1
|
Ruan W, Ma X, Bang IH, Liang Y, Muehlschlegel JD, Tsai KL, Mills TW, Yuan X, Eltzschig HK. The Hypoxia-Adenosine Link during Myocardial Ischemia-Reperfusion Injury. Biomedicines 2022; 10:1939. [PMID: 36009485 PMCID: PMC9405579 DOI: 10.3390/biomedicines10081939] [Citation(s) in RCA: 18] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2022] [Revised: 07/28/2022] [Accepted: 08/01/2022] [Indexed: 11/16/2022] Open
Abstract
Despite increasing availability and more successful interventional approaches to restore coronary reperfusion, myocardial ischemia-reperfusion injury is a substantial cause of morbidity and mortality worldwide. During myocardial ischemia, the myocardium becomes profoundly hypoxic, thus causing stabilization of hypoxia-inducible transcription factors (HIF). Stabilization of HIF leads to a transcriptional program that promotes adaptation to hypoxia and cellular survival. Transcriptional consequences of HIF stabilization include increases in extracellular production and signaling effects of adenosine. Extracellular adenosine functions as a signaling molecule via the activation of adenosine receptors. Several studies implicated adenosine signaling in cardioprotection, particularly through the activation of the Adora2a and Adora2b receptors. Adenosine receptor activation can lead to metabolic adaptation to enhance ischemia tolerance or dampen myocardial reperfusion injury via signaling events on immune cells. Many studies highlight that clinical strategies to target the hypoxia-adenosine link could be considered for clinical trials. This could be achieved by using pharmacologic HIF activators or by directly enhancing extracellular adenosine production or signaling as a therapy for patients with acute myocardial infarction, or undergoing cardiac surgery.
Collapse
Affiliation(s)
- Wei Ruan
- Department of Anesthesiology, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, TX 77030, USA
- Department of Anesthesiology, Second Xiangya Hospital, Central South University, Changsha 410011, China
| | - Xinxin Ma
- Department of Anesthesiology, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, TX 77030, USA
| | - In Hyuk Bang
- Department of Anesthesiology, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, TX 77030, USA
| | - Yafen Liang
- Department of Anesthesiology, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, TX 77030, USA
| | - Jochen Daniel Muehlschlegel
- Department of Anesthesiology, Perioperative, and Pain Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Kuang-Lei Tsai
- Department of Biochemistry and Molecular Biology, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, TX 77030, USA
| | - Tingting W. Mills
- Department of Biochemistry and Molecular Biology, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, TX 77030, USA
| | - Xiaoyi Yuan
- Department of Anesthesiology, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, TX 77030, USA
| | - Holger K. Eltzschig
- Department of Anesthesiology, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, TX 77030, USA
| |
Collapse
|
2
|
Härter M, Kalthof B, Delbeck M, Lustig K, Gerisch M, Schulz S, Kast R, Meibom D, Lindner N. Novel non-xanthine antagonist of the A 2B adenosine receptor: From HTS hit to lead structure. Eur J Med Chem 2018; 163:763-778. [PMID: 30576906 DOI: 10.1016/j.ejmech.2018.11.045] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2018] [Revised: 11/16/2018] [Accepted: 11/19/2018] [Indexed: 11/17/2022]
Abstract
The A2B adenosine receptor is a G protein-coupled receptor that belongs to the four member family of adenosine receptors: A1, A2A, A2B, A3. While adenosine-mediated A2B receptor signaling attenuates acute inflammation, facilitates tissue adaptation to hypoxia, and induces increased ischemia tolerance under conditions of an acute insult, persistently elevated adenosine levels and A2B receptor signaling are characteristics of a number of chronic disease states. In this report we describe the discovery of certain thienouracils (thieno[2,3-d]pyrimidine-2,4(1H,3H)-diones) as antagonists of the A2B adenosine receptor by high-throughput screening from our corporate substance collection. The structure optimization of the initial screening hits led to BAY-545, an A2B receptor antagonist that was suitable for in vivo testing. The structure optimization work, SAR that was derived from there, as well as the properties of BAY-545 are also described. In vivo efficacy of BAY-545 was demonstrated in two models of lung fibrosis and data is presented.
Collapse
Affiliation(s)
- Michael Härter
- Small Molecules Innovation, Research & Development, Bayer Pharmaceuticals, Wuppertal, Germany.
| | - Bernd Kalthof
- Small Molecules Innovation, Research & Development, Bayer Pharmaceuticals, Wuppertal, Germany
| | - Martina Delbeck
- Preclinical Research, Research & Development, Bayer Pharmaceuticals, Wuppertal, Germany
| | - Klemens Lustig
- Translational Sciences, Research & Development, Bayer Pharmaceuticals, Wuppertal, Germany
| | - Michael Gerisch
- Translational Sciences, Research & Development, Bayer Pharmaceuticals, Wuppertal, Germany
| | - Simone Schulz
- Translational Sciences, Research & Development, Bayer Pharmaceuticals, Wuppertal, Germany
| | - Raimund Kast
- Preclinical Research, Research & Development, Bayer Pharmaceuticals, Wuppertal, Germany
| | - Daniel Meibom
- Small Molecules Innovation, Research & Development, Bayer Pharmaceuticals, Wuppertal, Germany
| | - Niels Lindner
- Small Molecules Innovation, Research & Development, Bayer Pharmaceuticals, Wuppertal, Germany
| |
Collapse
|
3
|
Structures of Human A 1 and A 2A Adenosine Receptors with Xanthines Reveal Determinants of Selectivity. Structure 2017; 25:1275-1285.e4. [DOI: 10.1016/j.str.2017.06.012] [Citation(s) in RCA: 141] [Impact Index Per Article: 20.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2017] [Revised: 04/13/2017] [Accepted: 06/15/2017] [Indexed: 01/06/2023]
|
4
|
Busse H, Bitzinger D, Höcherl K, Seyfried T, Gruber M, Graf BM, Zausig YA. Adenosine A2A and A2B Receptor Substantially Attenuate Ischemia/Reperfusion Injury in Septic rat Hearts. Cardiovasc Drugs Ther 2017; 30:551-558. [PMID: 27757725 PMCID: PMC5156668 DOI: 10.1007/s10557-016-6693-y] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Introduction Mechanical and morphological ischemia and reperfusion (I/R) injury is reduced in septic hearts. The mechanism behind this “cardioprotection” is less well understood. As adenosine receptors play a major role for cardioprotection in non-septic hearts, we investigated the influence of adenosine receptors in a model of I/R in septic hearts. Methods SHAM operation or cecal ligation and puncture (CLP) was performed in adult male Wistar rats (n = 60). After 24 h of incubation, hearts were isolated and randomly assigned to a group with or without adenosine receptor (Ador) antagonists (SCH 58261 and MRS 1706) administered before reperfusion. Ischemia and reperfusion lasted for 40 min each. Cardiac function of the heart was determined by measuring left ventricular pressure (LVP). Results Before I/R, CLP hearts showed a significant mechanical left ventricular impairment (CLP: 63 ± 5 mmHg vs. SHAM: 104 ± 6 mmHg. After I/R, left ventricular function was significantly reduced in SHAM (24 ± 32 mmHg), but not in CLP hearts (65 ± 13 mmHg). mRNA expression for the AdorA2a and AdorA2b was significantly increased in CLP, but not in SHAM hearts. LVP of CLP hearts deteriorated when AdorA2a and AdorA2b were blocked. Conclusions The morphological and functional I/R injury in septic animals is less pronounced compared to non-septic animals. By a combined blockade of AdorA2a and AdorA2b this “cardioprotective” effect is nearly abolished in septic hearts. This is the first study showing, that AdorA2a and AdorA2b may play an important role for a reduced functional I/R injury in the septic heart.
