1
|
AlSiraj Y, Thatcher SE, Liang CL, Ali H, Ensor M, Cassis LA. Therapeutic Assessment of Combination Therapy with a Neprilysin Inhibitor and Angiotensin Type 1 Receptor Antagonist on Angiotensin II-Induced Atherosclerosis, Abdominal Aortic Aneurysms, and Hypertension. J Pharmacol Exp Ther 2021; 377:326-335. [PMID: 33707301 PMCID: PMC8140395 DOI: 10.1124/jpet.121.000525] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2021] [Accepted: 03/05/2021] [Indexed: 12/21/2022] Open
Abstract
Combined neprilysin (NEP) inhibition (sacubitril) and angiotensin type 1 receptor (AT1R) antagonism (valsartan) is used in the treatment of congestive heart failure and is gaining interest for other angiotensin II (AngII)-related cardiovascular diseases. In addition to heart failure, AngII promotes hypertension, atherosclerosis, and abdominal aortic aneurysms (AAAs). Similarly, NEP substrates or products have broad effects on the cardiovascular system. In this study, we examined NEP inhibition (with sacubitril) and AT1R antagonism (with valsartan) alone or in combination on AngII-induced hypertension, atherosclerosis, or AAAs in male low-density lipoprotein receptor-deficient mice. Preliminary studies assessed drug delivery via osmotic minipumps for simultaneous release of sacubitril and/or valsartan with AngII over 28 days. Mice were infused with AngII (1000 ng/kg per minute) in the absence (vehicle) or presence of sacubitril (1, 6, or 9 mg/kg per day), valsartan (0.3, 0.5, 1, 6, or 20 mg/kg per day), or the combination thereof (1 and 0.3, or 9 or 0.5 mg/kg per day of sacubitril and valsartan, respectively). Plasma AngII and renin concentrations increased 4-fold at higher valsartan doses, indicative of removal of AngII negative feedback on renin. Sacubitril doubled plasma AngII concentrations at lower doses (1 mg/kg per day). Valsartan dose-dependently decreased systolic blood pressure, aortic atherosclerosis, and AAAs of AngII-infused mice, whereas sacubitril had no effect on atherosclerosis or AAAs but reduced blood pressure of AngII-infused mice. Combination therapy with sacubitril and valsartan did not provide additive benefits. These results suggest limited effects of combination therapy with NEP inhibition and AT1R antagonism against AngII-induced hypertension, atherosclerosis, or AAAs. SIGNIFICANCE STATEMENT: The combination of valsartan (angiotensin type 1 receptor antagonist) and sacubitril (neprilysin inhibitor) did not provide benefit above valsartan alone on AngII-induced hypertension, atherosclerosis, or abdominal aortic aneurysms in low-density lipoprotein receptor-deficient male mice. These results do not support this drug combination in therapy of these AngII-induced cardiovascular diseases.
Collapse
Affiliation(s)
- Yasir AlSiraj
- Department of Pharmacology and Nutritional Sciences, University of Kentucky, Lexington, Kentucky
| | - Sean E Thatcher
- Department of Pharmacology and Nutritional Sciences, University of Kentucky, Lexington, Kentucky
| | - Ching Ling Liang
- Department of Pharmacology and Nutritional Sciences, University of Kentucky, Lexington, Kentucky
| | - Heba Ali
- Department of Pharmacology and Nutritional Sciences, University of Kentucky, Lexington, Kentucky
| | - Mark Ensor
- Department of Pharmacology and Nutritional Sciences, University of Kentucky, Lexington, Kentucky
| | - Lisa A Cassis
- Department of Pharmacology and Nutritional Sciences, University of Kentucky, Lexington, Kentucky
| |
Collapse
|
2
|
Oatmen KE, Zile MR, Burnett JC, Spinale FG. Bioactive Signaling in Next-Generation Pharmacotherapies for Heart Failure: A Review. JAMA Cardiol 2019; 3:1232-1243. [PMID: 30484834 DOI: 10.1001/jamacardio.2018.3789] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/01/2023]
Abstract
Importance The standard pharmacotherapy for heart failure (HF), particularly HF with reduced ejection fraction (HFrEF), is primarily through the use of receptor antagonists, notably inhibition of the renin-angiotensin system by either angiotensin-converting enzyme inhibition or angiotensin II receptor blockade (ARB). However, the completed Prospective Comparison of ARNI With an ACE-Inhibitor to Determine Impact on Global Mortality and Morbidity in Heart Failure (PARADIGM-HF) trial identified that the use of a single molecule (sacubitril/valsartan), which is an ARB and the neutral endopeptidase inhibitor (NEPi) neprilysin, yielded improved clinical outcomes in HFrEF compared with angiotensin-converting enzyme inhibition alone. Observations This review examined specific bioactive signaling pathways that would be potentiated by NEPi and how these would affect key cardiovascular processes relevant to HFrEF. It also addressed potential additive/synergistic effects of ARB. A number of biological signaling pathways that may be potentiated by sacubitril/valsartan were identified, including some novel candidate molecules, which will act in a synergistic manner to favorably alter the natural history of HFrEF. Conclusions and Relevance This review identified that activation rather than inhibition of specific receptor pathways provided favorable cardiovascular effects that cannot be achieved by renin-angiotensin system inhibition alone. Thus, an entirely new avenue of translational and clinical research lies ahead in which HF pharmacotherapies will move beyond receptor antagonist strategies.
Collapse
Affiliation(s)
- Kelsie E Oatmen
- Cardiovascular Translational Research Center, University of South Carolina School of Medicine, Columbia
| | - Michael R Zile
- Medical University of South Carolina, Charleston.,Ralph H. Johnson Department of VA Medical Center, Charleston, South Carolina
| | - John C Burnett
- Cardiorenal Research Laboratory, Mayo Clinic, Rochester, Minnesota
| | - Francis G Spinale
- Cardiovascular Translational Research Center, University of South Carolina School of Medicine, Columbia.,William Jennings Bryan Dorn VA Medical Center, Columbia, South Carolina
| |
Collapse
|
3
|
Abstract
Neprilysin has a major role in both the generation and degradation of bioactive peptides. LCZ696 (valsartan/sacubitril, Entresto), the first of the new ARNI (dual-acting angiotensin-receptor-neprilysin inhibitor) drug class, contains equimolar amounts of valsartan, an angiotensin-receptor blocker, and sacubitril, a prodrug for the neprilysin inhibitor LBQ657. LCZ696 reduced blood pressure more than valsartan alone in patients with hypertension. In the PARADIGM-HF study, LCZ696 was superior to the angiotensin-converting enzyme inhibitor enalapril for the treatment of heart failure with reduced ejection fraction, and LCZ696 was approved by the FDA for this purpose in 2015. This approval was the first for chronic neprilysin inhibition. The many peptides metabolized by neprilysin suggest many potential consequences of chronic neprilysin inhibitor therapy, both beneficial and adverse. Moreover, LBQ657 might inhibit enzymes other than neprilysin. Chronic neprilysin inhibition might have an effect on angio-oedema, bronchial reactivity, inflammation, and cancer, and might predispose to polyneuropathy. Additionally, inhibition of neprilysin metabolism of amyloid-β peptides might have an effect on Alzheimer disease, age-related macular degeneration, and cerebral amyloid angiopathy. Much of the evidence for possible adverse consequences of chronic neprilysin inhibition comes from studies in animal models, and the relevance of this evidence to humans is unknown. This Review summarizes current knowledge of neprilysin function and possible consequences of chronic neprilysin inhibition that indicate a need for vigilance in the use of neprilysin inhibitor therapy.
