1
|
Wölkart G, Gissing S, Stessel H, Fassett EK, Klösch B, Greene RW, Mayer B, Fassett JT. An adenosinergic positive feedback loop extends pharmacological cardioprotection duration. Br J Pharmacol 2024; 181:4920-4936. [PMID: 39256947 DOI: 10.1111/bph.17331] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2024] [Revised: 06/25/2024] [Accepted: 07/30/2024] [Indexed: 09/12/2024] Open
Abstract
BACKGROUND AND PURPOSE Adenosine receptor activation induces delayed, sustained cardioprotection against ischaemia-reperfusion (IR) injury (24-72 h), but the mechanisms underlying extended cardioprotection duration remain unresolved. We hypothesized that a positive feedback loop involving adenosine receptor-induced proteasomal degradation of adenosine kinase (ADK) and decreased myocardial adenosine metabolism extends the duration of cardioprotection. EXPERIMENTAL APPROACH Mice were administered an ADK inhibitor, ABT-702, to induce endogenous adenosine signalling. Cardiac ADK protein and mRNA levels were analysed 24-120 h later. Theophylline or bortezomib was administered 24 h after ABT-702 to examine the late roles of adenosine receptors or proteasomal activity, respectively, in ADK expression and cardioprotection at 72 h. Coronary flow and IR tolerance were analysed by Langendorff technique. The potential for continuous adenosinergic cardioprotection was examined using heterozygous, cardiac-specific ADK KO (cADK+/-) mice. Cardiac ADK expression was also examined after A1 or A3 receptor agonist, phenylephrine, lipopolysaccharide or sildenafil administration. KEY RESULTS ABT-702 treatment decreased ADK protein content and provided cardioprotection from 24 to 72 h. ADK mRNA upregulation restored ADK protein after 96-120 h. Adenosine receptor or proteasome inhibition at 24 h reversed ABT-702-induced ADK protein deficit and cardioprotection at 72 h. cADK+/- hearts exhibited continuous cardioprotection. Diverse preconditioning agents also diminished cardiac ADK protein expression. CONCLUSION AND IMPLICATIONS A positive feedback loop driven by adenosine receptor-induced ADK degradation and renewed adenosine signalling extends the duration of cardioprotection by ABT-702 and possibly other preconditioning agents. The therapeutic potential of continuous adenosinergic cardioprotection is demonstrated in cADK+/- hearts.
Collapse
Affiliation(s)
- Gerald Wölkart
- Department of Pharmacology and Toxicology, University of Graz, Graz, Austria
| | - Simon Gissing
- Department of Pharmacology and Toxicology, University of Graz, Graz, Austria
| | - Heike Stessel
- Department of Pharmacology and Toxicology, University of Graz, Graz, Austria
| | - Erin K Fassett
- Department of Pharmacology and Toxicology, University of Graz, Graz, Austria
| | - Burkhard Klösch
- Department of Pharmacology and Toxicology, University of Graz, Graz, Austria
- Ludwig Boltzmann Institute for Arthritis and Rehabilitation, University of Graz, Graz, Austria
| | - Robert W Greene
- Department of Psychiatry and Neuroscience, Peter O'Donnell Brain Institute, UTSW Medical Center, Dallas, Texas, USA
| | - Bernd Mayer
- Department of Pharmacology and Toxicology, University of Graz, Graz, Austria
| | - John T Fassett
- Department of Pharmacology and Toxicology, University of Graz, Graz, Austria
| |
Collapse
|
2
|
Hoebart C, Kiss A, Podesser BK, Tahir A, Fischer MJM, Heber S. Sensory Neurons Release Cardioprotective Factors in an In Vitro Ischemia Model. Biomedicines 2024; 12:1856. [PMID: 39200320 PMCID: PMC11351881 DOI: 10.3390/biomedicines12081856] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2024] [Revised: 08/07/2024] [Accepted: 08/13/2024] [Indexed: 09/02/2024] Open
Abstract
Sensory neurons densely innervate the myocardium. The role of their sensing and response to acute and prolonged ischemia is largely unclear. In a cellular model of ischemia-reperfusion injury, the presence of sensory neurons increases cardiomyocyte survival. Here, after the exclusion of classical neurotransmitter release, and measurement of cytokine release, we modified the experiment from a direct co-culture of primary murine cardiomyocytes and sensory neurons to a transfer of the supernatant. Sensory neurons were exposed to ischemia and the resulting conditioned supernatant was transferred onto cardiomyocytes. This approach largely increased the tolerance of cardiomyocytes to ischemia and reperfusion. Towards the identification of the mechanism, it was demonstrated that after ten-fold dilution, the conditioned solution lost its protective effect. The effect remained after removal of extracellular vesicles by ultracentrifugation, and was not affected by exposure to protease activity, and fractionation pointed towards a hydrophilic agent. Solutions conditioned by HEK293t cells or 3T3 fibroblasts also increase cardiomyocyte survival, but to a lower degree. A metabolomic search identified 64 at least two-fold changed metabolites and lipids. Many of these could be identified and are involved in essential cellular functions. In the presented model for ischemia-reperfusion, sensory neurons secrete one or more cardioprotective substances that can improve cardiomyocyte survival.
