1
|
Agostini LDC, Silva NNT, Belo VDA, Luizon MR, Lima AA, da Silva GN. Pharmacogenetics of angiotensin-converting enzyme inhibitors (ACEI) and angiotensin II receptor blockers (ARB) in cardiovascular diseases. Eur J Pharmacol 2024; 981:176907. [PMID: 39154825 DOI: 10.1016/j.ejphar.2024.176907] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2024] [Revised: 07/29/2024] [Accepted: 08/15/2024] [Indexed: 08/20/2024]
Abstract
Cardiovascular diseases (CVDs) have a high mortality rate, and despite the several available therapeutic targets, non-response to antihypertensives remains a common problem. Angiotensin-converting enzyme inhibitors (ACEIs) and angiotensin receptor blockers (ARBs) are important classes of drugs recommended as first-line therapy for several CVDs. However, response to ACEIs and ARBs varies among treated patients. Pharmacogenomics assesses how an individual's genetic characteristics affect their likely response to drug therapy. Currently, numerous studies suggest that genetic polymorphisms may contribute to variability in drug response. Moreover, further studies evaluating gene-gene interactions within signaling pathways in response to antihypertensives might help to unravel potential genetic predictors for antihypertensive response. This review summarizes the pharmacogenetic data for ACEIs and ARBs in patients with CVD, and discusses the potential pharmacogenetics of these classes of antihypertensives in clinical practice. However, replication studies in different populations are needed. In addition, studies that evaluate gene-gene interactions that share signaling pathways in the response to antihypertensive drugs might facilitate the discovery of genetic predictors for antihypertensive response.
Collapse
Affiliation(s)
- Lívia da Cunha Agostini
- Programa de Pós-Graduação em Ciências Farmacêuticas (CiPharma), Escola de Farmácia, Universidade Federal de Ouro Preto, CEP 35400-000, Ouro Preto, Minas Gerais, Brazil
| | - Nayara Nascimento Toledo Silva
- Departamento de Análises Clínicas (DEACL), Escola de Farmácia, Universidade Federal de Ouro Preto, CEP 35400-000, Ouro Preto, Minas Gerais, Brazil
| | - Vanessa de Almeida Belo
- Departamento de Farmácia (DEFAR), Escola de Farmácia, Universidade Federal de Ouro Preto, CEP 35400-000, Ouro Preto, Minas Gerais, Brazil
| | - Marcelo Rizzatti Luizon
- Departamento de Genética, Ecologia e Evolução, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, CEP 31270-901, Belo Horizonte, MG, Brazil
| | - Angelica Alves Lima
- Departamento de Análises Clínicas (DEACL), Escola de Farmácia, Universidade Federal de Ouro Preto, CEP 35400-000, Ouro Preto, Minas Gerais, Brazil
| | - Glenda Nicioli da Silva
- Departamento de Análises Clínicas (DEACL), Escola de Farmácia, Universidade Federal de Ouro Preto, CEP 35400-000, Ouro Preto, Minas Gerais, Brazil.
| |
Collapse
|
2
|
Pop D, Dădârlat-Pop A, Tomoaia R, Zdrenghea D, Caloian B. Updates on the Renin-Angiotensin-Aldosterone System and the Cardiovascular Continuum. Biomedicines 2024; 12:1582. [PMID: 39062156 PMCID: PMC11274767 DOI: 10.3390/biomedicines12071582] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2024] [Revised: 07/03/2024] [Accepted: 07/15/2024] [Indexed: 07/28/2024] Open
Abstract
The cardiovascular continuum describes how several cardiovascular risk factors contribute to the development of atherothrombosis, ischemic heart disease, and peripheral arteriopathy, leading to cardiac and renal failure and ultimately death. Due to its multiple valences, the renin-angiotensin-aldosterone system plays an important role in all stages of the cardiovascular continuum, starting from a cluster of cardiovascular risk factors, and continuing with the development of atherosclerosis thorough various mechanisms, and culminating with heart failure. Therefore, this article aims to analyze how certain components of the renin-angiotensin-aldosterone system (converting enzymes, angiotensin, angiotensin receptors, and aldosterone) are involved in the underlying pathophysiology of the cardiovascular continuum and the possible arrest of its progression.
Collapse
Affiliation(s)
- Dana Pop
- 4th Department of Internal Medicine, Faculty of Medicine, “Iuliu Hațieganu” University of Medicine and Pharmacy, 400347 Cluj-Napoca, Romania; (D.P.); (R.T.); (D.Z.); (B.C.)
- Cardiology Department, Rehabilitation Hospital, 400012 Cluj-Napoca, Romania
| | - Alexandra Dădârlat-Pop
- 4th Department of Internal Medicine, Faculty of Medicine, “Iuliu Hațieganu” University of Medicine and Pharmacy, 400347 Cluj-Napoca, Romania; (D.P.); (R.T.); (D.Z.); (B.C.)
- Cardiology Department, Heart Institute “N. Stăncioiu”, 400001 Cluj-Napoca, Romania
| | - Raluca Tomoaia
- 4th Department of Internal Medicine, Faculty of Medicine, “Iuliu Hațieganu” University of Medicine and Pharmacy, 400347 Cluj-Napoca, Romania; (D.P.); (R.T.); (D.Z.); (B.C.)
- Cardiology Department, Rehabilitation Hospital, 400012 Cluj-Napoca, Romania
| | - Dumitru Zdrenghea
- 4th Department of Internal Medicine, Faculty of Medicine, “Iuliu Hațieganu” University of Medicine and Pharmacy, 400347 Cluj-Napoca, Romania; (D.P.); (R.T.); (D.Z.); (B.C.)
| | - Bogdan Caloian
- 4th Department of Internal Medicine, Faculty of Medicine, “Iuliu Hațieganu” University of Medicine and Pharmacy, 400347 Cluj-Napoca, Romania; (D.P.); (R.T.); (D.Z.); (B.C.)
- Cardiology Department, Rehabilitation Hospital, 400012 Cluj-Napoca, Romania
| |
Collapse
|
3
|
Vear A, Thalmann C, Youngs K, Hannan N, Gaspari T, Chai SY. Development of a sandwich ELISA to detect circulating, soluble IRAP as a potential disease biomarker. Sci Rep 2023; 13:17565. [PMID: 38001104 PMCID: PMC10673851 DOI: 10.1038/s41598-023-44038-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2023] [Accepted: 10/03/2023] [Indexed: 11/26/2023] Open
Abstract
There is growing interest in the use of the enzyme, insulin regulated aminopeptidase (IRAP), as a biomarker for conditions such as cardio-metabolic diseases and ischemic stroke, with upregulation in its tissue expression in these conditions. However, quantification of circulating IRAP has been hampered by difficulties in detecting release of the truncated, soluble form of this enzyme into the blood stream. The current study aimed to develop a sandwich ELISA using novel antibodies directed towards the soluble portion of IRAP (sIRAP), to improve accuracy in detection and quantification of low levels of sIRAP in plasma. A series of novel anti-IRAP antibodies were developed and found to be highly specific for sIRAP in Western blots. A sandwich ELISA was then optimised using two distinct antibody combinations to detect sIRAP in the low nanogram range (16-500 ng/ml) with a sensitivity of 9 ng/ml and intra-assay variability < 10%. Importantly, the clinical validity of the ELISA was verified by the detection of significant increases in the levels of sIRAP throughout gestation in plasma samples from pregnant women. The specific and sensitive sandwich ELISA described in this study has the potential to advance the development of IRAP as a biomarker for certain diseases.
Collapse
Affiliation(s)
- Anika Vear
- Department of Physiology, Biomedicine Discovery Institute, Monash University, Clayton, VIC, 3800, Australia
| | - Claudia Thalmann
- Department of Physiology, Biomedicine Discovery Institute, Monash University, Clayton, VIC, 3800, Australia
| | - Kristina Youngs
- Department of Physiology, Biomedicine Discovery Institute, Monash University, Clayton, VIC, 3800, Australia
| | - Natalie Hannan
- Department of Obstetrics and Gynaecology, University of Melbourne, Heidelberg, VIC, 3084, Australia
- Mercy Perinatal, Mercy Hospital for Women, Heidelberg, VIC, 3084, Australia
| | - Tracey Gaspari
- Department of Pharmacology, Biomedicine Discovery Institute, Monash University, Clayton, VIC, 3800, Australia
| | - Siew Yeen Chai
- Department of Physiology, Biomedicine Discovery Institute, Monash University, Clayton, VIC, 3800, Australia.
| |
Collapse
|
4
|
Alcocer LA, Bryce A, De Padua Brasil D, Lara J, Cortes JM, Quesada D, Rodriguez P. The Pivotal Role of Angiotensin-Converting Enzyme Inhibitors and Angiotensin II Receptor Blockers in Hypertension Management and Cardiovascular and Renal Protection: A Critical Appraisal and Comparison of International Guidelines. Am J Cardiovasc Drugs 2023; 23:663-682. [PMID: 37668854 PMCID: PMC10625506 DOI: 10.1007/s40256-023-00605-5] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 08/06/2023] [Indexed: 09/06/2023]
Abstract
Arterial hypertension is the main preventable cause of premature mortality worldwide. Across Latin America, hypertension has an estimated prevalence of 25.5-52.5%, although many hypertensive patients remain untreated. Appropriate treatment, started early and continued for the remaining lifespan, significantly reduces the risk of complications and mortality. All international and most regional guidelines emphasize a central role for renin-angiotensin-aldosterone system inhibitors (RAASis) in antihypertensive treatment. The two main RAASi options are angiotensin-converting enzyme inhibitors (ACEis) and angiotensin II receptor blockers (ARBs). Although equivalent in terms of blood pressure reduction, ACEis are preferably recommended by some guidelines to manage other cardiovascular comorbidities, with ARBs considered as an alternative when ACEis are not tolerated. This review summarizes the differences between ACEis and ARBs and their place in the international guidelines. It provides a critical appraisal of the guidelines based on available evidence from randomized controlled trials (RCTs) and meta-analyses, especially considering that hypertensive patients in daily practice often have other comorbidities. The observed differences in cardiovascular and renal outcomes in RCTs may be attributed to the different mechanisms of action of ACEis and ARBs, including increased bradykinin levels, potentiated bradykinin response, and stimulated nitric oxide production with ACEis. It may therefore be appropriate to consider ACEis and ARBs as different antihypertensive drugs classes within the same RAASi group. Although guideline recommendations only differentiate between ACEis and ARBs in patients with cardiovascular comorbidities, clinical evidence suggests that ACEis provide benefits in many hypertensive patients, as well as those with other cardiovascular conditions.
