1
|
Bahreyni A, Saeedi N, Al-Asady AM, Soleimani A, Ghorbani E, Khazaei M, Alaei M, Hanaei R, Ryzhikov M, Avan A, Hassanian SM. Therapeutic potency of A1 adenosine receptor antagonists in the treatment of cardiovascular diseases, current status and perspectives. Mol Biol Rep 2024; 51:358. [PMID: 38400849 DOI: 10.1007/s11033-024-09246-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2023] [Accepted: 01/11/2024] [Indexed: 02/26/2024]
Abstract
BACKGROUND Cardiomyocytes form, transport, and metabolize the omnipresent metabolite adenosine. Depending upon the adenosine concentrations and the pharmacological properties of receptor subtypes, adenosine exerts (patho)physiological responses in the cardiovascular system. The objective of this review is to present different protective mechanisms of A1-adenosine receptor inhibitors in cardiovascular diseases. METHODS AND RESULTS Literature references were collected and sorted using relevant keywords and key phrases as search terms in scientific databases such as Web of Science, PubMed and Google Scholar. A1 adenosine receptor regulates free fatty acid metabolism, lipolysis, heart rate, blood pressure, and cardiovascular toxicity. The evidence clearly supporting the therapeutic potency of pharmacological A1 adenosine receptors agonists and antagonists in modulating cardiovascular risk factor parameters and treatment of cardiovascular diseases. CONCLUSION This review summarizes the protective role of pharmacological A1-adenosine receptor regulators in the pathogenesis of cardiovascular diseases for a better management of cardiovascular diseases.
Collapse
Affiliation(s)
- Amirhossein Bahreyni
- Department of Clinical Biochemistry, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Nikoo Saeedi
- Student Research Committee, Islamic Azad University, Mashhad Branch, Mashhad, Iran
| | - Abdulridha Mohammed Al-Asady
- Department of Medical Sciences, Faculty of Nursing, University of Warith Al-Anbiyaa, Karbala, Iraq
- Department of pharmacology, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
- Department of Medical Sciences, Faculty of Dentistry, University of Kerbala, Karbala, Iraq
| | - Atena Soleimani
- Department of Clinical Biochemistry, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Elnaz Ghorbani
- Department of Medical Microbiology and virology, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Majid Khazaei
- Department of Medical Physiology, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
- Metabolic Syndrome Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Maryam Alaei
- Department of Clinical Biochemistry, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Reyhane Hanaei
- Metabolic Syndrome Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
| | | | - Amir Avan
- Metabolic Syndrome Research Center, Mashhad University of Medical Sciences, Mashhad, Iran.
- Department of Human Genetics, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran.
| | - Seyed Mahdi Hassanian
- Department of Clinical Biochemistry, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran.
- Metabolic Syndrome Research Center, Mashhad University of Medical Sciences, Mashhad, Iran.
- Department of Medical Biochemistry, School of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran.
| |
Collapse
|
2
|
Vilahur G, Gutiérrez M, Casani L, Varela L, Capdevila A, Pons-Lladó G, Carreras F, Carlsson L, Hidalgo A, Badimon L. Protective Effects of Ticagrelor on Myocardial Injury After Infarction. Circulation 2016; 134:1708-1719. [PMID: 27789556 DOI: 10.1161/circulationaha.116.024014] [Citation(s) in RCA: 96] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/15/2016] [Accepted: 10/08/2016] [Indexed: 11/16/2022]
Abstract
BACKGROUND The P2Y12 receptor antagonist ticagrelor has been shown to be clinically superior to clopidogrel. Although the underlying mechanisms remain elusive, ticagrelor may exert off-target effects through adenosine-related mechanisms. We aimed to investigate whether ticagrelor reduces myocardial injury to a greater extent than clopidogrel after myocardial infarction (MI) at a similar level of platelet inhibition and to determine the underlying mechanisms. METHODS Pigs received the following before MI induction: (1) placebo-control; (2) a loading dose of clopidogrel (600 mg); (3) a loading dose of ticagrelor (180 mg); or (4) a loading dose of ticagrelor followed by an adenosine A1/A2-receptor antagonist [8-(p-sulfophenyl)theophylline, 4 mg/kg intravenous] to determine the potential contribution of adenosine in ticagrelor-related cardioprotection. Animals received the corresponding maintenance doses of the antiplatelet agents during the following 24 hours and underwent 3T-cardiac MRI analysis. Platelet inhibition was monitored by ADP-induced platelet aggregation. In the myocardium, we assessed the expression and activation of proteins known to modulate edema formation, including aquaporin-4 and AMP-activated protein kinase and its downstream effectors CD36 and endothelial nitric oxide synthase and cyclooxygenase-2 activity. RESULTS Clopidogrel and ticagrelor exerted a high and consistent antiplatelet effect (68.2% and 62.2% of platelet inhibition, respectively, on challenge with 20 μmol/L ADP) that persisted up to 24 hours post-MI (P<0.05). All groups showed comparable myocardial area-at-risk and cardiac worsening after MI induction. 3T-Cardiac MRI analysis revealed that clopidogrel- and ticagrelor-treated animals had a significantly smaller extent of MI than placebo-control animals (15.7 g left ventricle and 12.0 g left ventricle versus 22.8 g left ventricle, respectively). Yet, ticagrelor reduced infarct size to a significantly greater extent than clopidogrel (further 23.5% reduction; P=0.0026), an effect supported by troponin-I assessment and histopathologic analysis (P=0.0021). Furthermore, in comparison with clopidogrel, ticagrelor significantly diminished myocardial edema by 24.5% (P=0.004), which correlated with infarct mass (r=0.73; P<0.001). 8-(p-Sulfophenyl)theophylline administration abolished the cardioprotective effects of ticagrelor over clopidogrel. At a molecular level, aquaporin-4 expression decreased and the expression and activation of AMP-activated protein kinase signaling and cyclooxygenase-2 increased in the ischemic myocardium of ticagrelor- versus clopidogrel-treated animals (P<0.05). These protein changes were not observed in those animals administered the adenosine receptor blocker 8-(p-sulfophenyl)theophylline. CONCLUSIONS Ticagrelor, beyond its antiplatelet efficacy, exerts cardioprotective effects by reducing necrotic injury and edema formation via adenosine-dependent mechanisms.
Collapse
Affiliation(s)
- Gemma Vilahur
- From Cardiovascular Research Center (CSIC-ICCC), IIB-HSCSP, Barcelona, Spain (G.V., L.C., L.V., L.B.); Radiology Unit, Hospital de la Santa Creu i Sant Pau (HSCSP), Barcelona, Spain (M.G., A.C., A.H.); Cardiology Unit. Hospital de la Santa Creu i Sant Pau (HSCSP), Barcelona, Spain (G.P.-L., F.C.); Cardiovascular and Metabolic Diseases, Innovative Medicines and Early Development Biotech Unit, AstraZeneca, Mölndal, Sweden (L.C.); and Cardiovascular Research Chair UAB (Autonomous University of Barcelona), Spain (L.B.)
| | - Manuel Gutiérrez
- From Cardiovascular Research Center (CSIC-ICCC), IIB-HSCSP, Barcelona, Spain (G.V., L.C., L.V., L.B.); Radiology Unit, Hospital de la Santa Creu i Sant Pau (HSCSP), Barcelona, Spain (M.G., A.C., A.H.); Cardiology Unit. Hospital de la Santa Creu i Sant Pau (HSCSP), Barcelona, Spain (G.P.-L., F.C.); Cardiovascular and Metabolic Diseases, Innovative Medicines and Early Development Biotech Unit, AstraZeneca, Mölndal, Sweden (L.C.); and Cardiovascular Research Chair UAB (Autonomous University of Barcelona), Spain (L.B.)
| | - Laura Casani
- From Cardiovascular Research Center (CSIC-ICCC), IIB-HSCSP, Barcelona, Spain (G.V., L.C., L.V., L.B.); Radiology Unit, Hospital de la Santa Creu i Sant Pau (HSCSP), Barcelona, Spain (M.G., A.C., A.H.); Cardiology Unit. Hospital de la Santa Creu i Sant Pau (HSCSP), Barcelona, Spain (G.P.-L., F.C.); Cardiovascular and Metabolic Diseases, Innovative Medicines and Early Development Biotech Unit, AstraZeneca, Mölndal, Sweden (L.C.); and Cardiovascular Research Chair UAB (Autonomous University of Barcelona), Spain (L.B.)