Collapse
Affiliation(s)
- Clara Hoebart
- Institute of Physiology, Center for Physiology and Pharmacology, Medical University of Vienna, 1090 Vienna, Austria; (C.H.); (S.H.)
| | - Attila Kiss
- Center for Biomedical Research and Translational Surgery, Medical University of Vienna, 1090 Vienna, Austria; (A.K.); (B.K.P.)
| | - Bruno K. Podesser
- Center for Biomedical Research and Translational Surgery, Medical University of Vienna, 1090 Vienna, Austria; (A.K.); (B.K.P.)
| | - Ammar Tahir
- Division of Pharmacognosy, University of Vienna, 1090 Vienna, Austria;
| | - Michael J. M. Fischer
- Institute of Physiology, Center for Physiology and Pharmacology, Medical University of Vienna, 1090 Vienna, Austria; (C.H.); (S.H.)
| | - Stefan Heber
- Institute of Physiology, Center for Physiology and Pharmacology, Medical University of Vienna, 1090 Vienna, Austria; (C.H.); (S.H.)
| |
Collapse
|
3
|
Gao ZG, Haddad M, Jacobson KA. A 2B adenosine receptor signaling and regulation. Purinergic Signal 2024:10.1007/s11302-024-10025-y. [PMID: 38833181 DOI: 10.1007/s11302-024-10025-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2023] [Accepted: 05/20/2024] [Indexed: 06/06/2024] Open
Abstract
The A2B adenosine receptor (A2BR) is one of the four adenosine-activated G protein-coupled receptors. In addition to adenosine, protein kinase C (PKC) was recently found to activate the A2BR. The A2BR is coupled to both Gs and Gi, as well as Gq proteins in some cell types. Many primary cells and cell lines, such as bladder and breast cancer, bronchial smooth muscle, skeletal muscle, and fat cells, express the A2BR endogenously at high levels, suggesting its potentially important role in asthma, cancer, diabetes, and other conditions. The A2BR has been characterized as both pro- and anti-inflammatory, inducing cell type-dependent secretion of IL-6, IL-8, and IL-10. Theophylline and enprofylline have long been used for asthma treatment, although it is still not entirely clear if their A2BR antagonism contributes to their therapeutic effects or side effects. The A2BR is required in ischemic cardiac preconditioning by adenosine. Both A2BR and protein kinase C (PKC) contribute to cardioprotection, and both modes of A2BR signaling can be blocked by A2BR antagonists. Inhibitors of PKC and A2BR are in clinical cancer trials. Sulforaphane and other isothiocyanates from cruciferous vegetables such as broccoli and cauliflower have been reported to inhibit A2BR signaling via reaction with an intracellular A2BR cysteine residue (C210). A full, A2BR-selective agonist, critical to elucidate many controversial roles of the A2BR, is still not available, although agonist-bound A2BR structures have recently been reported.
Collapse
Affiliation(s)
- Zhan-Guo Gao
- Molecular Recognition Section, Laboratory of Bioorganic Chemistry, NIDDK, National Institutes of Health, 9000 Rockville Pike, Bethesda, MD, 20892, USA.
| | - Mansour Haddad
- Faculty of Pharmacy, Yarmouk University, Irbid, 21163, Jordan
| | - Kenneth A Jacobson
- Molecular Recognition Section, Laboratory of Bioorganic Chemistry, NIDDK, National Institutes of Health, 9000 Rockville Pike, Bethesda, MD, 20892, USA.
| |
Collapse
|
4
|
Galli M, Niccoli G, De Maria G, Brugaletta S, Montone RA, Vergallo R, Benenati S, Magnani G, D'Amario D, Porto I, Burzotta F, Abbate A, Angiolillo DJ, Crea F. Coronary microvascular obstruction and dysfunction in patients with acute myocardial infarction. Nat Rev Cardiol 2024; 21:283-298. [PMID: 38001231 DOI: 10.1038/s41569-023-00953-4] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 10/23/2023] [Indexed: 11/26/2023]
Abstract
Despite prompt epicardial recanalization in patients presenting with ST-segment elevation myocardial infarction (STEMI), coronary microvascular obstruction and dysfunction (CMVO) is still fairly common and is associated with poor prognosis. Various pharmacological and mechanical strategies to treat CMVO have been proposed, but the positive results reported in preclinical and small proof-of-concept studies have not translated into benefits in large clinical trials conducted in the modern treatment setting of patients with STEMI. Therefore, the optimal management of these patients remains a topic of debate. In this Review, we appraise the pathophysiological mechanisms of CMVO, explore the evidence and provide future perspectives on strategies to be implemented to reduce the incidence of CMVO and improve prognosis in patients with STEMI.
