal muscle atrophy in part by increased muscle-enriched E3 ubiquitin ligase muscle RING-finger-1 (MuRF1) expression, which may involve protein kinase D1 (PKD1). OBJECTIVE To elucidate the molecular mechanism of Ang II-induced skeletal muscle wasting. METHODS AND RESULTS A cDNA expression screen identified the lysosomal hydrolase-coordinating transcription factor EB (TFEB) as novel regulator of the human MuRF1 promoter. TFEB played a key role in regulating Ang II-induced skeletal muscle atrophy by transcriptional control of MuRF1 via conserved E-box elements. Inhibiting TFEB with small interfering RNA prevented Ang II-induced MuRF1 expression and atrophy. The histone deacetylase-5 (HDAC5), which was directly bound to and colocalized with TFEB, inhibited TFEB-induced MuRF1 expression. The inhibition of TFEB by HDAC5 was reversed by PKD1, which was associated with HDAC5 and mediated its nuclear export. Mice lacking PKD1 in skeletal myocytes were resistant to Ang II-induced muscle wasting. CONCLUSION We propose that elevated Ang II serum concentrations, as occur in patients with congestive heart failure, could activate the PKD1/HDAC5/TFEB/MuRF1 pathway to induce skeletal muscle wasting.
Collapse
Affiliation(s)
- Philipp Du Bois
- From the Department of Molecular Cardiology, Experimental and Clinical Research Center (ECRC), a Cooperation between Max-Delbrück-Centrum and Charité Universitätsmedizin Berlin, Campus Buch, Berlin, Germany (P.D.B., C.P.T., D.L., M.K., F.S., S.S., J.F.); Department of Cardiology, Charité Universitätsmedizin Berlin, Campus Virchow, Berlin, Germany (J.F.); and Department of Molecular Biology, University of Texas Southwestern Medical Center, Dallas (K.S., R.B.-D., E.N.O.)
| | - Cristina Pablo Tortola
- From the Department of Molecular Cardiology, Experimental and Clinical Research Center (ECRC), a Cooperation between Max-Delbrück-Centrum and Charité Universitätsmedizin Berlin, Campus Buch, Berlin, Germany (P.D.B., C.P.T., D.L., M.K., F.S., S.S., J.F.); Department of Cardiology, Charité Universitätsmedizin Berlin, Campus Virchow, Berlin, Germany (J.F.); and Department of Molecular Biology, University of Texas Southwestern Medical Center, Dallas (K.S., R.B.-D., E.N.O.)
| | - Doerte Lodka
- From the Department of Molecular Cardiology, Experimental and Clinical Research Center (ECRC), a Cooperation between Max-Delbrück-Centrum and Charité Universitätsmedizin Berlin, Campus Buch, Berlin, Germany (P.D.B., C.P.T., D.L., M.K., F.S., S.S., J.F.); Department of Cardiology, Charité Universitätsmedizin Berlin, Campus Virchow, Berlin, Germany (J.F.); and Department of Molecular Biology, University of Texas Southwestern Medical Center, Dallas (K.S., R.B.-D., E.N.O.)
| | - Melanie Kny
- From the Department of Molecular Cardiology, Experimental and Clinical Research Center (ECRC), a Cooperation between Max-Delbrück-Centrum and Charité Universitätsmedizin Berlin, Campus Buch, Berlin, Germany (P.D.B., C.P.T., D.L., M.K., F.S., S.S., J.F.); Department of Cardiology, Charité Universitätsmedizin Berlin, Campus Virchow, Berlin, Germany (J.F.); and Department of Molecular Biology, University of Texas Southwestern Medical Center, Dallas (K.S., R.B.-D., E.N.O.)
| | - Franziska Schmidt
- From the Department of Molecular Cardiology, Experimental and Clinical Research Center (ECRC), a Cooperation between Max-Delbrück-Centrum and Charité Universitätsmedizin Berlin, Campus Buch, Berlin, Germany (P.D.B., C.P.T., D.L., M.K., F.S., S.S., J.F.); Department of Cardiology, Charité Universitätsmedizin Berlin, Campus Virchow, Berlin, Germany (J.F.); and Department of Molecular Biology, University of Texas Southwestern Medical Center, Dallas (K.S., R.B.-D., E.N.O.)
| | - Kunhua Song
- From the Department of Molecular Cardiology, Experimental and Clinical Research Center (ECRC), a Cooperation between Max-Delbrück-Centrum and Charité Universitätsmedizin Berlin, Campus Buch, Berlin, Germany (P.D.B., C.P.T., D.L., M.K., F.S., S.S., J.F.); Department of Cardiology, Charité Universitätsmedizin Berlin, Campus Virchow, Berlin, Germany (J.F.); and Department of Molecular Biology, University of Texas Southwestern Medical Center, Dallas (K.S., R.B.-D., E.N.O.)