Collapse
Affiliation(s)
- Hendrik Busse
- Department of Anesthesiology, University hospital of Regensburg, Franz-Josef-Strauss-Allee 11, 93053, Regensburg, Germany.
| | - Diane Bitzinger
- Department of Anesthesiology, University hospital of Regensburg, Franz-Josef-Strauss-Allee 11, 93053, Regensburg, Germany
| | - Klaus Höcherl
- Institute of Experimental and Clinical Pharmacology and Toxicology, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), Erlangen, Germany
| | - Timo Seyfried
- Department of Anesthesiology, University hospital of Regensburg, Franz-Josef-Strauss-Allee 11, 93053, Regensburg, Germany
| | - Michael Gruber
- Department of Anesthesiology, University hospital of Regensburg, Franz-Josef-Strauss-Allee 11, 93053, Regensburg, Germany
| | - Bernhard M Graf
- Department of Anesthesiology, University hospital of Regensburg, Franz-Josef-Strauss-Allee 11, 93053, Regensburg, Germany
| | - York A Zausig
- Department of Anesthesiology, University hospital of Regensburg, Franz-Josef-Strauss-Allee 11, 93053, Regensburg, Germany
| |
Collapse
|
5
|
Tofovic SP, Salah EM, Smits GJ, Whalley ET, Ticho B, Deykin A, Jackson EK. Dual A1/A2B Receptor Blockade Improves Cardiac and Renal Outcomes in a Rat Model of Heart Failure with Preserved Ejection Fraction. J Pharmacol Exp Ther 2016; 356:333-40. [PMID: 26585572 PMCID: PMC4727158 DOI: 10.1124/jpet.115.228841] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2015] [Accepted: 11/17/2015] [Indexed: 12/19/2022] Open
Abstract
Heart failure with preserved ejection fraction (HFpEF) is prevalent and often accompanied by metabolic syndrome. Current treatment options are limited. Here, we test the hypothesis that combined A1/A2B adenosine receptor blockade is beneficial in obese ZSF1 rats, an animal model of HFpEF with metabolic syndrome. The combined A1/A2B receptor antagonist 3-[4-(2,6-dioxo-1,3-dipropyl-7H-purin-8-yl)-1-bicyclo[2.2.2]octanyl]propanoic acid (BG9928) was administered orally (10 mg/kg/day) to obese ZSF1 rats (n = 10) for 24 weeks (from 20 to 44 weeks of age). Untreated ZSF1 rats (n = 9) served as controls. After 24 weeks of administration, BG9928 significantly lowered plasma triglycerides (in mg/dl: control group, 4351 ± 550; BG9928 group, 2900 ± 551) without adversely affecting plasma cholesterol or activating renin release. BG9928 significantly decreased 24-hour urinary glucose excretion (in mg/kg/day: control group, 823 ± 179; BG9928 group, 196 ± 80) and improved oral glucose tolerance, polydipsia, and polyuria. BG9928 significantly augmented left ventricular diastolic function in association with a reduction in cardiac vasculitis and cardiac necrosis. BG9928 significantly reduced 24-hour urinary protein excretion (in mg/kg/day: control group, 1702 ± 263; BG9928 group, 1076 ± 238), and this was associated with a reduction in focal segmental glomerulosclerosis, tubular atrophy, tubular dilation, and deposition of proteinaceous material in the tubules. These findings show that, in a model of HFpEF with metabolic syndrome, A1/A2B receptor inhibition improves hyperlipidemia, exerts antidiabetic actions, reduces HFpEF, improves cardiac histopathology, and affords renal protection. We conclude that chronic administration of combined A1/A2B receptor antagonists could be beneficial in patients with HFpEF, in particular those with comorbidities such as obesity, diabetes, and dyslipidemias.
Collapse
Affiliation(s)
- Stevan P Tofovic
- Vascular Medicine Institute (S.P.T.) and the Departments of Medicine (S.P.T., E.K.J.), Pathology (E.M.S.), and Pharmacology and Chemical Biology (E.K.J.), University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania; and Biogen Idec, Inc., Cambridge, Massachusetts (G.J.S., E.T.W., B.T., A.D.)
| | - Eman M Salah
- Vascular Medicine Institute (S.P.T.) and the Departments of Medicine (S.P.T., E.K.J.), Pathology (E.M.S.), and Pharmacology and Chemical Biology (E.K.J.), University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania; and Biogen Idec, Inc., Cambridge, Massachusetts (G.J.S., E.T.W., B.T., A.D.)
| | - Glenn J Smits
- Vascular Medicine Institute (S.P.T.) and the Departments of Medicine (S.P.T., E.K.J.), Pathology (E.M.S.), and Pharmacology and Chemical Biology (E.K.J.), University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania; and Biogen Idec, Inc., Cambridge, Massachusetts (G.J.S., E.T.W., B.T., A.D.)
| | - Eric T Whalley
- Vascular Medicine Institute (S.P.T.) and the Departments of Medicine (S.P.T., E.K.J.), Pathology (E.M.S.), and Pharmacology and Chemical Biology (E.K.J.), University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania; and Biogen Idec, Inc., Cambridge, Massachusetts (G.J.S., E.T.W., B.T., A.D.)
| | - Barry Ticho
- Vascular Medicine Institute (S.P.T.) and the Departments of Medicine (S.P.T., E.K.J.), Pathology (E.M.S.), and Pharmacology and Chemical Biology (E.K.J.), University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania; and Biogen Idec, Inc., Cambridge, Massachusetts (G.J.S., E.T.W., B.T., A.D.)
| | - Aaron Deykin
- Vascular Medicine Institute (S.P.T.) and the Departments of Medicine (S.P.T., E.K.J.), Pathology (E.M.S.), and Pharmacology and Chemical Biology (E.K.J.), University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania; and Biogen Idec, Inc., Cambridge, Massachusetts (G.J.S., E.T.W., B.T., A.D.)
| | - Edwin K Jackson
- Vascular Medicine Institute (S.P.T.) and the Departments of Medicine (S.P.T., E.K.J.), Pathology (E.M.S.), and Pharmacology and Chemical Biology (E.K.J.), University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania; and Biogen Idec, Inc., Cambridge, Massachusetts (G.J.S., E.T.W., B.T., A.D.)
| |
Collapse
|
6
|
Berger MM, Köhne H, Hotz L, Hammer M, Schommer K, Bärtsch P, Mairbäurl H. Remote ischemic preconditioning delays the onset of acute mountain sickness in normobaric hypoxia. Physiol Rep 2015; 3:3/3/e12325. [PMID: 25742960 PMCID: PMC4393159 DOI: 10.14814/phy2.12325] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023] Open
Abstract
Acute mountain sickness (AMS) is a neurological disorder occurring when ascending too fast, too high. Remote ischemic preconditioning (RIPC) is a noninvasive intervention protecting remote organs from subsequent hypoxic damage. We hypothesized that RIPC protects against AMS and that this effect is related to reduced oxidative stress. Fourteen subjects were exposed to 18 hours of normoxia (21% oxygen) and 18 h of normobaric hypoxia (12% oxygen, equivalent to 4500 m) on different days in a blinded, randomized order. RIPC consisted of four cycles of lower limb ischemia (5 min) and 5 min of reperfusion, and was performed immediately before the study room was entered. A control group was exposed to hypoxia (12% oxygen, n = 14) without RIPC. AMS was evaluated by the Lake Louise score (LLS) and the AMS-C score of the Environmental Symptom Questionnaire. Plasma concentrations of ascorbate radicals, oxidized sulfhydryl (SH) groups, and electron paramagnetic resonance (EPR) signal intensity were measured as biomarkers of oxidative stress. RIPC reduced AMS scores (LLS: 1.9 ± 0.4 vs. 3.2 ± 0.5; AMS-C score: 0.4 ± 0.1 vs. 0.8 ± 0.2), ascorbate radicals (27 ± 7 vs. 65 ± 18 nmol/L), oxidized SH groups (3.9 ± 1.4 vs. 14.3 ± 4.6 μmol/L), and EPR signal intensity (0.6 ± 0.2 vs. 1.5 ± 0.4 × 10(6)) after 5 h in hypoxia (all P < 0.05). After 18 hours in hypoxia there was no difference in AMS and oxidative stress between RIPC and control. AMS and plasma markers of oxidative stress did not correlate. This study demonstrates that RIPC transiently reduces symptoms of AMS and that this effect is not associated with reduced plasma levels of reactive oxygen species.