Collapse
Affiliation(s)
| | | | - David De Padua Brasil
- Departamento de Medicina, Faculdade de Ciências da Saúde (FCS), Universidade Federal de Lavras (UFLA), Lavras, Minas Gerais, Brazil
| | - Joffre Lara
- Hospital Juan Tanca Marengo, Guayaquil, Ecuador
| | | | | | - Pablo Rodriguez
- Instituto Cardiovascular de Buenos Aires, Sanatorio Dr. Julio Méndez, Av del Libertador 6302, C1428ART, Buenos Aires, Argentina.
| |
Collapse
|
5
|
Arumugam TV, Alli-Shaik A, Liehn EA, Selvaraji S, Poh L, Rajeev V, Cho Y, Cho Y, Kim J, Kim J, Swa HLF, Hao DTZ, Rattanasopa C, Fann DYW, Mayan DC, Ng GYQ, Baik SH, Mallilankaraman K, Gelderblom M, Drummond GR, Sobey CG, Kennedy BK, Singaraja RR, Mattson MP, Jo DG, Gunaratne J. Multiomics analyses reveal dynamic bioenergetic pathways and functional remodeling of the heart during intermittent fasting. eLife 2023; 12:RP89214. [PMID: 37769126 PMCID: PMC10538958 DOI: 10.7554/elife.89214] [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] [Indexed: 09/30/2023] Open
Abstract
Intermittent fasting (IF) has been shown to reduce cardiovascular risk factors in both animals and humans, and can protect the heart against ischemic injury in models of myocardial infarction. However, the underlying molecular mechanisms behind these effects remain unclear. To shed light on the molecular and cellular adaptations of the heart to IF, we conducted comprehensive system-wide analyses of the proteome, phosphoproteome, and transcriptome, followed by functional analysis. Using advanced mass spectrometry, we profiled the proteome and phosphoproteome of heart tissues obtained from mice that were maintained on daily 12- or 16 hr fasting, every-other-day fasting, or ad libitum control feeding regimens for 6 months. We also performed RNA sequencing to evaluate whether the observed molecular responses to IF occur at the transcriptional or post-transcriptional levels. Our analyses revealed that IF significantly affected pathways that regulate cyclic GMP signaling, lipid and amino acid metabolism, cell adhesion, cell death, and inflammation. Furthermore, we found that the impact of IF on different metabolic processes varied depending on the length of the fasting regimen. Short IF regimens showed a higher correlation of pathway alteration, while longer IF regimens had an inverse correlation of metabolic processes such as fatty acid oxidation and immune processes. Additionally, functional echocardiographic analyses demonstrated that IF enhances stress-induced cardiac performance. Our systematic multi-omics study provides a molecular framework for understanding how IF impacts the heart's function and its vulnerability to injury and disease.
Collapse
Affiliation(s)
- Thiruma V Arumugam
- Centre for Cardiovascular Biology and Disease Research, Department of Microbiology, Anatomy, Physiology and Pharmacology, School of Agriculture, Biomedicine and Environment, La Trobe UniversityMelbourneAustralia
- Department of Physiology, Yong Loo Lin School Medicine, National University of SingaporeSingaporeSingapore
- School of Pharmacy, Sungkyunkwan UniversitySuwonRepublic of Korea
| | - Asfa Alli-Shaik
- Translational Biomedical Proteomics Laboratory, Institute of Molecular and Cell Biology, Agency for Science, Technology and ResearchSingaporeSingapore
| | - Elisa A Liehn
- National Heart Research Institute, National Heart Centre SingaporeSingaporeSingapore
- Institute for Molecular Medicine, University of Southern DenmarkOdenseDenmark
- National Institute of Pathology "Victor Babes"BucharestRomania
| | - Sharmelee Selvaraji
- Department of Physiology, Yong Loo Lin School Medicine, National University of SingaporeSingaporeSingapore
- NUS Graduate School for Integrative Sciences and Engineering, National University of SingaporeSingaporeSingapore
| | - Luting Poh
- Department of Physiology, Yong Loo Lin School Medicine, National University of SingaporeSingaporeSingapore
| | - Vismitha Rajeev
- Department of Physiology, Yong Loo Lin School Medicine, National University of SingaporeSingaporeSingapore
| | - Yoonsuk Cho
- School of Pharmacy, Sungkyunkwan UniversitySuwonRepublic of Korea
| | - Yongeun Cho
- School of Pharmacy, Sungkyunkwan UniversitySuwonRepublic of Korea
| | - Jongho Kim
- School of Pharmacy, Sungkyunkwan UniversitySuwonRepublic of Korea
| | - Joonki Kim
- Department of Physiology, Yong Loo Lin School Medicine, National University of SingaporeSingaporeSingapore
- Natural Products Research Center, Korea Institute of Science and TechnologyGangneungRepublic of Korea
| | - Hannah LF Swa
- Translational Biomedical Proteomics Laboratory, Institute of Molecular and Cell Biology, Agency for Science, Technology and ResearchSingaporeSingapore
| | - David Tan Zhi Hao
- Department of Physiology, Yong Loo Lin School Medicine, National University of SingaporeSingaporeSingapore
| | - Chutima Rattanasopa
- Translational Laboratories in Genetic Medicine, Agency for Science, Technology and ResearchSingaporeSingapore
- Cardiovascular and Metabolic Disorders Program, Duke-National University of SingaporeSingaporeSingapore
| | - David Yang-Wei Fann
- Department of Physiology, Yong Loo Lin School Medicine, National University of SingaporeSingaporeSingapore
| | - David Castano Mayan
- Translational Laboratories in Genetic Medicine, Agency for Science, Technology and ResearchSingaporeSingapore
| | - Gavin Yong-Quan Ng
- Department of Physiology, Yong Loo Lin School Medicine, National University of SingaporeSingaporeSingapore
| | - Sang-Ha Baik
- Department of Physiology, Yong Loo Lin School Medicine, National University of SingaporeSingaporeSingapore
| | - Karthik Mallilankaraman
- Department of Physiology, Yong Loo Lin School Medicine, National University of SingaporeSingaporeSingapore
| | - Mathias Gelderblom
- Department of Neurology, University Medical Center Hamburg-EppendorfHamburgGermany
| | - Grant R Drummond
- Centre for Cardiovascular Biology and Disease Research, Department of Microbiology, Anatomy, Physiology and Pharmacology, School of Agriculture, Biomedicine and Environment, La Trobe UniversityMelbourneAustralia
| | - Christopher G Sobey
- Centre for Cardiovascular Biology and Disease Research, Department of Microbiology, Anatomy, Physiology and Pharmacology, School of Agriculture, Biomedicine and Environment, La Trobe UniversityMelbourneAustralia
| | - Brian K Kennedy
- Department of Physiology, Yong Loo Lin School Medicine, National University of SingaporeSingaporeSingapore
- Department of Biochemistry, Yong Loo Lin School Medicine, National University of SingaporeSingaporeSingapore
| | - Roshni R Singaraja
- Department of Medicine, Yong Loo Lin School of Medicine, National University of SingaporeSingaporeSingapore
| | - Mark P Mattson
- Department of Neuroscience, Johns Hopkins University School of MedicineBaltimoreUnited States
| | - Dong-Gyu Jo
- School of Pharmacy, Sungkyunkwan UniversitySuwonRepublic of Korea
| | - Jayantha Gunaratne
- Translational Biomedical Proteomics Laboratory, Institute of Molecular and Cell Biology, Agency for Science, Technology and ResearchSingaporeSingapore
- Department of Anatomy, Yong Loo Lin School of Medicine, National University of SingaporeSingaporeSingapore
| |
Collapse
|
6
|
Swiderski J, Gadanec LK, Apostolopoulos V, Moore GJ, Kelaidonis K, Matsoukas JM, Zulli A. Role of Angiotensin II in Cardiovascular Diseases: Introducing Bisartans as a Novel Therapy for Coronavirus 2019. Biomolecules 2023; 13:787. [PMID: 37238657 PMCID: PMC10216788 DOI: 10.3390/biom13050787] [Citation(s) in RCA: 18] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2023] [Revised: 04/26/2023] [Accepted: 04/27/2023] [Indexed: 05/28/2023] Open
Abstract
Cardiovascular diseases (CVDs) are the main contributors to global morbidity and mortality. Major pathogenic phenotypes of CVDs include the development of endothelial dysfunction, oxidative stress, and hyper-inflammatory responses. These phenotypes have been found to overlap with the pathophysiological complications of coronavirus disease 2019 (COVID-19). CVDs have been identified as major risk factors for severe and fatal COVID-19 states. The renin-angiotensin system (RAS) is an important regulatory system in cardiovascular homeostasis. However, its dysregulation is observed in CVDs, where upregulation of angiotensin type 1 receptor (AT1R) signaling via angiotensin II (AngII) leads to the AngII-dependent pathogenic development of CVDs. Additionally, the interaction between the spike protein of severe acute respiratory syndrome coronavirus 2 with angiotensin-converting enzyme 2 leads to the downregulation of the latter, resulting in the dysregulation of the RAS. This dysregulation favors AngII/AT1R toxic signaling pathways, providing a mechanical link between cardiovascular pathology and COVID-19. Therefore, inhibiting AngII/AT1R signaling through angiotensin receptor blockers (ARBs) has been indicated as a promising therapeutic approach to the treatment of COVID-19. Herein, we review the role of AngII in CVDs and its upregulation in COVID-19. We also provide a future direction for the potential implication of a novel class of ARBs called bisartans, which are speculated to contain multifunctional targeting towards COVID-19.