| | - Sibylle Schmidt
- From the Department of Molecular Cardiology, Experimental and Clinical Research Center (ECRC), a Cooperation between Max-Delbrück-Centrum and Charité Universitätsmedizin Berlin, Campus Buch, Berlin, Germany (P.D.B., C.P.T., D.L., M.K., F.S., S.S., J.F.); Department of Cardiology, Charité Universitätsmedizin Berlin, Campus Virchow, Berlin, Germany (J.F.); and Department of Molecular Biology, University of Texas Southwestern Medical Center, Dallas (K.S., R.B.-D., E.N.O.)
| | - Rhonda Bassel-Duby
- From the Department of Molecular Cardiology, Experimental and Clinical Research Center (ECRC), a Cooperation between Max-Delbrück-Centrum and Charité Universitätsmedizin Berlin, Campus Buch, Berlin, Germany (P.D.B., C.P.T., D.L., M.K., F.S., S.S., J.F.); Department of Cardiology, Charité Universitätsmedizin Berlin, Campus Virchow, Berlin, Germany (J.F.); and Department of Molecular Biology, University of Texas Southwestern Medical Center, Dallas (K.S., R.B.-D., E.N.O.)
| | - Eric N Olson
- From the Department of Molecular Cardiology, Experimental and Clinical Research Center (ECRC), a Cooperation between Max-Delbrück-Centrum and Charité Universitätsmedizin Berlin, Campus Buch, Berlin, Germany (P.D.B., C.P.T., D.L., M.K., F.S., S.S., J.F.); Department of Cardiology, Charité Universitätsmedizin Berlin, Campus Virchow, Berlin, Germany (J.F.); and Department of Molecular Biology, University of Texas Southwestern Medical Center, Dallas (K.S., R.B.-D., E.N.O.)
| | - Jens Fielitz
- From the Department of Molecular Cardiology, Experimental and Clinical Research Center (ECRC), a Cooperation between Max-Delbrück-Centrum and Charité Universitätsmedizin Berlin, Campus Buch, Berlin, Germany (P.D.B., C.P.T., D.L., M.K., F.S., S.S., J.F.); Department of Cardiology, Charité Universitätsmedizin Berlin, Campus Virchow, Berlin, Germany (J.F.); and Department of Molecular Biology, University of Texas Southwestern Medical Center, Dallas (K.S., R.B.-D., E.N.O.).
| |
Collapse
|
45
|
|
46
|
Schwenk MH, Hirsch JS, Bomback AS. Aldosterone blockade in CKD: emphasis on pharmacology. Adv Chronic Kidney Dis 2015; 22:123-32. [PMID: 25704349 DOI: 10.1053/j.ackd.2014.08.003] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2014] [Accepted: 08/11/2014] [Indexed: 12/16/2022]
Abstract
Besides its epithelial effect on sodium retention and potassium excretion in the distal tubule, aldosterone promotes inflammation and fibrosis in the heart, kidneys, and blood vessels. As glomerular filtration rate falls, aldosterone is inappropriately elevated relative to extracellular fluid expansion. In addition, studies in CKD patients on angiotensin-converting enzyme inhibitors, angiotensin receptor blockers, and/or direct renin inhibitors have shown that aldosterone levels paradoxically rise in approximately 30% to 40% of patients on these renin-angiotensin system-blocking drugs. Hence, there is interest in using mineralocorticoid receptor blockers that directly target the inflammatory and fibrotic effects of aldosterone in CKD patients. This interest, however, is tempered by a number of unresolved issues, including the safety of using such drugs in advanced CKD and ESRD populations, and the potential for differences in drug efficacy according to race and ethnicity of patient populations. A better understanding of mineralocorticoid receptor blocker pharmacology should help inform future research directions and clinical practice decisions as to how best to use these agents in CKD.
