51
|
Adenosine Receptor Adora2b Plays a Mechanistic Role in the Protective Effect of the Volatile Anesthetic Sevoflurane during Liver Ischemia/Reperfusion. Anesthesiology 2016; 125:547-60. [DOI: 10.1097/aln.0000000000001234] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
Abstract
Background
Liver ischemia/reperfusion (IR) injury is characterized by hepatic tissue damage and an inflammatory response. This is accompanied by the formation and vascular sequestration of platelet–neutrophil conjugates (PNCs). Signaling through Adora2b adenosine receptors can provide liver protection. Volatile anesthetics may interact with adenosine receptors. This study investigates potential antiinflammatory effects of the volatile anesthetic sevoflurane during liver IR.
Methods
Experiments were performed ex vivo with human blood and in a liver IR model with wild-type, Adora2a−/−, and Adora2b−/− mice. The effect of sevoflurane on platelet activation, PNC formation and sequestration, cytokine release, and liver damage (alanine aminotransferase release) was analyzed using flow cytometry, luminometry, and immunofluorescence. Adenosine receptor expression in liver tissue was analyzed using immunohistochemistry and real-time polymerase chain reaction.
Results
Ex vivo experiments indicate that sevoflurane inhibits platelet and leukocyte activation (n = 5). During liver IR, sevoflurane (2 Vol%) decreased PNC formation 2.4-fold in wild-type (P < 0.05) but not in Adora2b−/− mice (n ≥ 5). Sevoflurane reduced PNC sequestration 1.9-fold (P < 0.05) and alanine aminotransferase release 3.5-fold (P < 0.05) in wild-type but not in Adora2b−/− mice (n = 5). In Adora2a−/− mice, sevoflurane also inhibited PNC formation and cytokine release. Sevoflurane diminished cytokine release (n ≥ 3) and increased Adora2b transcription and expression in liver tissue of wild-types (n = 4).
Conclusions
Our experiments highlight antiinflammatory and tissue-protective properties of sevoflurane during liver IR and reveal a mechanistic role of Adora2b in sevoflurane-associated effects. The targeted use of sevoflurane not only as an anesthetic but also to prevent IR damage is a promising approach in the treatment of critically ill patients.
Collapse
|
52
|
Kleyman I, Weimer LH. Syncope: Case Studies. Neurol Clin 2016; 34:525-45. [PMID: 27445240 DOI: 10.1016/j.ncl.2016.04.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022]
Abstract
Syncope, or the sudden loss of consciousness, is a common presenting symptom for evaluation by neurologists. It is not a unique diagnosis but rather a common manifestation of disorders with diverse mechanisms. Loss of consciousness is typically preceded by a prodrome of symptoms and sometimes there is a clear trigger. This article discusses several cases that illustrate the various causes of syncope. Reflex syncope is the most common type and includes neurally mediated, vasovagal, situational, carotid sinus hypersensitivity, and atypical forms. Acute and chronic autonomic neuropathies and neurodegenerative disorders can also present with syncope.
Collapse
Affiliation(s)
- Inna Kleyman
- Department of Neurology, Columbia University College of Physicians and Surgeons, Neurological Institute of New York, 710 West 168th Street, New York, NY 10032, USA
| | - Louis H Weimer
- Department of Neurology, Columbia University College of Physicians and Surgeons, Neurological Institute of New York, 710 West 168th Street, New York, NY 10032, USA.
| |
Collapse
|
53
|
Abstract
Syncope due to idiopathic AV block is characterized by: 1) ECG documentation (usually by means of prolonged ECG monitoring) of paroxysmal complete AV block with one or multiple consecutive pauses, without P-P cycle lengthening or PR interval prolongation, not triggered by atrial or ventricular premature beats nor by rate variations; 2) long history of recurrent syncope without prodromes; 3) absence of cardiac and ECG abnormalities; 4) absence of progression to persistent forms of AV block; 5) efficacy of cardiac pacing therapy. The patients affected by idiopathic AV block have low baseline adenosine plasma level values and show an increased susceptibility to exogenous adenosine. The APL value of the patients with idiopathic AV block is much lower than patients affected by vasovagal syncope who have high adenosine values.
Collapse
Affiliation(s)
- Michele Brignole
- Department of Cardiology, Arrhythmologic Center, Ospedali del Tigullio, Via Don Bobbio 25, Lavagna 16033, Italy.
| | - Jean-Claude Deharo
- Department of Cardiology, Timone University Hospital, 264, rue Saint Pierre 13385, Marseille, France
| | - Regis Guieu
- Laboratory of Biochemistry and Molecular Biology, Timone University Hospital, Unité Mixte de Recherche Ministere de la Defense, Aix Marseille Université, Boulevard P Dramard, Marseille 13015, France
| |
Collapse
|
54
|
Liu H, Xia Y. Beneficial and detrimental role of adenosine signaling in diseases and therapy. J Appl Physiol (1985) 2015; 119:1173-82. [PMID: 26316513 DOI: 10.1152/japplphysiol.00350.2015] [Citation(s) in RCA: 55] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2015] [Accepted: 08/25/2015] [Indexed: 12/17/2022] Open
Abstract
Adenosine is a major signaling nucleoside that orchestrates cellular and tissue adaptation under energy depletion and ischemic/hypoxic conditions by activation of four G protein-coupled receptors (GPCR). The regulation and generation of extracellular adenosine in response to stress are critical in tissue protection. Both mouse and human studies reported that extracellular adenosine signaling plays a beneficial role during acute states. However, prolonged excess extracellular adenosine is detrimental and contributes to the development and progression of various chronic diseases. In recent years, substantial progress has been made to understand the role of adenosine signaling in different conditions and to clarify its significance during the course of disease progression in various organs. These efforts have and will identify potential therapeutic possibilities for protection of tissue injury at acute stage by upregulation of adenosine signaling or attenuation of chronic disease progression by downregulation of adenosine signaling. This review is to summarize current progress and the importance of adenosine signaling in different disease stages and its potential therapeutic effects.
Collapse
Affiliation(s)
- Hong Liu
- Department of Biochemistry and Molecular Biology, The University of Texas Health Science Center at Houston, Houston, Texas; Graduate School of Biomedical Science, University of Texas Health Science Center at Houston, Houston, Texas; Department of Otolaryngology, Xiangya Hospital, Central South University, Changsha, Hunan, China; and
| | - Yang Xia
- Department of Biochemistry and Molecular Biology, The University of Texas Health Science Center at Houston, Houston, Texas; Graduate School of Biomedical Science, University of Texas Health Science Center at Houston, Houston, Texas; Department of Nephrology, Xiangya Hospital, Central South University, Changsha, Hunan, China
| |
Collapse
|
55
|
Bonney S, Hughes K, Eckle T. Anesthetic cardioprotection: the role of adenosine. Curr Pharm Des 2015; 20:5690-5. [PMID: 24502579 DOI: 10.2174/1381612820666140204102524] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2013] [Accepted: 02/03/2014] [Indexed: 12/25/2022]
Abstract
Brief periods of cardiac ischemia and reperfusion exert a protective effect against subsequent longer ischemic periods, a phenomenon coined ischemic preconditioning. Similarly, repeated brief episodes of coronary occlusion and reperfusion at the onset of reperfusion, called post-conditioning, dramatically reduce infarct sizes. Interestingly, both effects can be achieved by the administration of any volatile anesthetic. In fact, cardio-protection by volatile anesthetics is an older phenomenon than ischemic pre- or post-conditioning. Although the mechanism through which anesthetics can mimic ischemic pre- or post-conditioning is still unknown, adenosine generation and signaling are the most redundant triggers in ischemic pre- or post-conditioning. In fact, adenosine signaling has been implicated in isoflurane-mediated cardioprotection. Adenosine acts via four receptors designated as A1, A2a, A2b, and A3. Cardioprotection has been associated with all subtypes, although the role of each remains controversial. Much of the controversy stems from the abundance of receptor agonists and antagonists that are, in fact, capable of interacting with multiple receptor subtypes. Recently, more specific receptor agonists and new genetic animal models have become available paving way towards new discoveries. As such, the adenosine A2b receptor was shown to be the only one of the adenosine receptors whose cardiac expression is induced by ischemia in both mice and humans and whose function is implicated in ischemic pre- or post-conditioning. In the current review, we will focus on adenosine signaling in the context of anesthetic cardioprotection and will highlight new discoveries, which could lead to new therapeutic concepts to treat myocardial ischemia using anesthetic preconditioning.