Collapse
Affiliation(s)
- Mattia Galli
- Department of Cardiology, Maria Cecilia Hospital, GVM Care & Research, Cotignola, Italy
| | | | - Gianluigi De Maria
- Oxford Heart Centre, John Radcliffe Hospital, Oxford University Hospitals NHS Foundation Trust, Oxford, UK
| | - Salvatore Brugaletta
- Institut Clinic Cardiovascular, Hospital Clínic de Barcelona, University of Barcelona, Barcelona, Spain
| | - Rocco A Montone
- Department of Cardiovascular Sciences, Fondazione Policlinico Universitario A. Gemelli IRCCS, Rome, Italy
| | - Rocco Vergallo
- Department of Internal Medicine, University of Genoa, Genoa, Italy
- Cardiovascular Disease Unit, IRCCS Ospedale Policlinico San Martino, IRCCS Italian Cardiology Network, Genova, Italy
| | - Stefano Benenati
- Oxford Heart Centre, John Radcliffe Hospital, Oxford University Hospitals NHS Foundation Trust, Oxford, UK
- Cardiovascular Disease Unit, IRCCS Ospedale Policlinico San Martino, IRCCS Italian Cardiology Network, Genova, Italy
| | - Giulia Magnani
- Department of Cardiology, University of Parma, Parma, Italy
| | - Domenico D'Amario
- Department of Translational Medicine, University of Eastern Piedmont, Novara, Italy
- Division of Cardiology, Azienda Ospedaliero Universitaria 'Maggiore Della Carita', Novara, Italy
| | - Italo Porto
- Department of Internal Medicine, University of Genoa, Genoa, Italy
- Cardiovascular Disease Unit, IRCCS Ospedale Policlinico San Martino, IRCCS Italian Cardiology Network, Genova, Italy
| | - Francesco Burzotta
- Department of Cardiovascular Sciences, Fondazione Policlinico Universitario A. Gemelli IRCCS, Rome, Italy
- Department of Cardiovascular Sciencies, Catholic University of the Sacred Heart, Rome, Italy
| | - Antonio Abbate
- Robert M. Berne Cardiovascular Research Center, Division of Cardiology - Heart and Vascular Center, University of Virginia, Charlottesville, VA, USA
| | - Dominick J Angiolillo
- Division of Cardiology, University of Florida College of Medicine - Jacksonville, Jacksonville, FL, USA.
| | - Filippo Crea
- Department of Cardiovascular Sciencies, Catholic University of the Sacred Heart, Rome, Italy
| |
Collapse
|
5
|
Etzion O, Bareket-Samish A, Yardeni D, Fishman P. Namodenoson at the Crossroad of Metabolic Dysfunction-Associated Steatohepatitis and Hepatocellular Carcinoma. Biomedicines 2024; 12:848. [PMID: 38672201 PMCID: PMC11047856 DOI: 10.3390/biomedicines12040848] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2024] [Revised: 04/02/2024] [Accepted: 04/08/2024] [Indexed: 04/28/2024] Open
Abstract
Namodenoson (CF102) is a small, orally available, anti-inflammatory, and anti-cancer drug candidate currently in phase 2B trial for the treatment of metabolic dysfunction-associated steatohepatitis (MASH; formerly known as non-alcoholic steatohepatitis (NASH)) and in phase 3 pivotal clinical trial for the treatment of hepatocellular carcinoma (HCC). In both MASH and HCC, the mechanism-of-action of namodenoson involves targeting the A3 adenosine receptor (A3AR), resulting in deregulation of downstream signaling pathways and leading to inhibition of inflammatory cytokines (TNF-α, IL-1, IL-6, and IL-8) and stimulation of positive cytokines (G-CSF and adiponectin). Subsequently, inhibition of liver inflammation, steatosis, and fibrosis were documented in MASH experimental models, and inhibition of HCC growth was observed in vitro, in vivo, and in clinical studies. This review discusses the evidence related to the multifaceted mechanism of action of namodenoson, and how this mechanism is reflected in the available clinical data in MASH and HCC.