Collapse
Affiliation(s)
- Marc M Berger
- Department of Anesthesiology, University of Heidelberg, Heidelberg, Germany Department of Anesthesiology, Perioperative and General Critical Care Medicine, Salzburg General Hospital Paracelsus Medical University, Salzburg, Austria
| | - Hannah Köhne
- Department of Anesthesiology, University of Heidelberg, Heidelberg, Germany
| | - Lorenz Hotz
- Department of Anesthesiology, University of Heidelberg, Heidelberg, Germany Department of Anesthesiology, Perioperative and General Critical Care Medicine, Salzburg General Hospital Paracelsus Medical University, Salzburg, Austria
| | - Moritz Hammer
- Department of Internal Medicine VII, Division of Sports Medicine, University of Heidelberg, Heidelberg, Germany
| | - Kai Schommer
- Department of Internal Medicine VII, Division of Sports Medicine, University of Heidelberg, Heidelberg, Germany
| | - Peter Bärtsch
- Department of Internal Medicine VII, Division of Sports Medicine, University of Heidelberg, Heidelberg, Germany
| | - Heimo Mairbäurl
- Department of Internal Medicine VII, Division of Sports Medicine, University of Heidelberg, Heidelberg, Germany
| |
Collapse
|
7
|
Foster GP, Giri PC, Rogers DM, Larson SR, Anholm JD. Ischemic preconditioning improves oxygen saturation and attenuates hypoxic pulmonary vasoconstriction at high altitude. High Alt Med Biol 2014; 15:155-61. [PMID: 24949710 DOI: 10.1089/ham.2013.1137] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
Exposure to hypoxic environments is associated with decreased arterial oxygen saturation and increased pulmonary artery pressures. Ischemic preconditioning of an extremity (IPC) is a procedure that stimulates vasoactive and inflammatory pathways that protect remote organ systems from ongoing or future ischemic injury. To test the effects of IPC on oxygen saturation and pulmonary artery pressures at high altitude, 12 healthy adult volunteers were evaluated in a randomized cross-over trial. IPC was administered utilizing a standardized protocol. IPC or placebo was administered daily for 5 days prior to ascent to altitude. All participants were evaluated twice at 4342 m altitude (placebo and IPC conditions separated by 4 weeks, randomized). The pulmonary artery systolic pressure (PASP) at 4342 m was significantly lower in the IPC condition than the placebo condition (36 ± 6.0 mmHg vs. 38.1 ± 7.6 mmHg, respectively, p = 0.035). Oxygen saturation at 4342 m was significantly higher with IPC compared to placebo (80.3 ± 8.7% vs. 75.3 ± 9.6%, respectively, p = 0.003). Prophylactic IPC treatment is associated with improved oxygen saturation and attenuation of the normal hypoxic increase in pulmonary artery pressures following ascent to high altitude.
Collapse
Affiliation(s)
- Gary P Foster
- 1 Cardiology and Pulmonary/Critical Care Sections, Medical Service VA Loma Linda Healthcare System , Loma Linda, California
| | | | | | | | | |
Collapse
|
8
|
Yu H, Liu Z, Zhou H, Dai W, Chen S, Shu Y, Feng J. JAK-STAT pathway modulates the roles of iNOS and COX-2 in the cytoprotection of early phase of hydrogen peroxide preconditioning against apoptosis induced by oxidative stress. Neurosci Lett 2012; 529:166-71. [PMID: 22995181 DOI: 10.1016/j.neulet.2012.09.013] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2012] [Revised: 08/23/2012] [Accepted: 09/04/2012] [Indexed: 10/27/2022]
Abstract
Our previous studies have demonstrated that preconditioning with hydrogen peroxide (H(2)O(2)) activated the JAK-STAT pathway that played an important role in the cytoprotection, and inducible nitric oxide synthase (iNOS) and cyclooxygenase-2 (COX-2) mediated the late phase of cytoprotection induced by high concentration of H(2)O(2) after preconditioning. Here we sought to identify the downstream targets of the JAK-STAT axis that mediated H(2)O(2) preconditioning and the expression of iNOS and COX-2 in the early phase of H(2)O(2) preconditioning. It was shown that (1) Preconditioning with H(2)O(2) at 100 μmol/L for 90 min in PC12 cells induced significant expression of iNOS and COX-2. (2) Pretreatment with the iNOS inhibitor AG (10 μmol/L) or the COX-2 inhibitor NS-398 (10 μmol/L) respectively 20min before H(2)O(2) preconditioning not only inhibits the increased expression of iNOS or COX-2 but also abrogates the protective effects of H(2)O(2) preconditioning against apoptosis induced by oxidative stress. (3) Pretreatment with the JAK inhibitor AG-490 (10 μmol/L) 20 min before H(2)O(2) preconditioning obviously inhibits the up-regulation of iNOS or COX-2 induced by H(2)O(2) preconditioning. These results suggested that JAK-STAT pathway modulates the roles of iNOS and COX-2 in the cytoprotection of early phase of H(2)O(2) preconditioning.
Collapse
Affiliation(s)
- Huimin Yu
- Department of Pathogenic Biology & Immunology, Medical College, Shenzhen University, China.
| | | | | | | | | | | | | |
Collapse
|
9
|
Foster GP, Westerdahl DE, Foster LA, Hsu JV, Anholm JD. Ischemic preconditioning of the lower extremity attenuates the normal hypoxic increase in pulmonary artery systolic pressure. Respir Physiol Neurobiol 2011; 179:248-53. [PMID: 21924386 DOI: 10.1016/j.resp.2011.09.001] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2011] [Revised: 08/31/2011] [Accepted: 09/01/2011] [Indexed: 11/24/2022]
Abstract
Ischemic pre-condition of an extremity (IPC) induces effects on local and remote tissues that are protective against ischemic injury. To test the effects of IPC on the normal hypoxic increase in pulmonary pressures and exercise performance, 8 amateur cyclists were evaluated under normoxia and hypoxia (13% F(I)O(2)) in a randomized cross-over trial. IPC was induced using an arterial occlusive cuff to one thigh for 5 min followed by deflation for 5 min for 4 cycles. In the control condition, the resting pulmonary artery systolic pressure (PASP) increased from a normoxic value of 25.6±2.3 mmHg to 41.8±7.2 mmHg following 90 min of hypoxia. In the IPC condition, the PASP increased to only 32.4±3.1 mmHg following hypoxia, representing a 72.8% attenuation (p=0.003). No significant difference was detected in cycle ergometer time trial duration between control and IPC conditions with either normoxia or hypoxia. IPC administered prior to hypoxic exposure was associated with profound attenuation of the normal hypoxic increase of pulmonary artery systolic pressure.