Collapse
Affiliation(s)
| | | | - Tobias Eckle
- Department of Anesthesiology, University of Colorado Denver, 12700 E 19th Avenue, Mailstop B112, RC 2, Room 7121, Aurora, CO 80045.
| |
Collapse
|
56
|
Burnstock G, Pelleg A. Cardiac purinergic signalling in health and disease. Purinergic Signal 2015; 11:1-46. [PMID: 25527177 PMCID: PMC4336308 DOI: 10.1007/s11302-014-9436-1] [Citation(s) in RCA: 103] [Impact Index Per Article: 11.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2014] [Accepted: 11/25/2014] [Indexed: 01/09/2023] Open
Abstract
This review is a historical account about purinergic signalling in the heart, for readers to see how ideas and understanding have changed as new experimental results were published. Initially, the focus is on the nervous control of the heart by ATP as a cotransmitter in sympathetic, parasympathetic, and sensory nerves, as well as in intracardiac neurons. Control of the heart by centers in the brain and vagal cardiovascular reflexes involving purines are also discussed. The actions of adenine nucleotides and nucleosides on cardiomyocytes, atrioventricular and sinoatrial nodes, cardiac fibroblasts, and coronary blood vessels are described. Cardiac release and degradation of ATP are also described. Finally, the involvement of purinergic signalling and its therapeutic potential in cardiac pathophysiology is reviewed, including acute and chronic heart failure, ischemia, infarction, arrhythmias, cardiomyopathy, syncope, hypertrophy, coronary artery disease, angina, diabetic cardiomyopathy, as well as heart transplantation and coronary bypass grafts.
Collapse
Affiliation(s)
- Geoffrey Burnstock
- Autonomic Neuroscience Centre, University College Medical School, Rowland Hill Street, London, NW3 2PF, UK,
| | | |
Collapse
|
57
|
Idzko M, Ferrari D, Riegel AK, Eltzschig HK. Extracellular nucleotide and nucleoside signaling in vascular and blood disease. Blood 2014; 124:1029-37. [PMID: 25001468 PMCID: PMC4133480 DOI: 10.1182/blood-2013-09-402560] [Citation(s) in RCA: 107] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2013] [Accepted: 06/16/2014] [Indexed: 12/17/2022] Open
Abstract
Nucleotides and nucleosides-such as adenosine triphosphate (ATP) and adenosine-are famous for their intracellular roles as building blocks for the genetic code or cellular energy currencies. In contrast, their function in the extracellular space is different. Here, they are primarily known as signaling molecules via activation of purinergic receptors, classified as P1 receptors for adenosine or P2 receptors for ATP. Because extracellular ATP is rapidly converted to adenosine by ectonucleotidase, nucleotide-phosphohydrolysis is important for controlling the balance between P2 and P1 signaling. Gene-targeted mice for P1, P2 receptors, or ectonucleotidase exhibit only very mild phenotypic manifestations at baseline. However, they demonstrate alterations in disease susceptibilities when exposed to a variety of vascular or blood diseases. Examples of phenotypic manifestations include vascular barrier dysfunction, graft-vs-host disease, platelet activation, ischemia, and reperfusion injury or sickle cell disease. Many of these studies highlight that purinergic signaling events can be targeted therapeutically.
Collapse
Affiliation(s)
- Marco Idzko
- Department of Pneumology, Freiburg University Medical Center, Albert-Ludwigs-University, Freiburg, Germany
| | - Davide Ferrari
- Department of Morphology, Surgery and Experimental Medicine, Section of General Pathology, Oncology and Experimental Biology, University of Ferrara, Ferrara, Italy; and
| | - Ann-Kathrin Riegel
- Organ Protection Program, Department of Anesthesiology, University of Colorado School of Medicine, Aurora, CO
| | - Holger K Eltzschig
- Organ Protection Program, Department of Anesthesiology, University of Colorado School of Medicine, Aurora, CO
| |
Collapse
|
58
|
Lou Q, Hansen BJ, Fedorenko O, Csepe TA, Kalyanasundaram A, Li N, Hage LT, Glukhov AV, Billman GE, Weiss R, Mohler PJ, Györke S, Biesiadecki BJ, Carnes CA, Fedorov VV. Upregulation of adenosine A1 receptors facilitates sinoatrial node dysfunction in chronic canine heart failure by exacerbating nodal conduction abnormalities revealed by novel dual-sided intramural optical mapping. Circulation 2014; 130:315-24. [PMID: 24838362 DOI: 10.1161/circulationaha.113.007086] [Citation(s) in RCA: 55] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/02/2023]
Abstract
BACKGROUND Although sinoatrial node (SAN) dysfunction is a hallmark of human heart failure (HF), the underlying mechanisms remain poorly understood. We aimed to examine the role of adenosine in SAN dysfunction and tachy-brady arrhythmias in chronic HF. METHODS AND RESULTS We applied multiple approaches to characterize SAN structure, SAN function, and adenosine A1 receptor expression in control (n=17) and 4-month tachypacing-induced chronic HF (n=18) dogs. Novel intramural optical mapping of coronary-perfused right atrial preparations revealed that adenosine (10 μmol/L) markedly prolonged postpacing SAN conduction time in HF by 206 ± 99 milliseconds (versus 66 ± 21 milliseconds in controls; P=0.02). Adenosine induced SAN intranodal conduction block or microreentry in 6 of 8 dogs with HF versus 0 of 7 controls (P=0.007). Adenosine-induced SAN conduction abnormalities and automaticity depression caused postpacing atrial pauses in HF versus control dogs (17.1 ± 28.9 versus 1.5 ± 1.3 seconds; P<0.001). Furthermore, 10 μmol/L adenosine shortened atrial repolarization and led to pacing-induced atrial fibrillation in 6 of 7 HF versus 0 of 7 control dogs (P=0.002). Adenosine-induced SAN dysfunction and atrial fibrillation were abolished or prevented by adenosine A1 receptor antagonists (50 μmol/L theophylline/1 μmol/L 8-cyclopentyl-1,3-dipropylxanthine). Adenosine A1 receptor protein expression was significantly upregulated during HF in the SAN (by 47 ± 19%) and surrounding atrial myocardium (by 90 ± 40%). Interstitial fibrosis was significantly increased within the SAN in HF versus control dogs (38 ± 4% versus 23 ± 4%; P<0.001). CONCLUSIONS In chronic HF, adenosine A1 receptor upregulation in SAN pacemaker and atrial cardiomyocytes may increase cardiac sensitivity to adenosine. This effect may exacerbate conduction abnormalities in the structurally impaired SAN, leading to SAN dysfunction, and potentiate atrial repolarization shortening, thereby facilitating atrial fibrillation. Atrial fibrillation may further depress SAN function and lead to tachy-brady arrhythmias in HF.
Collapse
Affiliation(s)
- Qing Lou
- From the Department of Physiology and Cell Biology (Q.L., B.J.H., O.F., T.A.C., A.K., N.L., L.T.H., A.V.G., G.E.B., P.J.M., S.G., B.J.B., V.V.F.), Davis Heart & Lung Research Institute (Q.L., A.K., G.E.B., R.W., P.J.M., S.G., B.J.B., C.A.C., V.V.F.), and Department of Internal Medicine (R.W., P.J.M.), The Ohio State University Wexner Medical Center, Columbus, OH; Mental Health Research Institute and National Research Tomsk Polytechnic University, Tomsk, Russia (O.F.); and College of Pharmacy, The Ohio State University, Columbus (C.A.C.)
| | - Brian J Hansen
- From the Department of Physiology and Cell Biology (Q.L., B.J.H., O.F., T.A.C., A.K., N.L., L.T.H., A.V.G., G.E.B., P.J.M., S.G., B.J.B., V.V.F.), Davis Heart & Lung Research Institute (Q.L., A.K., G.E.B., R.W., P.J.M., S.G., B.J.B., C.A.C., V.V.F.), and Department of Internal Medicine (R.W., P.J.M.), The Ohio State University Wexner Medical Center, Columbus, OH; Mental Health Research Institute and National Research Tomsk Polytechnic University, Tomsk, Russia (O.F.); and College of Pharmacy, The Ohio State University, Columbus (C.A.C.)