Collapse
|
47
|
Yao X, Li N, Zhang Y, Zhang J, Abulikm S, Zhang D, Chang G, Zhou K, Kong J. Plasma aldosterone concentration is positively associated with pulse pressure in patients with primary hypertension. Medicine (Baltimore) 2015; 94:e614. [PMID: 25761186 PMCID: PMC4602476 DOI: 10.1097/md.0000000000000614] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/26/2022] Open
Abstract
Increasing evidence showed a link between arterial elasticity and stiffness and pulse pressure (PP), in which plasma aldosterone may play a role. The observational study aimed to explore the potential relations between plasma aldosterone concentration (PAC) and PP in patients with hypertension. We evaluated the relation between PP and PAC in supine, seated, and upright positions in 195 patients with primary hypertension who underwent postural stimulation test. They were divided into 3 groups by tertiles of PP: PP ≤ 44 mm Hg (n = 70), 44 mm Hg < PP ≤ 51 mm Hg (n = 63), and PP ≥ 51 mm Hg (n = 62). The PAC in different postures was compared, respectively. The results showed the following. First, segregated by tertiles of PP, serum K⁺, 24-hour systolic blood pressure, 24-hour diastolic blood pressure, sex, upright PAC, and seated PAC showed statistically significant differences in groups. Second, the PAC were significantly different in 3 levels of PP regardless of postures, the individuals with PP ≥ 51 mm Hg had the highest PAC. On contrast, the patients with PAC > 12 ng/dL showed greater PP than those with PAC ≤ 12 ng/dL. Third, weak associations between PP and upright (r = 0.288, P < 0.001), seated (r = 0.265, P < 0.001), and supine postures (r = 0.191, P = 0.008) were detected by simple correlation analysis. After corrected serum K⁺, age, and sex, the partial correlation coefficients did not change greatly. Fourth, the logistic regression model was constructed with PP ≥ 40 mm Hg or PP < 40 mm Hg as the dependent variable; the serum K⁺[OR = 0.043, 95% CI: 1.09(1.00-1.12)] and PAC [OR = 0.025, 95%CI: 0.35(0.13-0.88)] were included as significant contributing factors. The results showed that higher PAC was weakly, but significantly, correlated to greater PP regardless of different postures, suggesting that higher PAC may be a risk factor of reduced arterial elasticity in patients with hypertension.
Collapse
Affiliation(s)
- Xiaoguang Yao
- From The Center for Hypertension of the People's Hospital of Xinjiang Uygur Autonomous Region; the Center for Diagnosis, Treatment and Research of Hypertension in Xinjiang, Urumqi, China
| | | | | | | | | | | | | | | | | |
Collapse
|
48
|
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: 1.1] [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
|
49
|
Lu X, Li M, Zhou L, Jiang H, Wang H, Chen J. Urinary serum- and glucocorticoid-inducible kinase SGK1 reflects renal injury in patients with immunoglobulin A nephropathy. Nephrology (Carlton) 2015; 19:307-17. [PMID: 24602173 DOI: 10.1111/nep.12225] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
AIM Serum- and glucocorticoid-inducible kinase SGK1 functions as an important regulator of transepithelial sodium transport by activating epithelial sodium channel in renal tubules. Considerable evidence demonstrated that SGK1 was associated with hypertension and fibrosing diseases, such as diabetic nephropathy and glomerulonephritis. The present study was performed to evaluate the role of SGK1 played in immunoglobulin A (IgA) nephropathy. METHODS Seventy-six patients of biopsy-proven IgA nephropathy and 33 healthy volunteers were enrolled in this study. All patients and healthy volunteers' urinary and serum samples were tested for SGK1 expression by indirect enzyme-linked immunosorbent assay. Meanwhile all patients' renal tissues were semi-quantified for SGK1 expression by immunohistochemistry assay. The relationships between SGK1 expressions and clinical or pathological parameters were also assessed. RESULTS SGK1 expression was upregulated in urine and renal tubules in patients of Oxford classification T1 and T2, whereas its expression in serum did not increase significantly. Relationship analysis indicated that urinary and tissue SGK1 expressions were associated with heavy proteinuria and renal insufficiency in patients with IgA nephropathy. On the other hand, RAS blockades would reduce the SGK1 levels both in urine and renal tissues. CONCLUSION These results suggested that urinary SGK1 should be a good indicator of tubulointerstitial damage in patients of IgA nephropathy. SGK1 expressions in urine and renal tissues were associated with the activity of renin-angiotensin-aldosterone system.
Collapse
Affiliation(s)
- Xiaoqian Lu
- Kidney Disease Center, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, China; Kidney Disease Immunology Laboratory, The Third Grade Laboratory, State Administration of Traditional Chinese Medicine of China, Hangzhou, China; Key Laboratory of Multiple Organ Transplantation, Ministry of Health of China, Hangzhou, China; Key Laboratory of Nephropathy of Zhejiang Province, Hangzhou, China
| | | | | | | | | | | |
Collapse
|
50
|
Mochel JP, Fink M, Peyrou M, Soubret A, Giraudel JM, Danhof M. Pharmacokinetic/Pharmacodynamic Modeling of Renin-Angiotensin Aldosterone Biomarkers Following Angiotensin-Converting Enzyme (ACE) Inhibition Therapy with Benazepril in Dogs. Pharm Res 2014; 32:1931-46. [DOI: 10.1007/s11095-014-1587-9] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2014] [Accepted: 11/24/2014] [Indexed: 12/21/2022]
|