Collapse
Affiliation(s)
- Jordan Swiderski
- Institute for Health and Sport, Victoria University, Melbourne, VIC 3030, Australia; (J.S.); (L.K.G.); (V.A.)
| | - Laura Kate Gadanec
- Institute for Health and Sport, Victoria University, Melbourne, VIC 3030, Australia; (J.S.); (L.K.G.); (V.A.)
| | - Vasso Apostolopoulos
- Institute for Health and Sport, Victoria University, Melbourne, VIC 3030, Australia; (J.S.); (L.K.G.); (V.A.)
- Immunology Program, Australian Institute for Musculoskeletal Science, Melbourne, VIC 3021, Australia
| | - Graham J. Moore
- Pepmetics Incorporated, 772 Murphy Place, Victoria, BC V8Y 3H4, Canada;
- Department of Physiology and Pharmacology, Cumming School of Medicine, University of Calgary, Calgary, AB T2N 4N1, Canada
| | | | - John M. Matsoukas
- Institute for Health and Sport, Victoria University, Melbourne, VIC 3030, Australia; (J.S.); (L.K.G.); (V.A.)
- Department of Physiology and Pharmacology, Cumming School of Medicine, University of Calgary, Calgary, AB T2N 4N1, Canada
- NewDrug PC, Patras Science Park, 26500 Patras, Greece;
- Department of Chemistry, University of Patras, 26504 Patras, Greece
| | - Anthony Zulli
- Institute for Health and Sport, Victoria University, Melbourne, VIC 3030, Australia; (J.S.); (L.K.G.); (V.A.)
| |
Collapse
|
7
|
Al-Kouh A, Babiker F, Al-Bader M. Renin-Angiotensin System Antagonism Protects the Diabetic Heart from Ischemia/Reperfusion Injury in Variable Hyperglycemia Duration Settings by a Glucose Transporter Type 4-Mediated Pathway. Pharmaceuticals (Basel) 2023; 16:238. [PMID: 37259385 PMCID: PMC9967344 DOI: 10.3390/ph16020238] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2022] [Revised: 01/15/2023] [Accepted: 02/01/2023] [Indexed: 11/07/2023] Open
Abstract
BACKGROUND Diabetes mellitus (DM) is a risk factor for cardiovascular diseases, specifically, the ischemic heart diseases (IHD). The renin-angiotensin system (RAS) affects the heart directly and indirectly. However, its role in the protection of the heart against I/R injury is not completely understood. The aim of the current study was to evaluate the efficacy of the angiotensin-converting enzyme (ACE) inhibitor and Angiotensin II receptor (AT1R) blocker or a combination thereof in protection of the heart from I/R injury. METHODS Hearts isolated from adult male Wistar rats (n = 8) were subjected to high glucose levels; acute hyperglycemia or streptozotocin (STZ)-induced diabetes were used in this study. Hearts were subjected to I/R injury, treated with Captopril, an ACE inhibitor; Losartan, an AT1R antagonist; or a combination thereof. Hemodynamics data were measured using a suitable software for that purpose. Additionally, infarct size was evaluated using 2,3,5-Triphenyltetrazolium chloride (TTC) staining. The levels of apoptosis markers (caspase-3 and -8), antioxidant enzymes, superoxide dismutase (SOD) and catalase (CAT), nitric oxide synthase (eNOS), and glucose transporter type 4 (GLUT-4) protein levels were evaluated by Western blotting. Pro-inflammatory and anti-inflammatory cytokines levels were evaluated by enzyme-linked immunosorbent assay (ELISA). RESULTS Captopril and Losartan alone or in combination abolished the effect of I/R injury in hearts subjected to acute hyperglycemia or STZ-induced diabetes. There was a significant (p < 0.05) recovery in hemodynamics, infarct size, and apoptosis markers following the treatment with Captopril, Losartan, or their combination. Treatment with Captopril, Losartan, or their combination significantly (p < 0.05) reduced pro-inflammatory cytokines and increased GLUT-4 protein levels. CONCLUSIONS The blockade of the RAS system protected the diabetic heart from I/R injury. This protection followed a pathway that utilizes GLUT-4 to decrease the apoptosis markers, pro-inflammatory cytokines, and to increase the anti-inflammatory cytokines. This protection seems to employ a pathway which is not involving ERK1/2 and eNOS.
Collapse
Affiliation(s)
| | - Fawzi Babiker
- Department of Physiology, Faculty of Medicine, Kuwait University, P.O. Box 24923, Kuwait City 13110, Kuwait
| | | |
Collapse
|
8
|
Asante I, Lu A, Mitchell BI, Boisvert WA, Shikuma CM, Chow DC, Louie SG. Alterations in Renin-Angiotensin System (RAS) Peptide Levels in Patients with HIV. Metabolites 2022; 13:61. [PMID: 36676986 PMCID: PMC9860813 DOI: 10.3390/metabo13010061] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2022] [Revised: 12/21/2022] [Accepted: 12/28/2022] [Indexed: 01/04/2023] Open
Abstract
Chronic HIV infection has long been associated with an increased risk for cardiovascular diseases. The metabolites of the renin−angiotensin system (RAS) such as angiotensin II (AngII) play an important role in regulating blood pressure and fluid dynamics. Cross-sectional analysis of HIV-positive individuals (n = 71, age > 40 years, stable ART > 3 months with HIV viral load < 50 copies/mL) were compared to a similar HIV seronegative group (n = 72). High-resolution B-mode ultrasound images of the right carotid bifurcation (RBIF) and right common carotid artery (RCCA) were conducted to measure the extent of carotid atherosclerotic vascular disease. Plasma RAS peptide levels were quantified using a liquid chromatography-mass spectrometry-based metabolomics assay. RAS peptide concentrations were compared between persons with HIV and persons without HIV, correlating their association with clinical and cardiac measures. Median precursor peptides (Ang(1-12) and AngI) were significantly higher in the HIV-positive group compared to the HIV-negative. Analyses of the patient subgroup not on antihypertensive medication revealed circulating levels of AngII to be four-fold higher in the HIV-positive subgroup. AngII and TNF-alpha levels were found to have a positive association with RCCA, and AngI/Ang(1-12) ratio and TNF-alpha levels were found to have a positive association with RBIF. In both predictive models, AngIII had a negative association with either RCCA or RBIF, which may be attributed to its ability to bind onto AT2R and thus oppose pro-inflammatory events. These results reveal systemic alterations in RAS as a result of chronic HIV infection, which may lead to the activation of inflammatory pathways associated with carotid thickening. RAS peptide levels and cytokine markers were associated with RCCA and RBIF measurements.
Collapse
Affiliation(s)
- Isaac Asante
- Department of Ophthalmology, Keck School of Medicine, University of Southern California, Los Angeles, CA 90033, USA
| | - Angela Lu
- Alfred Mann School of Pharmacy and Pharmaceutical Sciences, University of Southern California, Los Angeles, CA 90089, USA
| | - Brooks I. Mitchell
- John A. Burns School of Medicine, University of Hawai’i at Manoa, Honolulu, HI 96813, USA
| | - William A. Boisvert
- John A. Burns School of Medicine, University of Hawai’i at Manoa, Honolulu, HI 96813, USA
| | - Cecilia M. Shikuma
- John A. Burns School of Medicine, University of Hawai’i at Manoa, Honolulu, HI 96813, USA
| | - Dominic C. Chow
- John A. Burns School of Medicine, University of Hawai’i at Manoa, Honolulu, HI 96813, USA
| | - Stan G. Louie
- Department of Ophthalmology, Keck School of Medicine, University of Southern California, Los Angeles, CA 90033, USA
- Alfred Mann School of Pharmacy and Pharmaceutical Sciences, University of Southern California, Los Angeles, CA 90089, USA
| |
Collapse
|
9
|
Zhang M, Sui W, Xing Y, Cheng J, Cheng C, Xue F, Zhang J, Wang X, Zhang C, Hao P, Zhang Y. Angiotensin IV attenuates diabetic cardiomyopathy via suppressing FoxO1-induced excessive autophagy, apoptosis and fibrosis. Theranostics 2021; 11:8624-8639. [PMID: 34522203 PMCID: PMC8419053 DOI: 10.7150/thno.48561] [Citation(s) in RCA: 97] [Impact Index Per Article: 24.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2020] [Accepted: 07/17/2021] [Indexed: 12/27/2022] Open
Abstract
Rationale: The rennin-angiotensin-aldosterone system (RAAS) plays a critical role in the pathogenesis of diabetic cardiomyopathy, but the role of a member of RAAS, angiotensin IV (Ang IV), in this disease and its underlying mechanism are unclear. This study was aimed to clarify the effects of Ang IV and its downstream mediator forkhead box protein O1 (FoxO1) on diabetic cardiomyopathy. Methods:In vivo, diabetic mice were treated with low-, medium- and high-dose Ang IV, AT4R antagonist divalinal, FoxO1 inhibitor AS1842856 (AS), or their combinations. In vitro, H9C2 cardiomyocytes and cardiac fibroblasts were treated with different concentrations of glucose, low-, medium- and high-dose Ang IV, divalinal, FoxO1-overexpression plasmid (FoxO1-OE), AS, or their combinations. Results: Ang IV treatment dose-dependently attenuated left ventricular dysfunction, fibrosis, and myocyte apoptosis in diabetic mice. Besides, enhanced autophagy and FoxO1 protein expression by diabetes were dose-dependently suppressed by Ang IV treatment. However, these cardioprotective effects of Ang IV were completely abolished by divalinal administration. Bioinformatics analysis revealed that the differentially expressed genes were enriched in autophagy, apoptosis, and FoxO signaling pathways among control, diabetes, and diabetes+high-dose Ang IV groups. Similar to Ang IV, AS treatment ameliorated diabetic cardiomyopathy in mice. In vitro, high glucose stimulation increased collagen expression, apoptosis, overactive autophagy flux and FoxO1 nuclear translocation in cardiomyocytes, and upregulated collagen and FoxO1 expression in cardiac fibroblasts, which were substantially attenuated by Ang IV treatment. However, these protective effects of Ang IV were completely blocked by the use of divalinal or FoxO1-OE, and these detrimental effects were reversed by the additional administration of AS. Conclusions: Ang IV treatment dose-dependently attenuated left ventricular dysfunction and remodeling in a mouse model of diabetic cardiomyopathy, and the mechanisms involved stimulation of AT4R and suppression of FoxO1-mediated fibrosis, apoptosis, and overactive autophagy.