| | - Olga Fedorenko
- From the Department of Physiology and Cell Biology (Q.L., B.J.H., O.F., T.A.C., A.K., N.L., L.T.H., A.V.G., G.E.B., P.J.M., S.G., B.J.B., V.V.F.), Davis Heart & Lung Research Institute (Q.L., A.K., G.E.B., R.W., P.J.M., S.G., B.J.B., C.A.C., V.V.F.), and Department of Internal Medicine (R.W., P.J.M.), The Ohio State University Wexner Medical Center, Columbus, OH; Mental Health Research Institute and National Research Tomsk Polytechnic University, Tomsk, Russia (O.F.); and College of Pharmacy, The Ohio State University, Columbus (C.A.C.)
| | - Thomas A Csepe
- From the Department of Physiology and Cell Biology (Q.L., B.J.H., O.F., T.A.C., A.K., N.L., L.T.H., A.V.G., G.E.B., P.J.M., S.G., B.J.B., V.V.F.), Davis Heart & Lung Research Institute (Q.L., A.K., G.E.B., R.W., P.J.M., S.G., B.J.B., C.A.C., V.V.F.), and Department of Internal Medicine (R.W., P.J.M.), The Ohio State University Wexner Medical Center, Columbus, OH; Mental Health Research Institute and National Research Tomsk Polytechnic University, Tomsk, Russia (O.F.); and College of Pharmacy, The Ohio State University, Columbus (C.A.C.)
| | - Anuradha Kalyanasundaram
- From the Department of Physiology and Cell Biology (Q.L., B.J.H., O.F., T.A.C., A.K., N.L., L.T.H., A.V.G., G.E.B., P.J.M., S.G., B.J.B., V.V.F.), Davis Heart & Lung Research Institute (Q.L., A.K., G.E.B., R.W., P.J.M., S.G., B.J.B., C.A.C., V.V.F.), and Department of Internal Medicine (R.W., P.J.M.), The Ohio State University Wexner Medical Center, Columbus, OH; Mental Health Research Institute and National Research Tomsk Polytechnic University, Tomsk, Russia (O.F.); and College of Pharmacy, The Ohio State University, Columbus (C.A.C.)
| | - Ning Li
- From the Department of Physiology and Cell Biology (Q.L., B.J.H., O.F., T.A.C., A.K., N.L., L.T.H., A.V.G., G.E.B., P.J.M., S.G., B.J.B., V.V.F.), Davis Heart & Lung Research Institute (Q.L., A.K., G.E.B., R.W., P.J.M., S.G., B.J.B., C.A.C., V.V.F.), and Department of Internal Medicine (R.W., P.J.M.), The Ohio State University Wexner Medical Center, Columbus, OH; Mental Health Research Institute and National Research Tomsk Polytechnic University, Tomsk, Russia (O.F.); and College of Pharmacy, The Ohio State University, Columbus (C.A.C.)
| | - Lori T Hage
- From the Department of Physiology and Cell Biology (Q.L., B.J.H., O.F., T.A.C., A.K., N.L., L.T.H., A.V.G., G.E.B., P.J.M., S.G., B.J.B., V.V.F.), Davis Heart & Lung Research Institute (Q.L., A.K., G.E.B., R.W., P.J.M., S.G., B.J.B., C.A.C., V.V.F.), and Department of Internal Medicine (R.W., P.J.M.), The Ohio State University Wexner Medical Center, Columbus, OH; Mental Health Research Institute and National Research Tomsk Polytechnic University, Tomsk, Russia (O.F.); and College of Pharmacy, The Ohio State University, Columbus (C.A.C.)
| | - Alexey V Glukhov
- From the Department of Physiology and Cell Biology (Q.L., B.J.H., O.F., T.A.C., A.K., N.L., L.T.H., A.V.G., G.E.B., P.J.M., S.G., B.J.B., V.V.F.), Davis Heart & Lung Research Institute (Q.L., A.K., G.E.B., R.W., P.J.M., S.G., B.J.B., C.A.C., V.V.F.), and Department of Internal Medicine (R.W., P.J.M.), The Ohio State University Wexner Medical Center, Columbus, OH; Mental Health Research Institute and National Research Tomsk Polytechnic University, Tomsk, Russia (O.F.); and College of Pharmacy, The Ohio State University, Columbus (C.A.C.)
| | - George E Billman
- From the Department of Physiology and Cell Biology (Q.L., B.J.H., O.F., T.A.C., A.K., N.L., L.T.H., A.V.G., G.E.B., P.J.M., S.G., B.J.B., V.V.F.), Davis Heart & Lung Research Institute (Q.L., A.K., G.E.B., R.W., P.J.M., S.G., B.J.B., C.A.C., V.V.F.), and Department of Internal Medicine (R.W., P.J.M.), The Ohio State University Wexner Medical Center, Columbus, OH; Mental Health Research Institute and National Research Tomsk Polytechnic University, Tomsk, Russia (O.F.); and College of Pharmacy, The Ohio State University, Columbus (C.A.C.)
| | - Raul Weiss
- From the Department of Physiology and Cell Biology (Q.L., B.J.H., O.F., T.A.C., A.K., N.L., L.T.H., A.V.G., G.E.B., P.J.M., S.G., B.J.B., V.V.F.), Davis Heart & Lung Research Institute (Q.L., A.K., G.E.B., R.W., P.J.M., S.G., B.J.B., C.A.C., V.V.F.), and Department of Internal Medicine (R.W., P.J.M.), The Ohio State University Wexner Medical Center, Columbus, OH; Mental Health Research Institute and National Research Tomsk Polytechnic University, Tomsk, Russia (O.F.); and College of Pharmacy, The Ohio State University, Columbus (C.A.C.)
| | - Peter J Mohler
- From the Department of Physiology and Cell Biology (Q.L., B.J.H., O.F., T.A.C., A.K., N.L., L.T.H., A.V.G., G.E.B., P.J.M., S.G., B.J.B., V.V.F.), Davis Heart & Lung Research Institute (Q.L., A.K., G.E.B., R.W., P.J.M., S.G., B.J.B., C.A.C., V.V.F.), and Department of Internal Medicine (R.W., P.J.M.), The Ohio State University Wexner Medical Center, Columbus, OH; Mental Health Research Institute and National Research Tomsk Polytechnic University, Tomsk, Russia (O.F.); and College of Pharmacy, The Ohio State University, Columbus (C.A.C.)
| | - Sándor Györke
- From the Department of Physiology and Cell Biology (Q.L., B.J.H., O.F., T.A.C., A.K., N.L., L.T.H., A.V.G., G.E.B., P.J.M., S.G., B.J.B., V.V.F.), Davis Heart & Lung Research Institute (Q.L., A.K., G.E.B., R.W., P.J.M., S.G., B.J.B., C.A.C., V.V.F.), and Department of Internal Medicine (R.W., P.J.M.), The Ohio State University Wexner Medical Center, Columbus, OH; Mental Health Research Institute and National Research Tomsk Polytechnic University, Tomsk, Russia (O.F.); and College of Pharmacy, The Ohio State University, Columbus (C.A.C.)
| | - Brandon J Biesiadecki
- From the Department of Physiology and Cell Biology (Q.L., B.J.H., O.F., T.A.C., A.K., N.L., L.T.H., A.V.G., G.E.B., P.J.M., S.G., B.J.B., V.V.F.), Davis Heart & Lung Research Institute (Q.L., A.K., G.E.B., R.W., P.J.M., S.G., B.J.B., C.A.C., V.V.F.), and Department of Internal Medicine (R.W., P.J.M.), The Ohio State University Wexner Medical Center, Columbus, OH; Mental Health Research Institute and National Research Tomsk Polytechnic University, Tomsk, Russia (O.F.); and College of Pharmacy, The Ohio State University, Columbus (C.A.C.)
| | - Cynthia A Carnes
- From the Department of Physiology and Cell Biology (Q.L., B.J.H., O.F., T.A.C., A.K., N.L., L.T.H., A.V.G., G.E.B., P.J.M., S.G., B.J.B., V.V.F.), Davis Heart & Lung Research Institute (Q.L., A.K., G.E.B., R.W., P.J.M., S.G., B.J.B., C.A.C., V.V.F.), and Department of Internal Medicine (R.W., P.J.M.), The Ohio State University Wexner Medical Center, Columbus, OH; Mental Health Research Institute and National Research Tomsk Polytechnic University, Tomsk, Russia (O.F.); and College of Pharmacy, The Ohio State University, Columbus (C.A.C.)