Collapse
Affiliation(s)
- Gary P Foster
- Cardiology Section, Medical Service, VA Loma Linda HealthCare System, Loma Linda, CA 92357, United States.
| | | | | | | | | |
Collapse
|
10
|
Interplay of hypoxia and A2B adenosine receptors in tissue protection. ADVANCES IN PHARMACOLOGY 2011; 61:145-86. [PMID: 21586359 DOI: 10.1016/b978-0-12-385526-8.00006-0] [Citation(s) in RCA: 58] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
That adenosine signaling can elicit adaptive tissue responses during conditions of limited oxygen availability (hypoxia) is a long-suspected notion that recently gained general acceptance from genetic and pharmacologic studies of the adenosine signaling pathway. As hypoxia and inflammation share an interdependent relationship, these studies have demonstrated that adenosine signaling events can be targeted to dampen hypoxia-induced inflammation. Here, we build on the hypothesis that particularly the A(2B) adenosine receptor (ADORA(2B)) plays a central role in tissue adaptation to hypoxia. In fact, the ADORA(2B) requires higher adenosine concentrations than any of the other adenosine receptors. However, during conditions of hypoxia or ischemia, the hypoxia-elicited rise in extracellular adenosine is sufficient to activate the ADORA(2B). Moreover, several studies have demonstrated very robust induction of the ADORA(2B) elicited by transcriptional mechanisms involving hypoxia-dependent signaling pathways and the transcription factor "hypoxia-induced factor" 1. In the present chapter, genetic and pharmacologic evidence is presented to support our hypothesis of a tissue protective role of ADORA(2B) signaling during hypoxic conditions, including hypoxia-elicited vascular leakage, organ ischemia, or acute lung injury. All these disease models are characterized by hypoxia-elicited tissue inflammation. As such, the ADORA(2B) has emerged as a therapeutic target for dampening hypoxia-induced inflammation and tissue adaptation to limited oxygen availability.
Collapse
|
11
|
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: 6.8] [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
|
12
|
Ensor CR, Russell SD. Tonapofylline: a selective adenosine-1 receptor antagonist for the treatment of heart failure. Expert Opin Pharmacother 2010; 11:2405-15. [DOI: 10.1517/14656566.2010.514605] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
|
13
|
Leshem-Lev D, Hochhauser E, Chanyshev B, Isak A, Shainberg A. Adenosine A1 and A3 receptor agonists reduce hypoxic injury through the involvement of P38 MAPK. Mol Cell Biochem 2010; 345:153-60. [DOI: 10.1007/s11010-010-0568-5] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2010] [Accepted: 07/29/2010] [Indexed: 01/09/2023]
|
14
|
Abstract
The study of the A(3) adenosine receptor (A(3)AR) represents a rapidly growing and intense area of research in the adenosine field. The present chapter will provide an overview of the expression patterns, molecular pharmacology and functional role of this A(3)AR subtype under pathophysiological conditions. Through studies utilizing selective A(3)AR agonists and antagonists, or A(3)AR knockout mice, it is now clear that this receptor plays a critical role in the modulation of ischemic diseases as well as in inflammatory and autoimmune pathologies. Therefore, the potential therapeutic use of agonists and antagonists will also be described. The discussion will principally address the use of such compounds in the treatment of brain and heart ischemia, asthma, sepsis and glaucoma. The final part concentrates on the molecular basis of A(3)ARs in autoimmune diseases such as rheumatoid arthritis, and includes a description of clinical trials with the selective agonist CF101. Based on this chapter, it is evident that continued research to discover agonists and antagonists for the A(3)AR subtype is warranted.
Collapse
|
15
|
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
|
16
|
Abstract
Extracellular adenosine is produced in a coordinated manner from cells following cellular challenge or tissue injury. Once produced, it serves as an autocrine- and paracrine-signaling molecule through its interactions with seven-membrane-spanning G-protein-coupled adenosine receptors. These signaling pathways have widespread physiological and pathophysiological functions. Immune cells express adenosine receptors and respond to adenosine or adenosine agonists in diverse manners. Extensive in vitro and in vivo studies have identified potent anti-inflammatory functions for all of the adenosine receptors on many different inflammatory cells and in various inflammatory disease processes. In addition, specific proinflammatory functions have also been ascribed to adenosine receptor activation. The potent effects of adenosine signaling on the regulation of inflammation suggest that targeting specific adenosine receptor activation or inactivation using selective agonists and antagonists could have important therapeutic implications in numerous diseases. This review is designed to summarize the current status of adenosine receptor signaling in various inflammatory cells and in models of inflammation, with an emphasis on the advancement of adenosine-based therapeutics to treat inflammatory disorders.
Collapse
Affiliation(s)
- Michael R Blackburn
- Department of Biochemistry and Molecular Biology, The University of Texas-Houston Medical School, Houston, TX 77030, USA.
| | | | | | | |
Collapse
|
17
|
Kiesman WF, Elzein E, Zablocki J. A1 adenosine receptor antagonists, agonists, and allosteric enhancers. Handb Exp Pharmacol 2009:25-58. [PMID: 19639278 DOI: 10.1007/978-3-540-89615-9_2] [Citation(s) in RCA: 72] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/28/2023]
Abstract
Intense efforts of many pharmaceutical companies and academicians in the A(1) adenosine receptor (AR) field have led to the discovery of clinical candidates that are antagonists, agonists, and allosteric enhancers. The A(1)AR antagonists currently in clinical development are KW3902, BG9928, and SLV320. All three have high affinity for the human (h) A(1)AR subtype (hA(1) K (i) < 10 nM), > 200-fold selectivity over the hA(2A) subtype, and demonstrate renal protective effects in multiple animal models of disease and pharmacologic effects in human subjects. In the A(1)AR agonist area, clinical candidates have been discovered for the following conditions: atrial arrhythmias (tecadenoson, selodenoson and PJ-875); Type II diabetes and insulin sensitizing agents (GR79236, ARA, RPR-749, and CVT-3619); and angina (BAY 68-4986). The challenges associated with the development of any A(1)AR agonist are to obtain tissue-specific effects but avoid off-target tissue side effects and A(1)AR desensitization leading to tachyphylaxis. For the IV antiarrhythmic agents that act as ventricular rate control agents, a selective response can be accomplished by careful IV dosing paradigms. The treatment of type II diabetes using A(1)AR agonists in the clinic has met with limited success due to cardiovascular side effects and a well-defined desensitization of full agonists in human trials (GR79236, ARA, and RPR 749). However, new partial A(1)AR agonists are in development, including CVT-3619 hA(1) AR K(i) = 55nM, hA(2A:hA2B:hA(3))1,000:20, CV Therapeutics), which have the potential to provide enhanced insulin sensitivity without cardiovascular side effects and tachyphylaxis. The nonnucleosidic A(1)AR agonist BAY 68-4986 (capadenoson) represents a novel approach to angina wherein both animal studies and early human studies are promising. T-62 is an A(1)AR allosteric enhancer that is currently being evaluated in clinical trials as a potential treatment for neuropathic pain. The challenges associated with developing A(1)AR antagonists, agonists, or allosteric enhancers for therapeutic intervention are now well defined in humans. Significant progress has been made in identifying A(1)AR antagonists for the treatment of edema associated with congestive heart failure (CHF), A(1)AR agonists for the treatment of atrial arrhythmias, type II diabetes and angina, and A(1)AR allosteric enhancers for the treatment of neuropathic pain.
Collapse
|
18
|
Abstract
Adenosine acts as a cytoprotective modulator in response to stress to an organ or tissue. Although short-lived in the circulation, it can activate four subtypes of G protein-coupled adenosine receptors (ARs): A(1), A(2A), A(2B), and A(3). The alkylxanthines caffeine and theophylline are the prototypical antagonists of ARs, and their stimulant actions occur primarily through this mechanism. For each of the four AR subtypes, selective agonists and antagonists have been introduced and used to develop new therapeutic drug concepts. ARs are notable among the GPCR family in the number and variety of agonist therapeutic candidates that have been proposed. The selective and potent synthetic AR agonists, which are typically much longer lasting in the body than adenosine, have potential therapeutic applications based on their anti-inflammatory (A(2A) and A(3)), cardioprotective (preconditioning by A(1) and A(3) and postconditioning by A(2B)), cerebroprotective (A(1) and A(3)), and antinociceptive (A(1)) properties. Potent and selective AR antagonists display therapeutic potential as kidney protective (A(1)), antifibrotic (A(2A)), neuroprotective (A(2A)), and antiglaucoma (A(3)) agents. AR agonists for cardiac imaging and positron-emitting AR antagonists are in development for diagnostic applications. Allosteric modulators of A(1) and A(3) ARs have been described. In addition to the use of selective agonists/antagonists as pharmacological tools, mouse strains in which an AR has been genetically deleted have aided in developing novel drug concepts based on the modulation of ARs.