| | - Lourdes Varela
- From Cardiovascular Research Center (CSIC-ICCC), IIB-HSCSP, Barcelona, Spain (G.V., L.C., L.V., L.B.); Radiology Unit, Hospital de la Santa Creu i Sant Pau (HSCSP), Barcelona, Spain (M.G., A.C., A.H.); Cardiology Unit. Hospital de la Santa Creu i Sant Pau (HSCSP), Barcelona, Spain (G.P.-L., F.C.); Cardiovascular and Metabolic Diseases, Innovative Medicines and Early Development Biotech Unit, AstraZeneca, Mölndal, Sweden (L.C.); and Cardiovascular Research Chair UAB (Autonomous University of Barcelona), Spain (L.B.)
| | - Antoni Capdevila
- From Cardiovascular Research Center (CSIC-ICCC), IIB-HSCSP, Barcelona, Spain (G.V., L.C., L.V., L.B.); Radiology Unit, Hospital de la Santa Creu i Sant Pau (HSCSP), Barcelona, Spain (M.G., A.C., A.H.); Cardiology Unit. Hospital de la Santa Creu i Sant Pau (HSCSP), Barcelona, Spain (G.P.-L., F.C.); Cardiovascular and Metabolic Diseases, Innovative Medicines and Early Development Biotech Unit, AstraZeneca, Mölndal, Sweden (L.C.); and Cardiovascular Research Chair UAB (Autonomous University of Barcelona), Spain (L.B.)
| | - Guillem Pons-Lladó
- From Cardiovascular Research Center (CSIC-ICCC), IIB-HSCSP, Barcelona, Spain (G.V., L.C., L.V., L.B.); Radiology Unit, Hospital de la Santa Creu i Sant Pau (HSCSP), Barcelona, Spain (M.G., A.C., A.H.); Cardiology Unit. Hospital de la Santa Creu i Sant Pau (HSCSP), Barcelona, Spain (G.P.-L., F.C.); Cardiovascular and Metabolic Diseases, Innovative Medicines and Early Development Biotech Unit, AstraZeneca, Mölndal, Sweden (L.C.); and Cardiovascular Research Chair UAB (Autonomous University of Barcelona), Spain (L.B.)
| | - Francesc Carreras
- From Cardiovascular Research Center (CSIC-ICCC), IIB-HSCSP, Barcelona, Spain (G.V., L.C., L.V., L.B.); Radiology Unit, Hospital de la Santa Creu i Sant Pau (HSCSP), Barcelona, Spain (M.G., A.C., A.H.); Cardiology Unit. Hospital de la Santa Creu i Sant Pau (HSCSP), Barcelona, Spain (G.P.-L., F.C.); Cardiovascular and Metabolic Diseases, Innovative Medicines and Early Development Biotech Unit, AstraZeneca, Mölndal, Sweden (L.C.); and Cardiovascular Research Chair UAB (Autonomous University of Barcelona), Spain (L.B.)
| | - Leif Carlsson
- From Cardiovascular Research Center (CSIC-ICCC), IIB-HSCSP, Barcelona, Spain (G.V., L.C., L.V., L.B.); Radiology Unit, Hospital de la Santa Creu i Sant Pau (HSCSP), Barcelona, Spain (M.G., A.C., A.H.); Cardiology Unit. Hospital de la Santa Creu i Sant Pau (HSCSP), Barcelona, Spain (G.P.-L., F.C.); Cardiovascular and Metabolic Diseases, Innovative Medicines and Early Development Biotech Unit, AstraZeneca, Mölndal, Sweden (L.C.); and Cardiovascular Research Chair UAB (Autonomous University of Barcelona), Spain (L.B.)
| | - Alberto Hidalgo
- From Cardiovascular Research Center (CSIC-ICCC), IIB-HSCSP, Barcelona, Spain (G.V., L.C., L.V., L.B.); Radiology Unit, Hospital de la Santa Creu i Sant Pau (HSCSP), Barcelona, Spain (M.G., A.C., A.H.); Cardiology Unit. Hospital de la Santa Creu i Sant Pau (HSCSP), Barcelona, Spain (G.P.-L., F.C.); Cardiovascular and Metabolic Diseases, Innovative Medicines and Early Development Biotech Unit, AstraZeneca, Mölndal, Sweden (L.C.); and Cardiovascular Research Chair UAB (Autonomous University of Barcelona), Spain (L.B.)
| | - Lina Badimon
- From Cardiovascular Research Center (CSIC-ICCC), IIB-HSCSP, Barcelona, Spain (G.V., L.C., L.V., L.B.); Radiology Unit, Hospital de la Santa Creu i Sant Pau (HSCSP), Barcelona, Spain (M.G., A.C., A.H.); Cardiology Unit. Hospital de la Santa Creu i Sant Pau (HSCSP), Barcelona, Spain (G.P.-L., F.C.); Cardiovascular and Metabolic Diseases, Innovative Medicines and Early Development Biotech Unit, AstraZeneca, Mölndal, Sweden (L.C.); and Cardiovascular Research Chair UAB (Autonomous University of Barcelona), Spain (L.B.).
| |
Collapse
|
3
|
Downey JM, Cohen MV. A2B or not 2B: that is the question: AUTHORS' RETROSPECTIVE. Cardiovasc Res 2012. [DOI: 10.1093/cvr/cvs214] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/14/2022] Open
|
4
|
McIntosh VJ, Lasley RD. Adenosine receptor-mediated cardioprotection: are all 4 subtypes required or redundant? J Cardiovasc Pharmacol Ther 2011; 17:21-33. [PMID: 21335481 DOI: 10.1177/1074248410396877] [Citation(s) in RCA: 88] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/24/2023]
Abstract
Adenosine is a purine nucleoside, which is produced primarily through the metabolism of adenosine triphosphate (ATP), therefore its levels increase during stressful situations when ATP utilization increases. Adenosine exerts potent cardioprotective effects on the ischemic/reperfused heart, reducing reversible and irreversible myocardial injury. Adenosine receptors (ARs) are G-protein-coupled receptors, and 4 subtypes exist--A(1), A(2A), A(2B), and A(3), all of which have been shown to be cardioprotective. Adenosine receptors are expressed on multiple cardiac cells, including fibroblasts, endothelial cells, smooth muscle cells, and myocytes. Activation of both A(1) and A(3) receptors prior to ischemia has been shown in multiple experimental models to reduce ischemia/reperfusion-induced cardiac injury. Additionally, activation of the A(2A) receptor at the onset of reperfusion has been shown to reduce injury. Most recently, there is evidence that the A(2B) receptor has cardioprotective effects upon its activation. However, controversy remains regarding the precise timing of activation of these receptors required to induce cardioprotection, as well as their involvement in ischemic preconditioning and postconditioning. Adenosine receptors have been suggested to reduce cell death through actions at the mitochondrial ATP-dependent potassium (K(ATP)) channel, as well as protein kinase C and mitogen-activated protein kinase (MAPK) signaling. Additionally, the ability of ARs to interact has been documented, and several recent reports suggest that these interactions play a role in AR-mediated cardioprotection. This review summarizes the current knowledge of the cardioprotective effects of each AR subtype, as well as the proposed mechanisms of AR cardioprotection. Additionally, the role of AR interactions in cardioprotection is discussed.