Collapse
Affiliation(s)
- Duncan J Campbell
- St Vincent's Institute of Medical Research, 41 Victoria Parade, Fitzroy, Victoria 3065, Australia.,University of Melbourne, Parkville, Melbourne, Victoria 3010, Australia
| |
Collapse
|
4
|
Weckler N, Leitzbach D, Kalinowski L, Malinski T, Busch AE, Linz W, Kalinowski L. Effect of chronic treatment with the vasopeptidase inhibitor AVE 7688 and ramipril on endothelial function in atherogenic diet rabbits. J Renin Angiotensin Aldosterone Syst 2016; 4:191-6. [PMID: 14608526 DOI: 10.3317/jraas.2003.031] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/01/2022] Open
Abstract
Cardiovascular disease is the major cause of death in Western nations, although improved possibilities regarding diagnosis and therapy now exist. Endothelial dysfunction is triggered by cardiovascular risk factors such as hypercholesterolaemia, hypertension, adiposity and smoking, contributing to the common endpoint of atherosclerosis. This study examined the pharmacological effects of angiotensin-converting enzyme (ACE) and combined ACE-neutral endopeptidase (NEP) (vasopeptidase) inhibitors on endothelial dysfunction in the model of hyperlipidaemic rabbits. The focus of the study was to assess endothelial function after treatment with the ACE-NEP inhibitor AVE 7688 (30 mg/kg/day) in comparison to the ACE inhibitor (ACE-I) ramipril (1 mg/kg/day). Different parameters, such as endothelial function, blood pressure (BP), expansion of plaques, endothelial nitric oxide (NO) and superoxide (O2 ) release and plasma levels of various lipidaemic parameters were analysed. Control groups consisted of one group fed only with normal diet, one group fed only with atherogenic diet and the direct control group fed with varied diets (six weeks atherogenic diet followed by 12 weeks normal diet). Since for the treatment of atherosclerosis, a change in feeding is absolutely necessary, in the present study, at the start of the treatments with AVE 7688 and ramipril, the rabbits food was changed to a normal diet. At the end of the study, mean arterial blood pressure (MAP) was measured in the anaesthetised animals. The values in standard, atherogenic and varied diet-fed rabbits were around 73±2 mmHg. Angiotensin I (Ang I) given intravenous (i.v.) induced a strong increase in MAP of about 20%. In both the treated groups Ang I-induced BP increase was inhibited. In contrast, i.v. bradykinin led to a strong reduction in MAP in both the treated groups of around 50%. Six weeks' feeding with an atherogenic diet in the rabbits induced an enduring endothelial dysfunction despite the food subsequently being changed to a normal chow. All measured parameters indicated a significant favourable effect on endothelial dysfunction as a result of the two treatment regimens. Endothelial function measured in the organ chamber showed somewhat greater improvement in the ACE-NEP treated group than in the ACE-I treated group. The treatment with ramipril, as well as with AVE 7688, restored endothelial function by increasing the ratio of NO to O2- concentration and bioavailability of NO. In this study, a similar protective effect on endothelial function was shown by ACE-NEP inhibition as already seen with ACE inhibitors in an animal model of atherosclerosis.
Collapse
Affiliation(s)
- Nadine Weckler
- DG Cardiovascular Diseases, Aventis Pharma, Frankfurt, 65926, Germany
| | | | | | | | | | | | | |
Collapse
|
5
|
Talukdar HA, Foroughi Asl H, Jain RK, Ermel R, Ruusalepp A, Franzén O, Kidd BA, Readhead B, Giannarelli C, Kovacic JC, Ivert T, Dudley JT, Civelek M, Lusis AJ, Schadt EE, Skogsberg J, Michoel T, Björkegren JLM. Cross-Tissue Regulatory Gene Networks in Coronary Artery Disease. Cell Syst 2016; 2:196-208. [PMID: 27135365 PMCID: PMC4855300 DOI: 10.1016/j.cels.2016.02.002] [Citation(s) in RCA: 101] [Impact Index Per Article: 12.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2015] [Revised: 12/02/2015] [Accepted: 01/30/2016] [Indexed: 01/23/2023]
Abstract
Inferring molecular networks can reveal how genetic perturbations interact with environmental factors to cause common complex diseases. We analyzed genetic and gene expression data from seven tissues relevant to coronary artery disease (CAD) and identified regulatory gene networks (RGNs) and their key drivers. By integrating data from genome-wide association studies, we identified 30 CAD-causal RGNs interconnected in vascular and metabolic tissues, and we validated them with corresponding data from the Hybrid Mouse Diversity Panel. As proof of concept, by targeting the key drivers AIP, DRAP1, POLR2I, and PQBP1 in a cross-species-validated, arterial-wall RGN involving RNA-processing genes, we re-identified this RGN in THP-1 foam cells and independent data from CAD macrophages and carotid lesions. This characterization of the molecular landscape in CAD will help better define the regulation of CAD candidate genes identified by genome-wide association studies and is a first step toward achieving the goals of precision medicine.
Collapse
Affiliation(s)
- Husain A Talukdar
- Cardiovascular Genomics Group, Division of Vascular Biology, Department of Medical Biochemistry and Biophysics, Karolinska Institutet, 171 77 Stockholm, Sweden
| | - Hassan Foroughi Asl
- Cardiovascular Genomics Group, Division of Vascular Biology, Department of Medical Biochemistry and Biophysics, Karolinska Institutet, 171 77 Stockholm, Sweden
| | - Rajeev K Jain
- Department of Physiology, Institute of Biomedicine and Translation Medicine, University of Tartu, 51014 Tartu, Estonia
| | - Raili Ermel
- Department of Physiology, Institute of Biomedicine and Translation Medicine, University of Tartu, 51014 Tartu, Estonia; Department of Cardiac Surgery, Tartu University Hospital, 51014 Tartu, Estonia
| | - Arno Ruusalepp
- Department of Physiology, Institute of Biomedicine and Translation Medicine, University of Tartu, 51014 Tartu, Estonia; Department of Cardiac Surgery, Tartu University Hospital, 51014 Tartu, Estonia; Clinical Gene Networks AB, 114 44 Stockholm, Sweden
| | - Oscar Franzén
- Clinical Gene Networks AB, 114 44 Stockholm, Sweden; Department of Genetics & Genomic Sciences, Institute of Genomics and Multiscale Biology, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Brian A Kidd
- Department of Genetics & Genomic Sciences, Institute of Genomics and Multiscale Biology, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Ben Readhead
- Department of Genetics & Genomic Sciences, Institute of Genomics and Multiscale Biology, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Chiara Giannarelli
- Department of Genetics & Genomic Sciences, Institute of Genomics and Multiscale Biology, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; The Zena and Michael A. Wiener Cardiovascular Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Jason C Kovacic
- The Zena and Michael A. Wiener Cardiovascular Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Torbjörn Ivert
- Department of Molecular Medicine and Surgery, Karolinska Institutet, 171 77 Stockholm, Sweden; Department of Thoracic Surgery, Karolinska University Hospital, 171 76 Stockholm, Sweden
| | - Joel T Dudley
- Department of Genetics & Genomic Sciences, Institute of Genomics and Multiscale Biology, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Mete Civelek
- Departments of Medicine, Cardiology, Human Genetics, Microbiology, Immunology & Molecular Genetics, University of California, Los Angeles, Los Angeles, CA 90024, USA
| | - Aldons J Lusis
- Departments of Medicine, Cardiology, Human Genetics, Microbiology, Immunology & Molecular Genetics, University of California, Los Angeles, Los Angeles, CA 90024, USA
| | - Eric E Schadt
- Clinical Gene Networks AB, 114 44 Stockholm, Sweden; Department of Genetics & Genomic Sciences, Institute of Genomics and Multiscale Biology, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Josefin Skogsberg
- Cardiovascular Genomics Group, Division of Vascular Biology, Department of Medical Biochemistry and Biophysics, Karolinska Institutet, 171 77 Stockholm, Sweden
| | - Tom Michoel
- Clinical Gene Networks AB, 114 44 Stockholm, Sweden; Division of Genetics and Genomics, The Roslin Institute, University of Edinburgh, Edinburgh EH25 9RG, UK
| | - Johan L M Björkegren
- Cardiovascular Genomics Group, Division of Vascular Biology, Department of Medical Biochemistry and Biophysics, Karolinska Institutet, 171 77 Stockholm, Sweden; Department of Physiology, Institute of Biomedicine and Translation Medicine, University of Tartu, 51014 Tartu, Estonia; Clinical Gene Networks AB, 114 44 Stockholm, Sweden; Department of Genetics & Genomic Sciences, Institute of Genomics and Multiscale Biology, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; Cardiovascular Institute, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA.