| | - Vadim V Fedorov
- From the Department of Physiology and Cell Biology (Q.L., B.J.H., O.F., T.A.C., A.K., N.L., L.T.H., A.V.G., G.E.B., P.J.M., S.G., B.J.B., V.V.F.), Davis Heart & Lung Research Institute (Q.L., A.K., G.E.B., R.W., P.J.M., S.G., B.J.B., C.A.C., V.V.F.), and Department of Internal Medicine (R.W., P.J.M.), The Ohio State University Wexner Medical Center, Columbus, OH; Mental Health Research Institute and National Research Tomsk Polytechnic University, Tomsk, Russia (O.F.); and College of Pharmacy, The Ohio State University, Columbus (C.A.C.).
| |
Collapse
|
59
|
Kashihara T, Hirose M, Shimojo H, Nakada T, Gomi S, Hongo M, Yamada M. β(2)-Adrenergic and M(2)-muscarinic receptors decrease basal t-tubular L-type Ca2+ channel activity and suppress ventricular contractility in heart failure. Eur J Pharmacol 2013; 724:122-31. [PMID: 24389135 DOI: 10.1016/j.ejphar.2013.12.037] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2013] [Revised: 11/29/2013] [Accepted: 12/15/2013] [Indexed: 01/01/2023]
Abstract
L-type Ca(2+) channels (LTCC) play a crucial role in cardiac excitation-contraction coupling. We previously found that in failing ventricular myocytes of mice chronically treated with isoproterenol, basal t-tubular (TT) LTCC activity was halved by activation of protein phosphatase (PP)2A whereas basal surface sarcolemmal (SS) LTCC activity was doubled by inhibition of PP1. Interestingly, chronic treatment of these mice with pertussis toxin almost completely normalized TT and SS LTCC densities and cardiac contractility. In the present study, we therefore sought to identify the Gi/o protein-coupled receptors in cardiac myocytes (i.e. β2-adrenergic, M2-muscarinic and A1-adenosine receptors) that are responsible for these abnormalities in heart failure by chronically administrating mice a selective antagonist of each receptor (ICI118,551, atropine and 8-cyclopentyl-1,3-dipropilxanthine (DPCPX), respectively) with isoproterenol. Compared with mice treated with isoproterenol alone, mice treated with isoproterenol plus ICI118,551 or atropine, but not DPCPX showed significantly lower lung weight/tibial length, higher fractional shortening, lower left ventricular end-diastolic pressure and higher dP/dtmax and dP/dtmin. In addition, ventricular myocytes of mice treated with isoproterenol plus ICI118,551 or atropine, but not DPCPX exhibited significantly higher TT and lower SS LTCC current densities than those of mice treated with isoproterenol alone due to normalization of the PP activities. These results indicate that β2-adrenergic, M2-muscarinic, but not A1-adenosine receptors contribute to reduced ventricular contractility at least partially by decreasing basal TT LTCC activity in heart failure. Therefore, antagonists of β2-adrenergic and/or M2-muscarinic receptors can be good adjuncts to β1-adrenergic receptor antagonists in the treatment of heart failure.
Collapse
Affiliation(s)
- Toshihide Kashihara
- Department of Molecular Pharmacology, Shinshu University School of Medicine, 3-1-1 Asahi, Matsumoto, Nagano 390-8621, Japan
| | - Masamichi Hirose
- Department of Molecular and Cellular Pharmacology, School of Pharmaceutical Sciences, Iwate Medical University, Morioka, Iwate, Japan
| | - Hisashi Shimojo
- Department of Pathology, Shinshu University School of Medicine, Matsumoto, Nagano, Japan
| | - Tsutomu Nakada
- Department of Molecular Pharmacology, Shinshu University School of Medicine, 3-1-1 Asahi, Matsumoto, Nagano 390-8621, Japan
| | - Simmon Gomi
- Department of Molecular Pharmacology, Shinshu University School of Medicine, 3-1-1 Asahi, Matsumoto, Nagano 390-8621, Japan; Department of Cardiovascular Medicine, Shinshu University School of Medicine, Matsumoto, Nagano, Japan
| | - Minoru Hongo
- Department of Cardiovascular Medicine, Shinshu University School of Health Science, Matsumoto, Nagano, Japan
| | - Mitsuhiko Yamada
- Department of Molecular Pharmacology, Shinshu University School of Medicine, 3-1-1 Asahi, Matsumoto, Nagano 390-8621, Japan.
| |
Collapse
|
60
|
Bonney S, Kominsky D, Brodsky K, Eltzschig H, Walker L, Eckle T. Cardiac Per2 functions as novel link between fatty acid metabolism and myocardial inflammation during ischemia and reperfusion injury of the heart. PLoS One 2013; 8:e71493. [PMID: 23977055 PMCID: PMC3748049 DOI: 10.1371/journal.pone.0071493] [Citation(s) in RCA: 60] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2013] [Accepted: 07/01/2013] [Indexed: 01/06/2023] Open
Abstract
Disruption of peripheral circadian rhyme pathways dominantly leads to metabolic disorders. Studies on circadian rhythm proteins in the heart indicated a role for Clock or Per2 in cardiac metabolism. In contrast to Clock−/−, Per2−/− mice have larger infarct sizes with deficient lactate production during myocardial ischemia. To test the hypothesis that cardiac Per2 represents an important regulator of cardiac metabolism during myocardial ischemia, we measured lactate during reperfusion in Per1−/−, Per2−/− or wildtype mice. As lactate measurements in whole blood indicated an exclusive role of Per2 in controlling lactate production during myocardial ischemia, we next performed gene array studies using various ischemia-reperfusion protocols comparing wildtype and Per2−/− mice. Surprisingly, high-throughput gene array analysis revealed dominantly lipid metabolism as the differentially regulated pathway in wildtype mice when compared to Per2−/−. In all ischemia-reperfusion protocols used, the enzyme enoyl-CoA hydratase, which is essential in fatty acid beta-oxidation, was regulated in wildtype animals only. Studies using nuclear magnet resonance imaging (NMRI) confirmed altered fatty acid populations with higher mono-unsaturated fatty acid levels in hearts from Per2−/− mice. Unexpectedly, studies on gene regulation during reperfusion revealed solely pro inflammatory genes as differentially regulated ‘Per2-genes’. Subsequent studies on inflammatory markers showed increasing IL-6 or TNFα levels during reperfusion in Per2−/− mice. In summary, these studies reveal an important role of cardiac Per2 for fatty acid metabolism and inflammation during myocardial ischemia and reperfusion, respectively.
Collapse
Affiliation(s)
- Stephanie Bonney
- Department of Anesthesiology and Mucosal Inflammation Program, University of Colorado Denver, Aurora, Colorado, United States of America
| | - Doug Kominsky
- Department of Anesthesiology and Mucosal Inflammation Program, University of Colorado Denver, Aurora, Colorado, United States of America
| | - Kelley Brodsky
- Department of Anesthesiology and Mucosal Inflammation Program, University of Colorado Denver, Aurora, Colorado, United States of America
| | - Holger Eltzschig
- Department of Anesthesiology and Mucosal Inflammation Program, University of Colorado Denver, Aurora, Colorado, United States of America
| | - Lori Walker
- Division of Cardiology, Department of Medicine, University of Colorado Denver, Aurora, United States of America
| | - Tobias Eckle
- Department of Anesthesiology and Mucosal Inflammation Program, University of Colorado Denver, Aurora, Colorado, United States of America
- * E-mail:
| |
Collapse
|
61
|
|
62
|
Detrimental role of the airway mucin Muc5ac during ventilator-induced lung injury. Mucosal Immunol 2013; 6:762-75. [PMID: 23187315 PMCID: PMC3890100 DOI: 10.1038/mi.2012.114] [Citation(s) in RCA: 58] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
Acute lung injury (ALI) is associated with high morbidity and mortality in critically ill patients. At present, the functional contribution of airway mucins to ALI is unknown. We hypothesized that excessive mucus production could be detrimental during lung injury. Initial transcriptional profiling of airway mucins revealed a selective and robust induction of MUC5AC upon cyclic mechanical stretch exposure of pulmonary epithelia (Calu-3). Additional studies confirmed time- and stretch-dose-dependent induction of MUC5AC transcript or protein during cyclic mechanical stretch exposure in vitro or during ventilator-induced lung injury in vivo. Patients suffering from ALI showed a 58-fold increase in MUC5AC protein in their bronchoalveolar lavage. Studies of the MUC5AC promoter implicated nuclear factor κB in Muc5ac induction during ALI. Moreover, mice with gene-targeted deletion of Muc5ac⁻/⁻ experience attenuated lung inflammation and pulmonary edema during injurious ventilation. We observed that neutrophil trafficking into the lungs of Muc5ac⁻/⁻ mice was selectively attenuated. This implicates that endogenous Muc5ac production enhances pulmonary neutrophil trafficking during lung injury. Together, these studies reveal a detrimental role for endogenous Muc5ac production during ALI and suggest pharmacological strategies to dampen mucin production in the treatment of lung injury.