Collapse
Affiliation(s)
- Meng Zhang
- The Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education, Chinese National Health Commission and Chinese Academy of Medical Sciences, The State and Shandong Province Joint Key Laboratory of Translational Cardiovascular Medicine, Department of Cardiology, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan 250012, Shandong, China
| | - Wenhai Sui
- The Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education, Chinese National Health Commission and Chinese Academy of Medical Sciences, The State and Shandong Province Joint Key Laboratory of Translational Cardiovascular Medicine, Department of Cardiology, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan 250012, Shandong, China
| | - Yanqiu Xing
- Department of Geriatrics, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Key Laboratory of Cardiovascular Proteomics of Shandong Province, Jinan 250012, Shandong, China
| | - Jing Cheng
- Beijing Chaoyang Hospital, Capital Medical University, Beijing 100020, China
| | - Cheng Cheng
- The Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education, Chinese National Health Commission and Chinese Academy of Medical Sciences, The State and Shandong Province Joint Key Laboratory of Translational Cardiovascular Medicine, Department of Cardiology, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan 250012, Shandong, China
| | - Fei Xue
- The Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education, Chinese National Health Commission and Chinese Academy of Medical Sciences, The State and Shandong Province Joint Key Laboratory of Translational Cardiovascular Medicine, Department of Cardiology, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan 250012, Shandong, China
| | - Jie Zhang
- The Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education, Chinese National Health Commission and Chinese Academy of Medical Sciences, The State and Shandong Province Joint Key Laboratory of Translational Cardiovascular Medicine, Department of Cardiology, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan 250012, Shandong, China
| | - Xiaohong Wang
- The Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education, Chinese National Health Commission and Chinese Academy of Medical Sciences, The State and Shandong Province Joint Key Laboratory of Translational Cardiovascular Medicine, Department of Cardiology, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan 250012, Shandong, China
| | - Cheng Zhang
- The Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education, Chinese National Health Commission and Chinese Academy of Medical Sciences, The State and Shandong Province Joint Key Laboratory of Translational Cardiovascular Medicine, Department of Cardiology, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan 250012, Shandong, China
| | - Panpan Hao
- The Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education, Chinese National Health Commission and Chinese Academy of Medical Sciences, The State and Shandong Province Joint Key Laboratory of Translational Cardiovascular Medicine, Department of Cardiology, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan 250012, Shandong, China
| | - Yun Zhang
- The Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education, Chinese National Health Commission and Chinese Academy of Medical Sciences, The State and Shandong Province Joint Key Laboratory of Translational Cardiovascular Medicine, Department of Cardiology, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan 250012, Shandong, China
| |
Collapse
|
10
|
Mascolo A, Urbanek K, De Angelis A, Sessa M, Scavone C, Berrino L, Rosano GMC, Capuano A, Rossi F. Angiotensin II and angiotensin 1-7: which is their role in atrial fibrillation? Heart Fail Rev 2021; 25:367-380. [PMID: 31375968 DOI: 10.1007/s10741-019-09837-7] [Citation(s) in RCA: 30] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Atrial fibrillation (AF) is a significant cause of morbidity and mortality as well as a public health burden considering the high costs of AF-related hospitalizations. Pre-clinical and clinical evidence showed a potential role of the renin angiotensin system (RAS) in the etiopathogenesis of AF. Among RAS mediators, angiotensin II (AII) and angiotensin 1-7 (A1-7) have been mostly investigated in AF. Specifically, the stimulation of the pathway mediated by AII or the inhibition of the pathway mediated by A1-7 may participate in inducing and sustaining AF. In this review, we summarize the evidence showing that both RAS pathways may balance the onset of AF through different biological mechanisms involving inflammation, epicardial adipose tissue (EAT) accumulation, and electrical cardiac remodeling. EAT is a predictor for AF as it may induce its onset through direct (infiltration of epicardial adipocytes into the underlying atrial myocardium) and indirect (release of inflammatory adipokines, the stimulation of oxidative stress, macrophage phenotype switching, and AF triggers) mechanisms. Classic RAS blockers such as angiotensin converting enzyme inhibitors (ACE-I) and angiotensin receptor blockers (ARB) may prevent AF by affecting the accumulation of the EAT, representing a useful therapeutic strategy for preventing AF especially in patients with heart failure and known left ventricular dysfunction. Further studies are necessary to prove this benefit in patients with other cardiovascular diseases. Finally, the possibility of using the A1-7 or ACE2 analogues, to enlarge current therapeutic options for AF, may represent an important field of research.
Collapse
Affiliation(s)
- Annamaria Mascolo
- Department of Experimental Medicine, Section of Pharmacology L. Donatelli, University of Campania "Luigi Vanvitelli", Via Santa Maria di Costantinopoli 16, 80138, Naples, Italy.
| | - Konrad Urbanek
- Department of Experimental Medicine, Section of Pharmacology L. Donatelli, University of Campania "Luigi Vanvitelli", Via Santa Maria di Costantinopoli 16, 80138, Naples, Italy
| | - Antonella De Angelis
- Department of Experimental Medicine, Section of Pharmacology L. Donatelli, University of Campania "Luigi Vanvitelli", Via Santa Maria di Costantinopoli 16, 80138, Naples, Italy
| | - Maurizio Sessa
- Department of Experimental Medicine, Section of Pharmacology L. Donatelli, University of Campania "Luigi Vanvitelli", Via Santa Maria di Costantinopoli 16, 80138, Naples, Italy
| | - Cristina Scavone
- Department of Experimental Medicine, Section of Pharmacology L. Donatelli, University of Campania "Luigi Vanvitelli", Via Santa Maria di Costantinopoli 16, 80138, Naples, Italy
| | - Liberato Berrino
- Department of Experimental Medicine, Section of Pharmacology L. Donatelli, University of Campania "Luigi Vanvitelli", Via Santa Maria di Costantinopoli 16, 80138, Naples, Italy
| | - Giuseppe Massimo Claudio Rosano
- IRCCS San Raffaele Pisana, Rome, Italy.,Cardiovascular and Cell Sciences Research Institute, St. George's, University of London, London, UK
| | - Annalisa Capuano
- Department of Experimental Medicine, Section of Pharmacology L. Donatelli, University of Campania "Luigi Vanvitelli", Via Santa Maria di Costantinopoli 16, 80138, Naples, Italy
| | - Francesco Rossi
- Department of Experimental Medicine, Section of Pharmacology L. Donatelli, University of Campania "Luigi Vanvitelli", Via Santa Maria di Costantinopoli 16, 80138, Naples, Italy
| |
Collapse
|
11
|
Vear A, Gaspari T, Thompson P, Chai SY. Is There an Interplay Between the Functional Domains of IRAP? Front Cell Dev Biol 2020; 8:585237. [PMID: 33134302 PMCID: PMC7550531 DOI: 10.3389/fcell.2020.585237] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2020] [Accepted: 09/08/2020] [Indexed: 01/16/2023] Open
Abstract
As a member of the M1 family of aminopeptidases, insulin regulated aminopeptidase (IRAP) is characterized by distinct binding motifs at the active site in the C-terminal domain that mediate the catalysis of peptide substrates. However, what makes IRAP unique in this family of enzymes is that it also possesses trafficking motifs at the N-terminal domain which regulate the movement of IRAP within different intracellular compartments. Research on the role of IRAP has focused predominantly on the C-terminus catalytic domain in different physiological and pathophysiological states ranging from pregnancy to memory loss. Many of these studies have utilized IRAP inhibitors, that bind competitively to the active site of IRAP, to explore the functional significance of its catalytic activity. However, it is unknown whether these inhibitors are able to access intracellular sites where IRAP is predominantly located in a basal state as the enzyme may need to be at the cell surface for the inhibitors to mediate their effects. This property of IRAP has often been overlooked. Interestingly, in some pathophysiological states, the distribution of IRAP is altered. This, together with the fact that IRAP possesses trafficking motifs, suggest the localization of IRAP may play an important role in defining its physiological or pathological functions and provide insights into the interplay between the two functional domains of the protein.