Collapse
Affiliation(s)
- Ohad Etzion
- Department of Gastroenterology and Liver Diseases, Sorkoa University Medical Center, Beer Sheva 84101, Israel; (O.E.); (D.Y.)
| | | | - David Yardeni
- Department of Gastroenterology and Liver Diseases, Sorkoa University Medical Center, Beer Sheva 84101, Israel; (O.E.); (D.Y.)
| | | |
Collapse
|
6
|
Cruz FF, Pereira TCB, da Costa KM, Bonan CD, Bogo MR, Morrone FB. Effect of adenosine treatment on ionizing radiation toxicity in zebrafish early life stages. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2024; 397:521-534. [PMID: 37480487 DOI: 10.1007/s00210-023-02617-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/11/2023] [Accepted: 07/07/2023] [Indexed: 07/24/2023]
Abstract
The danger of ionizing radiation exposure to human health is a concern. Since its wide use in medicine and industry, the development of radioprotectors has been very significant. Adenosine exerts anti-inflammatory actions and promotes tissue protection and repair, by activating the P1 receptors (A1, A2A, A2B, and A3). Zebrafish (Danio rerio) is an appropriate tool in the fields of toxicology and pharmacology, including the evaluation of radiobiological outcomes and in the search for radioprotector agents. This study aims to evaluate the effect of adenosine in the toxicity induced by radiation in zebrafish. Embryos were treated with 1, 10, or 100 µM adenosine, 30 min before the exposure to 15 Gy of gamma radiation. Adenosine potentiated the effects of radiation in heart rate, body length, and pericardial edema. We evaluated oxidative stress, tissue remodeling and inflammatory. It was seen that 100 µM adenosine reversed the inflammation induced by radiation, and that A2A2 and A2B receptors are involved in these anti-inflammatory effects. Our results indicate that P1R activation could be a promising pharmacological strategy for radioprotection.
Collapse
Affiliation(s)
- Fernanda Fernandes Cruz
- Programa de Pós-Graduação em Biologia Celular e Molecular, Escola de Ciências da Saúde e da Vida, Pontifícia Universidade Católica do Rio Grande do Sul, Porto Alegre, Rio Grande do Sul, Brazil
- Laboratório de Farmacologia Aplicada, Escola de Ciências da Saúde e da Vida, Pontifícia Universidade Católica do Rio Grande do Sul, Porto Alegre, Rio Grande do Sul, Brazil
| | - Talita Carneiro Brandão Pereira
- Programa de Pós-Graduação em Biologia Celular e Molecular, Escola de Ciências da Saúde e da Vida, Pontifícia Universidade Católica do Rio Grande do Sul, Porto Alegre, Rio Grande do Sul, Brazil
- Laboratório de Biologia Genômica e Molecular, Escola de Ciências da Saúde e da Vida, Pontifícia Universidade Católica do Rio Grande do Sul, Porto Alegre, Rio Grande do Sul, Brazil
| | - Kesiane Mayra da Costa
- Laboratório de Biologia Genômica e Molecular, Escola de Ciências da Saúde e da Vida, Pontifícia Universidade Católica do Rio Grande do Sul, Porto Alegre, Rio Grande do Sul, Brazil
- Programa de Pós-Graduação em Medicina e Ciências da Saúde, Escola de Medicina, Pontifícia Universidade Católica do Rio Grande do Sul, Porto Alegre, Rio Grande do Sul, Brazil
| | - Carla Denise Bonan
- Programa de Pós-Graduação em Biologia Celular e Molecular, Escola de Ciências da Saúde e da Vida, Pontifícia Universidade Católica do Rio Grande do Sul, Porto Alegre, Rio Grande do Sul, Brazil
- Programa de Pós-Graduação em Medicina e Ciências da Saúde, Escola de Medicina, Pontifícia Universidade Católica do Rio Grande do Sul, Porto Alegre, Rio Grande do Sul, Brazil
| | - Maurício Reis Bogo
- Programa de Pós-Graduação em Biologia Celular e Molecular, Escola de Ciências da Saúde e da Vida, Pontifícia Universidade Católica do Rio Grande do Sul, Porto Alegre, Rio Grande do Sul, Brazil
- Laboratório de Biologia Genômica e Molecular, Escola de Ciências da Saúde e da Vida, Pontifícia Universidade Católica do Rio Grande do Sul, Porto Alegre, Rio Grande do Sul, Brazil
- Programa de Pós-Graduação em Medicina e Ciências da Saúde, Escola de Medicina, Pontifícia Universidade Católica do Rio Grande do Sul, Porto Alegre, Rio Grande do Sul, Brazil
| | - Fernanda Bueno Morrone
- Programa de Pós-Graduação em Biologia Celular e Molecular, Escola de Ciências da Saúde e da Vida, Pontifícia Universidade Católica do Rio Grande do Sul, Porto Alegre, Rio Grande do Sul, Brazil.