Collapse
Affiliation(s)
- Kenneth A Jacobson
- Molecular Recognition Section, Laboratory of Biooorganic Chemistry, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, MD 20892-0810, USA.
| |
Collapse
|
19
|
Abstract
Adenosine A1 antagonists are being developed for the treatment of renal dysfunction in patients with congestive heart failure. After early small studies prompted hope that these agents could increase urine output without worsening the glomerular filtration rate, larger studies published and presented in 2007 confirmed their beneficial impact on weight and renal function. However, in many studies the renal benefits disappear with higher doses, suggesting that specificity may be lost with higher doses of these drugs. Investigations in animals indicate that there may also be direct benefits on the myocardium and in the lung. Although studies have not shown adverse effects at optimal dosing, the widespread actions of adenosine mandate that safety be established. Ongoing studies should be able to demonstrate whether adenosine A1 antagonists can be used to improve renal function without adversely affecting patients with heart failure.
Collapse
Affiliation(s)
- Stephen S Gottlieb
- University of Maryland School of Medicine, 22 South Greene Street, Baltimore, MD 21201, USA.
| |
Collapse
|
20
|
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
|
21
|
Dip RG. Adenosine receptor modulation: potential implications in veterinary medicine. Vet J 2007; 179:38-49. [PMID: 17904398 DOI: 10.1016/j.tvjl.2007.08.005] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2007] [Revised: 07/27/2007] [Accepted: 08/02/2007] [Indexed: 12/01/2022]
Abstract
Adenosine is a purine nucleoside whose concentration increases during inflammation and hypoxia and the many roles of this molecule are becoming better understood. Increased reactivity to adenosine of the airways of asthmatic but not of normal subjects underlines the role of adenosine in airway inflammation. The identification and pharmacological characterisation of different adenosine receptors have stimulated the search for subtype-specific ligands able to modulate the effects of this molecule in a directed way. Several compounds of different chemical classes have been identified as having potential drawbacks, including side effects resulting from the broad distribution of the receptors across the organism, have prevented clinical application. In this article, the effects of adenosine's different receptors and the intracellular signalling pathways are reviewed. The potential of adenosine receptor modulation as a therapeutic target for chronic airway inflammation is considered, taking equine recurrent airway disease and feline asthma as examples of naturally occurring airway obstructive diseases. Other potential applications for adenosine receptor modulation are also discussed. As the intrinsic molecular events of adenosine's mechanism of action become uncovered, new concrete therapeutic approaches will become available for the treatment of various conditions in veterinary medicine.
Collapse
Affiliation(s)
- Ramiro G Dip
- Institute of Pharmacology and Toxicology, University of Zurich-Vetsuisse, Winterthurerstrasse 260, CH-8057 Zurich, Switzerland.
| |
Collapse
|
22
|
El-Ani D, Zimlichman R, Mashiach Y, Shainberg A. Adenosine and TNF-alpha exert similar inotropic effect on heart cultures, suggesting a cardioprotective mechanism against hypoxia. Life Sci 2007; 81:803-13. [PMID: 17764703 DOI: 10.1016/j.lfs.2007.06.030] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2007] [Revised: 06/20/2007] [Accepted: 06/27/2007] [Indexed: 11/20/2022]
Abstract
When cardiomyocytes were subjected to hypoxia, tumor necrosis factor-alpha (TNF-alpha; 3-50 ng/ml) or adenosine (1-100 microM), decreased hypoxic damage as was detected by lactate dehydrogenase (LDH) release, MTT (3-[4,5-Dimethylthiazol-2-yl]-2,5-diphenyltetrazolium bromide) absorbance, ROS (reactive oxygen species) measurement or desmin immunostaining. This cardioprotection was not prevented in TNF-alpha-treated cultures by 5-hydroxydecanoic acid (5-HD). Our aim was to elucidate whether adenosine and TNF-alpha mediate a similar protective mechanism against hypoxia in primary heart cultures and in H9c2 cardiomyocytes. Adenosine and TNF-alpha are known for their negative inotropic effects on the heart. We have suggested that deoxyglucose uptake reflects heart contractility in cell cultures; therefore, we assayed its accumulation under various conditions. Treatment for 20 min with adenosine, R-PIA [(-)-N(6)-phenylisopropyladenosine] (10 microM), or TNF-alpha reduced (3)H-deoxyglucose uptake in primary heart cultures and also in H9c2 cardiomyocytes by 30-50%. Isoproterenol accelerated (3)H-deoxyglucose uptake by 50%. Adenosine, R-PIA, or TNF-alpha attenuated the stimulatory effect of isoproterenol on (3)H-deoxyglucose uptake to control levels. Hypoxia reduced (3)H-deoxyglucose uptake by 50%, as in the treatment of the hypoxic cultures with TNF-alpha or adenosine. Glibenclamide (2 microM), 5-HD (300 microM), or diazoxide (50 microM) increased (3)H-deoxyglucose uptake by 50-80%. Adenosine (100 microM) and TNF-alpha (50 ng/ml) stimulated (86)Rb efflux. Glibenclamide attenuated this effect. We demonstrate that TNF-alpha, like adenosine, accelerated Ca(2+) uptake into the sarcoplasmic reticulum (SR) by 50-100% and therefore prevented cardiomyocyte Ca(2+) overload. Our findings further suggest that TNF-alpha, as well as adenosine, may mediate an adaptive effect in the heart by preventing Ca(2+) overload via activation of SR Ca-ATPase (SERCA(2)a).
Collapse
Affiliation(s)
- Dalia El-Ani
- The Mina & Everard Goodman Faculty of Life Sciences, Bar-Ilan University, Ramat-Gan, Israel
| | | | | | | |
Collapse
|
23
|
Kalk P, Eggert B, Relle K, Godes M, Heiden S, Sharkovska Y, Fischer Y, Ziegler D, Bielenberg GW, Hocher B. The adenosine A1 receptor antagonist SLV320 reduces myocardial fibrosis in rats with 5/6 nephrectomy without affecting blood pressure. Br J Pharmacol 2007; 151:1025-32. [PMID: 17558436 PMCID: PMC2042943 DOI: 10.1038/sj.bjp.0707319] [Citation(s) in RCA: 49] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022] Open
Abstract
BACKGROUND AND PURPOSE Myocardial fibrosis is an unwanted effect associated with chronic renal failure. The adenosine system is involved in cardiac and renal function. Therefore, we investigated the novel selective adenosine A(1) receptor antagonist SLV320 focusing on its potential in preventing cardiomyopathy in rats with 5/6 nephrectomy. EXPERIMENTAL APPROACH Male Sprague-Dawley rats were allocated to 4 groups of 12 rats each: 5/6 nephrectomy (5/6 NX), 5/6 NX plus SLV320 (10 mg kg(-1) d(-1) mixed with food), sham and sham plus SLV320. Study duration was 12 weeks, blood pressure was assessed repeatedly. At study end kidney function was assessed, blood samples and hearts were taken for histology/immunohistochemistry. Pharmacological properties of SLV320 were assessed using receptor binding and enzyme assays and in vivo. KEY RESULTS SLV320 is a selective and potent adenosine A(1) antagonist in vitro (Ki=1 nM) with a selectivity factor of at least 200 versus other adenosine receptor subtypes. Functional A(1) antagonism was demonstrated in vivo. In rats with 5/6 NX SLV320 significantly decreased albuminuria by about 50%, but did not alter glomerular filtration rate (GFR). SLV320 normalized cardiac collagen I+III contents in 5/6 NX rats. SLV320 prevented nephrectomy-dependent rise in plasma levels of creatinine kinase (CK), ALT and AST. Blood pressure did not differ between study groups. CONCLUSION SLV320 suppresses cardiac fibrosis and attenuates albuminuria without affecting blood pressure in rats with 5/6 nephrectomy, indicating that selective A(1) receptor antagonists may be beneficial in uraemic cardiomyopathy.