Collapse
Affiliation(s)
- Anika Vear
- Department of Physiology, Monash Biomedicine Discovery Institute, Monash University, Clayton, VIC, Australia
| | - Tracey Gaspari
- Department of Pharmacology, Monash Biomedicine Discovery Institute, Monash University, Clayton, VIC, Australia
| | - Philip Thompson
- Department of Medicinal Chemistry, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, VIC, Australia
| | - Siew Yeen Chai
- Department of Physiology, Monash Biomedicine Discovery Institute, Monash University, Clayton, VIC, Australia
| |
Collapse
|
12
|
Larouche‐Lebel É, Loughran KA, Oyama MA, Solter PF, Laughlin DS, Sánchez MD, Assenmacher C, Fox PR, Fries RC. Plasma and tissue angiotensin-converting enzyme 2 activity and plasma equilibrium concentrations of angiotensin peptides in dogs with heart disease. J Vet Intern Med 2019; 33:1571-1584. [PMID: 31254308 PMCID: PMC6639469 DOI: 10.1111/jvim.15548] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2018] [Accepted: 06/18/2019] [Indexed: 12/20/2022] Open
Abstract
BACKGROUND Angiotensin-converting enzyme 2 (ACE2) is a homologue of angiotensin-converting enzyme (ACE) and produces angiotensin peptides (APs), such as angiotensin 1-9 and 1-7 that are vasodilatory and natriuretic, and act to counterbalance angiotensin II. HYPOTHESIS Evidence of ACE2 can be found in tissues and plasma of dogs. Equilibrium concentrations of renin angiotensin aldosterone system (RAAS) APs differ in dogs with heart disease compared to healthy dogs and recombinant human ACE2 (rhACE2) alters relative concentrations of APs. ANIMALS Forty-nine dogs with and 34 dogs without heart disease. METHODS Immunohistochemistry and assays for tissue and plasma ACE2 activity and equilibrium concentrations of plasma RAAS APs were performed. RESULTS Immunolabeling for ACE2 was present in kidney and myocardial tissue. Median plasma ACE2 activity was significantly increased in dogs with congestive heart failure (CHF; 6.9 mU/mg; interquartile range [IQR], 5.1-12.1) as compared to control (2.2 mU/mg; IQR, 1.8-3.0; P = .0003). Plasma equilibrium analysis of RAAS APs identified significant increases in the median concentrations of beneficial APs, such as angiotensin 1-7, in dogs with CHF (486.7 pg/mL; IQR, 214.2-1168) as compared to those with preclinical disease (41.0 pg/mL; IQR, 27.4-45.1; P < .0001) or control (11.4 pg/mL; IQR, 7.1-25.3; P = .01). Incubation of plasma samples from dogs with CHF with rhACE2 increased beneficial APs, such as angiotensin 1-9 (preincubation, 10.3 pg/mL; IQR, 4.4-37.2; postincubation, 2431 pg/mL; IQR, 1355-3037; P = .02), while simultaneously decreasing maladaptive APs, such as angiotensin II (preincubation, 53.4 pg/mL; IQR, 28.6-226.4; postincubation, 2.4 pg/mL; IQR, 0.50-5.8; P = .02). CONCLUSIONS AND CLINICAL IMPORTANCE Recognition of the ACE2 system expands the conventional view of the RAAS in the dog and represents an important potential therapeutic target.
Collapse
Affiliation(s)
- Éva Larouche‐Lebel
- Department of Clinical Sciences and Advanced Medicine, School of Veterinary MedicineUniversity of PennsylvaniaPhiladelphiaPennsylvania
| | - Kerry A. Loughran
- Department of Clinical Sciences and Advanced Medicine, School of Veterinary MedicineUniversity of PennsylvaniaPhiladelphiaPennsylvania
| | - Mark A. Oyama
- Department of Clinical Sciences and Advanced Medicine, School of Veterinary MedicineUniversity of PennsylvaniaPhiladelphiaPennsylvania
- Institute for Translational Medicine and Therapeutics, Perelman School of MedicineUniversity of PennsylvaniaPhiladelphiaPennsylvania
| | - Phil F. Solter
- Department of Pathobiology, College of Veterinary MedicineUniversity of Illinois at Urbana‐ChampaignUrbanaIllinois
| | - Danielle S. Laughlin
- Department of Clinical Sciences and Advanced Medicine, School of Veterinary MedicineUniversity of PennsylvaniaPhiladelphiaPennsylvania
| | - Melissa D. Sánchez
- Department of Pathobiology, School of Veterinary MedicineUniversity of PennsylvaniaPhiladelphiaPennsylvania
| | | | | | - Ryan C. Fries
- Department of Veterinary Clinical Medicine, College of Veterinary MedicineUniversity of Illinois at Urbana‐ChampaignIllinois
| |
Collapse
|
13
|
Zhou J, He F, Sun B, Liu R, Gao Y, Ren H, Shu Y, Chen X, Liu Z, Zhou H, Deng S, Xu H, Li J, Xu L, Zhang W. Polytropic Influence of TRIB3 rs2295490 Genetic Polymorphism on Response to Antihypertensive Agents in Patients With Essential Hypertension. Front Pharmacol 2019; 10:236. [PMID: 30971918 PMCID: PMC6445854 DOI: 10.3389/fphar.2019.00236] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2018] [Accepted: 02/26/2019] [Indexed: 02/05/2023] Open
Abstract
Tribbles homolog 3 (TRIB3) mediating signaling pathways are closely related to blood pressure regulation. Our previous findings suggested a greater benefit on vascular outcomes in patients carrying TRIB3 (251, A > G, rs2295490) G allele with good glucose and blood pressure control. And TRIB3 (rs2295490) AG/GG genotypes were found to reduce primary vascular events in type 2 diabetic patients who received intensive glucose treatment as compared to those receiving standard glucose treatment. However, the effect of TRIB3 genetic variation on antihypertensives was not clear in essential hypertension patients. A total of 368 patients treated with conventional dosage of antihypertensives (6 groups, grouped by atenolol/bisoprolol, celiprolol, doxazosin, azelnidipine/nitrendipine, imidapril, and candesartan/irbesartan) were enrolled in our study. Genetic variations were successfully identified by sanger sequencing. A linear mixed model analysis was performed to evaluate blood pressures among TRIB3 (251, A > G) genotypes and adjusted for baseline age, gender, body mass index, systolic blood pressure (SBP), diastolic blood pressure (DBP), total cholesterol and other biochemical factors appropriately. Our data suggested that TRIB3 (251, A > G) AA genotype carriers showed better antihypertensive effect than the AG/GG genotype carriers [P = 0.014 for DBP and P = 0.042 for mean arterial pressure (MAP)], with a maximal reduction of DBP by 4.2 mmHg and MAP by 3.56 mmHg after azelnidipine or nitrendipine treatment at the 4th week. Similar tendency of DBP-change and MAP-change was found for imidapril (ACEI) treatment, in which marginally significances were achieved (P = 0.073 and 0.075, respectively). Against that, we found that TRIB3 (251, A > G) AG/GG genotype carriers benefited from antihypertensive therapy of ARBs with a larger DBP-change during the period of observation (P = 0.036). Additionally, stratified analysis revealed an obvious difference of the maximal blood pressure change (13 mmHg for the MAP between male and female patients with AA genotype who took ARBs). Although no significant difference in antihypertensive effect between TRIB3 (251, A > G) genotypes in patients treated with α, β-ADRs was observed, we found significant difference in age-, sex-dependent manner related to α, β-ADRs. In conclusion, our data supported that TRIB3 (251, A > G) genetic polymorphism may serve as a useful biomarker in the treatment of hypertension.
Collapse
Affiliation(s)
- Jiecan Zhou
- Department of Clinical Pharmacology, Xiangya Hospital, Central South University, Changsha, China.,Pharmacogenetics Research Institute, Institute of Clinical Pharmacology, Hunan Key Laboratory of Pharmacogenetics, Central South University, Changsha, China.,National Clinical Research Center for Geriatrics, Xiangya Hospital, Central South University, Changsha, China.,Pharmacy Department, The First Affiliated Hospital, University of South China, Hengyang, China
| | - Fazhong He
- Department of Clinical Pharmacology, Xiangya Hospital, Central South University, Changsha, China.,Pharmacogenetics Research Institute, Institute of Clinical Pharmacology, Hunan Key Laboratory of Pharmacogenetics, Central South University, Changsha, China.,National Clinical Research Center for Geriatrics, Xiangya Hospital, Central South University, Changsha, China
| | - Bao Sun
- Department of Clinical Pharmacology, Xiangya Hospital, Central South University, Changsha, China.,Pharmacogenetics Research Institute, Institute of Clinical Pharmacology, Hunan Key Laboratory of Pharmacogenetics, Central South University, Changsha, China.,National Clinical Research Center for Geriatrics, Xiangya Hospital, Central South University, Changsha, China
| | - Rong Liu
- Department of Clinical Pharmacology, Xiangya Hospital, Central South University, Changsha, China.,Pharmacogenetics Research Institute, Institute of Clinical Pharmacology, Hunan Key Laboratory of Pharmacogenetics, Central South University, Changsha, China.,National Clinical Research Center for Geriatrics, Xiangya Hospital, Central South University, Changsha, China
| | - Yongchao Gao
- Department of Clinical Pharmacology, Xiangya Hospital, Central South University, Changsha, China.,Pharmacogenetics Research Institute, Institute of Clinical Pharmacology, Hunan Key Laboratory of Pharmacogenetics, Central South University, Changsha, China.,National Clinical Research Center for Geriatrics, Xiangya Hospital, Central South University, Changsha, China
| | - Huan Ren
- Department of Clinical Pharmacology, Xiangya Hospital, Central South University, Changsha, China.,Pharmacogenetics Research Institute, Institute of Clinical Pharmacology, Hunan Key Laboratory of Pharmacogenetics, Central South University, Changsha, China.,National Clinical Research Center for Geriatrics, Xiangya Hospital, Central South University, Changsha, China
| | - Yan Shu
- Department of Pharmaceutical Sciences, School of Pharmacy, University of Maryland, Baltimore, MD, United States
| | - Xiaoping Chen
- Department of Clinical Pharmacology, Xiangya Hospital, Central South University, Changsha, China.,Pharmacogenetics Research Institute, Institute of Clinical Pharmacology, Hunan Key Laboratory of Pharmacogenetics, Central South University, Changsha, China.,National Clinical Research Center for Geriatrics, Xiangya Hospital, Central South University, Changsha, China
| | - Zhaoqian Liu
- Department of Clinical Pharmacology, Xiangya Hospital, Central South University, Changsha, China.,Pharmacogenetics Research Institute, Institute of Clinical Pharmacology, Hunan Key Laboratory of Pharmacogenetics, Central South University, Changsha, China.,National Clinical Research Center for Geriatrics, Xiangya Hospital, Central South University, Changsha, China
| | - Honghao Zhou
- Department of Clinical Pharmacology, Xiangya Hospital, Central South University, Changsha, China.,Pharmacogenetics Research Institute, Institute of Clinical Pharmacology, Hunan Key Laboratory of Pharmacogenetics, Central South University, Changsha, China.,National Clinical Research Center for Geriatrics, Xiangya Hospital, Central South University, Changsha, China
| | - Sheng Deng
- Department of Pharmacy, Xiangya Hospital, Central South University, Changsha, China
| | - Heng Xu
- Department of Laboratory Medicine, Precision Medicine Center, and Precision Medicine Key Laboratory of Sichuan Province, Collaborative Innovation Center, West China Hospital, Sichuan University, Chengdu, China
| | - Jianmin Li
- Department of Respiratory Medicine, Hunan Provincial People's Hospital, The First Affiliated Hospital of Hunan Normal University, Changsha, China
| | - Linyong Xu
- Department of Epidemiology and Health Statistics, Xiangya School of Public Health, Central South University, Changsha, China
| | - Wei Zhang
- Department of Clinical Pharmacology, Xiangya Hospital, Central South University, Changsha, China.,Pharmacogenetics Research Institute, Institute of Clinical Pharmacology, Hunan Key Laboratory of Pharmacogenetics, Central South University, Changsha, China.,National Clinical Research Center for Geriatrics, Xiangya Hospital, Central South University, Changsha, China
| |
Collapse
|
14
|
Ramírez-Expósito MJ, Martínez-Martos JM. Differential Effects of Doxazosin on Renin-Angiotensin-System- Regulating Aminopeptidase Activities in Neuroblastoma and Glioma Tumoral Cells. CNS & NEUROLOGICAL DISORDERS-DRUG TARGETS 2019; 18:29-36. [DOI: 10.2174/1871527317666181029111739] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/05/2018] [Revised: 07/13/2018] [Accepted: 10/10/2018] [Indexed: 11/22/2022]
Abstract
Background:
It has been described that doxazosin, an antihypertensive drug, also promotes
glioblastoma cells death by inhibiting cell proliferation, arresting cell cycle and inducing apoptosis.