Collapse
Affiliation(s)
- Victoria J McIntosh
- Department of Physiology and Cardiovascular Research Institute, Wayne State University School of Medicine, Detroit, MI, USA
| | | |
Collapse
|
5
|
AMP579 is revealed to be a potent A2b-adenosine receptor agonist in human 293 cells and rabbit hearts. Basic Res Cardiol 2010; 105:129-37. [PMID: 19730798 DOI: 10.1007/s00395-009-0056-9] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/26/2009] [Revised: 08/07/2009] [Accepted: 08/13/2009] [Indexed: 12/11/2022]
Abstract
The mixed A1/A2a-adenosine agonist AMP579 given at reperfusion is protective in animal models of myocardial infarction. Receptor-blocking studies have indicated that the protection came from an adenosine receptor (AR), but neither A1- nor A2a-selective agonists could duplicate its protection. We recently found that A2b-selective agonists given at reperfusion are protective, and, therefore, tested whether AMP579 might also be an A2b agonist. We used human embryonic kidney cells overexpressing human A2b receptors as an assay system. In these cells, A2b receptor occupancy causes phosphorylation of ERK. AMP579 induced ERK phosphorylation with an EC50 of 250 nM and this phosphorylation could be blocked by MRS1754 or PSB1115, two highly selective blockers of human A2b receptors. We attempted to confirm our A2b hypothesis in a rabbit heart model of ischemia-reperfusion. AMP579 (500 nM) for 1 h starting at reperfusion reduced infarct size in isolated rabbit hearts exposed to 30 min of regional ischemia and 2 h of reperfusion (12.9 +/- 2.2% infarction of risk zone vs. 32.0 +/- 1.9% in untreated hearts). PSB1115 (500 nM) given for the first 15 min of reperfusion blocked AMP579's protection (32.2 +/- 3.1% infarction) which is consistent with an A2b mechanism. We conclude that AMP579 is a non-selective, but potent A2b-AR agonist, and that its protection against infarction is through that receptor.
Collapse
|
6
|
Pharmacological targets revealed by myocardial postconditioning. Curr Opin Pharmacol 2009; 9:177-88. [DOI: 10.1016/j.coph.2008.11.009] [Citation(s) in RCA: 31] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2008] [Revised: 11/11/2008] [Accepted: 11/11/2008] [Indexed: 12/22/2022]
|
7
|
Abstract
Adenosine, a catabolite of ATP, exerts numerous effects in the heart, including modulation of the cardiac response to stress, such as that which occurs during myocardial ischemia and reperfusion. Over the past 20 years, substantial evidence has accumulated that adenosine, administered either prior to ischemia or during reperfusion, reduces both reversible and irreversible myocardial injury. The latter effect results in a reduction of both necrosis or myocardial infarction (MI) and apoptosis. These effects appear to be mediated via the activation of one or more G-protein-coupled receptors (GPCRs), referred to as A(1), A(2A), A(2B) and A(3) adenosine receptor (AR) subtypes. Experimental studies in different species and models suggest that activation of the A(1) or A(3)ARs prior to ischemia is cardioprotective. Further experimental studies reveal that the administration of A(2A)AR agonists during reperfusion can also reduce MI, and recent reports suggest that A(2B)ARs may also play an important role in modulating myocardial reperfusion injury. Despite convincing experimental evidence for AR-mediated cardioprotection, there have been only a limited number of clinical trials examining the beneficial effects of adenosine or adenosine-based therapeutics in humans, and the results of these studies have been equivocal. This review summarizes our current knowledge of AR-mediated cardioprotection, and the roles of the four known ARs in experimental models of ischemia-reperfusion. The chapter concludes with an examination of the clinical trials to date assessing the safety and efficacy of adenosine as a cardioprotective agent during coronary thrombolysis in humans.
Collapse
Affiliation(s)
- John P Headrick
- Heart Foundation Research Centre, School of Medical Science, Griffith University, Southport, Queensland, 4217, Australia.
| | | |
Collapse
|
8
|
Abstract
Adenosine, a purine nucleoside, is ubiquitous in the body, and is a critical component of ATP. Its concentration jumps 100-fold during periods of oxygen depletion and ischemia. There are four adenosine receptors: A(1) and A(3) coupled to G(i/o) and the high-affinity A(2A) and low-affinity A(2B) coupled to G(s). Adenosine is one of three autacoids released by ischemic tissue which are important triggers of ischemic preconditioning (IPC). It is the A(1) and to some extent A(3) receptors which participate in the intracellular signaling that triggers cardioprotection. Unlike bradykinin and opioids, the other two autacoids, adenosine is not dependent on opening of mitochondrial K(ATP) channels or release of reactive oxygen species (ROS), but rather activates phospholipase C and/or protein kinase C (PKC) directly. Another signaling cascade at reperfusion involves activated PKC which initiates binding to and activation of an A(2) adenosine receptor that we believe is the A(2B). Although the latter is the low-affinity receptor, its interaction with PKC increases its affinity and makes it responsive to the accumulated tissue adenosine. A(2B) agonists, but not adenosine or A(1) agonists, infused at reperfusion can initiate this second signaling cascade and mimic preconditioning's protection. The same A(2B) receptors are critical for postconditioning's protection. Thus adenosine is both an important trigger and a mediator of cardioprotection.
Collapse
|
9
|
Clark KL, Merkel L, Zannikos P, Kelley MF, Boutouyrie B, Perrone MH. AMP 579, a Novel Adenosine Agonist for the Treatment of Acute Myocardial Infarction. ACTA ACUST UNITED AC 2006. [DOI: 10.1111/j.1527-3466.2000.tb00043.x] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
|
10
|
Wang X, Wu BW, Wu DM. Effects of AMP579 and adenosine on L-type Ca2+ current in isolated rat ventricular myocytes. Acta Pharmacol Sin 2005; 26:559-62. [PMID: 15842773 DOI: 10.1111/j.1745-7254.2005.00107.x] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
Abstract
AIM To compare the effects of AMP579 and adenosine on L-type Ca2+ current (I(Ca-L)) in rat ventricular myocytes and explore the mechanism by which AMP579 acts on I(Ca-L). METHODS I(Ca-L) was recorded by patch-clamp technique in whole-cell configuration. RESULTS Adenosine (10 nmol/L to 50 micromol/L) showed no effect on basal I(Ca-L), but it inhibited the I(Ca-L) induced by isoproterenol 10 nmol/L in a concentration-dependent manner with the IC(50) of 13.06 micromol/L. Similar to adenosine, AMP579 also showed an inhibitory effect on the I(Ca-L) induced by isoproterenol. AMP579 and adenosine (both in 10 micromol/L) suppressed isoproterenol-induced ICa-L by 11.1% and 5.2%, respectively. In addition, AMP579 had a direct inhibitory effect on basal I(Ca-L) in a concentration-dependent manner with IC50 (1.17 micromol/L). PD116948 (30 micromol/L), an adenosine A1 receptor blocker, showed no action on the inhibitory effect of AMP579 on basal I(Ca-L). However, GF109203X (0.4 micromol/L), a special protein kinase C (PKC) blocker, could abolish the inhibitory effect of AMP579 on basal I(Ca-L). So the inhibitory effect of AMP579 on basal I(Ca-L) was induced through activating PKC, but not linked to adenosine A1 receptor. CONCLUSION AMP579 shows a stronger inhibitory effect than adenosine on the I(Ca-L) induced by isoproterenol. AMP579 also has a strong inhibitory effect on basal I(Ca-L) in rat ventricular myocytes. Activation of PKC is involved in the inhibitory effect of AMP579 on basal I(Ca-L) at downstream-mechanism.
Collapse
Affiliation(s)
- Xiong Wang
- Department of Physiology, Shanxi Medical University, Taiyuan 030001, China.