Collapse
|
63
|
Chen JF, Eltzschig HK, Fredholm BB. Adenosine receptors as drug targets--what are the challenges? Nat Rev Drug Discov 2013; 12:265-86. [PMID: 23535933 PMCID: PMC3930074 DOI: 10.1038/nrd3955] [Citation(s) in RCA: 662] [Impact Index Per Article: 60.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Adenosine signalling has long been a target for drug development, with adenosine itself or its derivatives being used clinically since the 1940s. In addition, methylxanthines such as caffeine have profound biological effects as antagonists at adenosine receptors. Moreover, drugs such as dipyridamole and methotrexate act by enhancing the activation of adenosine receptors. There is strong evidence that adenosine has a functional role in many diseases, and several pharmacological compounds specifically targeting individual adenosine receptors--either directly or indirectly--have now entered the clinic. However, only one adenosine receptor-specific agent--the adenosine A2A receptor agonist regadenoson (Lexiscan; Astellas Pharma)--has so far gained approval from the US Food and Drug Administration (FDA). Here, we focus on the biology of adenosine signalling to identify hurdles in the development of additional pharmacological compounds targeting adenosine receptors and discuss strategies to overcome these challenges.
Collapse
Affiliation(s)
- Jiang-Fan Chen
- Department of Neurology and Pharmacology, Boston University School of Medicine, Boston, Massachusetts 02118, USA
| | | | | |
Collapse
|
64
|
Eltzschig HK, Bonney SK, Eckle T. Attenuating myocardial ischemia by targeting A2B adenosine receptors. Trends Mol Med 2013; 19:345-54. [PMID: 23540714 DOI: 10.1016/j.molmed.2013.02.005] [Citation(s) in RCA: 89] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2012] [Revised: 02/12/2013] [Accepted: 02/20/2013] [Indexed: 02/06/2023]
Abstract
Myocardial ischemia is associated with profound tissue hypoxia due to an imbalance in oxygen supply and demand, and studies of hypoxia-elicited adaptive responses during myocardial ischemia revealed a cardioprotective role for the signaling molecule adenosine. In ischemic human hearts, the A2B adenosine receptor (ADORA2B) is selectively induced. Functional studies in genetic models show that ADORA2B signaling attenuates myocardial infarction by adapting metabolism towards more oxygen efficient utilization of carbohydrates. This adenosine-mediated cardio-adaptive response involves the transcription factor hypoxia-inducible factor HIF1α and the circadian rhythm protein PER2. In this article, we discuss advances in the understanding of adenosine-elicited cardioprotection with particular emphasis on ADORA2B, its downstream targets, and the implications for novel strategies to prevent or treat myocardial ischemia.
Collapse
Affiliation(s)
- Holger K Eltzschig
- Mucosal Inflammation Program, Department of Anesthesiology, University of Colorado School of Medicine, Aurora, CO, USA.
| | | | | |
Collapse
|
65
|
Affiliation(s)
- Holger K Eltzschig
- Mucosal Inflammation Program, Department of Anesthesiology, University of Colorado School of Medicine, Aurora, CO 80045, USA.
| | | | | |
Collapse
|
66
|
Iliff BW, Swoap SJ. Central adenosine receptor signaling is necessary for daily torpor in mice. Am J Physiol Regul Integr Comp Physiol 2012; 303:R477-84. [PMID: 22785425 DOI: 10.1152/ajpregu.00081.2012] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
When calorically restricted at cool ambient temperatures, mice conserve energy by entering torpor, during which metabolic rate (MR), body temperature (T(b)), heart rate (HR), and locomotor activity (LMA) decrease. Treatment with exogenous adenosine produces a similar hypometabolic state. In this study, we conducted a series of experiments using the nonspecific adenosine receptor antagonists aminophylline and 8-sulfophenyltheophylline (8-SPT) to test the hypothesis that adenosine signaling is necessary for torpor in fasted mice. In the first experiment, mice were subcutaneously infused with aminophylline while T(b), HR, and LMA were continuously monitored using implanted radiotelemeters. During a 23-h fast, saline-treated mice were torpid for 518 ± 43 min, whereas aminophylline-treated mice were torpid for significantly less time (54 ± 20 min). In a second experiment, aminophylline was infused subcutaneously into torpid mice to test the role of adenosine in the maintenance of torpor. Aminophylline reversed the hypometabolism, hypothermia, bradycardia, and hypoactivity of torpor, whereas saline did not. In the third and fourth experiments, the polar adenosine antagonist 8-SPT, which does not cross the blood-brain barrier, was infused either subcutaneously or intracerebroventricularly to test the hypothesis that both peripheral and central adenosine receptor signaling are necessary for the maintenance of torpor. Intracerebroventricular, but not subcutaneous, infusion of 8-SPT causes a return to euthermia. These findings support the hypothesis that adenosine is necessary for torpor in mice and further suggest that whereas peripheral adenosine signaling is not necessary for the maintenance of torpor, antagonism of central adenosine is sufficient to disrupt torpor.
Collapse
Affiliation(s)
- Benjamin W Iliff
- Department of Biology, Williams College, Williamstown, MA 01267, USA
| | | |
Collapse
|
67
|
Abstract
PURPOSE OF REVIEW During critical illness, alterations of intestinal blood supply and inflammatory activation can result in severe intestinal hypoxia (limited oxygen availability). Conditions of hypoxia lead to the activation of a transcriptional program that is under the control of the transcription factor hypoxia-inducible factor (HIF). In many instances, HIF-dependent alterations of gene expression represent endogenous adaptive responses that dampen pathologic inflammation and could be targeted to treat intestinal injury. RECENT FINDINGS Post-translational stabilization of the HIF transcription factor and corresponding changes in gene expression are central to the resolution of intestinal injury. Examples for such responses that we discuss in this review include hypoxia-elicited increases in extracellular adenosine production and signaling, particularly through the A2B adenosine receptor, and intestinal protection provided by hypoxia-inducible netrin-1. SUMMARY The present review focuses on HIF-elicited anti-inflammatory pathways that result in intestinal protection during critical illness. Many of these pathways represent novel therapeutic targets for attenuating multiorgan failure and critical illness. Whereas these therapeutic approaches are currently being investigated in cell culture models or in genetic mouse models, we are optimistic that at least some of these novel targets can be translated from bench to bedside in the near future.