Doxazosin has also demonstrated several modulator effects on renin-angiotensin system (RAS)-
regulating aminopeptidase activities, which are highly involved in tumor growth in experimental
glioma. Therefore, it remains to elucidate if the anti-tumoral effects of doxazosin could also be mediated
by the proteolytic regulatory components of the RAS.
Objective:
To analyze the effects of doxazosin on cell growth and on RAS-regulating proteolytic regulatory
aspartyl aminopeptidase (ASAP), aminopeptidase A (APA), aminopeptidase N (APN), aminopeptidase
B (APB) and insulin-regulated aminopeptidase (IRAP) specific activities in the human neuroblastoma
NB69 and astroglioma U373-MG tumoral cell lines.
Methods:
Human neuroblastoma NB69 and astroglioma U373-MG cell lines were treated with doxazosin
50-500 μM for 24h or 48h. The effects on cell growth and on RAS-regulating aminopeptidase
specific activities were analyzed.
Results:
Doxazosin treatments promote a concentration-dependent inhibition on cell growth in both
NB69 and U373-MG cells, being NB69 cells more sensitive to the drug than U373-MG cells. However,
its effects on RAS-regulating aminopeptidase specific activities depend on the concentration
used, the duration of the treatment and the cell type. These data confirm the existence of a different
dynamic progression of RAS cascade in each tumoral cell line as a consequence of the treatment with
doxazosin and time of action, which also implies a very dynamic metabolism of the peptides which
participate in each step of RAS cascade.
Conclusion:
Our results indicate that doxazosin modifies the proteolytic regulatory enzymes of RAS
cascade, modulating the bioactive efficacy of the different angiotensin peptides, and therefore, of their
functional roles as initiators/promoters of cell proliferation as autocrine/paracrine mediators.
Collapse
Affiliation(s)
- María Jesús Ramírez-Expósito
- Experimental and Clinical Physiopathology Research Group CTS1039, Department of Health Sciences, Faculty of Health Sciences, University of Jaen, Jaen, Spain
| | - José Manuel Martínez-Martos
- Experimental and Clinical Physiopathology Research Group CTS1039, Department of Health Sciences, Faculty of Health Sciences, University of Jaen, Jaen, Spain
| |
Collapse
|
15
|
Musial DC, Miranda-Ferreira R, Padin JF, Arranz-Tagarro JA, Parra-Vitela AJ, Jurkiewicz A, Garcia AG, Jurkiewicz NH. Function of AT1 and AT2 receptors in atrial contractions from spontaneous hypertensive and diabetic-induced streptozotocin rats. Clin Exp Pharmacol Physiol 2018; 45:1274-1285. [DOI: 10.1111/1440-1681.13019] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2017] [Revised: 07/23/2018] [Accepted: 07/24/2018] [Indexed: 01/11/2023]
Affiliation(s)
- Diego Castro Musial
- Departamento de Farmacología; Universidade Federal de São Paulo; São Paulo Brazil
- Centro de Ciências da Saúde e do Desporto; Universidade Federal do Acre; São Paulo Brazil
| | | | - Juan Fernando Padin
- Departamento Ciencias Médicas (Farmacología); Facultad de Medicina; Universidad de Castilla-La Mancha (UCLM); Ciudad Real España
| | - Juan Alberto Arranz-Tagarro
- Departamento de Farmacología; Facultad de Medicina; Universidad Autónoma de Madrid; Madrid Spain
- Instituto Teófilo Hernando; Madrid Spain
| | - Alberto J. Parra-Vitela
- THRCE - Tulane Hypertension and Renal Center of Excellence; Department of Physiology; School of Medicine; Tulane University; New Orleans Louisiana
| | - Aron Jurkiewicz
- Departamento de Farmacología; Universidade Federal de São Paulo; São Paulo Brazil
| | - Antonio Garcia Garcia
- Departamento de Farmacología; Facultad de Medicina; Universidad Autónoma de Madrid; Madrid Spain
- Instituto Teófilo Hernando; Madrid Spain
| | | |
Collapse
|
16
|
Qiu H, Wu Y, Wang Q, Liu C, Xue L, Wang H, Wu Q, Jiang Q. Effect of berberine on PPAR α-NO signalling pathway in vascular smooth muscle cell proliferation induced by angiotensin IV. PHARMACEUTICAL BIOLOGY 2017; 55:227-232. [PMID: 27927051 PMCID: PMC6130450 DOI: 10.1080/13880209.2016.1257642] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/04/2016] [Revised: 08/28/2016] [Accepted: 11/02/2016] [Indexed: 05/26/2023]
Abstract
CONTEXT The available treatments for the abnormal proliferation of vascular smooth muscle cells (VSMCs) are still dismal. Berberine has been demonstrated to possess extensive medicine activity, yet relatively little is known about its effect on VSMCs proliferation. Many studies showed that PPARα and NO participated in the process of VSMCs proliferation. OBJECTIVE To evaluate the effect of berberine and its possible influence on PPARα-NO pathway in angiotensin IV-stimulated VSMCs. MATERIALS AND METHODS The primary VSMCs were cultured with the tissue explants method, and the proliferation was characterized by MTT and protein content. Protein and mRNA expression were measured by Western blot and real-time RT-PCR, respectively. NO synthase (NOS) activity was measured using a spectrophotometric assay, and NO concentration was measured using the Griess assay. RESULTS Angiotensin IV (0.1 nmol/L)-induced VSMCs proliferation was evidenced by increasing the optical density at A490 and total protein content (p < 0.01), which was inhibited by berberine (10, 30 and 100 μmol/L) in a concentration-dependent manner (p < 0.05). Angiotensin IV decreased the expression of PPARα at mRNA and protein level (p < 0.05), which occurred in parallel with declining eNOS mRNA expression, NOS activity and NO concentration (p < 0.01). Berberine at 30 μmol/L reversed the effects of angiotensin IV in VSMCs (p < 0.05), which were abolished by MK 886 (0.3 μmol/L) (p < 0.05). DISCUSSION AND CONCLUSION The results support the therapeutic effects of berberine on angiotensin IV-induced proliferation in cultured VSMCs at least partially through targeting the PPARα-NO signalling pathway.