| |
Collapse
|
6
|
Moyes AJ, Khambata RS, Villar I, Bubb KJ, Baliga RS, Lumsden NG, Xiao F, Gane PJ, Rebstock AS, Worthington RJ, Simone MI, Mota F, Rivilla F, Vallejo S, Peiró C, Sánchez Ferrer CF, Djordjevic S, Caulfield MJ, MacAllister RJ, Selwood DL, Ahluwalia A, Hobbs AJ. Endothelial C-type natriuretic peptide maintains vascular homeostasis. J Clin Invest 2014; 124:4039-51. [PMID: 25105365 PMCID: PMC4151218 DOI: 10.1172/jci74281] [Citation(s) in RCA: 108] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2013] [Accepted: 06/19/2014] [Indexed: 01/13/2023] Open
Abstract
The endothelium plays a fundamental role in maintaining vascular homeostasis by releasing factors that regulate local blood flow, systemic blood pressure, and the reactivity of leukocytes and platelets. Accordingly, endothelial dysfunction underpins many cardiovascular diseases, including hypertension, myocardial infarction, and stroke. Herein, we evaluated mice with endothelial-specific deletion of Nppc, which encodes C-type natriuretic peptide (CNP), and determined that this mediator is essential for multiple aspects of vascular regulation. Specifically, disruption of CNP leads to endothelial dysfunction, hypertension, atherogenesis, and aneurysm. Moreover, we identified natriuretic peptide receptor-C (NPR-C) as the cognate receptor that primarily underlies CNP-dependent vasoprotective functions and developed small-molecule NPR-C agonists to target this pathway. Administration of NPR-C agonists promotes a vasorelaxation of isolated resistance arteries and a reduction in blood pressure in wild-type animals that is diminished in mice lacking NPR-C. This work provides a mechanistic explanation for genome-wide association studies that have linked the NPR-C (Npr3) locus with hypertension by demonstrating the importance of CNP/NPR-C signaling in preserving vascular homoeostasis. Furthermore, these results suggest that the CNP/NPR-C pathway has potential as a disease-modifying therapeutic target for cardiovascular disorders.
Collapse
Affiliation(s)
- Amie J. Moyes
- William Harvey Research Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London, United Kingdom. Wolfson Institute for Biomedical Research, University College London, London, United Kingdom. División de Cirugía Pediátrica, Hospital Universitario Ramón y Cajal, Madrid, Spain. Departamento de Farmacologia, Facultad de Medicina, Universidad Autonoma de Madrid, Madrid, Spain. Structural and Molecular Biology and Clinical Pharmacology, University College London, London, United Kingdom
| | - Rayomand S. Khambata
- William Harvey Research Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London, United Kingdom. Wolfson Institute for Biomedical Research, University College London, London, United Kingdom. División de Cirugía Pediátrica, Hospital Universitario Ramón y Cajal, Madrid, Spain. Departamento de Farmacologia, Facultad de Medicina, Universidad Autonoma de Madrid, Madrid, Spain. Structural and Molecular Biology and Clinical Pharmacology, University College London, London, United Kingdom
| | - Inmaculada Villar
- William Harvey Research Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London, United Kingdom. Wolfson Institute for Biomedical Research, University College London, London, United Kingdom. División de Cirugía Pediátrica, Hospital Universitario Ramón y Cajal, Madrid, Spain. Departamento de Farmacologia, Facultad de Medicina, Universidad Autonoma de Madrid, Madrid, Spain. Structural and Molecular Biology and Clinical Pharmacology, University College London, London, United Kingdom
| | - Kristen J. Bubb
- William Harvey Research Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London, United Kingdom. Wolfson Institute for Biomedical Research, University College London, London, United Kingdom. División de Cirugía Pediátrica, Hospital Universitario Ramón y Cajal, Madrid, Spain. Departamento de Farmacologia, Facultad de Medicina, Universidad Autonoma de Madrid, Madrid, Spain. Structural and Molecular Biology and Clinical Pharmacology, University College London, London, United Kingdom
| | - Reshma S. Baliga
- William Harvey Research Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London, United Kingdom. Wolfson Institute for Biomedical Research, University College London, London, United Kingdom. División de Cirugía Pediátrica, Hospital Universitario Ramón y Cajal, Madrid, Spain. Departamento de Farmacologia, Facultad de Medicina, Universidad Autonoma de Madrid, Madrid, Spain. Structural and Molecular Biology and Clinical Pharmacology, University College London, London, United Kingdom
| | - Natalie G. Lumsden
- William Harvey Research Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London, United Kingdom. Wolfson Institute for Biomedical Research, University College London, London, United Kingdom. División de Cirugía Pediátrica, Hospital Universitario Ramón y Cajal, Madrid, Spain. Departamento de Farmacologia, Facultad de Medicina, Universidad Autonoma de Madrid, Madrid, Spain. Structural and Molecular Biology and Clinical Pharmacology, University College London, London, United Kingdom
| | - Fang Xiao
- William Harvey Research Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London, United Kingdom. Wolfson Institute for Biomedical Research, University College London, London, United Kingdom. División de Cirugía Pediátrica, Hospital Universitario Ramón y Cajal, Madrid, Spain. Departamento de Farmacologia, Facultad de Medicina, Universidad Autonoma de Madrid, Madrid, Spain. Structural and Molecular Biology and Clinical Pharmacology, University College London, London, United Kingdom
| | - Paul J. Gane
- William Harvey Research Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London, United Kingdom. Wolfson Institute for Biomedical Research, University College London, London, United Kingdom. División de Cirugía Pediátrica, Hospital Universitario Ramón y Cajal, Madrid, Spain. Departamento de Farmacologia, Facultad de Medicina, Universidad Autonoma de Madrid, Madrid, Spain. Structural and Molecular Biology and Clinical Pharmacology, University College London, London, United Kingdom
| | - Anne-Sophie Rebstock
- William Harvey Research Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London, United Kingdom. Wolfson Institute for Biomedical Research, University College London, London, United Kingdom. División de Cirugía Pediátrica, Hospital Universitario Ramón y Cajal, Madrid, Spain. Departamento de Farmacologia, Facultad de Medicina, Universidad Autonoma de Madrid, Madrid, Spain. Structural and Molecular Biology and Clinical Pharmacology, University College London, London, United Kingdom
| | - Roberta J. Worthington
- William Harvey Research Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London, United Kingdom. Wolfson Institute for Biomedical Research, University College London, London, United Kingdom. División de Cirugía Pediátrica, Hospital Universitario Ramón y Cajal, Madrid, Spain. Departamento de Farmacologia, Facultad de Medicina, Universidad Autonoma de Madrid, Madrid, Spain. Structural and Molecular Biology and Clinical Pharmacology, University College London, London, United Kingdom
| | - Michela I. Simone
- William Harvey Research Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London, United Kingdom. Wolfson Institute for Biomedical Research, University College London, London, United Kingdom. División de Cirugía Pediátrica, Hospital Universitario Ramón y Cajal, Madrid, Spain. Departamento de Farmacologia, Facultad de Medicina, Universidad Autonoma de Madrid, Madrid, Spain. Structural and Molecular Biology and Clinical Pharmacology, University College London, London, United Kingdom
| | - Filipa Mota
- William Harvey Research Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London, United Kingdom. Wolfson Institute for Biomedical Research, University College London, London, United Kingdom. División de Cirugía Pediátrica, Hospital Universitario Ramón y Cajal, Madrid, Spain. Departamento de Farmacologia, Facultad de Medicina, Universidad Autonoma de Madrid, Madrid, Spain. Structural and Molecular Biology and Clinical Pharmacology, University College London, London, United Kingdom
| | - Fernando Rivilla
- William Harvey Research Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London, United Kingdom. Wolfson Institute for Biomedical Research, University College London, London, United Kingdom. División de Cirugía Pediátrica, Hospital Universitario Ramón y Cajal, Madrid, Spain. Departamento de Farmacologia, Facultad de Medicina, Universidad Autonoma de Madrid, Madrid, Spain. Structural and Molecular Biology and Clinical Pharmacology, University College London, London, United Kingdom
| | - Susana Vallejo
- William Harvey Research Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London, United Kingdom. Wolfson Institute for Biomedical Research, University College London, London, United Kingdom. División de Cirugía Pediátrica, Hospital Universitario Ramón y Cajal, Madrid, Spain. Departamento de Farmacologia, Facultad de Medicina, Universidad Autonoma de Madrid, Madrid, Spain. Structural and Molecular Biology and Clinical Pharmacology, University College London, London, United Kingdom