- Laboratório de Farmacologia Aplicada, Escola de Ciências da Saúde e da Vida, Pontifícia Universidade Católica do Rio Grande do Sul, Porto Alegre, Rio Grande do Sul, Brazil.
- Programa de Pós-Graduação em Medicina e Ciências da Saúde, Escola de Medicina, Pontifícia Universidade Católica do Rio Grande do Sul, Porto Alegre, Rio Grande do Sul, Brazil.
| |
Collapse
|
7
|
Fishman P, Stemmer SM, Bareket-Samish A, Silverman MH, Kerns WD. Targeting the A3 adenosine receptor to treat hepatocellular carcinoma: anti-cancer and hepatoprotective effects. Purinergic Signal 2023; 19:513-522. [PMID: 36781824 PMCID: PMC10539266 DOI: 10.1007/s11302-023-09925-2] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2022] [Accepted: 01/31/2023] [Indexed: 02/15/2023] Open
Abstract
The A3 adenosine receptor (A3AR) is over-expressed in human hepatocellular carcinoma (HCC) cells. Namodenoson, an A3AR agonist, induces de-regulation of the Wnt and NF-kB signaling pathways resulting in apoptosis of HCC cells. In a phase I healthy volunteer study and in a phase I/II study in patients with advanced HCC, namodenoson was safe and well tolerated. Preliminary evidence of antitumor activity was observed in the phase I/II trial in a subset of patients with advanced disease, namely patients with Child-Pugh B (CPB) hepatic dysfunction, whose median overall survival (OS) on namodenoson was 8.1 months. A phase II blinded, randomized, placebo-controlled trial was subsequently conducted in patients with advanced HCC and CPB cirrhosis. The primary endpoint of OS superiority over placebo was not met. However, subgroup analysis of CPB7 patients (34 namodenoson-treated, 22 placebo-treated) showed nonsignificant differences in OS/progression-free survival and a significant difference in 12-month OS (44% vs 18%, p = 0.028). Partial response was achieved in 9% of namodenoson-treated patients vs 0% in placebo-treated patients. Based on the positive efficacy signal in HCC CPB7 patients and the favorable safety profile of namodenoson, a phase III study is underway.
Collapse
Affiliation(s)
- Pnina Fishman
- Can-Fite BioPharma Ltd., 10 Bareket St., 49170, Petah Tikva, Israel.
| | - Salomon M Stemmer
- Davidoff Cancer Center, Petah Tikva and Sackler Faculty of Medicine, Rabin Medical Center-Beilinson Hospital, Tel Aviv, Israel
| | | | | | - William D Kerns
- Can-Fite BioPharma Ltd., 10 Bareket St., 49170, Petah Tikva, Israel
| |
Collapse
|
8
|
Brusini R, Tran NLL, Cailleau C, Domergue V, Nicolas V, Dormont F, Calet S, Cajot C, Jouran A, Lepetre-Mouelhi S, Laloy J, Couvreur P, Varna M. Assessment of Squalene-Adenosine Nanoparticles in Two Rodent Models of Cardiac Ischemia-Reperfusion. Pharmaceutics 2023; 15:1790. [PMID: 37513977 PMCID: PMC10384353 DOI: 10.3390/pharmaceutics15071790] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2023] [Revised: 06/13/2023] [Accepted: 06/15/2023] [Indexed: 07/30/2023] Open
Abstract
Reperfusion injuries after a period of cardiac ischemia are known to lead to pathological modifications or even death. Among the different therapeutic options proposed, adenosine, a small molecule with platelet anti-aggregate and anti-inflammatory properties, has shown encouraging results in clinical trials. However, its clinical use is severely limited because of its very short half-life in the bloodstream. To overcome this limitation, we have proposed a strategy to encapsulate adenosine in squalene-based nanoparticles (NPs), a biocompatible and biodegradable lipid. Thus, the aim of this study was to assess, whether squalene-based nanoparticles loaded with adenosine (SQAd NPs) were cardioprotective in a preclinical cardiac ischemia/reperfusion model. Obtained SQAd NPs were characterized in depth and further evaluated in vitro. The NPs were formulated with a size of about 90 nm and remained stable up to 14 days at both 4 °C and room temperature. Moreover, these NPs did not show any signs of toxicity, neither on HL-1, H9c2 cardiac cell lines, nor on human PBMC and, further retained their inhibitory platelet aggregation properties. In a mouse model with experimental cardiac ischemia-reperfusion, treatment with SQAd NPs showed a reduction of the area at risk, as well as of the infarct area, although not statistically significant. However, we noted a significant reduction of apoptotic cells on cardiac tissue from animals treated with the NPs. Further studies would be interesting to understand how and through which mechanisms these nanoparticles act on cardiac cells.