Collapse
Affiliation(s)
- P Kalk
- Department of Pharmacology and Toxicology, Center for Cardiovascular Research, Charite, Campus Mitte Berlin, Germany
- Institute of Vegetative Physiology, Charite Campus Mitte, Berlin
| | - B Eggert
- Department of Pharmacology and Toxicology, Center for Cardiovascular Research, Charite, Campus Mitte Berlin, Germany
| | - K Relle
- Department of Pharmacology and Toxicology, Center for Cardiovascular Research, Charite, Campus Mitte Berlin, Germany
- Institute of Vegetative Physiology, Charite Campus Mitte, Berlin
| | - M Godes
- Department of Pharmacology and Toxicology, Center for Cardiovascular Research, Charite, Campus Mitte Berlin, Germany
- Institute of Vegetative Physiology, Charite Campus Mitte, Berlin
| | - S Heiden
- Department of Pharmacology and Toxicology, Center for Cardiovascular Research, Charite, Campus Mitte Berlin, Germany
| | - Y Sharkovska
- Department of Pharmacology and Toxicology, Center for Cardiovascular Research, Charite, Campus Mitte Berlin, Germany
| | - Y Fischer
- Solvay Pharmaceuticals Research Laboratories Hannover, Germany
| | - D Ziegler
- Solvay Pharmaceuticals Research Laboratories Hannover, Germany
| | - G-W Bielenberg
- Solvay Pharmaceuticals Research Laboratories Hannover, Germany
| | - B Hocher
- Department of Pharmacology and Toxicology, Center for Cardiovascular Research, Charite, Campus Mitte Berlin, Germany
- Solvay Pharmaceuticals Research Laboratories Hannover, Germany
- Author for correspondence:
| |
Collapse
|
24
|
Peart JN, Headrick JP. Adenosinergic cardioprotection: Multiple receptors, multiple pathways. Pharmacol Ther 2007; 114:208-21. [PMID: 17408751 DOI: 10.1016/j.pharmthera.2007.02.004] [Citation(s) in RCA: 130] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2007] [Accepted: 02/08/2007] [Indexed: 11/18/2022]
Abstract
Adenosine, formed primarily via hydrolysis of 5'-AMP, has been historically dubbed a "retaliatory" metabolite due to enhanced local release and beneficial actions during cellular/metabolic stress. From a cardiovascular perspective, evidence indicates the adenosinergic system is essential in mediation of intrinsic protection (e.g., pre- and postconditioning) and determining myocardial resistance to insult. Modulation of adenosine and its receptors thus remains a promising, though as yet not well-realized, approach to amelioration of injury in ischemic-reperfused myocardium. Adenosine exerts effects through A(1), A(2A), A(2B), and A(3) adenosine receptor subtypes (A(1)AR, A(2A)AR, A(2B)AR, and A(3)AR), which are all expressed in myocardial and vascular cells, and couple to G proteins to trigger a range of responses (generally, but not always, beneficial). Adenosine can also enhance tolerance to injurious stimuli via receptor-independent metabolic effects. Given adenosines contribution to preconditioning, it is no surprise that postreceptor signaling typically mimics that associated with preconditioning. This involves activation/translocation of PKC, PI3 kinase, and MAPKs, with ultimate effects at the level of mitochondrial targets-the mitochondrial K(ATP) channel and/or the mitochondrial permeability transition pore (mPTP). Nonetheless, differences in cytoprotective signaling and actions of the different adenosine receptor subtypes have been recently revealed. Our understanding of adenosinergic cytoprotection continues to evolve, with roles for the A(2) subtypes emerging, together with evidence of essential receptor "cross-talk" in mediation of protection. This review focuses on current research into adenosine-mediated cardioprotection, highlighting recent findings which, together with a wealth of prior knowledge, may ultimately facilitate adenosinergic approaches to clinical cardiac protection.
Collapse
Affiliation(s)
- Jason N Peart
- Heart Foundation Research Center, Griffith University, PMB 50 Gold Coast Mail Center, QLD, 4217, Australia.
| | | |
Collapse
|
25
|
Cappello S, Angelone T, Tota B, Pagliaro P, Penna C, Rastaldo R, Corti A, Losano G, Cerra MC. Human recombinant chromogranin A-derived vasostatin-1 mimics preconditioning via an adenosine/nitric oxide signaling mechanism. Am J Physiol Heart Circ Physiol 2007; 293:H719-27. [PMID: 17416598 DOI: 10.1152/ajpheart.01352.2006] [Citation(s) in RCA: 52] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
The acidic protein chromogranin A (CgA) is the precursor of several regulatory peptides generated by specific proteolytic processes. Human recombinant CgA NH(2)-terminal fragment STA-CgA(1-78) (hrSTA-CgA(1-78)), containing vasostatin-1 (CgA(1-76)) domain, exerts a negative inotropic effect and counteracts the beta-adrenergic positive inotropic effect on the rat heart. We hypothesized an involvement of nitric oxide (NO)-dependent pathway in both cardiodepression and cardioprotection by hrSTA-CgA(1-78). We also hypothesized an involvement of adenosine A(1) receptor and protein kinase C (PKC) in cardioprotection by hrSTA-CgA(1-78). Therefore, we evaluated whether 1) the cardioinhibition mediated by hrSTA-CgA(1-78) involves the G(i/o) proteins/NO-dependent signal transduction cascade, 2) hrSTA-CgA(1-78) induces ischemic preconditioning-like protective effects on the myocardium, and 3) inhibition of NO synthase (NOS), adenosine A(1) receptor, or PKC affects hrSTA-CgA(1-78) protection. Using the isolated rat heart, we found that the reduction of left ventricular pressure (LVP), rate-pressure product, and maximal values of the first derivative of LVP elicited by hrSTA-CgA(1-78) at 33 nM is abolished by blocking G(i/o) proteins with pertussis toxin, scavenging NO with hemoglobin, and blocking NOS activity with N(G)-monomethyl-l-arginine or N(5)-(iminoethyl)-l-ornithine, soluble guanylate cyclase with 1H-[1,2,4]oxadiazole-[4,4-a]quinoxalin-1-one, and protein kinase (PKG) with KT5823. Data suggest the involvement of the G(i/o) proteins/NO-cGMP-PKG pathway in the hrSTA-CgA(1-78)-dependent cardioinhibition. When given before 30 min of ischemia, hrSTA-CgA(1-78) significantly reduced the size of the infarct from 64 +/- 4 to 32 +/- 3% of the left ventricular mass. This protective effect was abolished by either NOS inhibition or PKC blockade and was attenuated, but not suppressed, by the blockade of A(1) receptors. These results suggest that hrSTA-CgA(1-78) activity triggers two different pathways: one of these pathways is mediated by A(1) receptors, and the other is mediated by NO release. As with repeated brief preconditioning ischemia, hrSTA-CgA(1-78) may be considered a stimulus strong enough to trigger both pathways, which may converge on PKC.