| | - Concepción Peiró
- William Harvey Research Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London, United Kingdom. Wolfson Institute for Biomedical Research, University College London, London, United Kingdom. División de Cirugía Pediátrica, Hospital Universitario Ramón y Cajal, Madrid, Spain. Departamento de Farmacologia, Facultad de Medicina, Universidad Autonoma de Madrid, Madrid, Spain. Structural and Molecular Biology and Clinical Pharmacology, University College London, London, United Kingdom
| | - Carlos F. Sánchez Ferrer
- William Harvey Research Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London, United Kingdom. Wolfson Institute for Biomedical Research, University College London, London, United Kingdom. División de Cirugía Pediátrica, Hospital Universitario Ramón y Cajal, Madrid, Spain. Departamento de Farmacologia, Facultad de Medicina, Universidad Autonoma de Madrid, Madrid, Spain. Structural and Molecular Biology and Clinical Pharmacology, University College London, London, United Kingdom
| | - Snezana Djordjevic
- William Harvey Research Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London, United Kingdom. Wolfson Institute for Biomedical Research, University College London, London, United Kingdom. División de Cirugía Pediátrica, Hospital Universitario Ramón y Cajal, Madrid, Spain. Departamento de Farmacologia, Facultad de Medicina, Universidad Autonoma de Madrid, Madrid, Spain. Structural and Molecular Biology and Clinical Pharmacology, University College London, London, United Kingdom
| | - Mark J. Caulfield
- William Harvey Research Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London, United Kingdom. Wolfson Institute for Biomedical Research, University College London, London, United Kingdom. División de Cirugía Pediátrica, Hospital Universitario Ramón y Cajal, Madrid, Spain. Departamento de Farmacologia, Facultad de Medicina, Universidad Autonoma de Madrid, Madrid, Spain. Structural and Molecular Biology and Clinical Pharmacology, University College London, London, United Kingdom
| | - Raymond J. MacAllister
- William Harvey Research Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London, United Kingdom. Wolfson Institute for Biomedical Research, University College London, London, United Kingdom. División de Cirugía Pediátrica, Hospital Universitario Ramón y Cajal, Madrid, Spain. Departamento de Farmacologia, Facultad de Medicina, Universidad Autonoma de Madrid, Madrid, Spain. Structural and Molecular Biology and Clinical Pharmacology, University College London, London, United Kingdom
| | - David L. Selwood
- William Harvey Research Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London, United Kingdom. Wolfson Institute for Biomedical Research, University College London, London, United Kingdom. División de Cirugía Pediátrica, Hospital Universitario Ramón y Cajal, Madrid, Spain. Departamento de Farmacologia, Facultad de Medicina, Universidad Autonoma de Madrid, Madrid, Spain. Structural and Molecular Biology and Clinical Pharmacology, University College London, London, United Kingdom
| | - Amrita Ahluwalia
- William Harvey Research Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London, United Kingdom. Wolfson Institute for Biomedical Research, University College London, London, United Kingdom. División de Cirugía Pediátrica, Hospital Universitario Ramón y Cajal, Madrid, Spain. Departamento de Farmacologia, Facultad de Medicina, Universidad Autonoma de Madrid, Madrid, Spain. Structural and Molecular Biology and Clinical Pharmacology, University College London, London, United Kingdom
| | - Adrian J. Hobbs
- William Harvey Research Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London, United Kingdom. Wolfson Institute for Biomedical Research, University College London, London, United Kingdom. División de Cirugía Pediátrica, Hospital Universitario Ramón y Cajal, Madrid, Spain. Departamento de Farmacologia, Facultad de Medicina, Universidad Autonoma de Madrid, Madrid, Spain. Structural and Molecular Biology and Clinical Pharmacology, University College London, London, United Kingdom
| |
Collapse
|
7
|
Wang J, Ohno-Matsui K, Morita I. Cholesterol enhances amyloid β deposition in mouse retina by modulating the activities of Aβ-regulating enzymes in retinal pigment epithelial cells. Biochem Biophys Res Commun 2012; 424:704-9. [PMID: 22796523 DOI: 10.1016/j.bbrc.2012.07.014] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2012] [Accepted: 07/06/2012] [Indexed: 10/28/2022]
Abstract
Subretinally-deposited amyloid β (Aβ) is a main contributor of developing age-related macular degeneration (AMD). However, the mechanism causing Aβ deposition in AMD eyes is unknown. Hypercholesterolemia is a significant risk for developing AMD. Thus, we investigated the effects of cholesterol on Aβ production in retinal pigment epithelial (RPE) cells in vitro and in the mouse retina in vivo. RPE cells isolated from senescent (12-month-old) C57BL/6 mice were treated with 10μg/ml cholesterol for 48h. Aβ amounts in culture supernatants were measured by ELISA. Activity and expression of enzymes and proteins that regulate Aβ production were examined by activity assay and real time PCR. The retina of mice fed cholesterol-enriched diet was examined by transmission electron microscopy. Cholesterol significantly increased Aβ production in cultured RPE cells. Activities of Aβ degradation enzyme; neprilysin (NEP) and anti-amyloidogenic secretase; α-secretase were significantly decreased in cell lysates of cholesterol-treated RPE cells compared to non-treated cells, but there was no change in the activities of β- or γ-secretase. mRNA levels of NEP and α-secretase (ADAM10 and ADAM17) were significantly lower in cholesterol-treated RPE cells than non-treated cells. Senescent (12-month-old) mice fed cholesterol-enriched chow developed subRPE deposits containing Aβ, whereas age-matched mice fed standard rodent chow diet did not. Activities and mRNA levels of NEP and α-secretase were significantly lower in native RPE cells freshly isolated from cholesterol-enriched chow fed mice compared to standard rodent chow fed mice. These findings suggest that cholesterol enhances subretinal Aβ accumulation by modulating the activities of enzymes degrading and processing Aβ in RPE cells in senescent subjects.
Collapse
Affiliation(s)
- Jiying Wang
- Department of Ophthalmology and Visual Science, Tokyo Medical and Dental University, 1-5-45 Yushima, Bunkyo-ku, Tokyo 113-8519, Japan
| | | | | |
Collapse
|
8
|
Fitzpatrick PA, Guinan AF, Walsh TG, Murphy RP, Killeen MT, Tobin NP, Pierotti AR, Cummins PM. Down-regulation of neprilysin (EC3.4.24.11) expression in vascular endothelial cells by laminar shear stress involves NADPH oxidase-dependent ROS production. Int J Biochem Cell Biol 2009; 41:2287-94. [PMID: 19464387 DOI: 10.1016/j.biocel.2009.05.011] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2009] [Revised: 05/14/2009] [Accepted: 05/16/2009] [Indexed: 10/20/2022]
Abstract
Neprilysin (NEP, neutral endopeptidase, EC3.4.24.11), a zinc metallopeptidase expressed on the surface of endothelial cells, influences vascular homeostasis primarily through regulated inactivation of natriuretic peptides and bradykinin. Earlier in vivo studies reporting on the anti-atherosclerotic effects of NEP inhibition and on the atheroprotective effects of flow-associated laminar shear stress (LSS) have lead us to hypothesize that the latter hemodynamic stimulus may serve to down-regulate NEP levels within the vascular endothelium. To address this hypothesis, we have undertaken an investigation of the effects of LSS on NEP expression in vitro in bovine aortic endothelial cells (BAECs), coupled with an examination of the signalling mechanism putatively mediating these effects. BAECs were exposed to physiological levels of LSS (10 dynes/cm(2), 24h) and harvested for analysis of NEP expression using real-time PCR, Western blotting, and immunocytochemistry. Relative to unsheared controls, NEP mRNA and protein were substantially down-regulated by LSS (>or=50%), events which could be prevented by treatment of BAECs with either N-acetylcysteine, superoxide dismutase, or catalase, implicating reactive oxygen species (ROS) involvement. Employing pharmacological and molecular inhibition strategies, the signal transduction pathway mediating shear-dependent NEP suppression was also examined, and roles implicated for G beta gamma, Rac1, and NADPH oxidase activation in these events. Treatment of static BAECs with angiotensin-II, a potent stimulus for NADPH oxidase activation, mimicked the suppressive effects of shear on NEP expression, further supporting a role for NADPH oxidase-dependent ROS production. Interestingly, inhibition of receptor tyrosine kinase signalling had no effect. In conclusion, we confirm for the first time that NEP expression is down-regulated in vascular endothelial cells by physiological laminar shear, possibly via a mechanotransduction mechanism involving NADPH oxidase-induced ROS production.