| | | | | |
Collapse
|
11
|
Clark KL, Merkel L. Potential of adenosine receptor agonists for the prevention and treatment of coronary artery disease and acute myocardial infarction. ACTA ACUST UNITED AC 2005. [DOI: 10.1517/14728214.5.1.89] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
|
12
|
Kristo G, Yoshimura Y, Ferraris SP, Jahania SA, Mentzer RM, Lasley RD. The preischemic combination of the sodium–hydrogen exchanger inhibitor cariporide and the adenosine agonist AMP579 acts additively to reduce porcine myocardial infarct size. J Am Coll Surg 2004; 199:586-94. [PMID: 15454144 DOI: 10.1016/j.jamcollsurg.2004.05.274] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2003] [Revised: 05/05/2004] [Accepted: 05/11/2004] [Indexed: 11/13/2022]
Abstract
BACKGROUND We tested whether the combination of two known cardioprotective agents, the type-1 sodium-hydrogen exchanger inhibitor cariporide plus the adenosine A(1)/A(2a) receptor agonist AMP579 ([1S-[1a,2b,3b, 4a(S*)]]-4-[7-[[2-(3-chloro-2-thienyl)-1-methylpropyl]amino]-[(3)H]-imidazo[4,5-b]pyridyl-3-yl]cyclopentane carboxamide), acted additively to reduce myocardial infarct size. STUDY DESIGN Pigs underwent 1 hour of coronary artery occlusion and 3 hours reperfusion. Vehicle-treated controls were compared with animals treated before ischemia with low-dose and high-dose cariporide and AMP579, and low-dose cariporide plus AMP579. The effects of both agents, alone and in combination, were also tested in isolated porcine polymorphonuclear neutrophils (PMNs). The PMN respiratory burst was induced with phorbol 12-myristate 13-acetate and quantified by the increase in 2',7'-dichlorofluorescein fluorescence, measured by flow cytometry. RESULTS Infarct size in the control pigs was 65 +/- 1% of the area at risk. Cariporide dose-dependently reduced infarct size to 39 +/- 2% and 24 +/- 3% in the low- and high-dose groups, respectively. Infarct size was 54 +/- 3% in the low-dose AMP579 group and 47 +/- 3% with high dose. The combination of low doses of cariporide and AMP579 reduced infarction to 25 +/- 6% of the area at risk. In the PMN studies, cariporide and AMP579 alone had no effect on 2',7'-dichlorofluorescein fluorescence, but the combination of the two agents reduced the PMN 2',7'-dichlorofluorescein increase to 79 +/- 5% of the vehicle control response. CONCLUSIONS The preischemic combination of low doses of cariporide and AMP579 decreased myocardial infarct size more than either agent used alone in low concentration, indicating an additive effect of the two agents. Given the effects that cariporide plus AMP579 combination exerted on PMN activity, it appears that this combination has the potential to reduce PMN-mediated effects during myocardial reperfusion.
Collapse
Affiliation(s)
- Gentian Kristo
- Department of Surgery, University of Kentucky, College of Medicine, Lexington, KY 40536-0084, USA
| | | | | | | | | | | |
Collapse
|
13
|
Bosch MP, Campos F, Niubó I, Rosell G, Díaz JL, Brea J, Loza MI, Guerrero A. Synthesis and biological activity of new potential agonists for the human adenosine A2A receptor. J Med Chem 2004; 47:4041-53. [PMID: 15267242 DOI: 10.1021/jm031143+] [Citation(s) in RCA: 32] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
New adenosine derivatives have been synthesized and tested as putative agonists of adenosine receptors. Compounds 2-6 derive from the introduction of several types of substituents (electron donating, electron withdrawing, and halogens) in the para-position of the phenyl ring of the parent compound 1, and compound 7 lacks the hydroxyl group of amino alcohol 1. In radioligand binding assays using recombinant human A(1), A(2A), A(2B), and A(3) receptors, all compounds showed very low or negligible affinity for A(1) and A(2B) receptors but compounds 3, 5, and 7 displayed a remarkably potent affinity for the A(2A) receptor with K(i) values of 1-5 nM. Bromo derivative 3 displayed a selectivity A(1)/A(2A) = 62 and A(3)/A(2A) = 16 whereas the presence of a hydroxyl group (compound 5) improved the selectivity of A(1)/A(2A) and A(3)/A(2A) to 120- and 28-fold, respectively. When the methoxy derivative 4 lacks the hydroxyl group on the side chain (compound 7), the binding affinity for A(2A) is increased to 1 nM, improving selectivity ratios to 356- and 100-fold against A(1) and A(3), respectively. In Chinese hamster ovary cells transfected with human A(2A) and A(2B) receptors, most compounds showed a remarkable activity for the A(2A) receptor, except chloro derivative 2, with EC(50) values ranging from 1.4 to 8.8 nM. The compounds behaved as good A(2A) agonists, and all were more selective than 5'-(N-ethylcarboxamino)adenosine (NECA), with A(2B)/A(2A) ratios of cAMP accumulation ranging from 48 for compound 2 to 666 for compound 7 while the corresponding A(2B)/A(2A) ratio for NECA was only 9. Compounds 1, 3, 5, and 7 also displayed higher selectivities than NECA up to 100-fold in isolated aortas of rat and guinea pig. In guinea pig tracheal rings precontracted by carbachol, compounds 2 and 4 were more potent than adenosine (100-fold) and NECA (10-fold), whereas compounds 1 and 7 displayed similar effects to NECA. Pretreatment of the tracheal rings with A(2), A(2A), and A(2B) receptor antagonists 3,7-dimethyl-l-propargylxanthine, 8-(3-chlorostyryl)caffeine, and alloxazine produced a marked inhibition of the tracheal relaxations induced by compounds 1, 2, and 4, but none of the compounds showed selectivity toward any of the adenosine receptors.
Collapse
Affiliation(s)
- M Pilar Bosch
- Department of Biological Organic Chemistry, IIQAB (CSIC), 08034 Barcelona, Spain.
| | | | | | | | | | | | | | | |
Collapse
|
14
|
Kristo G, Yoshimura Y, Keith BJ, Stevens RM, Jahania SA, Mentzer RM, Lasley RD. Adenosine A1/A2a receptor agonist AMP-579 induces acute and delayed preconditioning against in vivo myocardial stunning. Am J Physiol Heart Circ Physiol 2004; 287:H2746-53. [PMID: 15271662 DOI: 10.1152/ajpheart.00493.2004] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
The purpose of this study was to determine whether the adenosine A1/A2a receptor agonist AMP-579 induces acute and delayed preconditioning against in vivo myocardial stunning. Regional stunning was produced by 15 min of coronary artery occlusion and 3 h of reperfusion (RP) in anesthetized open-chest pigs. In acute protection studies, animals were pretreated with saline, low-dose AMP-579 (15 microg/kg iv bolus 10 min before ischemia), or high-dose AMP-579 (50 microg/kg iv at 14 microg/kg bolus + 1.2 microg.kg(-1).min(-1) for 30 min before coronary occlusion). The delayed preconditioning effects of AMP-579 were evaluated 24 h after administration of saline vehicle or high-dose AMP-579 (50 microg/kg iv). Load-insensitive contractility was assessed by measuring regional preload recruitable stroke work (PRSW) and PRSW area. Acute preconditioning with AMP-579 dose dependently improved regional PRSW: 129 +/- 5 and 100 +/- 2% in high- and low-dose AMP-579 groups, respectively, and 78 +/- 5% in the control group at 3 h of RP. Administration of the adenosine A1 receptor antagonist 8-cyclopentyl-1,3-dipropylxanthine (0.7 mg/kg) blocked the acute protective effect of high-dose AMP-579, indicating that these effects are mediated through A1 receptor activation. Delayed preconditioning with AMP-579 significantly increased recovery of PRSW area: 64 +/- 5 vs. 33 +/- 5% in control at 3 h of RP. In isolated perfused rat heart studies, kinetics of the onset and washout of AMP-579 A1 and A2a receptor-mediated effects were distinct compared with those of other adenosine receptor agonists. The unique nature of the adenosine agonist AMP-579 may play a role in its ability to induce delayed preconditioning against in vivo myocardial stunning.
Collapse
Affiliation(s)
- Gentian Kristo
- Dept. of Surgery, Univ. of Kentucky College of Medicine, 800 Rose St., Lexington, KY 40536-0298, USA
| | | | | | | | | | | | | |
Collapse
|
15
|
Zhang SJ, Yang XM, Liu GS, Cohen MV, Pemberton K, Downey JM. CGX-1051, A Peptide from Conus Snail Venom, Attenuates Infarction in Rabbit Hearts When Administered at Reperfusion. J Cardiovasc Pharmacol 2003; 42:764-71. [PMID: 14639099 DOI: 10.1097/00005344-200312000-00011] [Citation(s) in RCA: 30] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
CGX-1051, isolated from the venom of the marine snail Conus purpurasens, was previously noted to interact with potassium channels. Since potassium channels play an important role in cardiac physiology, we assessed the effect of CGX-1051 on infarct size in a rabbit heart model of ischemia/reperfusion. A coronary branch was occluded for 30 minutes followed by 3 hours of reperfusion in in situ and 2 hours in in vitro preparations. Infarct size was measured with triphenyltetrazolium chloride staining and expressed as a percent of the risk zone. In in situ studies, a bolus intravenous injection of CGX-1051, either 10 or 100 microg/kg, administered 5 minutes before reperfusion, reduced infarct size from 40.4 +/- 2.8% of the risk zone in untreated animals to 19.8 +/- 3.8% and 15.0 +/- 1.9%, respectively. One microg/kg CGX-1051 was not protective. To see if the salvage was sustained, two groups of rabbits underwent 72 hours of reperfusion. The dose of 10 microg/kg infused 5 minutes before reperfusion reduced infarct size from 37.0 +/- 1.6% in untreated rabbits to 15.5 +/- 2.0%. When administered 10 minutes after reperfusion had begun, 100 microg/kg CGX-1051 had no effect. CGX-1051 also reduced infarct size in crystalloid-perfused, isolated rabbit hearts suggesting that protection did not depend on circulating leukocytes. The mitochondrial KATP inhibitors glibenclamide and 5-hydroxydecanoate and the MEK(1/2), ERK and hence, inhibitor PD 98059 aborted protection from CGX-1051. These data indicate that functionally active ERK and mitochondrial KATP channels are necessary for protection. CGX-1051 caused no hemodynamic alterations at any dose tested. We conclude that CGX-1051 has a powerful anti-infarct effect when given just before reperfusion.