Collapse
Affiliation(s)
- Romain Brusini
- Université Paris-Saclay, Institut Galien Paris-Saclay, CNRS UMR 8612, Pole Biologie-Pharmacie-Chimie, Bâtiment Henri Moissan, 6 Rue d'Arsonval, 91400 Orsay, France
| | - Natalie Lan Linh Tran
- Université Paris-Saclay, Institut Galien Paris-Saclay, CNRS UMR 8612, Pole Biologie-Pharmacie-Chimie, Bâtiment Henri Moissan, 6 Rue d'Arsonval, 91400 Orsay, France
- Namur Nanosafety Centre, Department of Pharmacy, Namur Research Institute for Life Sciences (NARILIS), University of Namur (UNamur), 5000 Namur, Belgium
| | - Catherine Cailleau
- Université Paris-Saclay, Institut Galien Paris-Saclay, CNRS UMR 8612, Pole Biologie-Pharmacie-Chimie, Bâtiment Henri Moissan, 6 Rue d'Arsonval, 91400 Orsay, France
| | - Valérie Domergue
- Université Paris-Saclay, Inserm, CNRS, Ingénierie et Plateformes au Service de l'Innovation Thérapeutique, ANIMEX, 17 Avenue des Sciences, 91400 Orsay, France
| | - Valérie Nicolas
- Université Paris-Saclay, Inserm, CNRS, Ingénierie et Plateformes au Service de l'Innovation Thérapeutique, MIPSIT, 17 Avenue des Sciences, 91400 Orsay, France
| | - Flavio Dormont
- Université Paris-Saclay, Institut Galien Paris-Saclay, CNRS UMR 8612, Pole Biologie-Pharmacie-Chimie, Bâtiment Henri Moissan, 6 Rue d'Arsonval, 91400 Orsay, France
| | - Serge Calet
- Holochem, Rue du Moulin de la Canne, 45300 Pithiviers, France
| | - Caroline Cajot
- Quality Assistance S.A, Technoparc de Thudinie 2, 6536 Thuin, Belgium
| | - Albin Jouran
- Quality Assistance S.A, Technoparc de Thudinie 2, 6536 Thuin, Belgium
| | - Sinda Lepetre-Mouelhi
- Université Paris-Saclay, Institut Galien Paris-Saclay, CNRS UMR 8612, Pole Biologie-Pharmacie-Chimie, Bâtiment Henri Moissan, 6 Rue d'Arsonval, 91400 Orsay, France
| | - Julie Laloy
- Namur Nanosafety Centre, Department of Pharmacy, Namur Research Institute for Life Sciences (NARILIS), University of Namur (UNamur), 5000 Namur, Belgium
| | - Patrick Couvreur
- Université Paris-Saclay, Institut Galien Paris-Saclay, CNRS UMR 8612, Pole Biologie-Pharmacie-Chimie, Bâtiment Henri Moissan, 6 Rue d'Arsonval, 91400 Orsay, France
| | - Mariana Varna
- Université Paris-Saclay, Institut Galien Paris-Saclay, CNRS UMR 8612, Pole Biologie-Pharmacie-Chimie, Bâtiment Henri Moissan, 6 Rue d'Arsonval, 91400 Orsay, France
| |
Collapse
|
9
|
Yacouba Moukeila MB, Thokerunga E, He F, Bongolo CC, Xia Y, Wang F, Gado AF, Mamoudou H, Khan S, Ousseina B, Ousmane HA, Diarra D, Ke J, Zhang Z, Wang Y. Adenosine 2 receptor regulates autophagy and apoptosis to alleviate ischemia reperfusion injury in type 2 diabetes via IRE-1 signaling. BMC Cardiovasc Disord 2023; 23:154. [PMID: 36964482 PMCID: PMC10039586 DOI: 10.1186/s12872-023-03116-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2022] [Accepted: 02/08/2023] [Indexed: 03/26/2023] Open
Abstract
PURPOSE This study aimed to determine the effect and mechanism of action of adenosine 2 receptor (A2R) activation on myocardial ischemia reperfusion injury (MIRI) under diabetic conditions. METHODS MIRI type 2 diabetic rats and H9C2 cardiomyocytes were treated with A2R agonist and then subjected to hypoxia for 6 h and reoxygenation for 18 h. Myocardial damage, and infarct size were determined by cardiac ultrasound. Indicators of cardiomyocyte injury, creatine kinase-MB and cardiac troponin I were detected by Enzyme Linked Immunosorbent Assay. Endoplasmic reticulum stress (ERS) was determined through measuring the expression levels of ERS related genes GRP78, p-IRE1/IRE1, and p-JNKJNK. The mechanism of A2R cardio protection in MIRI through regulating ERS induced autophagy was determined by investigating the ER resident protein IRE-1. The ER-stress inducer Tunicamycin, and the IRE-1 inhibitor STF in combination with the A2R agonist NECA were used, and the cellular responses were assessed through autophagy proteins expression Beclin-1, p62, LC3 and apoptosis. RESULTS NECA improved left ventricular function post MIRI, limited myocardial infarct size, reduced myocardial damage, decreased cardiomyocytes apoptosis, and attenuated ERS induced autophagy through regulating the IRE-XBP1s-CHOP pathway. These actions resulted into overall protection of the myocardium against MIRI. CONCLUSION In summary, A2R activation by NECA prior to ischemia attenuates apoptosis, reduces ERS induced autophagy and restores left ventricular function. This protective effect occurs through regulating the IRE1-XBPs-CHOP related mechanisms. NECA is thus a potential target for the treatment of MIRI in patient with type 2 diabetes.