Collapse
Affiliation(s)
- Paul A Fitzpatrick
- School of Biotechnology, Dublin City University, Glasnevin, Dublin 9, Ireland
| | | | | | | | | | | | | | | |
Collapse
|
9
|
Leuranguer V, Vanhoutte PM, Verbeuren T, Félétou M. C-type natriuretic peptide and endothelium-dependent hyperpolarization in the guinea-pig carotid artery. Br J Pharmacol 2007; 153:57-65. [PMID: 17906681 PMCID: PMC2199393 DOI: 10.1038/sj.bjp.0707476] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022] Open
Abstract
BACKGROUND AND PURPOSE C-type natriuretic peptide (CNP) has been proposed to make a fundamental contribution in arterial endothelium-dependent hyperpolarization to acetylcholine. The present study was designed to address this hypothesis in the guinea-pig carotid artery. EXPERIMENTAL APPROACH The membrane potential of vascular smooth muscle cells was recorded in isolated arteries with intracellular microelectrodes. KEY RESULTS Acetylcholine induced endothelium-dependent hyperpolarizations in the presence or absence of N (G)-nitro-L-arginine, indomethacin and/or thiorphan, inhibitors of NO-synthases, cyclooxygenases or neutral endopeptidase, respectively. Acetycholine hyperpolarized smooth muscle cells in resting arteries and produced repolarizations in phenylephrine-stimulated arteries. CNP produced hyperpolarizations with variable amplitude. They were observed only in the presence of inhibitors of NO-synthases and cyclooxygenases and were endothelium-independent, maintained in phenylephrine-depolarized carotid arteries, and not affected by the additional presence of thiorphan. In arteries with endothelium, the hyperpolarizations produced by CNP were always significantly smaller than those induced by acetylcholine. Upon repeated administration, a significant tachyphylaxis of the hyperpolarizing effect of CNP was observed, while consecutive administration of acetycholine produced sustained responses. The hyperpolarizations evoked by acetylcholine were abolished by the combination of apamin plus charybdotoxin, but unaffected by glibenclamide or tertiapin. In contrast, CNP-induced hyperpolarizations were abolished by glibenclamide and unaffected by the combination of apamin plus charybdotoxin. CONCLUSIONS AND IMPLICATIONS In the isolated carotid artery of the guinea-pig, CNP activates K(ATP) and is a weak hyperpolarizing agent. In this artery, the contribution of CNP to EDHF-mediated responses is unlikely.
Collapse
Affiliation(s)
| | - P M Vanhoutte
- Department of Pharmacology, Faculty of Medicine, University of Hong Kong Hong Kong, China
| | - T Verbeuren
- Institut Recherches Servier Suresnes, France
| | - M Félétou
- Institut Recherches Servier Suresnes, France
- Author for correspondence:
| |
Collapse
|
10
|
McDowell G, Nicholls DP. The Therapeutic Potential of Candoxatril, a Neutral Endopeptidase Inhibitor, in Humans. ACTA ACUST UNITED AC 2006. [DOI: 10.1111/j.1527-3466.2000.tb00051.x] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
|
11
|
Jandeleit-Dahm K, Lassila M, Davis BJ, Candido R, Johnston CI, Allen TJ, Burrell LM, Cooper ME. Anti-atherosclerotic and renoprotective effects of combined angiotensin-converting enzyme and neutral endopeptidase inhibition in diabetic apolipoprotein E-knockout mice. J Hypertens 2006; 23:2071-82. [PMID: 16208151 DOI: 10.1097/01.hjh.0000184747.41565.a1] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
OBJECTIVE To investigate the effects of the combined angiotensin-converting enzyme (ACE)/neutral endopeptidase (NEP) inhibitor omapatrilat on atherosclerosis and renal injury in a model of diabetes-associated accelerated atherosclerosis and renal injury. DESIGN The study was performed using diabetic apolipoprotein E-knockout (apo E-KO) mice, a model combining hyperlipidemia and hyperglycemia, which leads to accelerated atherosclerosis and renal injury. METHODS Diabetes was induced by the injection of streptozotocin in 6-week old apo E-KO mice. Diabetic animals received no treatment (n = 12) or treatment with the ACE/NEP inhibitor omapatrilat (30 mg/kg per day, via gavage, n = 12) or quinapril (10 mg/kg per day, in drinking water, n = 12) for 20 weeks. Non-diabetic apo E-KO mice (n = 12) served as controls. RESULTS Omapatrilat reduced atherosclerosis and protected the mice from renal structural injury and albuminuria. The protective effects were associated with tissue inhibition of aortic and renal ACE and NEP as well as a significant reduction in blood pressure. Omapatrilat had similar anti-atherosclerotic effects compared with the ACE inhibitor quinapril in association with an almost complete inhibition of aortic ACE activity by both drugs. Omapatrilat conferred superior renoprotection in the diabetic apo E-KO mouse compared with quinapril in the context of greater renal ACE inhibition by omapatrilat than seen with quinapril, additional renal NEP inhibition and a modestly enhanced antihypertensive response. CONCLUSIONS These studies demonstrate the anti-atherosclerotic and renoprotective effects of omapatrilat in diabetic apo E-KO mice, a model of accelerated atherosclerosis and renal injury. These effects were observed in association with the local inhibition of ACE and NEP at the tissue level in the aorta and kidney. These results suggest that the anti-atherosclerotic effect conferred by omapatrilat treatment in the diabetic apo E-KO mouse is predominantly mediated by its capacity to inhibit local vascular ACE. By contrast, in the kidney, local renal ACE and NEP inhibition and the superior antihypertensive effect of omapatrilat all contribute to the renoprotective effect conferred by omapatrilat treatment in the diabetic apo E-KO mouse.
Collapse
Affiliation(s)
- Karin Jandeleit-Dahm
- The Baker Heart Research Institute, Vascular Division, Danielle Alberti Memorial Centre for Diabetes Complications, Melbourne, Australia.
| | | | | | | | | | | | | | | |
Collapse
|
12
|
Campese VM, Nadim MK. Natriuretic Peptides. Hypertension 2005. [DOI: 10.1016/b978-0-7216-0258-5.50108-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2022]
|
13
|
Bratasz A, Kuter I, Konior R, Gościński I, Łukiewicz S. Nitric oxide as a prognostic marker for neurological diseases. Antioxid Redox Signal 2004; 6:613-7. [PMID: 15130288 DOI: 10.1089/152308604773934378] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
The potential value of the nitric oxide (NO) level as a prognostic marker in human brain diseases is investigated. Cerebrospinal fluid (CSF) collected from neurological patients was examined for NO content using electron paramagnetic resonance (EPR) spectroscopy. In adult patients with meningitis, the level of NO was higher than that in other groups of brain disorders, such as brain traumas and brain tumors. Very high levels of NO in the CSF appeared to be correlated with a high incidence of fatal outcomes. In children with meningitis, it was possible to differentiate between viral and bacterial origin of the disease as evidenced by the EPR analysis of the CSF. The results indicated that NO levels in the CSF can be a useful prognostic marker in neurological diseases.