Collapse
Affiliation(s)
- Shi Jun Zhang
- Department of Physiology, University of South Alabama, College of Medicine, Mobile, Alabama 36688, USA
| | | | | | | | | | | |
Collapse
|
16
|
Xu Z, Downey JM, Cohen MV. Timing and Duration of Administration Are Crucial for Antiinfarct Effect of AMP 579 Infused at Reperfusion in Rabbit Heart. ACTA ACUST UNITED AC 2003; 5:368-71. [PMID: 14633317 DOI: 10.1097/01.hdx.0000098614.29006.a7] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
The adenosine A(1)/A(2) receptor agonist AMP 579 has been reported to protect the heart against infarction even when administered after the onset of ischemia. The present study explored both the timing and the duration of treatment required to limit infarct size in in situ rabbit hearts subjected to 30 min of regional ischemia and 3 hours of reperfusion. In groups 1 and 2, AMP 579 was infused from 10 min before reperfusion and continued for either 30 or 40 min. In group 3, AMP 579 was begun at the onset of reperfusion and continued for 70 min. In group 4, AMP 579 was also infused for 70 min but begun 10 min after reperfusion. In untreated hearts 36.4 +/- 3.1% of the risk zone infarcted. Protection was observed only in hearts having a 70-minute infusion of AMP 579 starting at reperfusion (13.0 +/- 1.9%, P < 0.05). Therefore, AMP 579 must be present at the moment of reperfusion and have a continued presence of more than 30 min thereafter to protect. Importantly, because AMP 579 can protect when administered up to the time of reperfusion, it likely prevents a reperfusion injury, and, therefore, has impressive clinical potential.
Collapse
Affiliation(s)
- Zhelong Xu
- Departments of Physiology,University of South Alabama, College of Medicine, Mobile, AL 36688, USA
| | | | | |
Collapse
|
17
|
Auchampach JA, Ge ZD, Wan TC, Moore J, Gross GJ. A3 adenosine receptor agonist IB-MECA reduces myocardial ischemia-reperfusion injury in dogs. Am J Physiol Heart Circ Physiol 2003; 285:H607-13. [PMID: 12689858 PMCID: PMC3860822 DOI: 10.1152/ajpheart.01001.2002] [Citation(s) in RCA: 51] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/16/2023]
Abstract
We examined the effect of the A3 adenosine receptor (AR) agonist IB-MECA on infarct size in an open-chest anesthetized dog model of myocardial ischemia-reperfusion injury. Dogs were subjected to 60 min of left anterior descending (LAD) coronary artery occlusion and 3 h of reperfusion. Infarct size and regional myocardial blood flow were assessed by macrohistochemical staining with triphenyltetrazolium chloride and radioactive microspheres, respectively. Four experimental groups were studied: vehicle control (50% DMSO in normal saline), IB-MECA (100 microg/kg iv bolus) given 10 min before the coronary occlusion, IB-MECA (100 microg/kg iv bolus) given 5 min before initiation of reperfusion, and IB-MECA (100 microg/kg iv bolus) given 10 min before coronary occlusion in dogs pretreated 15 min earlier with the ATP-dependent potassium channel antagonist glibenclamide (0.3 mg/kg iv bolus). Administration of IB-MECA had no effect on any hemodynamic parameter measured including heart rate, first derivative of left ventricular pressure, aortic pressure, LAD coronary blood flow, or coronary collateral blood flow. Nevertheless, pretreatment with IB-MECA before coronary occlusion produced a marked reduction in infarct size ( approximately 40% reduction) compared with the control group (13.0 +/- 3.2% vs. 25.2 +/- 3.7% of the area at risk, respectively). This effect of IB-MECA was blocked completely in dogs pretreated with glibenclamide. An equivalent reduction in infarct size was observed when IB-MECA was administered immediately before reperfusion (13.1 +/- 3.9%). These results are the first to demonstrate efficacy of an A3AR agonist in a large animal model of myocardial infarction by mechanisms that are unrelated to changes in hemodynamic parameters and coronary blood flow. These data also demonstrate in an in vivo model that IB-MECA is effective as a cardioprotective agent when administered at the time of reperfusion.
Collapse
Affiliation(s)
- John A Auchampach
- Cardiovascular Research Center, Department of Pharmacology and Toxicology, Medical College of Wisconsin, 8701 Watertown Plank Rd., Milwaukee, WI 53226, USA.
| | | | | | | | | |
Collapse
|
18
|
Kopecky SL, Aviles RJ, Bell MR, Lobl JK, Tipping D, Frommell G, Ramsey K, Holland AE, Midei M, Jain A, Kellett M, Gibbons RJ. A randomized, double-blinded, placebo-controlled, dose-ranging study measuring the effect of an adenosine agonist on infarct size reduction in patients undergoing primary percutaneous transluminal coronary angioplasty: the ADMIRE (AmP579 Delivery for Myocardial Infarction REduction) study. Am Heart J 2003; 146:146-52. [PMID: 12851624 DOI: 10.1016/s0002-8703(03)00172-8] [Citation(s) in RCA: 78] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
BACKGROUND Evidence suggests that myocardial ischemic preconditioning and reperfusion injury may be mediated by adenosine A1 and A2 receptors. AMP579 is a mixed adenosine agonist with both A1 and A2 effects. In animal models of acute myocardial infarction (MI), AMP579 reduced infarct size at serum levels of 15 to 24 ng/mL. METHODS The AMP579 Delivery for Myocardial Infarction REduction study evaluated AMP579 in a double-blind, multicenter, placebo-controlled trial of 311 patients undergoing primary percutaneous transluminal coronary angioplasty (PTCA) after acute ST-segment elevation MI. Patients were randomly assigned to placebo or to 3 different doses of AMP579 continuously infused over 6 hours. The primary end point was final MI size measured by technetium Tc-99m sestamibi scanning at 120 to 216 hours after PTCA. Secondary end points included myocardial salvage and salvage index at the same time interval (in a subset of patients who underwent baseline technetium Tc-99m sestamibi scan), left ventricular ejection fraction and heart failure at 4 to 6 weeks, duration of hospitalization, and cardiac events at 4 weeks and 6 months. RESULTS Final infarct size did not differ among the placebo group and the active treatment groups for either anterior MI or nonanterior MI. In patients with anterior MI, median myocardial salvage was increasingly higher in the groups receiving ascending dosages of AMP579 plus PTCA. Serum levels approaching levels shown to reduce infarct size in animal models were achieved only in the 60-mcg/kg treatment group. CONCLUSION AMP579 was safe at the doses tested, but it did not reduce infarct size. There was a trend toward greater myocardial salvage in treated patients with anterior MI.
Collapse
Affiliation(s)
- Stephen L Kopecky
- Mayo Alliance for Clinical Trials, Mayo Clinic and Mayo Foundation, Rochester, MN 55902, USA.