Collapse
Affiliation(s)
| | - Erick Thokerunga
- Program and Department of Clinical Laboratory Medicine, Center for Gene Diagnosis, Zhongnan Hospital of Wuhan University, Wuhan, 430071, Hubei, China
| | - Feng He
- Department of Anesthesiology, Zhongnan Hospital of Wuhan University, Wuhan, 430071, Hubei, China
| | - Christian Cedric Bongolo
- Program and Department of Clinical Laboratory Medicine, Center for Gene Diagnosis, Zhongnan Hospital of Wuhan University, Wuhan, 430071, Hubei, China
| | - Yun Xia
- Department of Anesthesiology, Zhongnan Hospital of Wuhan University, Wuhan, 430071, Hubei, China
| | - Fuyu Wang
- Department of Anesthesiology, Zhongnan Hospital of Wuhan University, Wuhan, 430071, Hubei, China
| | - Adamou Foumakoye Gado
- Department of Anesthesia and Intensive Care, Hôpital Général de Référence Niamey, Niamey, Niger
| | - Hama Mamoudou
- Department of Anesthesia and Intensive Care, National Hospital of Niamey, Niamey, Niger
| | - Shahzad Khan
- Department of Pathology, Zhongnan Hospital of Wuhan University, Wuhan, 430071, Hubei, China
| | - Bonkano Ousseina
- Department of Cardiovascular and Internal Medicine, Niamey Amirou Boubacar Diallo National Hospital, Abdou Moumouni University, Niamey, Niger
| | | | - Drissa Diarra
- Department of Oral and Maxillofacial Surgery, School and Hospital of Stomatology, Wuhan University, Wuhan, China
| | - Jianjuan Ke
- Department of Anesthesiology, Zhongnan Hospital of Wuhan University, Wuhan, 430071, Hubei, China.
| | - Zongze Zhang
- Department of Anesthesiology, Zhongnan Hospital of Wuhan University, Wuhan, 430071, Hubei, China.
| | - Yanlin Wang
- Department of Anesthesiology, Zhongnan Hospital of Wuhan University, Wuhan, 430071, Hubei, China.
| |
Collapse
|
10
|
Zhuang Y, Yu ML, Lu SF. Purinergic signaling in myocardial ischemia-reperfusion injury. Purinergic Signal 2023; 19:229-243. [PMID: 35254594 PMCID: PMC9984618 DOI: 10.1007/s11302-022-09856-4] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2021] [Accepted: 02/18/2022] [Indexed: 10/18/2022] Open
Abstract
Purines and their derivatives, extensively distributed in the body, act as a class of extracellular signaling molecules via a rich array of receptors, also known as purinoceptors (P1, P2X, and P2Y). They mediate multiple intracellular signal transduction pathways and participate in various physiological and pathological cell behaviors. Since the function in myocardial ischemia-reperfusion injury (MIRI), this review summarized the involvement of purinergic signal transduction in diversified pathological processes, including energy metabolism disorder, oxidative stress injury, calcium overload, inflammatory immune response, platelet aggregation, coronary vascular dysfunction, and cell necrosis and apoptosis. Moreover, increasing evidence suggests that purinergic signaling also mediates the prevention and treatment of MIRI, such as ischemic conditioning, pharmacological intervention, and some other therapies. In conclusion, this review exhibited that purinergic signaling mediates the complex processes of MIRI which shows its promising application and prospecting in the future.