Collapse
Affiliation(s)
- Anna Bratasz
- Laboratory of Radiobiology and Radiospectroscopy of Cancer, Faculty of Biotechnology of the Jagiellonian University, Kraków, Poland
| | | | | | | | | |
Collapse
|
14
|
Newaz MA, Yousefipour Z, Hercule H, Truong L, Oyekan A. Chronic endopeptidase inhibition in DOCA-salt hypertension: mechanism of cardiovascular protection. Clin Exp Hypertens 2003; 25:335-47. [PMID: 12940471 DOI: 10.1081/ceh-120023543] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/03/2022]
Abstract
These studies examined the interactions of neutral endopeptidase (NEP), endothelin-1 (ET-1), and nitric oxide (NO) in deoxycorticosterone acetate (DOCA)-induced hypertension. Male Sprague-Dawley rats (n = 35) were uninephrectomized (UNx) or uninephrectomized and treated with DOCA (25 mg pellet implanted subcutaneously). Candoxatril (30 mg/kg day(-1)), a NEP inhibitor, was given orally for 3 weeks in UNx or DOCA rats. Sham nephrectomized rats (SHAM) served as controls. Except SHAM, all other groups received 1% NaCl in drinking water ad libitum. Measurements were taken of systolic blood pressure (SBP), left ventricle (LV), and aortic weight (AW), plasma ET-1, and urinary excretion of nitrite and Na+. Whole body vascular hypertrophy and morphometric analysis of histological sections of the heart were also determined. In DOCA rats, SBP increased from 113 +/- 5 to 170 +/- 5 mmHg without significant changes in body weight (BW). Candoxatril reduced the increase in SBP to 135 +/- 9 mmHg (P < 0.05), abolished the increased LV wall thickness (P < 0.05), and increased the reduced LV lumen diameter (P < 0.05) in DOCA-salt rats. Candoxatril also reduced plasma ET-1 by 88 +/- 9% and 89 +/- 17% (P < 0.05) in UNx and DOCA rats, respectively, and elicited increases in urinary excretion of nitrite. These effects were accompanied by a marked increase in urinary excretion of Na+ (U(Na)V) (P < 0.05) and a blunting of the proteinuria (32 +/- 5%; P < 0.05) in DOCA rats. We conclude that endopeptidase inhibition in DOCA-salt hypertension reduced the increase in blood pressure and the attendant tissue hypertrophy and renal injury. These effects suggest a correlation between endopeptidase-related reduction in ET-1 production and protection in DOCA-salt hypertension.
Collapse
Affiliation(s)
- Mohammad A Newaz
- Center for Cardiovascular Diseases, College of Pharmacy and Health Sciences, Texas Southern University, Houston, Texas 77004, USA
| | | | | | | | | |
Collapse
|
15
|
Rossi GP. Dual ACE and NEP inhibitors: a review of the pharmacological properties of MDL 100240. CARDIOVASCULAR DRUG REVIEWS 2003; 21:51-66. [PMID: 12595917 DOI: 10.1111/j.1527-3466.2003.tb00105.x] [Citation(s) in RCA: 19] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
The Angiotensin I converting enzyme (ACE, EC 3.4.14.1, kininase II) and neutral endopeptidases (NEP, NEP 24.11) are mechanistically related metallopeptidases. They play a key role in the regulation of blood pressure, body fluid homeostasis and cell growth. Therefore, they are implicated in the pathogenesis of arterial hypertension, congestive heart failure, left ventricular remodeling after myocardial infarction and other cardiovascular diseases. Furthermore, since these two metallopeptidases possess some subsite and substrate similarities, as indicated by their interaction with certain mercaptoalkanoyl inhibitors, they are regarded as an important common target for pharmacological inhibition with a single drug. MDL 100240 is a pro-drug that, upon conversion to MDL 100173, acts as a potent dual inhibitor of ACE and NEP with a balanced activity on both enzymes. Only very limited pharmacokinetic studies with MDL 100240 have been published. These studies used a high pressure liquid chromatography method with UV absorbance detection to quantify the drug. According to the studies in dogs the terminal t(1/2) of MDL 100173 was 35.7 h. The area under the curve for total MDL 100173 was nearly 10-fold greater than the sum of the areas under the curve for MDL 100240 and for unconjugated MDL 100173. These results support the hypothesis that MDL 100240 is hydrolyzed in plasma to the active thiol, MDL 100173, which is rapidly conjugated with endogenous plasma thiols thus providing a pathway for elimination. Studies in vivo in experimental models of hypertension and congestive heart failure confirmed the vasodilatory and natriuretic effects of MDL, which appear to be independent of the degree of activation of the renin-angiotensin-aldosterone system. In addition, MDL 100240 showed an impressive effectiveness both in preventing and in regressing hypertension-induced vascular remodeling and cardiac hypertrophy. Accordingly, MDL 100240 is being developed for the treatment of cardiovascular diseases, including hypertension and congestive heart failure. If the promises of this novel therapeutic strategy are fulfilled, clinical trials are expected to demonstrate advantages of MDL 100240 over pure ACE inhibitors.
Collapse
Affiliation(s)
- Gian Paolo Rossi
- Department of Clinical and Experimental Medicine, University of Padova, Italy.
| |
Collapse
|
16
|
Rossi GP, Cavallin M, Rizzoni D, Bova S, Mazzocchi G, Agabiti-Rosei E, Nussdorfer GG, Pessina AC. Dual ACE and NEP inhibitor MDL-100,240 prevents and regresses severe angiotensin II-dependent hypertension partially through bradykinin type 2 receptor. J Hypertens 2002; 20:1451-9. [PMID: 12131544 DOI: 10.1097/00004872-200207000-00034] [Citation(s) in RCA: 16] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
OBJECTIVE To investigate the effects of the dual angiotensin-converting enzyme (ACE) + neutral endopeptidase (NEP) inhibitor, MDL-100,240 (MDL), on hypertension and cardiovascular damage in male heterozygous transgenic Ren2 rats. METHODS Blood-pressure-matched 5-week-old transgenic rats were allocated to receive a placebo, MDL (40 mg/kg body weight) or ramipril (5 mg/kg body weight) for 8 weeks. During the last 4 weeks, the bradykinin B2 receptor antagonist, icatibant (0.5 mg/kg body weight), was also administered subcutaneously via osmotic minipumps to 50% of the transgenic rats receiving MDL or ramipril. We measured blood pressure, heart weight, structural changes in the aorta and small resistance mesenteric arteries, and the plasma concentrations of adrenomedullin, aldosterone, atrial natriuretic peptide and cGMP. To verify if MDL could regress long-standing hypertension and full-blown cardiovascular damage, 3-month-old transgenic rats received MDL subcutaneously (3 and 10 mg/kg body weight, osmotic minipumps) for 4 weeks. RESULTS Compared with placebo, MDL decreased blood pressure (P < 0.001) and prevented left ventricular hypertrophy (P < 0.001), being as effective as ramipril. Hypertrophy and dilatation of the aorta and hypertrophy of the resistance arterioles were all prevented by MDL. Plasma aldosterone was decreased by MDL (P < 0.001), but not by ramipril. Icatibant blunted the decrease in blood pressure (P < 0.001), decreased cGMP concentrations and blunted the decrease in cross-sectional area of the resistance arteries in MDL-treated, but not in ramipril-treated, transgenic rats. In 3-month-old transgenic rats, MDL normalized blood pressure, regressed left ventricular hypertrophy and decreased adrenomedullin concentrations. CONCLUSIONS The dual ACE+NEP inhibitor MDL prevented and regressed severe hypertension and cardiovascular damage, even in this model of severe angiotensin II-dependent hypertension with pronounced cardiovascular damage. Enhancement of the effects of bradykinin has a role in such favourable outcomes.