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
19
|
Bès S, Ponsard B, El Asri M, Tissier C, Vandroux D, Rochette L, Athias P. Assessment of the cytoprotective role of adenosine in an in vitro cellular model of myocardial ischemia. Eur J Pharmacol 2002; 452:145-54. [PMID: 12354564 DOI: 10.1016/s0014-2999(02)02295-1] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
This work aimed to detect functional adenosine receptors in isolated rat cardiomyocytes and to study the influence of stimulation of these receptors in an in vitro model of ischemia. Cultures of cardiomyocytes were prepared from newborn rat ventricles. The contractions were photometrically monitored. In this preparation, adenosine induced a positive chronotropic response. This effect was reproduced by CGS 21680 (2-(4-[2-carboxyethyl]-phen-ethyl-amino) adenosine-5'N-ethylunosamide), a specific adenosine A(2) receptor agonist, and antagonized by DMPX (3,7-dimethyl-1-propargylxanthine), an adenosine A(2) receptor antagonist. However, R-PIA (R-N(6)-(2-phenylisopropyl)-adenosine; a specific adenosine A(1) receptor agonist) induced a negative chronotropic effect that was abolished by its corresponding adenosine A(1) antagonist DPCPX (1,3-dipropyl-8-cyclo-pentyl-adenosine). Substrate-free hypoxia, as simulation of ischemia, induced a progressive decrease and then arrest of spontaneous cell contractions. The spontaneous rhythmic contractile activity was restored during reoxygenation following simulated ischemia. Adenosine A(1) receptor stimulation with R-PIA induced a decrease of hypoxia-induced damage. This effect was antagonized by DPCPX, an adenosine A(1) receptor antagonist. Conversely, the cells treated with CGS 21680 did not display complete recovery after reoxygenation. In addition, this effect was abolished by DMPX, since the cells recovered normal function after reoxygenation. To conclude, it appeared that cardiomyocytes possess both functional adenosine A(1) and A(2) receptors and that only the activation of adenosine A(1) receptor had a cytoprotective effect against simulated ischemia-induced cardiac cell injury.
Collapse
Affiliation(s)
- Sandrine Bès
- Laboratory of Physiopathology and Pharmacology, Institute of Cardiovascular Research, University Hospital Center, 2 Boulevard Maréchal de Lattre de Tassigny, 21034 Cedex, Dijon, France
| | | | | | | | | | | | | |
Collapse
|
20
|
Xu Z, Jiao Z, Cohen MV, Downey JM. Protection from AMP 579 can be added to that from either cariporide or ischemic preconditioning in ischemic rabbit heart. J Cardiovasc Pharmacol 2002; 40:510-8. [PMID: 12352312 DOI: 10.1097/00005344-200210000-00003] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
AMP 579, an adenosine A /A receptor agonist, is cardioprotective when administered at reperfusion. Pretreatment with the Na /H exchanger inhibitor cariporide or ischemic preconditioning (PC) also limits infarct size. To gain insight into the mechanism of AMP 579 we investigated whether its protection could be added to that from either cariporide or PC. rabbit hearts were subjected to 45 min of regional ischemia followed by 3 h of reperfusion. Infarct size in the control group was 55.8 +/- 3.9% of the risk zone. PC significantly reduced infarct size to 26.0 +/- 6.7% (p<0.05). AMP 579 (30 micro g/kg) given just before reperfusion followed by 3 micro g/kg/min infusion for 70 min also limited infarct size (32.1 +/- 1.8%,) but the combination of AMP 579 and PC showed a significantly greater limitation of infarct size (5.5 +/- 2.7%, p < 0.05). Because cariporide pretreatment was so protective (8.5 +/- 3.7% infarction), we had to increase the ischemic insult to 60 min to test for any additive effect of the combination of AMP 579 + cariporide. Infarct size in the untreated group was 66.0 +/- 4.9% of the risk zone. Cariporide (0.5 mg/kg) 5 min prior to ischemia significantly reduced infarct size to 41.5 +/- 7.7%. When cariporide pre-treatment was combined with AMP 579 at reperfusion, infarction was further limited (14.2 +/- 4.5%). Because AMP 579's protection can be added to that of either cariporide or PC, AMP 579's mechanism of protection probably differs from either of them. The combination of AMP 579 + cariporide was particularly efficacious and could be useful in the surgical setting.
Collapse
Affiliation(s)
- Zhelong Xu
- Deparment of Physiology, University of South Alabama, College of Medicine, Mobile, AL 36688, USA
| | | | | | | |
Collapse
|
21
|
Hutchinson SA, Baker SP, Scammells PJ. New 2,N6-disubstituted adenosines: potent and selective A1 adenosine receptor agonists. Bioorg Med Chem 2002; 10:1115-22. [PMID: 11836122 DOI: 10.1016/s0968-0896(01)00384-4] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
A number of adenosine analogues substituted in the 2- and N6-positions were synthesized and evaluated for affinity, functional potency and intrinsic activity at the A1 and A2A adenosine receptors (AR). Three classes of N6-substituents were tested; norbornen-2-yl (series 1), norborn-2-yl (series 2) and 5,6-epoxynorborn-2-yl (series 3). The halogens; fluoro, bromo, and iodo were evaluated as C-2 substituents. All compounds showed relatively high affinity (nanomolar) for the A1AR and high potency for inhibiting (-)isoproterenol-stimulated cAMP accumulation in hamster smooth muscle DDT1 MF-2 cells with the 2-fluoro derivatives from each series having the highest affinity. All of the derivatives showed the same intrinsic activity as CPA. At the A2AAR, all of the derivatives showed relatively low affinity and potency (micromolar) for stimulating cAMP accumulation in rat pheochromocytoma PC-12 cells. The intrinsic activity of the derivatives compared to CGS 21680 was dependent upon the halogen substituent in the C-2 position with most showing partial agonist activity. Of particular interest is 2-iodo-N6-(2S-endo-norborn-2-yl)adenosine (5e), which is over 100-fold selective for the A1AR, is a full agonist at this receptor subtype and has no detectable agonist activity at the A2AAR.
Collapse
Affiliation(s)
- Sally A Hutchinson
- Centre for Chiral & Molecular Technologies, Deakin University, VIC 3217, Geelong, Australia
| | | | | |
Collapse
|
22
|
Scherpereel A, Rome JJ, Wiewrodt R, Watkins SC, Harshaw DW, Alder S, Christofidou-Solomidou M, Haut E, Murciano JC, Nakada M, Albelda SM, Muzykantov VR. Platelet-endothelial cell adhesion molecule-1-directed immunotargeting to cardiopulmonary vasculature. J Pharmacol Exp Ther 2002; 300:777-86. [PMID: 11861781 DOI: 10.1124/jpet.300.3.777] [Citation(s) in RCA: 56] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Therapeutic molecules conjugated with antibodies to the platelet-endothelial cell adhesion molecule-1 (PECAM-1) accumulate in the pulmonary endothelium after i.v. injection in mice. In this study, we characterized PECAM-directed targeting to the lung and heart after local versus systemic intravascular administration in a large animal model, pigs. Radiolabel tracing showed that 1 h post-i.v. injection, 35% of anti-PECAM versus 2.5% of control IgG had accumulated in the lungs. Infusion of anti-PECAM via a catheter placed in the right pulmonary artery (RPA) resulted in a 3-fold elevation of the uptake in the right lower lobe and 2-fold reduction of uptake in the left lobes in the lung. Cardiac uptake of anti-PECAM was negligible after i.v. and RPA infusion. In contrast, delivery with a catheter placed in the right coronary artery (RCA) resulted in a 4-fold elevation of cardiac uptake of anti-PECAM, but not IgG, compared with i.v. injection. To estimate the targeting of an active reporter enzyme, streptavidin-conjugated beta-galactosidase (beta-Gal) was coupled to anti-PECAM or IgG (anti-PECAM/beta-Gal and IgG/beta-Gal) and injected into the RCA. Beta-Gal activity was markedly elevated in the heart and lungs (5- and 25-fold increased, respectively) after injection of anti-PECAM/beta-Gal, but not IgG/beta-Gal. Image analysis confirmed endothelial targeting of anti-PECAM/beta-Gal in the heart and lung. In summary, anti-PECAM antibody conjugates deliver agents to the pulmonary endothelium regardless of injection route, whereas local arterial infusion permits targeting to the cardiac vasculature. This paradigm may be useful for drug targeting to endothelium in lungs, heart, and possibly other organs.