Collapse
Affiliation(s)
- Sandra Cappello
- Dipartimento di Neuroscienze, Sezione di Fisiologia, Università di Torino, Corso Raffaello 30, 10125 Turin, Italy
| | | | | | | | | | | | | | | | | |
Collapse
|
26
|
Eckle T, Krahn T, Grenz A, Köhler D, Mittelbronn M, Ledent C, Jacobson MA, Osswald H, Thompson LF, Unertl K, Eltzschig HK. Cardioprotection by ecto-5'-nucleotidase (CD73) and A2B adenosine receptors. Circulation 2007; 115:1581-90. [PMID: 17353435 DOI: 10.1161/circulationaha.106.669697] [Citation(s) in RCA: 364] [Impact Index Per Article: 21.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
BACKGROUND Ecto-5'-nucleotidase (CD73)-dependent adenosine generation has been implicated in tissue protection during acute injury. Once generated, adenosine can activate cell-surface adenosine receptors (A1 AR, A2A AR, A2B AR, A3 AR). In the present study, we define the contribution of adenosine to cardioprotection by ischemic preconditioning. METHODS AND RESULTS On the basis of observations of CD73 induction by ischemic preconditioning, we found that inhibition or targeted gene deletion of cd73 abolished infarct size-limiting effects. Moreover, 5'-nucleotidase treatment reconstituted cd73-/- mice and attenuated infarct sizes in wild-type mice. Transcriptional profiling of adenosine receptors suggested a contribution of A2B AR because it was selectively induced by ischemic preconditioning. Specifically, in situ ischemic preconditioning conferred cardioprotection in A1 AR-/-, A2A AR-/-, or A3 AR-/- mice but not in A2B AR-/- mice or in wild-type mice after inhibition of the A2B AR. Moreover, A2B AR agonist treatment significantly reduced infarct sizes after ischemia. CONCLUSIONS Taken together, pharmacological and genetic evidence demonstrate the importance of CD73-dependent adenosine generation and signaling through A2B AR for cardioprotection by ischemic preconditioning and suggests 5'-nucleotidase or A2B AR agonists as therapy for myocardial ischemia.
Collapse
Affiliation(s)
- Tobias Eckle
- Department of Anesthesiology and Intensive Care Medicine, Tübingen University Hospital, Hoppe-Seyler-Str 3, D-72076 Tübingen, Germany
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
27
|
Kawasumi H, Satoh N, Kitada Y. Caldaret, an intracellular Ca2+ handling modulator, limits infarct size of reperfused canine heart. J Pharmacol Sci 2007; 103:222-33. [PMID: 17299242 DOI: 10.1254/jphs.fp0060765] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/23/2022] Open
Abstract
The cardioprotective effect of caldaret, a novel intracellular Ca(2+) handling modulator that acts through reverse-mode Na(+)/Ca(2+) exchanger inhibition and potential sarcoplasmic reticulum (SR) Ca(2+) uptake enhancement, against reperfusion injury was investigated. We employed a canine model of myocardial infarction induced by 90-min occlusion of left circumflex (LCX) coronary artery followed by 4 h of reperfusion. Intravenously infused caldaret (3 or 30 microg/kg per hour) for 30 min at LCX-reperfusion markedly reduced infarct size (by 51.3% or 71.9%, respectively). This cardioprotection was accompanied by an acceleration of left ventricular (LV) contraction and relaxation during reperfusion, but not by an increase in ischemic regional transmural myocardial blood flow (TMBF) or endocardial/epicardial blood flow ratio (Endo/Epi ratio) or a reduction in double-product throughout the protocol. Diltiazem (2000 microg/kg per hour) also reduced infarct size (by 36.1%), but unlike caldaret, was accompanied by the significant increase in Endo/Epi ratio in the ischemic region and decrease in double-product. There were significant inverse relationships between infarct size and ischemic regional TMBF in all groups. Caldaret, but not diltiazem shifted the regression line downward with a flatter slope. These results suggest that the amelioration of intracellular Ca(2+) handling dysfunction achieved by caldaret leads to cardioprotective effects against reperfusion injury following prolonged ischemia.
Collapse
Affiliation(s)
- Hisashi Kawasumi
- Research Laboratory II, Cardiovascular Section, Mitsubishi Pharma Corporation, 1000 Kamoshida, Aoba, Yokohama 227-0053, Japan.
| | | | | |
Collapse
|
28
|
Gross GJ, Auchampach JA. Reperfusion injury: does it exist? J Mol Cell Cardiol 2006; 42:12-8. [PMID: 17069848 PMCID: PMC1876792 DOI: 10.1016/j.yjmcc.2006.09.009] [Citation(s) in RCA: 85] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/28/2006] [Revised: 09/01/2006] [Accepted: 09/21/2006] [Indexed: 11/23/2022]
Abstract
It is well established that reperfusion of the heart is the optimal method of salvaging previously ischemic myocardium. However, the idea of reperfusion injury, i.e. injury caused by the process of reperfusion per se, still remains a controversial issue. In this review, we present mounting evidence supporting the concept that reperfusion injury exists, based on work conducted with adenosine and opioid receptor ligands, and the discovery of two new concepts regarding reperfusion injury: 'postconditioning' (POC) and the reperfusion injury salvage kinase (RISK) signaling pathway.
Collapse
Affiliation(s)
- Garrett J Gross
- Medical College of Wisconsin, Department of Pharmacology and Toxicology, 8701 Watertown Plank Road, Milwaukee, WI 53226, USA.
| | | |
Collapse
|
29
|
Lasley RD, Kristo G, Keith BJ, Mentzer RM. The A2a/A2b receptor antagonist ZM-241385 blocks the cardioprotective effect of adenosine agonist pretreatment in in vivo rat myocardium. Am J Physiol Heart Circ Physiol 2006; 292:H426-31. [PMID: 16980350 DOI: 10.1152/ajpheart.00675.2006] [Citation(s) in RCA: 35] [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: 11/22/2022]
Abstract
There is increasing evidence for interactions among adenosine receptor subtypes in the brain and heart. The purpose of this study was to determine whether the adenosine A(2a) receptor modulates the infarct size-reducing effect of preischemic administration of adenosine receptor agonists in intact rat myocardium. Adult male rats were submitted to in vivo regional myocardial ischemia (25 min) and 2 h reperfusion. Vehicle-treated rats were compared with rats pretreated with the A(1) agonist 2-chloro-N(6)-cyclopentyladenosine (CCPA, 10 mug/kg), the nonselective agonist 5'-N-ethylcarboxamidoadenosine (NECA, 10 mug/kg), or the A(2a) agonist 2-[4-(2-carboxyethyl)phenethylamino]-5'-N-methylcarboxamidoadenosine (CGS-21680, 20 mug/kg). Additional CCPA- and NECA-treated rats were pretreated with the A(1) antagonist 8-cyclopentyl-1,3-dipropylxanthine (DPCPX, 100 mug/kg), the A(2a)/A(2b) antagonist 4-(-2-[7-amino-2-{2-furyl}{1,2,4}triazolo{2,3-a} {1,3,5}triazin-5-yl-amino]ethyl)phenol (ZM-241385, 1.5 mg/kg) or the A(3) antagonist 3-propyl-6-ethyl-5[(ethylthio)carbonyl]-2-phenyl-4-propyl-3-pyridine carboxylate (MRS-1523, 2 mg/kg). CCPA and NECA reduced myocardial infarct size by 50% and 35%, respectively, versus vehicle, but CGS-21680 had no effect. DPCPX blunted the bradycardia associated with CCPA and NECA, whereas ZM-241385 attenuated their hypotensive effects. Both DPCPX and ZM-241385 blocked the protective effects of CCPA and NECA. The A(3) antagonist did not alter the hemodynamic effects of CCPA or NECA, nor did it alter adenosine agonist cardioprotection. None of the antagonists alone altered myocardial infarct size. These findings suggest that although preischemic administration of an A(2a) receptor agonist does not induce cardioprotection, antagonism of the A(2a) and/or the A(2b) receptor blocks the cardioprotection associated with adenosine agonist pretreatment.