Collapse
MESH Headings
- Angiotensin II/analogs & derivatives
- Angiotensin II/pharmacology
- Animals
- Berberine/pharmacology
- Cell Proliferation/drug effects
- Cells, Cultured
- Dose-Response Relationship, Drug
- Female
- Indoles/pharmacology
- Male
- Muscle, Smooth, Vascular/drug effects
- Muscle, Smooth, Vascular/metabolism
- Myocytes, Smooth Muscle/drug effects
- Myocytes, Smooth Muscle/metabolism
- Nitric Oxide/metabolism
- Nitric Oxide Synthase Type III/genetics
- Nitric Oxide Synthase Type III/metabolism
- PPAR alpha/agonists
- PPAR alpha/genetics
- PPAR alpha/metabolism
- Primary Cell Culture
- RNA, Messenger/genetics
- RNA, Messenger/metabolism
- Rats, Sprague-Dawley
- Signal Transduction/drug effects
Collapse
Affiliation(s)
- Hongmei Qiu
- Department of Pharmacology, Chongqing Key Laboratory of Biochemistry and Molecular Pharmacology, Chongqing Medical University, Chongqing, P.R. China
| | - Yang Wu
- Department of Pharmacology, Chongqing Key Laboratory of Biochemistry and Molecular Pharmacology, Chongqing Medical University, Chongqing, P.R. China
| | - Quanhua Wang
- Department of Pharmacology, Chongqing Key Laboratory of Biochemistry and Molecular Pharmacology, Chongqing Medical University, Chongqing, P.R. China
| | - Changqing Liu
- Department of Pharmacology, Chongqing Key Laboratory of Biochemistry and Molecular Pharmacology, Chongqing Medical University, Chongqing, P.R. China
| | - Lai Xue
- Department of Pharmacology, Chongqing Key Laboratory of Biochemistry and Molecular Pharmacology, Chongqing Medical University, Chongqing, P.R. China
| | - Hong Wang
- Department of Pharmacology, Chongqing Key Laboratory of Biochemistry and Molecular Pharmacology, Chongqing Medical University, Chongqing, P.R. China
| | - Qin Wu
- Department of Pharmacology, Key Laboratory for Basic Pharmacology of Ministry of Education, Zunyi Medical College, Guizhou, P.R. China
| | - Qingsong Jiang
- Department of Pharmacology, Chongqing Key Laboratory of Biochemistry and Molecular Pharmacology, Chongqing Medical University, Chongqing, P.R. China
| |
Collapse
|
17
|
Hussain M, Awan FR. Hypertension regulating angiotensin peptides in the pathobiology of cardiovascular disease. Clin Exp Hypertens 2017; 40:344-352. [PMID: 29190205 DOI: 10.1080/10641963.2017.1377218] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
Abstract
Renin angiotensin system (RAS) is an endogenous hormone system involved in the control of blood pressure and fluid volume. Dysregulation of RAS has a pathological role in causing cardiovascular diseases through hypertension. Among several key components of RAS, angiotensin peptides, varying in amino acid length and biological function, have important roles in preventing or promoting hypertension, cardiovascular diseases, stroke, vascular remodeling etc. These peptides are generated by the metabolism of inactive angiotensinogen or its derived peptides by hydrolyzing action of certain enzymes. Angiotensin II, angiotensin (1-12), angiotensin A and angiotensin III bind primarily to angiotensin II type 1 receptor and cause vasoconstriction, accumulation of inflammatory markers to sub-endothelial region of blood vessels and activate smooth muscle cell proliferation. Moreover, when bound to angiotensin II type 2 receptor, angiotensin II works as cardio-protective peptide and halt pathological cell signals. Other peptides like angiotensin (1-9), angiotensin (1-7), alamandine and angiotensin IV also help in protecting from cardiovascular diseases by binding to their respective receptors.
Collapse
Affiliation(s)
- Misbah Hussain
- a Diabetes and Cardio-Metabolic disorders Lab, Health Biotechnology Division , National Institute for Biotechnology and Genetic Engineering (NIBGE) , Faisalabad , Pakistan.,b Pakistan Institute of Engineering and Applied Sciences (PIEAS) , Nilore , Islamabad , Pakistan
| | - Fazli Rabbi Awan
- a Diabetes and Cardio-Metabolic disorders Lab, Health Biotechnology Division , National Institute for Biotechnology and Genetic Engineering (NIBGE) , Faisalabad , Pakistan.,b Pakistan Institute of Engineering and Applied Sciences (PIEAS) , Nilore , Islamabad , Pakistan
| |
Collapse
|
18
|
Taskin E, Tuncer KA, Guven C, Kaya ST, Dursun N. Inhibition of Angiotensin-II Production Increases Susceptibility to Acute Ischemia/Reperfusion Arrhythmia. Med Sci Monit 2016; 22:4587-4595. [PMID: 27889788 PMCID: PMC5142587 DOI: 10.12659/msm.896350] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/29/2023] Open
Abstract
BACKGROUND Myocardial ischemia and reperfusion lead to impairment of electrolyte balance and, eventually, lethal arrhythmias. The aim of this study was to investigate the effects of pharmacological inhibition of angiotensin-II (Ang-II) production on heart tissue with ischemia-reperfusion damage, arrhythmia, and oxidative stress. MATERIAL AND METHODS Rats were divided into 4 groups: only ischemia/reperfusion (MI/R), captopril (CAP), aliskiren (AL), and CAP+AL. The drugs were given by gavage 30 min before anesthesia. Blood pressure and electrocardiography (ECG) were recorded during MI/R procedures. The heart tissue and plasma was kept so as to evaluate the total oxidant (TOS), antioxidant status (TAS), and creatine kinase-MB (CK-MB). RESULTS Creatine kinase-MB was not different among the groups. Although TAS was not affected by inhibition of Ang-II production, TOS was significantly lower in the CAP and/or AL groups than in the MI/R group. Furthermore, oxidative stress index was significantly attenuated in the CAP and/or AL groups. Captopril significantly increased the duration of VT during ischemia; however, it did not have any effect on the incidence of arrhythmias. During reperfusion periods, aliskiren and its combinations with captopril significantly reduced the incidence of other types of arrhythmias. Captopril alone had no effect on the incidence of arrhythmias, but significantly increased arrhythmias score and durations of arrhythmias during reperfusion. MAP and heart rate did not show changes in any groups during ischemic and reperfusion periods. CONCLUSIONS Angiotensin-II production appears to be associated with elevated levels of reactive oxygen species, but Ang-II inhibitions increases arrhythmia, mainly by initiating ventricular ectopic beats.
Collapse
Affiliation(s)
- Eylem Taskin
- Department of Physiotherapy and Rehabilitation, School of Health Sciences, Istanbul Bilim University, Istanbul, Turkey
| | - Kadir Ali Tuncer
- Department of Physiology, Faculty of Medicine, University of Erciyes, Kayseri, Turkey
| | - Celal Guven
- Department of Biophysics, Faculty of Medicine, University of Adiyaman, Adiyaman, Turkey
| | - Salih Tunc Kaya
- Department of Biology, Faculty of Science and Arts, Düzce University, Düzce, Turkey
| | - Nurcan Dursun
- Department of Physiology, Faculty of Medicine, University of Erciyes, Kayseri, Turkey
| |
Collapse
|
19
|
Park BM, Cha SA, Lee SH, Kim SH. Angiotensin IV protects cardiac reperfusion injury by inhibiting apoptosis and inflammation via AT4R in rats. Peptides 2016; 79:66-74. [PMID: 27038740 DOI: 10.1016/j.peptides.2016.03.017] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/17/2015] [Revised: 03/17/2016] [Accepted: 03/29/2016] [Indexed: 02/07/2023]
Abstract
Angiotensin IV (Ang IV) is formed by aminopeptidase N from Ang III by removing the first N-terminal amino acid. Previously, we reported that Ang III has some cardioprotective effects against global ischemia in Langendorff heart. However, it is not clear whether Ang IV has cardioprotective effects. The aim of the present study was to evaluate the effect of Ang IV on myocardial ischemia-reperfusion (I/R) injury in rats. Before ischemia, male Sprague-Dawley rats received Ang IV (1mg/kg/day) for 3 days. Anesthetized rats were subjected to 45min of ischemia by ligation of left anterior descending coronary artery followed by reperfusion and then, sacrificed 1 day or 1 week after reperfusion. Plasma creatine kinase (CK) and lactate dehydrogenase (LDH) concentrations, and infarct size were measured. Quantitative analysis of apoptotic and inflammatory proteins in ventricles were performed using Western blotting. Pretreatment with Ang IV attenuated I/R-induced increases in plasma CK and LDH levels, and infarct size, which were blunted by Ang IV receptor (AT4R) antagonist and but not by antagonist for AT1R, AT2R, or Mas receptor. I/R increased Bax, caspase-3 and caspase-9 protein levels, and decreased Bcl-2 protein level in ventricles, which were blunted by Ang IV. I/R-induced increases in TNF-α, MMP-9, and VCAM-1 protein levels in ventricles were also blunted by Ang IV. Ang IV increased the phosphorylation of Akt and mTOR. These effects were attenuated by co-treatment with AT4R antagonist or inhibitors of downstream signaling pathway. Myocardial dysfunction after reperfusion was improved by Ang IV. These results suggest that Ang IV has cardioprotective effect against I/R injury by inhibiting apoptosis via AT4R and PI3K-Akt-mTOR pathway.
Collapse
Affiliation(s)
- Byung Mun Park
- Department of Physiology, Research Institute for Endocrine Sciences, Chonbuk National University Medical School, Jeonju, Republic of Korea
| | - Seung Ah Cha
- Department of Physiology, Research Institute for Endocrine Sciences, Chonbuk National University Medical School, Jeonju, Republic of Korea
| | - Sun Hwa Lee
- Internal Medicine, Research Institute for Endocrine Sciences, Chonbuk National University Medical School, Jeonju, Republic of Korea
| | - Suhn Hee Kim
- Department of Physiology, Research Institute for Endocrine Sciences, Chonbuk National University Medical School, Jeonju, Republic of Korea.
| |
Collapse
|
20
|
NOD2 contributes to myocardial ischemia/reperfusion injury by regulating cardiomyocyte apoptosis and inflammation. Life Sci 2016; 149:10-7. [DOI: 10.1016/j.lfs.2016.02.039] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2015] [Revised: 01/05/2016] [Accepted: 02/09/2016] [Indexed: 01/06/2023]
|
21
|
He F, Luo J, Zhang Z, Luo Z, Fan L, He Y, Wen J, Zhu D, Gao J, Wang Y, Qian Y, Zhou H, Chen X, Zhang W. The RGS2 (-391, C>G) genetic variation correlates to antihypertensive drug responses in Chinese patients with essential hypertension. PLoS One 2015; 10:e0121483. [PMID: 25849301 PMCID: PMC4388730 DOI: 10.1371/journal.pone.0121483] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2014] [Accepted: 02/01/2015] [Indexed: 01/09/2023] Open
Abstract
OBJECTIVE Regulators of G-protein signaling protein 2 (RGS2) play an irreplaceable role in the control of normal blood pressure (BP). One RGS2 (-391, C>G) genetic variation markedly changes its mRNA expression levels. This study explored the relationship between this genetic variation and the responses to antihypertensive drugs in Chinese patients with essential hypertension. METHODS Genetic variations of RGS2 were successfully identified in 367 specimens using polymerase chain reaction-restriction fragment length polymorphism (PCR-RFLP) assays. All patients were treated with conventional doses of antihypertensives after a 2-week run-in period and followed-up according to our protocol. A general linear model multivariate analysis of variance (ANOVA) was used for the data analysis. RESULTS A significant difference in the mean systolic BP change was observed between RGS2 (-391, C>G) CC/CG (n = 82) and GG (n = 38) genotype carriers (-13.6 vs. -19.9 mmHg, P = 0.043) who were treated with candesartan, irbesartan or imidapril at the end of 6 weeks. In addition, the patients' BP responses to α,β-adrenergic receptor blockers exhibited an age-specific association with the RGS2 (-391, C>G) genetic variation at the end of 4 weeks. CONCLUSION The RGS2 (-391, C>G) genetic polymorphism may serve as a biomarker to predict a patient's response to antihypertensive drug therapy, but future studies need to confirm this.