Collapse
Affiliation(s)
- Arnaud Scherpereel
- Division of Pulmonary, Allergy, and Critical Care, Department of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania 19104-6068, USA
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
23
|
Xu Z, Cohen MV, Downey JM, Vanden Hoek TL, Yao Z. Attenuation of oxidant stress during reoxygenation by AMP 579 in cardiomyocytes. Am J Physiol Heart Circ Physiol 2001; 281:H2585-9. [PMID: 11709426 DOI: 10.1152/ajpheart.2001.281.6.h2585] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
AMP 579, an adenosine A(1)/A(2) receptor agonist, has a strong anti-infarct effect when administered just before reperfusion. Because oxidative stress has been proposed to contribute to myocardial reperfusion injury, we tested whether AMP 579 can reduce the production of reactive oxidant species (ROS) during reoxygenation in cultured chick embryonic cardiomyocytes. The intracellular fluorescent probe 2',7'-dichlorofluorescin diacetate (DCFH) was used to detect ROS. The cells were subjected to 60 min of simulated ischemia, followed by either 15 min or 3 h of reoxygenation. AMP 579 (0.5 and 1 microM), when started 10 min before reoxygenation, significantly reduced ROS generation from 4.86 +/- 0.30 (arbitrary units) in untreated cells to 2.72 +/- 0.31 and 1.85 +/- 0.14, respectively (P < 0.05). Cell death that was assessed by propidium iodide uptake was markedly reduced by AMP 579 (49.6 +/- 4.7% of control cells vs. 25.4 +/- 2.4%, P < 0.05). In contrast, adenosine did not alter ROS generation or cell death. Attenuation of ROS production by AMP 579 was completely prevented by simultaneous exposure of cells to the selective adenosine A(2) antagonist 8-(13-chlorostyryl) caffeine. These results indicate that AMP 579 directly protects cardiomyocytes from reperfusion injury by a mechanism that attenuates intracellular oxidant stress. Furthermore, adenosine could not duplicate these effects.
Collapse
Affiliation(s)
- Z Xu
- Department of Physiology, College of Medicine, University of South Alabama, Mobile, Alabama 36688, USA
| | | | | | | | | |
Collapse
|
24
|
Xu Z, Downey JM, Cohen MV. Amp 579 reduces contracture and limits infarction in rabbit heart by activating adenosine A2 receptors. J Cardiovasc Pharmacol 2001; 38:474-81. [PMID: 11486252 DOI: 10.1097/00005344-200109000-00016] [Citation(s) in RCA: 45] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
To determine the mechanism by which AMP 579, an adenosine A1/A2 agonist, administered at reperfusion protects ischemic myocardium, buffer-perfused rabbit hearts were subjected to 30 min of global ischemia and 2 h of reperfusion. AMP 579 (500 nM) was included in the reperfusate for the first 70 min. Average left ventricular diastolic pressure during reperfusion in hearts receiving AMP 579 was lower than that in control hearts (17.9 +/- 2.4 vs. 39.0 +/- 6.5 mm Hg, p < 0.05), indicating attenuation of contracture. Left ventricular developed pressure and coronary flow during reperfusion were also significantly improved with AMP 579 treatment. AMP 579's anti-contracture effect was blocked by the adenosine A2-receptor antagonist 8-(3-chlorostyryl)caffeine (CSC), but not by the A1 antagonist 8-cyclopentyl-1,3-dipropylxanthine (DPCPX). CSC, but not DPCPX, also blocked AMP 579's ability to preserve developed pressure and coronary flow in these hearts. AMP 579 significantly reduced infarction in isolated hearts subjected to regional ischemia. The anti-infarct effect again was abolished by CSC but not by DPCPX. Finally, we tested whether 5'-(N-ethylcarboxamido)adenosine (NECA), another A1/A2 agonist, also administered for the initial 70 min of reperfusion, could duplicate the anti-infarct effect of AMP 579. One-hundred-nanomolar NECA duplicated the protection, but neither 50 nM CGS21680, a selective A2 agonist, nor 100 microM adenosine was protective. Therefore, AMP 579 given at reperfusion reduces contracture and infarction. Anti-contracture and anti-infarct effects require the adenosine A2, but not the A1, receptor suggesting that prevention of contracture and tissue salvage are mechanistically related. Not all A2 agonists were able to duplicate the anti-infarct effect, suggesting something unique about AMP579.
Collapse
Affiliation(s)
- Z Xu
- Department of Physiology, University of South Alabama, College of Medicine, Mobile, Alabama, USA
| | | | | |
Collapse
|
25
|
Liang BT, Stewart D, Jacobson KA. Adenosine A 1 and A 3 Receptors: Distinct Cardioprotection. Drug Dev Res 2001; 52:366-378. [PMID: 39741902 PMCID: PMC11687615 DOI: 10.1002/ddr.1136] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2025]
Abstract
Adenosine is released in large amounts during myocardial ischemia and exerts potent cardioprotective effects in the heart. Although these observations on adenosine have been known for a long time, how adenosine acts to achieve its antiischemic effect remains incompletely understood. Recent advances in the chemistry and pharmacology of adenosine receptor ligands have provided important and novel information on the function of adenosine receptor subtypes in the cardiovascular system. The development of model systems for the cardiac actions of adenosine has yielded important insights into its mechanism of action and have begun to elucidate the sequence of signaling events from receptor activation to the actual exertion of its cardioprotective effect. The goal of the current article is to review recent advances on the cellular and molecular mechanisms that mediate the cardiac actions of adenosine and to show the cardioprotective effect of novel adenosine ligands.
Collapse
Affiliation(s)
- Bruce T. Liang
- Department of Medicine, Cardiovascular Division, University of Pennsylvania Medical Center, Philadelphia, Pennsylvania
| | - Douglas Stewart
- Department of Medicine, Cardiovascular Division, University of Pennsylvania Medical Center, Philadelphia, Pennsylvania
| | - Kenneth A. Jacobson
- Laboratory of Bioorganic Chemistry, National Institutes of Health, Bethesda, Maryland
| |
Collapse
|
26
|
Xu Z, Yang XM, Cohen MV, Neumann T, Heusch G, Downey JM. Limitation of infarct size in rabbit hearts by the novel adenosine receptor agonist AMP 579 administered at reperfusion. J Mol Cell Cardiol 2000; 32:2339-47. [PMID: 11113009 DOI: 10.1006/jmcc.2000.1264] [Citation(s) in RCA: 52] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
The novel A(1)/A(2)adenosine receptor agonist AMP 579 has been reported to reduce myocardial infarct size in pig and dog. The present study tested the effect of AMP 579 in two rabbit models. In open-chest rabbits undergoing 30 min of regional ischemia and 3 h of reperfusion AMP 579 (3 microg/min/kg) reduced infarct size when treatment was started either 10 min before ischemia or 10 min prior to reperfusion from 36.4+/-3.1% of the risk zone in untreated hearts to 11.8+/-4.4 and 12.3+/-1.0%, respectively. To determine whether protection observed when the drug was administered shortly before reperfusion represented a long-lasting effect rather than merely a transient delay of necrosis, the chest wound was closed in layers and the rabbits permitted to recover. After 3 days the hearts were removed to evaluate infarct size. Continued limitation of infarct size after 3 days of reperfusion (8.2+/-2.8% of the risk zone) confirmed that sustained tissue salvage had been conferred by the drug. In isolated, buffer-perfused rabbit hearts undergoing 30 min of regional ischemia and 2 h of reperfusion, AMP 579 again limited infarct size (8.6+/-2.9% of the risk zone) when treatment started 10 min prior to reperfusion, arguing against an anti-leukocyte mechanism of protection. AMP 579's protective effect in this in vitro model was abrogated by 8-(p-sulfophenyl)theophylline, indicating that it was mediated through adenosine receptors. We conclude that AMP 579 given just prior to reperfusion may be an effective anti-infarct intervention.