Collapse
Affiliation(s)
- Robert D Lasley
- Dept. of Physiology, Wayne State Univ. School of Medicine, 1107 Elliman Bldg., 421 E. Canfield, Detroit, MI 48201, USA.
| | | | | | | |
Collapse
|
30
|
Lankford AR, Yang JN, Rose'Meyer R, French BA, Matherne GP, Fredholm BB, Yang Z. Effect of modulating cardiac A1adenosine receptor expression on protection with ischemic preconditioning. Am J Physiol Heart Circ Physiol 2006; 290:H1469-73. [PMID: 16299262 DOI: 10.1152/ajpheart.00181.2005] [Citation(s) in RCA: 56] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Activation of A1adenosine receptors (A1ARs) may be a crucial step in protection against myocardial ischemia-reperfusion (I/R) injury; however, the use of pharmacological A1AR antagonists to inhibit myocardial protection has yielded inconclusive results. In the current study, we have used mice with genetically modified A1AR expression to define the role of A1AR in intrinsic protection and ischemic preconditioning (IPC) against I/R injury. Normal wild-type (WT) mice, knockout mice with deleted (A1KO−/−) or single-copy (A1KO+/−) A1AR, and transgenic mice (A1TG) with increased cardiac A1AR expression underwent 45 min of left anterior descending coronary artery occlusion, followed by 60 min of reperfusion. Subsets of each group were preconditioned with short durations of ischemia (3 cycles of 5 min of occlusion and 5 min of reperfusion) before index ischemia. Infarct size (IF) in WT, A1KO+/−, and A1KO−/−mice was (in % of risk region) 58 ± 3, 60 ± 4, and 61 ± 2, respectively, and was less in A1TG mice (39 ± 4, P < 0.05). A strong correlation was observed between A1AR expression level and response to IPC. IF was significantly reduced by IPC in WT mice (35 ± 3, P < 0.05 vs. WT), A1KO+/−+ IPC (48 ± 4, P < 0.05 vs. A1KO+/−), and A1TG + IPC mice (24 ± 2, P < 0.05 vs. A1TG). However, IPC did not decrease IF in A1KO−/−+ IPC mice (63 ± 2). In addition, A1KO−/−hearts subjected to global I/R injury demonstrated diminished recovery of developed pressure and diastolic function compared with WT controls. These findings demonstrate that A1ARs are critical for protection from myocardial I/R injury and that cardioprotection with IPC is relative to the level of A1AR gene expression.
Collapse
Affiliation(s)
- Amy R Lankford
- Dept. of Pediatrics, University of Virginia Health System, Charlottesville, VA 22908, USA.
| | | | | | | | | | | | | |
Collapse
|
31
|
Abstract
Adenosine receptors are major targets of caffeine, the most commonly consumed drug in the world. There is growing evidence that they could also be promising therapeutic targets in a wide range of conditions, including cerebral and cardiac ischaemic diseases, sleep disorders, immune and inflammatory disorders and cancer. After more than three decades of medicinal chemistry research, a considerable number of selective agonists and antagonists of adenosine receptors have been discovered, and some have been clinically evaluated, although none has yet received regulatory approval. However, recent advances in the understanding of the roles of the various adenosine receptor subtypes, and in the development of selective and potent ligands, as discussed in this review, have brought the goal of therapeutic application of adenosine receptor modulators considerably closer.
Collapse
Affiliation(s)
- Kenneth 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
|
32
|
Peart JN, Gross GJ. Cardioprotection following adenosine kinase inhibition in rat hearts. Basic Res Cardiol 2005; 100:328-36. [PMID: 15795795 DOI: 10.1007/s00395-005-0526-7] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/08/2004] [Revised: 03/07/2005] [Accepted: 03/09/2005] [Indexed: 10/25/2022]
Abstract
Adenosine kinase phosphorylates adenosine to AMP, the primary pathway for adenosine metabolism under basal conditions. Inhibition of adenosine kinase results in a site-specific increase in interstitial adenosine. Using a rat model of myocardial infarction, we examined the protective effects of adenosine kinase inhibition. Male Sprague-Dawley rats underwent 30 min regional occlusion followed by 90 min reperfusion. Infarct size, expressed as a percent of the area-at-risk, IS/AAR(%), was 58.0 +/- 2.1 % in untreated rats. Pretreatment with the adenosine kinase inhibitor, 5-iodotubercidin (1 mg/kg), limited infarct development to 37.5+/-3.7% (P < 0.001). The A(1) adenosine receptor (A(1)AR) antagonist, DPCPX (100 microg/kg), abolished the infarct-sparing effect of 5-iodotubercidin (IS, 62.8 +/- 1.3%). Similarly, the A(3) adenosine receptor (A(3)AR) antagonist, MRS-1523 (2 mg/kg), and the delta-opioid receptor (DOR) antagonist, BNTX, (1 mg/kg) abolished the reduction of IS produced by iodotubercidin. Pretreatment with the ROS scavenger, 2-MPG (20 mg/kg), or the PKC-delta antagonist, rottlerin (0.3 mg/kg) also abolished iodotubercidin-mediated cardioprotection. Furthermore, pretreatment with 5-HD, a mitochondrial K(ATP) (mitoK(ATP)) channel inhibitor, but not the sarcolemmal K(ATP) channel blocker, HMR-1098, abrogated the beneficial effects of adenosine kinase inhibition (IS, 59.5 +/- 3.8%). These data suggest that inhibition of adenosine kinase is effective in reducing infarct development via A(1)AR, A(3)AR and DOR activation. Data also suggest that this protection is mediated via ROS, PKC-delta and mitoK(ATP) channels.
Collapse
Affiliation(s)
- J N Peart
- Department of Pharmacology & Toxicology, Medical College of Wisconsin, 8701 Watertown Plank Rd, Milwaukee, WI 53226, USA.
| | | |
Collapse
|
33
|
Reichelt ME, Willems L, Molina JG, Sun CX, Noble JC, Ashton KJ, Schnermann J, Blackburn MR, Headrick JP. Genetic Deletion of the A
1
Adenosine Receptor Limits Myocardial Ischemic Tolerance. Circ Res 2005; 96:363-7. [PMID: 15653569 DOI: 10.1161/01.res.0000156075.00127.c3] [Citation(s) in RCA: 47] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Adenosine receptors may be important determinants of intrinsic ischemic tolerance. Genetically modified mice were used to examine effects of global A
1
adenosine receptor (A
1
AR) knockout (KO) on function and ischemic tolerance in perfused mouse hearts. Baseline contractile function and heart rate were unaltered by A
1
AR KO, which was shown to abolish the negative chronotropic effects of 2-chloroadenosine (A
1
AR-mediated) without altering A
2
adenosine receptor–mediated coronary dilation. Tolerance to 25 minutes global normothermic ischemia (followed by 45 minutes reperfusion) was significantly limited by A
1
AR KO, with impaired contractile recovery (reduced by ≈25%) and enhanced lactate dehydrogenase (LDH) efflux (increased by ≈100%). Functional effects of A
1
AR KO involved worsened systolic pressure development with little to no change in diastolic dysfunction. In contrast, cardiac specific A
1
AR overexpression enhanced ischemic tolerance with a primary action on diastolic dysfunction. Nonselective receptor agonism (10 μmol/L 2-chloroadenosine) protected wild-type and also A
1
AR KO hearts (albeit to a lesser extent), implicating protection via subtypes additional to A
1
ARs. However, A
1
AR KO abrogated effects of 2-chloroadenosine on ischemic contracture and diastolic dysfunction. These data are the first demonstrating global deletion of the A
1
AR limits intrinsic myocardial resistance to ischemia. Data indicate the function of intrinsically activated A
1
ARs appears primarily to be enhancement of postischemic contractility and limitation of cell death.
Collapse
Affiliation(s)
- Melissa E Reichelt
- Heart Foundation Research Centre, Griffith University, Southport, QLD, Australia
| | | | | | | | | | | | | | | | | |
Collapse
|