Collapse
Affiliation(s)
- Gian Paolo Rossi
- Department of Medical and Surgical Sciences, University of Brescia, Brescia, Italy.
| | | | | | | | | | | | | | | |
Collapse
|
17
|
Rossi GP, Bova S, Sacchetto A, Rizzoni D, Agabiti-Rosei E, Neri G, Nussdorfer GG, Pessina AC. Comparative effects of the dual ACE-NEP inhibitor MDL-100,240 and ramipril on hypertension and cardiovascular disease in endogenous angiotensin II-dependent hypertension. Am J Hypertens 2002; 15:181-8. [PMID: 11863255 DOI: 10.1016/s0895-7061(01)02278-6] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
We investigated the effects of MDL-100,240 in a transgenic rat model (TGRen2) of hypertension with severe cardiovascular damage (CVD) due to enhanced tissue synthesis of angiotensin II (Ang II). Male heterozygous TGRen2 rats (5 weeks old) were allocated to receive MDL-100,240, ramipril (RAM) or placebo (PLAC) for 4 weeks, during which blood pressure (BP) was measured. We then evaluated: 1) left ventricle (LV) and right ventricle (RV), brain, kidney and adrenals weight; 2) structural changes in the aorta and the mesenteric arterioles wall; 3) tension responses of segments of the aorta to phenylephrine, KCl, and endothelin-1; and 4) creatinine, aldosterone, atrial natriuretic peptide (ANP), and cyclic GMP (cGMP) plasma levels. Compared to PLAC, both MDL-100,240 and RAM significantly (P < .001) lowered BP (after 4 weeks: 255 +/- 15 mm Hg PLAC, v 174 +/- 6 MDL-100,240, v 166 +/- 5 RAM). They hindered LV hypertrophy (3.73 +/- 0.25 mg/g body weight (PLAC) v 2.71 +/- 0.22 (MDL-100,240) P < .001; v 2.36 +/- 0.2 (RAM), P < .001). MDL-100,240 also prevented aortic dilatation and hypertrophy of the mesenteric arterioles (media thickness, 25.3 +/- 0.5 microm PLAC, v 21.1 +/- 0.9 MDL-100,240, P < .007; v 20.2 +/- 1.5 RAM, P = .033) and lowered the tension responses to phenylephrine (P < .01), KCl (P < .01), and endothelin-1 (P < .001). Plasma aldosterone (710 +/- 153 pmol/L PLAC, v 237 +/- 61 MDL-100,240, v 180 +/- 22 RAM) and creatinine levels (0.69 +/- 0.33 mg/dL PLAC, v 0.41 +/- 0.02 MDL-100,240, v 0.41 +/- 0.04 RAM) were also decreased (P < or = .001). Compared to PLAC, plasma ANP levels were 11% and 2.4% higher in MDL-100,240 and RAM, respectively (both P = not significant); cGMP levels were unaffected. Thus, severe hypertension and related CVD were regressed by MDL-100,240, which resulted to be as effective as a full dosage of ramipril in TGRen2.
Collapse
Affiliation(s)
- Gian Paolo Rossi
- Department of Clinical and Experimental Medicine--Clinica Medica 4, University of Padova, University Hospital, Italy.
| | | | | | | | | | | | | | | |
Collapse
|
18
|
Abstract
Studies using both in vitro and in vivo techniques have repeatedly shown that endothelium-dependent vasodilation (EDV) is impaired in different forms of experimental as well as human hypercholesterolemia. Clearly this impaired EDV can be reversed by lowering cholesterol levels by diet or medical therapy. Competitive blocking of L-arginine, changes in nitric oxide synthase activity, increased release of endothelin-1, and inactivation of nitric oxide due to superoxide ions all contribute to the impairment in EDV during dyslipidemia. The oxidation of low density lipoprotein, with its compound lysophosphatidylcholine, plays a critical role in these events. However, data on the role of triglycerides and fat-rich meals regarding EDV are not so consistent as data for cholesterol, although a view that the compositions of individual fatty acids and antioxidants are of major importance is emerging. Thus, this review shows that while impaired EDV is a general feature of hypercholesterolemia, the mechanisms involved and the therapeutic opportunities available still have to be investigated. Furthermore, discrepancies regarding the role of triglycerides and fat content in food may be explained by divergent effects of different fatty acids on the endothelium.
Collapse
Affiliation(s)
- Lars Lind
- Department of Medicine, Uppsala University Hospital and AstraZeneca R&D, Möndal, Sweden.
| |
Collapse
|
19
|
Arnal JF, Castano C, Maupas E, Mugniot A, Darblade B, Gourdy P, Michel JB, Bayard F. Omapatrilat, a dual angiotensin-converting enzyme and neutral endopeptidase inhibitor, prevents fatty streak deposit in apolipoprotein E-deficient mice. Atherosclerosis 2001; 155:291-5. [PMID: 11254898 DOI: 10.1016/s0021-9150(00)00565-7] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
Angiotensin-converting enzyme (ACE) is mainly responsible for converting angiotensin I (AI) to angiotensin II (AII), and ACE inhibitors prevent atherosclerosis in animal models. Neutral endopeptidase 24.11 (NEP) degrades substance P, kinins and atrial natriuretic peptide (ANP), and aortic wall NEP activity was found to be increased in atherosclerosis. In the present study, we have evaluated the effect of candoxatril, a NEP inhibitor, and of omapatrilat, a dual ACE and NEP inhibitor, on the development of fatty streak in apolipoprotein E (apoE)-deficient mice. Groups of ten male apoE-deficient mice were given either placebo, candoxatril 50 mg/kg per day, or omapatrilat 10, or 100 mg/kg per day for 4 months. None of the treatments influenced body weight, serum total or HDL-cholesterol. Compared with the placebo, candoxatril did not protect the mice from fatty streak deposit. In contrast, omapatrilat dose dependently inhibited the constitution of fatty streak in apoE-deficient mice. The precise advantages of the dual ACE and NEP inhibition versus the inhibition of only ACE should now be considered in the prevention of atherosclerosis as well as in the occurrence of its complications.
Collapse
Affiliation(s)
- J F Arnal
- INSERM U397 et laboratoire de Physiologie, Institut L. Bugnard, CHU Rangueil, 31403, Toulouse, France.
| | | | | | | | | | | | | | | |
Collapse
|
20
|
Affiliation(s)
- J C Burnett
- Department of Medicine, Mayo Clinic and Foundation, Rochester, Minnesota 55905, USA
| |
Collapse
|
21
|
Farina NK, Johnston CI, Burrell LM. Reversal of cardiac hypertrophy and fibrosis by S21402, a dual inhibitor of neutral endopeptidase and angiotensin converting enzyme in SHRs. J Hypertens 2000; 18:749-55. [PMID: 10872560 DOI: 10.1097/00004872-200018060-00013] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
OBJECTIVE The major advantage of dual inhibitors of neutral endopeptidase (NEP) and angiotensin converting enzyme (ACE) is their ability to lower blood pressure irrespective of renin or volume status. The aim of this study was to determine whether dual NEP/ACE inhibition produces different effects on cardiovascular structure and fibrosis, hormonal parameters and inhibition of tissue enzymes compared with selective inhibition of ACE and NEP in the spontaneously hypertensive rat (SHR). METHODS Male SHRs received the dual NEP/ACE inhibitor (S21402, 100 mg/kg per day), the ACE inhibitor (captopril, 50 mg/kg per day), the NEP inhibitor (SCH42495, 60 mg/kg per day) or vehicle for 2 weeks. RESULTS S21402 produced equivalent blood pressure lowering effects to captopril (vehicle, 220 +/- 1 mmHg; S21402, 189 +/- 2 mmHg; captopril, 187 +/- 3 mmHg), but was a more effective antihypertensive agent than SCH42495 (214 +/- 2 mmHg, P< 0.01). All treatments reduced left ventricular mass (P< 0.05) and cardiac fibrosis (P< 0.01). S21402 inhibited renal NEP and ACE (P< 0.01), SCH42495 inhibited renal NEP (P < 0.01), and captopril inhibited renal ACE (P< 0.01). Captopril and S21402 increased plasma renin activity (P< 0.05), but the rise with S21402 was attenuated compared with that caused by captopril (P< 0.01). All treatments reduced plasma aldosterone levels (P< 0.01), and NEP inhibition with SCH42495 and S21402 increased plasma atrial natriuretic peptide (ANP; P< 0.05). CONCLUSIONS These results indicate that selective NEP inhibition has major benefits in the regression of cardiac hypertrophy and reduction of fibrosis but has limited antihypertensive effects. The dual NEP/ACE inhibitor S21402 offered no advantage over the selective ACE inhibitor in terms of blood pressure reduction, or attenuation of cardiac hypertrophy and fibrosis, but did increase plasma ANP and blunted the reactive rise in renin with ACE inhibition. Further studies are needed to determine whether more complete blockade of the renin-angiotensin system with dual NEP/ACE inhibition results in additional benefits in terms of morbidity and mortality in cardiovascular disease.