Collapse
Affiliation(s)
- Almut Grenz
- Mucosal Inflammation Program, Department of Anesthesiology, University of Colorado, Aurora, Colorado 80045, USA.
| | | | | |
Collapse
|
68
|
Praman S, Mulvany MJ, Williams DE, Andersen RJ, Jansakul C. Hypotensive and cardio-chronotropic constituents of Tinospora crispa and mechanisms of action on the cardiovascular system in anesthetized rats. JOURNAL OF ETHNOPHARMACOLOGY 2012; 140:166-178. [PMID: 22265931 DOI: 10.1016/j.jep.2012.01.006] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/04/2011] [Revised: 12/11/2011] [Accepted: 01/07/2012] [Indexed: 05/31/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Tinospora crispa has been used in folkloric medicine for the control of blood pressure. We previously found that an extract of Tinospora crispa stems decreased the mean arterial blood pressure (MAP) with a transient decrease, followed by an increase in the heart rate (HR) in rats. AIM OF THE STUDY To identify the active components of the Tinospora crispa extract and investigate the mechanisms of action on blood pressure and heart rate in anesthetized rats. MATERIALS AND METHODS The active components of Tinospora crispa extract were separated by column chromatography and a preparative HPLC. The effects and mechanisms of the active compounds on blood pressure and heart rate were studied in anesthetized, normal and reserpinized rats in vivo. RESULTS 5 active compounds: adenosine, uridine, salsolinol, higenamine and tyramine were isolated. Adenosine decreased MAP and HR and this effect was inhibited by DMPX (A(2A) adenosine receptor antagonist). Uridine increased MAP and decreased HR and this was inhibited by suramin but not by DMPX. Salsolinol decreased the MAP and HR and this was inhibited by phentolamine but not by ICI-118,551 (β(2)-adrenoceptor antagonist) or atropine. In reserpinized rats, salsolinol had a hypertensive effect that was inhibited by prazosin and phentolamine, but not by atenolol, and caused an increase in HR that was inhibited by atenolol, but not by prazosin or phentolamine. Higenamine decreased the MAP with an increase in HR. The hypotensive effect was inhibited by ICI-118,551 or atenolol, whereas the increase in HR was not inhibited by ICI-118,551. Atenolol inhibited the increase in HR at a small dosage of higenamine but potentiated it at a higher dosage. In reserpinized rats, a small dosage of higenamine tended to potentiate the effect but at a higher dosage it caused inhibition. ICI-118,551 significantly inhibited this hypotensive effect. Tyramine caused an increase in MAP and HR and these effects almost disappeared in reserpinized rats. CONCLUSIONS The results demonstrate that these 5 compounds from Tinospora crispa acted in concert on the cardiovascular system of anesthetized rats. Salsolinol, tyramine and higenamine acted via the adrenoreceptors, whereas uridine and adenosine acted via the purinergic adenosine A(2) and P(2) receptors to decrease blood pressure with a transient decrease of HR followed by an increase.
Collapse
Affiliation(s)
- Siwaporn Praman
- Department of Physiology, Faculty of Science, Prince of Songkla University, Hat-Yai 90112, Thailand
| | | | | | | | | |
Collapse
|
69
|
Buscariollo DL, Breuer GA, Wendler CC, Rivkees SA. Caffeine acts via A1 adenosine receptors to disrupt embryonic cardiac function. PLoS One 2011; 6:e28296. [PMID: 22164264 PMCID: PMC3229565 DOI: 10.1371/journal.pone.0028296] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2011] [Accepted: 11/04/2011] [Indexed: 12/29/2022] Open
Abstract
Background Evidence suggests that adenosine acts via cardiac A1 adenosine receptors (A1ARs) to protect embryos against hypoxia. During embryogenesis, A1ARs are the dominant regulator of heart rate, and A1AR activation reduces heart rate. Adenosine action is inhibited by caffeine, which is widely consumed during pregnancy. In this study, we tested the hypothesis that caffeine influences developing embryos by altering cardiac function. Methodology/Principal Findings Effects of caffeine and adenosine receptor-selective antagonists on heart rate were studied in vitro using whole murine embryos at E9.5 and isolated hearts at E12.5. Embryos were examined in room air (21% O2) or hypoxic (2% O2) conditions. Hypoxia decreased heart rates of E9.5 embryos by 15.8% and in E12.5 isolated hearts by 27.1%. In room air, caffeine (200 µM) had no effect on E9.5 heart rates; however, caffeine increased heart rates at E12.5 by 37.7%. Caffeine abolished hypoxia-mediated bradycardia at E9.5 and blunted hypoxia-mediated bradycardia at E12.5. Real-time PCR analysis of RNA from isolated E9.5 and E12.5 hearts showed that A1AR and A2aAR genes were expressed at both ages. Treatment with adenosine receptor-selective antagonists revealed that SCH-58261 (A2aAR-specific antagonist) had no affects on heart function, whereas DPCPX (A1AR-specific antagonist) had effects similar to caffeine treatment at E9.5 and E12.5. At E12.5, embryonic hearts lacking A1AR expression (A1AR−/−) had elevated heart rates compared to A1AR+/− littermates, A1AR−/− heart rates failed to decrease to levels comparable to those of controls. Caffeine did not significantly affect heart rates of A1AR−/− embryos. Conclusions/Significance These data show that caffeine alters embryonic cardiac function and disrupts the normal cardiac response to hypoxia through blockade of A1AR action. Our results raise concern for caffeine exposure during embryogenesis, particularly in pregnancies with increased risk of embryonic hypoxia.
Collapse
Affiliation(s)
- Daniela L. Buscariollo
- Section of Developmental Endocrinology and Biology, Yale Child Health Research Center, Department of Pediatrics, Yale University School of Medicine, New Haven, Connecticut, United States of America
| | - Gregory A. Breuer
- Section of Developmental Endocrinology and Biology, Yale Child Health Research Center, Department of Pediatrics, Yale University School of Medicine, New Haven, Connecticut, United States of America
| | - Christopher C. Wendler
- Section of Developmental Endocrinology and Biology, Yale Child Health Research Center, Department of Pediatrics, Yale University School of Medicine, New Haven, Connecticut, United States of America
| | - Scott A. Rivkees
- Section of Developmental Endocrinology and Biology, Yale Child Health Research Center, Department of Pediatrics, Yale University School of Medicine, New Haven, Connecticut, United States of America
- * E-mail:
| |
Collapse
|
70
|
Colgan SP, Eltzschig HK. Adenosine and hypoxia-inducible factor signaling in intestinal injury and recovery. Annu Rev Physiol 2011; 74:153-75. [PMID: 21942704 DOI: 10.1146/annurev-physiol-020911-153230] [Citation(s) in RCA: 101] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
The gastrointestinal mucosa has proven to be an interesting tissue in which to investigate disease-related metabolism. In this review, we outline some of the evidence that implicates hypoxia-mediated adenosine signaling as an important signature within both healthy and diseased mucosa. Studies derived from cultured cell systems, animal models, and human patients have revealed that hypoxia is a significant component of the inflammatory microenvironment. These studies have revealed a prominent role for hypoxia-induced factor (HIF) and hypoxia signaling at several steps along the adenine nucleotide metabolism and adenosine receptor signaling pathways. Likewise, studies to date in animal models of intestinal inflammation have demonstrated an almost uniformly beneficial influence of HIF stabilization on disease outcomes. Ongoing studies to define potential similarities with and differences between innate and adaptive immune responses will continue to teach us important lessons about the complexity of the gastrointestinal tract. Such information has provided new insights into disease pathogenesis and, importantly, will provide insights into new therapeutic targets.
Collapse
Affiliation(s)
- Sean P Colgan
- Departments of Medicine and Anesthesiology and the Mucosal Inflammation Program, University of Colorado School of Medicine, Aurora, Colorado 80045, USA.
| | | |
Collapse
|
71
|
Gessi S, Merighi S, Fazzi D, Stefanelli A, Varani K, Borea PA. Adenosine receptor targeting in health and disease. Expert Opin Investig Drugs 2011; 20:1591-609. [PMID: 22017198 DOI: 10.1517/13543784.2011.627853] [Citation(s) in RCA: 63] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
INTRODUCTION The adenosine receptors A(1), A(2A), A(2B) and A(3) are important and ubiquitous mediators of cellular signaling that play vital roles in protecting tissues and organs from damage. In particular, adenosine triggers tissue protection and repair by different receptor-mediated mechanisms, including increasing the oxygen supply:demand ratio, pre-conditioning, anti-inflammatory effects and the stimulation of angiogenesis. AREAS COVERED The state of the art of the role of adenosine receptors which have been proposed as targets for drug design and discovery, in health and disease, and an overview of the ligands for these receptors in clinical development. EXPERT OPINION Selective ligands of A(1), A(2A), A(2B) and A(3) adenosine receptors are likely to find applications in the treatment of pain, ischemic conditions, glaucoma, asthma, arthritis, cancer and other disorders in which inflammation is a feature. The aim of this review is to provide an overview of the present knowledge regarding the role of these adenosine receptors in health and disease.