Collapse
Affiliation(s)
- Z Xu
- Department of Physiology, University of South Alabama, Mobile, AL 36688, USA
| | | | | | | | | | | |
Collapse
|
27
|
Sledeski AW, Kubiak GG, O'Brien MK, Powers MR, Powner TH, Truesdale LK. Efficient synthesis of AMP579, a novel adenosine A1/A2 receptor agonist. J Org Chem 2000; 65:8114-8. [PMID: 11073633 DOI: 10.1021/jo001066f] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Affiliation(s)
- A W Sledeski
- Process Chemistry, Rhône-Poulenc Rorer, Collegeville, Pennsylvania, USA.
| | | | | | | | | | | |
Collapse
|
28
|
Jacobson KA, Ji X, Li AH, Melman N, Siddiqui MA, Shin KJ, Marquez VE, Ravi RG. Methanocarba analogues of purine nucleosides as potent and selective adenosine receptor agonists. J Med Chem 2000; 43:2196-203. [PMID: 10841798 PMCID: PMC3471159 DOI: 10.1021/jm9905965] [Citation(s) in RCA: 104] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
Adenosine receptor agonists have cardioprotective, cerebroprotective, and antiinflammatory properties. We report that a carbocyclic modification of the ribose moiety incorporating ring constraints is a general approach for the design of A(1) and A(3) receptor agonists having favorable pharmacodynamic properties. While simple carbocyclic substitution of adenosine agonists greatly diminishes potency, methanocarba-adenosine analogues have now defined the role of sugar puckering in stabilizing the active adenosine receptor-bound conformation and thereby have allowed identification of a favored isomer. In such analogues a fused cyclopropane moiety constrains the pseudosugar ring of the nucleoside to either a Northern (N) or Southern (S) conformation, as defined in the pseudorotational cycle. In binding assays at A(1), A(2A), and A(3) receptors, (N)-methanocarba-adenosine was of higher affinity than the (S)-analogue, particularly at the human A(3) receptor (N/S affinity ratio of 150). (N)-Methanocarba analogues of various N(6)-substituted adenosine derivatives, including cyclopentyl and 3-iodobenzyl, in which the parent compounds are potent agonists at either A(1) or A(3) receptors, respectively, were synthesized. The N(6)-cyclopentyl derivatives were A(1) receptor-selective and maintained high efficacy at recombinant human but not rat brain A(1) receptors, as indicated by stimulation of binding of [(35)S]GTP-gamma-S. The (N)-methanocarba-N(6)-(3-iodobenzyl)adenosine and its 2-chloro derivative had K(i) values of 4.1 and 2.2 nM at A(3) receptors, respectively, and were highly selective partial agonists. Partial agonism combined with high functional potency at A(3) receptors (EC(50) < 1 nM) may produce tissue selectivity. In conclusion, as for P2Y(1) receptors, at least three adenosine receptors favor the ribose (N)-conformation.
Collapse
Affiliation(s)
- K A Jacobson
- Molecular Recognition Section, Laboratory of Bioorganic Chemistry, National Institute of Diabetes, Digestive and Kidney Diseases, National Institutes of Health, Bethesda, Maryland 20892-0810, USA.
| | | | | | | | | | | | | | | |
Collapse
|
29
|
Nakamura M, Zhao ZQ, Clark KL, Velez DV, Guyton RA, Vinten-Johansen J. A novel adenosine analog, AMP579, inhibits neutrophil activation, adherence and neutrophil-mediated injury to coronary vascular endothelium. Eur J Pharmacol 2000; 397:197-205. [PMID: 10844114 DOI: 10.1016/s0014-2999(00)00234-x] [Citation(s) in RCA: 16] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/16/2022]
Abstract
We hypothesized that 1S-[1a,2b,3b, 4a(S*)]-4-[7-[[1-[(3-chloro-2-thienyl)methylpropyl]propyl-amino]-3H-i midazo[4,5-b] pyridyl-3-yl]-N-ethyl-2,3-dihydroxycyclopentane carboxamide (AMP579), a new adenosine analog, inhibits superoxide anion (O(2)(-)) generation and degranulation from canine neutrophils, neutrophil adherence and neutrophil-induced dysfunction to canine coronary artery endothelium by adenosine receptor-mediated mechanisms. AMP579 inhibited O(2)(-) generation (nM/20x10(6) neutrophils) from platelet activating factor (PAF)-activated neutrophil in concentration-dependent manner (4.1+/-0.8 at 10 microM vs. 16.7+/-2.1 in PAF group, P<0.05). This inhibitory effect was blocked by the adenosine A(2A) receptor-selective antagonist, 4-(2-[7-Amino-2-(2-furyl)[1,2,4]triazolo[2,3-a][1,3, 5]triazin-5-ylamino]ethyl)phenol (ZM241385, 17.7+/-2.8, P<0.05), but not by either the adenosine A(1) receptor-selective antagonist, 8-Cyclopentyl-1.3-dipropylxanthine (DPCPX) or the adenosine A(3) receptor-selective antagonist, 9-Chloro-2-(2-furanyl)-5-[(phenylacetyl)amino][1,2,4]-triazolo[1, 5-c]quinazoline (MRS1220). AMP579 inhibited neutrophil degranulation dose-dependently by 38+/-2% at 10 microM (P<0.05). The inhibitory effect of AMP579 was not altered by either DPCPX or MRS1220, but was partially reversed by ZM241385 (69+/-8%, P<0.05 vs. AMP579 10 microM). A total of 10 microM AMP579 reduced neutrophil adherence to thrombin-stimulated endothelium (neutrophils/mm(2)) from 269+/-16 to 44+/-4 (P<0.05); this was reversed by ZM241385, but not by DPCPX or MRS1220. After coincubation of unstimulated neutrophil with thrombin-stimulated endothelium, concentration-relaxation responses to the endothelium receptor-dependent vasodilator, acetylcholine, were reduced (maximum 57+/-5% vs. 120+/-5% in controls, P<0.05). This endothelial dysfunction was attenuated by AMP579 (116+/-7%, P<0. 05). We conclude that AMP579 inhibits neutrophil activation and neutrophil-mediated coronary endothelial dysfunction, primarily by an adenosine A(2A) receptor mechanism.
Collapse
Affiliation(s)
- M Nakamura
- Department of Cardiothoracic Surgery, Emory University School of Medicine, Atlanta, GA 30365, USA
| | | | | | | | | | | |
Collapse
|
30
|
Meng H, McVey M, Perrone M, Clark KL. Intravenous AMP 579, a novel adenosine A(1)/A(2a) receptor agonist, induces a delayed protection against myocardial infarction in minipig. Eur J Pharmacol 2000; 387:101-5. [PMID: 10633167 DOI: 10.1016/s0014-2999(99)00804-3] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
The aim of the study was to probe if acute administration of [1S-[1a, 2b,3b, 4a(S*)]]-4-[7-[[2-(3-chloro-2-thienyl)-1-methylpropyl]amino]-3H-imida zo[4,5-b] pyridin-3-yl] cyclopentane carboxamide (AMP 579) could provide a delayed protection against myocardial ischemia-reperfusion injury after 24 h. Anesthetized Yucatan minipigs were given an intravenous (i.v.) loading dose (3 microg/kg) of AMP 579 in 2 min followed by a 68-min infusion (0.3 microg/kg/min) and were allowed to recover. After 24 h, the animals were subjected to an open-chest operation and the left anterior descending coronary artery was occluded for 40 min, followed by 3 h of reperfusion. Results indicated that there were no significant differences in hemodynamic parameters between vehicle- and drug-treated groups either during drug infusion or ischemia-reperfusion. Both groups had similar area at risk (24.9% for vehicle and 25.1% for AMP 579-treated). However, the infarct size was 36.5% of area at risk in vehicle group (n=8) and 12.5% in AMP 579 group (n=8), representing a 66% reduction of infarct size by AMP 579 (p=0.011). This is the first report to demonstrate that in a large animal model, a hemodynamically silent, single i.v. dose of an adenosine receptor agonist can result in a delayed protection against myocardial infarction.
Collapse
Affiliation(s)
- H Meng
- Cardiovascular Drug Discovery, Mail Stop NW4, Rhône-Poulenc Rorer, Collegeville, PA 19426, USA.
| | | | | | | |
Collapse
|
31
|
Zhao ZQ, Clark KL, Wang NP, Velez DA, Guyton RA, Vinten-Johansen J. Comparison of AMP579 and adenosine in inhibition of cell-cell interaction between human neutrophil and vascular endothelial cell. Drug Dev Res 2000. [DOI: 10.1002/1098-2299(200004)49:4<266::aid-ddr6>3.0.co;2-m] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
|