Collapse
Affiliation(s)
- Fazhong He
- Department of Clinical Pharmacology, Xiangya Hospital, Central South University, Changsha, P.R.C
- Institute of Clinical Pharmacology, Central South University, Changsha, P.R.C
- Hunan Key Laboratory of Pharmacogenetics, Changsha, P.R.C
| | - Jianquan Luo
- Department of Clinical Pharmacology, Xiangya Hospital, Central South University, Changsha, P.R.C
- Institute of Clinical Pharmacology, Central South University, Changsha, P.R.C
- Hunan Key Laboratory of Pharmacogenetics, Changsha, P.R.C
| | - Zhitao Zhang
- Second uropoiesis surgical department in Han Dan Central Hospital, Handan, P.R.C
| | - Zhiying Luo
- Department of Clinical Pharmacology, Xiangya Hospital, Central South University, Changsha, P.R.C
- Institute of Clinical Pharmacology, Central South University, Changsha, P.R.C
- Hunan Key Laboratory of Pharmacogenetics, Changsha, P.R.C
| | - Lan Fan
- Department of Clinical Pharmacology, Xiangya Hospital, Central South University, Changsha, P.R.C
- Institute of Clinical Pharmacology, Central South University, Changsha, P.R.C
- Hunan Key Laboratory of Pharmacogenetics, Changsha, P.R.C
| | - Yijing He
- Department of Clinical Pharmacology, Xiangya Hospital, Central South University, Changsha, P.R.C
- Institute of Clinical Pharmacology, Central South University, Changsha, P.R.C
- Hunan Key Laboratory of Pharmacogenetics, Changsha, P.R.C
| | - Jiagen Wen
- Department of Clinical Pharmacology, Xiangya Hospital, Central South University, Changsha, P.R.C
- Institute of Clinical Pharmacology, Central South University, Changsha, P.R.C
- Hunan Key Laboratory of Pharmacogenetics, Changsha, P.R.C
| | - Dingilang Zhu
- Shanghai Institute of Hypertension, Ruijin Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, P.R.C
| | - Jinping Gao
- Shanghai Institute of Hypertension, Ruijin Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, P.R.C
| | - Yan Wang
- Shanghai Institute of Hypertension, Ruijin Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, P.R.C
| | - Yuesheng Qian
- Shanghai Institute of Hypertension, Ruijin Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, P.R.C
| | - Honghao Zhou
- Department of Clinical Pharmacology, Xiangya Hospital, Central South University, Changsha, P.R.C
- Institute of Clinical Pharmacology, Central South University, Changsha, P.R.C
- Hunan Key Laboratory of Pharmacogenetics, Changsha, P.R.C
| | - Xiaoping Chen
- Department of Clinical Pharmacology, Xiangya Hospital, Central South University, Changsha, P.R.C
- Institute of Clinical Pharmacology, Central South University, Changsha, P.R.C
- Hunan Key Laboratory of Pharmacogenetics, Changsha, P.R.C
| | - Wei Zhang
- Department of Clinical Pharmacology, Xiangya Hospital, Central South University, Changsha, P.R.C
- Institute of Clinical Pharmacology, Central South University, Changsha, P.R.C
- Hunan Key Laboratory of Pharmacogenetics, Changsha, P.R.C
| |
Collapse
|
22
|
Yang H, Li N, Song LN, Wang L, Tian C, Tang CS, Du J, Li HH, Yu XH, Wang HX. Activation of NOD1 by DAP contributes to myocardial ischemia/reperfusion injury via multiple signaling pathways. Apoptosis 2015; 20:512-22. [DOI: 10.1007/s10495-015-1089-1] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
|
23
|
Ramírez-Expósito MJ, Mayas-Torres MD, Carrera-González MP, Jiménez-Pulido SB, Illán-Cabeza NA, Sánchez-Sánchez P, Hueso-Ureña F, Martínez-Martos JM, Moreno-Carretero MN. Silver(I)/6-hydroxyiminolumazine compounds differently modify renin–angiotensin system-regulating aminopeptidases A and N in human neuroblastoma and glioma cells. J Inorg Biochem 2014; 138:56-63. [DOI: 10.1016/j.jinorgbio.2014.04.019] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2014] [Revised: 04/27/2014] [Accepted: 04/28/2014] [Indexed: 12/28/2022]
|
24
|
Malinauskas M, Stankevičius E, Casselbrant A. Angiotensin IV induced contractions in human jejunal wall musculature in vitro. Peptides 2014; 59:63-9. [PMID: 25038512 DOI: 10.1016/j.peptides.2014.07.008] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/05/2014] [Revised: 07/07/2014] [Accepted: 07/08/2014] [Indexed: 10/25/2022]
Abstract
Angiotensin II (AngII) has been reported to mediate contractile actions in rats and human jejunal wall musculature. However, except for one report showing the angiotensin IV (AngIV) contractile effects on the internal anal sphincter of rats, no data is available describing the action of AngIV on smooth muscle in human small intestine. The aim of this study was to investigate the expression and localization of the enzymes responsible to AngIV formation, as well as the receptor, and to elucidate the contractile function of AngIV in the muscular layer of human jejunum in vitro. Jejunal smooth muscle was taken from 23 patients undergoing Roux-en-Y gastric bypass surgery and was used to record isometric tension in vitro in response to AngIV alone and in the presence of losartan or PD123319. ELISA, western blot and immunohistochemistry were used to investigate the expression and localization of key components for AngIV formation: the enzymes aminopeptidases-A, B, M, and the AngIV receptor insulin-regulated aminopeptidase (IRAP). AngIV elicited concentration-dependent contraction in both longitudinal and circular smooth-muscle preparation. Presence of losartan abolished AngIV-induced contraction, but not PD123319. The main peptide AngII, as well as the enzymes aminopeptidases-A, B and M was detected in all muscle samples. Immunohistochemistry localized the enzymes and IRAP in the myenteric plexus between longitudinal and circular muscle layers. The present study indicates that all enzymes necessary for AngIV formation exist in human jejunal smooth muscle and that the contractile action elicited by AngIV is primarily mediated through the AngII type 1 receptor.
Collapse
Affiliation(s)
- M Malinauskas
- Institute of Physiology and Pharmacology, Medical Academy, Lithuanian University of Health Sciences, Kaunas, Lithuania
| | - E Stankevičius
- Institute of Physiology and Pharmacology, Medical Academy, Lithuanian University of Health Sciences, Kaunas, Lithuania
| | - A Casselbrant
- Department of Gastrosurgical Research and Education, Institute of Clinical Sciences, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden.
| |
Collapse
|
25
|
Non-canonical signalling and roles of the vasoactive peptides angiotensins and kinins. Clin Sci (Lond) 2014; 126:753-74. [DOI: 10.1042/cs20130414] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
GPCRs (G-protein-coupled receptors) are among the most important targets for drug discovery due to their ubiquitous expression and participation in cellular events under both healthy and disease conditions. These receptors can be activated by a plethora of ligands, such as ions, odorants, small ligands and peptides, including angiotensins and kinins, which are vasoactive peptides that are classically involved in the pathophysiology of cardiovascular events. These peptides and their corresponding GPCRs have been reported to play roles in other systems and under pathophysiological conditions, such as cancer, central nervous system disorders, metabolic dysfunction and bone resorption. More recently, new mechanisms have been described for the functional regulation of GPCRs, including the transactivation of other signal transduction receptors and the activation of G-protein-independent pathways. The existence of such alternative mechanisms for signal transduction and the discovery of agonists that can preferentially trigger one signalling pathway over other pathways (called biased agonists) have opened new perspectives for the discovery and development of drugs with a higher specificity of action and, therefore, fewer side effects. The present review summarizes the current knowledge on the non-canonical signalling and roles of angiotensins and kinins.
Collapse
|
26
|
Plasma renin-angiotensin system-regulating aminopeptidase activities are modified in early stage Alzheimer's disease and show gender differences but are not related to apolipoprotein E genotype. Exp Gerontol 2013; 48:557-64. [PMID: 23500679 DOI: 10.1016/j.exger.2013.03.002] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2012] [Revised: 03/05/2013] [Accepted: 03/07/2013] [Indexed: 12/25/2022]
Abstract
Alterations in blood pressure and components of the renin-angiotensin system (RAS) contribute to the development and progression of Alzheimer's disease (AD), resulting in changes that can lead or contribute to cognitive decline. Aspartyl aminopeptidase (ASAP), aminopeptidase A (APA), aminopeptidase N (APN) and aminopeptidase B (APB) catabolise circulating angiotensins, whereas insulin-regulated aminopeptidase (IRAP) has been described as the AT4 receptor. We have found in AD patients a significant decrease of APA activity in men but not in women, and of APN, APB and IRAP in both genders, when compared with control subjects. No changes were found in ASAP activity. Also, APN, APB and IRAP but not APA correlated with the Mini-Mental test, but no relationship with APOE genotype was found. We conclude that several components of the RAS are modified in AD patients, with gender differences. Furthermore, ROC analysis indicates that APN, APB and IRAP activities could be useful non-invasive biomarkers of AD from the earliest stages.
Collapse
|