Collapse
Affiliation(s)
- Stefania Gessi
- University of Ferrara, Department of Clinical and Experimental Medicine, Pharmacology Section, 44100 Ferrara, Italy
| | | | | | | | | | | |
Collapse
|
72
|
Grenz A, Homann D, Eltzschig HK. Extracellular adenosine: a safety signal that dampens hypoxia-induced inflammation during ischemia. Antioxid Redox Signal 2011; 15:2221-34. [PMID: 21126189 PMCID: PMC3166177 DOI: 10.1089/ars.2010.3665] [Citation(s) in RCA: 74] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/16/2023]
Abstract
Traditionally, the single most unique feature of the immune system has been attributed to its capability to discriminate between self (e.g., host proteins) and nonself (e.g., pathogens). More recently, an emerging immunologic concept involves the notion that the immune system responds via a complex system for sensing signals of danger, such as pathogens or host-derived signals of cellular distress (e.g., ischemia), while remaining unresponsive to nondangerous motifs. Experimental studies have provided strong evidence that the production and signaling effects of extracellular adenosine are dramatically enhanced during conditions of limited oxygen availability as occurs during ischemia. As such, adenosine would fit the bill of signaling molecules that are enhanced during situations of cellular distress. In contrast to a danger signal, we propose here that extracellular adenosine operates as a countermeasure, in fact as a safety signal, to both restrain potentially harmful immune responses and to maintain and promote general tissue integrity during conditions of limited oxygen availability.
Collapse
Affiliation(s)
- Almut Grenz
- Mucosal Inflammation Program, Department of Anesthesiology, University of Colorado-Denver, Aurora, CO 80045, USA
| | | | | |
Collapse
|
73
|
Andersen H, Jaff MG, Høgh D, Vanhoutte P, Hansen PB. Adenosine elicits an eNOS-independent reduction in arterial blood pressure in conscious mice that involves adenosine A2A receptors. Acta Physiol (Oxf) 2011; 203:197-207. [PMID: 21062422 DOI: 10.1111/j.1748-1716.2010.02218.x] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/01/2022]
Abstract
AIMS Adenosine plays an important role in the regulation of heart rate (HR) and vascular reactivity. However, the mechanisms underlying the acute effect of adenosine on arterial blood pressure in conscious mice are unclear. Therefore, this study investigated the effect of the nucleoside on mean arterial blood pressure (MAP) and HR in conscious mice. METHODS Chronic indwelling catheters were placed in C57Bl/6J (WT) and endothelial nitric oxide synthase knockout (eNOS(-/-)) mice for continuous measurements of MAP and HR. Using PCR and myograph analysis, involvement of adenosine receptors was investigated in human and mouse renal blood vessels. RESULTS Bolus infusion of 0.5 mg kg(-1) adenosine elicited significant transient decreases in MAP (99.3 ± 2.3 to 70.4 ± 4.5 mmHg) and HR (603.2 ± 18.3 to 364.3 ± 49.2 min(-1)), which were inhibited by the A(2A) receptor antagonist ZM 241385. Activation of adenosine A(2A) receptors with CGS 21680 (0.02 mg kg(-1)) caused a significant reduction in MAP from 99.6 ± 1.2 to 73.1 ± 3.6 mmHg accompanied by tachycardia (610.5 ± 9.3 to 677.5 ± 9.5 min(-1)). The reduction in MAP observed after adenosine or CGS 21680 administrations was not significantly different in WT and eNOS(-/-) mice. In isolated human and mouse intrarenal arteries, adenosine caused a relaxation dependent on A(2A) adenosine receptor activation. A(2A) receptors were present in both human and mouse arteries whereas A(1) and A(2B) receptors were only present in mouse arteries. CONCLUSION In conclusion, acute adenosine administration and selective stimulation of adenosine A(2A) receptors results in an immediate, transient eNOS-independent reduction in MAP. A(2A) receptor activation causes relaxation of human and mouse arteries.
Collapse
Affiliation(s)
- H Andersen
- Department of Cardiovascular and Renal Research, Institute of Molecular Medicine, University of Southern Denmark, Odense, Denmark
| | | | | | | | | |
Collapse
|
74
|
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
|
75
|
Gessi S, Merighi S, Varani K, Borea PA. Adenosine receptors in health and disease. ADVANCES IN PHARMACOLOGY 2011; 61:41-75. [PMID: 21586355 DOI: 10.1016/b978-0-12-385526-8.00002-3] [Citation(s) in RCA: 55] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
The adenosine receptors A(1), A(2A), A(2B), and A(3) are important and ubiquitous mediators of cellular signaling, which play vital roles in protecting tissues and organs from damage. In particular, adenosine triggers tissue protection and repair by different receptor-mediated mechanisms, including an increase of oxygen supply/demand ratio, preconditioning, anti-inflammatory effects, and stimulation of angiogenesis. Considerable advances have been recently achieved in the pharmacological and molecular characterization of adenosine receptors, which have been proposed as targets for drug design and discovery. At the present time, it can be speculated that adenosine A(1), A(2A), A(2B), and A(3) receptor-selective ligands may show utility in the treatment of pain, ischemic conditions, glaucoma, asthma, arthritis, cancer, and other disorders in which inflammation is a feature. This chapter documents the present state of knowledge of adenosine receptors' role in health and disease.
Collapse
Affiliation(s)
- Stefania Gessi
- Department of Clinical and Experimental Medicine, Pharmacology Section, University of Ferrara, Italy
| | | | | | | |
Collapse
|
76
|
Koeppen M, Eckle T, Eltzschig HK. Pressure controlled ventilation to induce acute lung injury in mice. J Vis Exp 2011:2525. [PMID: 21587159 PMCID: PMC3197099 DOI: 10.3791/2525] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022] Open
Abstract
Murine models are extensively used to investigate acute injuries of different organs systems (1-34). Acute lung injury (ALI), which occurs with prolonged mechanical ventilation, contributes to morbidity and mortality of critical illness, and studies on novel genetic or pharmacological targets are areas of intense investigation (1-3, 5, 8, 26, 30, 33-36). ALI is defined by the acute onset of the disease, which leads to non-cardiac pulmonary edema and subsequent impairment of pulmonary gas exchange (36). We have developed a murine model of ALI by using a pressure-controlled ventilation to induce ventilator-induced lung injury (2). For this purpose, C57BL/6 mice are anesthetized and a tracheotomy is performed followed by induction of ALI via mechanical ventilation. Mice are ventilated in a pressure-controlled setting with an inspiratory peak pressure of 45 mbar over 1 - 3 hours. As outcome parameters, pulmonary edema (wet-to-dry ratio), bronchoalveolar fluid albumin content, bronchoalveolar fluid and pulmonary tissue myeloperoxidase content and pulmonary gas exchange are assessed (2). Using this technique we could show that it sufficiently induces acute lung inflammation and can distinguish between different treatment groups or genotypes (1-3, 5). Therefore this technique may be helpful for researchers who pursue molecular mechanisms involved in ALI using a genetic approach in mice with gene-targeted deletion.
Collapse
|
77
|
Eckle T, Koeppen M, Eltzschig H. Use of a hanging weight system for coronary artery occlusion in mice. J Vis Exp 2011:2526. [PMID: 21540816 PMCID: PMC3169250 DOI: 10.3791/2526] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
Murine studies of acute injury are an area of intense investigation, as knockout mice for different genes are becoming increasingly available. Cardioprotection by ischemic preconditioning (IP) remains an area of intense investigation. To further elucidate its molecular basis, the use of knockout mouse studies is particularly important. Despite the fact that previous studies have already successfully performed cardiac ischemia and reperfusion in mice, this model is technically very challenging. Particularly, visual identification of the coronary artery, placement of the suture around the vessel and coronary occlusion by tying off the vessel with a supported knot is technically difficult. In addition, re-opening the knot for intermittent reperfusion of the coronary artery during IP without causing surgical trauma adds additional challenge. Moreover, if the knot is not tied down strong enough, inadvertent reperfusion due to imperfect occlusion of the coronary may affect the results. In fact, this can easily occur due to the movement of the beating heart. Based on potential problems associated with using a knotted coronary occlusion system, we adopted a previously published model of chronic cardiomyopathy based on a hanging weight system for intermittent coronary artery occlusion during IP. In fact, coronary artery occlusion can thus be achieved without having to occlude the coronary by a knot. Moreover, reperfusion of the vessel can be easily achieved by supporting the hanging weights which are in a remote localization from cardiac tissues. We tested this system systematically, including variation of ischemia and reperfusion times, preconditioning regiments, body temperature and genetic backgrounds. In addition to infarct staining, we tested cardiac troponin I (cTnI) as a marker of myocardial infarction in this model. In fact, plasma levels of cTnI correlated with infarct sizes (R2=0.8). Finally, we could show in several studies that this technique yields highly reproducible infarct sizes during murine IP and myocardial infarction. Therefore, this technique may be helpful for researchers who pursue molecular mechanisms involved in cardioprotection by IP using a genetic approach in mice with targeted gene deletion. Further studies on cardiac IP using transgenic mice may consider this technique.