Collapse
Affiliation(s)
- N K Farina
- Department of Medicine, University of Melbourne, Austin and Repatriation Medical Centre, Heidelberg, Victoria, Australia
| | | | | |
Collapse
|
22
|
Schirger JA, Grantham JA, Kullo IJ, Jougasaki M, Wennberg PW, Chen HH, Lisy O, Miller V, Simari RD, Burnett JC. Vascular actions of brain natriuretic peptide: modulation by atherosclerosis and neutral endopeptidase inhibition. J Am Coll Cardiol 2000; 35:796-801. [PMID: 10716485 DOI: 10.1016/s0735-1097(99)00593-8] [Citation(s) in RCA: 46] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/17/2022]
Abstract
OBJECTIVES We sought to define the vascular actions of the cardiac hormone brain natriuretic peptide (BNP) on cellular proliferation and cyclic guanosine monophosphate (cGMP) in human aortic vascular smooth muscle cells (HAVSMCs). Secondly, we investigated BNP and acetylcholine (ACh) vasorelaxations in aortic rings from normal and atherosclerotic rabbits in the presence and absence of long-term oral inhibition of neutral endopeptidase (NEP). BACKGROUND The vascular actions of BNP are not well defined, despite the presence of its receptor in vascular smooth muscle and the upregulation of NEP, the ectoenzyme that degrades BNP, in the vascular wall in atherosclerosis. METHODS HAVSMCs stimulated with fetal calf serum (FCS) were pulsed with bromodeoxyuridine (BrdU) with and without BNP. The HAVSMCs were incubated in the presence and absence of BNP to assess cGMP. Vasorelaxations to BNP and ACh were assessed in rings in normal and atherosclerotic rabbits in the presence and absence of long-term oral inhibition of NEP, together with assessment of atheroma formation. RESULTS FCS-stimulated BrdU uptake in HAVSMCs was suppressed with BNP. BNP potentiated cGMP in HAVSMCs. BNP resulted in potent vasorelaxation in normal isolated aortic rings, which were impaired in atherosclerotic versus normal rabbits and preserved with NEP inhibition, which also decreased atheroma formation. Relaxations to ACh, which were also impaired in atherosclerosis, were preserved with inhibition of NEP. CONCLUSIONS We conclude that BNP potently inhibits vascular smooth muscle cell proliferation and potentiates the generation of cGMP. BNP potently relaxes the normal rabbit aorta, and this response is impaired in atherosclerosis but preserved with inhibition of NEP, together with a reduction in atheroma formation and preservation of relaxations to ACh.
Collapse
Affiliation(s)
- J A Schirger
- Department of Internal Medicine, Mayo Clinic and Foundation, Rochester, Minnesota 55905, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
23
|
Murohara T, Kugiyama K, Ota Y, Doi H, Ogata N, Ohgushi M, Yasue H. Effects of atrial and brain natriuretic peptides on lysophosphatidylcholine-mediated endothelial dysfunction. J Cardiovasc Pharmacol 1999; 34:870-8. [PMID: 10598132 DOI: 10.1097/00005344-199912000-00015] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
Lysophosphatidylcholine (LPC), a major atherogenic lysophospholipid contained in oxidized low-density lipoprotein (ox-LDL), induces endothelial dysfunction. Recent studies showed that natriuretic peptides (NPs) have antiatherogenic properties by inhibiting vascular smooth-muscle cell proliferation, but their effects on endothelial cells are little known. We examined whether atrial and brain NPs (ANP and BNP) have a protecting action against LPC-induced endothelial dysfunction. LPC (10 microM) significantly inhibited thrombin (0.001-1 U/ml)-induced endothelium-dependent relaxation without affecting endothelium-independent relaxation to sodium nitroprusside in isolated porcine coronary arteries. The impaired endothelium-dependent relaxation induced by LPC was prevented by treatment with ANP or BNP (i microM). In cultured bovine aortic endothelial cells (BAECs), LPC (10 microM) significantly attenuated bradykinin (1 microM)-stimulated nitric oxide (NO) release; however, this was prevented by ANP and BNP. Because LPC-induced endothelial dysfunction has been shown to be mediated at least in part by activation of the protein kinase C (PKC)-dependent signaling pathway, we also examined the effects of ANP and BNP on LPC-induced modulation of PKC activities in BAECs. LPC (10 microM) significantly stimulated PKC activity in BAECs. However, ANP or BNP significantly inhibited LPC (10 microM)-induced PKC activation. In conclusion, ANP and BNP protected endothelial cells from LPC-induced dysfunction in both isolated coronary arteries and cultured ECs. The mechanism appears to be at least in part related to the inhibition of LPC-induced PKC activation by NPs. These new actions of ANP and BNP against lysolipid-induced endothelial cytotoxicity may partly account for antiatherogenic properties of NPs.
Collapse
Affiliation(s)
- T Murohara
- Department of Cardiology, St. Elizabeth's Medical Center, Tufts University School of Medicine, Boston, Massachusetts, USA
| | | | | | | | | | | | | |
Collapse
|
24
|
Yamaguchi M, Shirai H, Sato R, Kawabe Y, Fukuda R, Kodama T, Hamakubo T. Characterization of cleavage enzymes for sterol regulatory element binding protein in hamster liver microsomes. Biochem Biophys Res Commun 1999; 258:542-7. [PMID: 10329422 DOI: 10.1006/bbrc.1999.0679] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Sterol regulatory element binding proteins (SREBP-1 and SREBP-2) are the key transcription factors for the regulation of the cellular cholesterol level. To identify proteolytic enzymes for SREBPs, a fluorogenic peptide substrate, MOCAc-GRSVLSFK(Dnp)rr-NH2, was synthesized according to the proposed cleavage site of human SREBP-2. In microsome fractions from hamster liver, we found a peptidase activity inhibitable by the synthetic inhibitor Ac-GRSVL-aldehyde with an IC50 of 40 nM. This peptidase separated into three peaks of approximately 400 kDa, 60 kDa, and 30 kDa (Mp400, Mp60 and Mp30 respectively) upon gel permeation chromatography. Mp30 was purified to apparent homogeneity with an Mr of 32 kDa. The partial amino acid sequence of Mp30 possessed homology to cathepsin B (EC 3.4.22.1). A 109 kDa protein band on SDS-PAGE which corresponded to Mp400 exhibited homology to neprilysin (EC 3.4.24.11) in partial amino acid sequence. These findings suggest several degradative pathways for SREBP in liver microsome membranes.
Collapse
Affiliation(s)
- M Yamaguchi
- Research Center for Advanced Science and Technology, University of Tokyo, Meguro, Tokyo, 153-8904, Japan
| | | | | | | | | | | | | |
Collapse
|
25
|
Affiliation(s)
- M R Wilkins
- Department of Clinical Pharmacology, Royal Postgraduate Medical School, London, UK
| | | | | |
Collapse
|