Collapse
Affiliation(s)
- Tobias Eckle
- Department of Anesthesiology, University of Colorado Denver, USA.
| | | | | |
Collapse
|
78
|
Hart ML, Grenz A, Gorzolla IC, Schittenhelm J, Dalton JH, Eltzschig HK. Hypoxia-inducible factor-1α-dependent protection from intestinal ischemia/reperfusion injury involves ecto-5'-nucleotidase (CD73) and the A2B adenosine receptor. THE JOURNAL OF IMMUNOLOGY 2011; 186:4367-74. [PMID: 21357264 DOI: 10.4049/jimmunol.0903617] [Citation(s) in RCA: 114] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Intestinal ischemia/reperfusion injury (IR) is characterized by intermittent loss of perfusion to the gut, resulting in dramatic increases in morbidity and mortality. Based on previous studies indicating an anti-inflammatory role for hypoxia-inducible factor (HIF)-1-elicited enhancement of extracellular adenosine production via ecto-5'-nucleotidase (CD73) and signaling through the A2B adenosine receptor (A2BAR), we targeted HIF-1 during IR using pharmacological or genetic approaches. Initial studies with pharmacological HIF activation indicated attenuation of intestinal injury with dimethyloxallyl glycine (DMOG) treatment during murine IR. Although DMOG treatment was associated with induction of CD73 transcript and protein, DMOG protection was abolished in cd73(-/-) mice. Similarly, DMOG treatment enhanced A2BAR transcript and protein levels, whereas DMOG protection was abolished in A2BAR(-/-) mice. Finally, studies of mice with conditional HIF-1α deletion in intestinal epithelia or pharmacological inhibition of HIF-1 with 17-(dimethylaminoethylamino)-17-demethoxygeldanamycin revealed enhanced tissue injury during IR. These studies indicated a tissue-protective role of HIF-dependent enhancement of intestinal adenosine generation and signaling during intestinal IR.
Collapse
Affiliation(s)
- Melanie L Hart
- Department of Anesthesiology and Critical Care Medicine, Tübingen University Hospital, D-72074 Tübingen, Germany.
| | | | | | | | | | | |
Collapse
|
79
|
Selective induction of endothelial P2Y6 nucleotide receptor promotes vascular inflammation. Blood 2010; 117:2548-55. [PMID: 21173118 DOI: 10.1182/blood-2010-10-313957] [Citation(s) in RCA: 95] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
During a systemic inflammatory response endothelial-expressed surface molecules have been strongly implicated in orchestrating immune responses. Previous studies have shown enhanced extracellular nucleotide release during acute inflammatory conditions. Therefore, we hypothesized that endothelial nucleotide receptors could play a role in vascular inflammation. To address this hypothesis, we performed screening experiments and exposed human microvascular endothelia to inflammatory stimuli, followed by measurements of P2Y or P2X transcriptional responses. These studies showed a selective induction of the P2Y(6) receptor (> 4-fold at 24 hours). Moreover, studies that used real-time reverse transcription-polymerase chain reaction, Western blot analysis, or immunofluorescence confirmed time- and dose-dependent induction of P2Y(6) with tumor necrosis factor α or Lipopolysaccharide (LPS) stimulation in vitro and in vivo. Studies that used MRS 2578 as P2Y(6) receptor antagonist showed attenuated nuclear factor κB reporter activity and proinflammatory gene expression in human microvascular endothelial cells in vitro. Moreover, pharmacologic or genetic in vivo studies showed attenuated inflammatory responses in P2Y(6)(-/-) mice or after P2Y(6) antagonist treatment during LPS-induced vascular inflammation. These studies show an important contribution of P2Y(6) signaling in enhancing vascular inflammation during systemic LPS challenge and implicate the P2Y(6) receptor as a therapeutic target during systemic inflammatory responses.
Collapse
|
80
|
Adenosine A1 receptors contribute to mitochondria vulnerability to pro-oxidant stressors. Mitochondrion 2010; 10:369-79. [DOI: 10.1016/j.mito.2010.03.004] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2009] [Revised: 02/01/2010] [Accepted: 03/04/2010] [Indexed: 11/17/2022]
|
81
|
Schingnitz U, Hartmann K, Macmanus CF, Eckle T, Zug S, Colgan SP, Eltzschig HK. Signaling through the A2B adenosine receptor dampens endotoxin-induced acute lung injury. THE JOURNAL OF IMMUNOLOGY 2010; 184:5271-9. [PMID: 20348420 DOI: 10.4049/jimmunol.0903035] [Citation(s) in RCA: 146] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Abstract] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Sepsis and septic acute lung injury are among the leading causes for morbidity and mortality of critical illness. Extracellular adenosine is a signaling molecule implicated in the cellular adaptation to hypoxia, ischemia, or inflammation. Therefore, we pursued the role of the A2B adenosine receptor (AR) as potential therapeutic target in endotoxin-induced acute lung injury. We gained initial insight from in vitro studies of cultured endothelia or epithelia exposed to inflammatory mediators showing time-dependent induction of the A2BAR (up to 12.9 + or - 3.4-fold, p < 0.05). Similarly, murine studies of endotoxin-induced lung injury identified an almost 4.6-fold induction of A2BAR transcript and corresponding protein induction with LPS exposure. Studies utilizing A2BAR promoter constructs and RNA protection assays indicated that A2BAR induction involved mRNA stability. Functional studies of LPS-induced lung injury revealed that pharmacological inhibition or genetic deletion of the A2BAR was associated with dramatic increases in lung inflammation and histologic tissue injury. Studies of A2BAR bone marrow chimeric mice suggested pulmonary A2BAR signaling in lung protection. Finally, studies with a specific A2BAR agonist (BAY 60-6583) demonstrated attenuation of lung inflammation and pulmonary edema in wild-type but not in gene-targeted mice for the A2BAR. These studies suggest the A2BAR as potential therapeutic target in the treatment of endotoxin-induced forms of acute lung injury.
Collapse
Affiliation(s)
- Ulrich Schingnitz
- Department of Anesthesiology and Intensive Care Medicine, Tübingen University Hospital, Tübingen, Germany
| | | | | | | | | | | | | |
Collapse
|
82
|
Anti-CD73 antibody therapy inhibits breast tumor growth and metastasis. Proc Natl Acad Sci U S A 2010; 107:1547-52. [PMID: 20080644 DOI: 10.1073/pnas.0908801107] [Citation(s) in RCA: 463] [Impact Index Per Article: 33.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022] Open
Abstract
Extracellular adenosine is a potent immunosuppressor that accumulates during tumor growth. We performed proof-of-concept studies investigating the therapeutic potential and mechanism of action of monoclonal antibody (mAb)-based therapy against CD73, an ecto-enzyme overexpressed on breast-cancer cells that catalyzes the dephosphorylation of adenosine monophosphates into adenosine. We showed that anti-CD73 mAb therapy significantly delayed primary 4T1.2 and E0771 tumor growth in immune-competent mice and significantly inhibited the development of spontaneous 4T1.2 lung metastases. Notably, anti-CD73 mAb therapy was essentially dependent on the induction of adaptive anti-tumor immune responses. Knockdown of CD73 in 4T1.2 tumor cells confirmed the tumor-promoting effects of CD73. In addition to its immunosuppressive effect, CD73 enhanced tumor-cell chemotaxis, suggesting a role for CD73-derived adenosine in tumor metastasis. Accordingly, administration of adenosine-5'-N-ethylcarboxamide to tumor-bearing mice significantly enhanced spontaneous 4T1.2 lung metastasis. Using selective adenosine-receptor antagonists, we showed that activation of A2B adenosine receptors promoted 4T1.2 tumor-cell chemotaxis in vitro and metastasis in vivo. In conclusion, our study identified tumor-derived CD73 as a mechanism of tumor immune escape and tumor metastasis, and it also established the proof of concept that targeted therapy against CD73 can trigger adaptive anti-tumor immunity and inhibit metastasis of breast cancer.
Collapse
|