Collapse
Affiliation(s)
- Yi Zhuang
- College of Acupuncture and Tuina, Nanjing University of Chinese Medicine, 138 Xian-lin Avenue, Qixia District, Nanjing, 210023, Jiangsu Province, China
| | - Mei-Ling Yu
- College of Acupuncture and Tuina, Nanjing University of Chinese Medicine, 138 Xian-lin Avenue, Qixia District, Nanjing, 210023, Jiangsu Province, China
| | - Sheng-Feng Lu
- College of Acupuncture and Tuina, Nanjing University of Chinese Medicine, 138 Xian-lin Avenue, Qixia District, Nanjing, 210023, Jiangsu Province, China. .,Key Laboratory of Acupuncture and Medicine Research of Ministry of Education, Nanjing University of Chinese Medicine, Nanjing, 210023, China.
| |
Collapse
|
11
|
Varlamova EG, Plotnikov EY, Turovsky EA. Neuronal Calcium Sensor-1 Protects Cortical Neurons from Hyperexcitation and Ca 2+ Overload during Ischemia by Protecting the Population of GABAergic Neurons. Int J Mol Sci 2022; 23:ijms232415675. [PMID: 36555318 PMCID: PMC9778989 DOI: 10.3390/ijms232415675] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2022] [Revised: 12/07/2022] [Accepted: 12/08/2022] [Indexed: 12/14/2022] Open
Abstract
A defection of blood circulation in the brain leads to ischemia, damage, and the death of nerve cells. It is known that individual populations of GABAergic neurons are the least resistant to the damaging factors of ischemia and therefore they die first of all, which leads to impaired inhibition in neuronal networks. To date, the neuroprotective properties of a number of calcium-binding proteins (calbindin, calretinin, and parvalbumin), which are markers of GABAergic neurons, are known. Neuronal calcium sensor-1 (NCS-1) is a signaling protein that is expressed in all types of neurons and is involved in the regulation of neurotransmission. The role of NCS-1 in the protection of neurons and especially their individual populations from ischemia and hyperexcitation has not been practically studied. In this work, using the methods of fluorescence microscopy, vitality tests, immunocytochemistry, and PCR analysis, the molecular mechanisms of the protective action of NCS-1 in ischemia/reoxygenation and hyperammonemia were established. Since NCS-1 is most expressed in GABAergic neurons, the knockdown of this protein with siRNA led to the most pronounced consequences in GABAergic neurons. The knockdown of NCS-1 (NCS-1-KD) suppressed the basic expression of protective proteins without significantly reducing cell viability. However, ischemia-like conditions (oxygen-glucose deprivation, OGD) and subsequent 24-h reoxygenation led to a more massive activation of apoptosis and necrosis in neurons with NCS-1-KD, compared to control cells. The mass death of NCS-1-KD cells during OGD and hyperammonemia has been associated with the induction of a more pronounced network hyperexcitation symptom, especially in the population of GABAergic neurons, leading to a global increase in cytosolic calcium ([Ca2+]i). The symptom of hyperexcitation of neurons with NCS-1-KD correlated with a decrease in the level of expression of the calcium-binding protein-parvalbumin. This was accompanied by an increase in the expression of excitatory ionotropic glutamate receptors, N-methyl-D-aspartate and α-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid receptors (NMDAR and AMPAR) against the background of suppression of the expression of glutamate decarboxylase (synthesis of γ-aminobutyric acid).
Collapse
Affiliation(s)
- Elena G. Varlamova
- Institute of Cell Biophysics of the Russian Academy of Sciences, Federal Research Center “Pushchino Scientific Center for Biological Research of the Russian Academy of Sciences”, 142290 Pushchino, Russia
- Correspondence: (E.G.V.); (E.A.T.)
| | - Egor Y. Plotnikov
- A.N. Belozersky Institute of Physico-Chemical Biology, Lomonosov Moscow State University, 119992 Moscow, Russia
- V.I. Kulakov National Medical Research Center of Obstetrics, Gynecology and Perinatology, 117997 Moscow, Russia
| | - Egor A. Turovsky
- Institute of Cell Biophysics of the Russian Academy of Sciences, Federal Research Center “Pushchino Scientific Center for Biological Research of the Russian Academy of Sciences”, 142290 Pushchino, Russia
- Correspondence: (E.G.V.); (E.A.T.)
| |
Collapse
|
12
|
Mangmool S, Kyaw ETH, Nuamnaichati N, Pandey S, Parichatikanond W. Stimulation of adenosine A 1 receptor prevents oxidative injury in H9c2 cardiomyoblasts: Role of Gβγ-mediated Akt and ERK1/2 signaling. Toxicol Appl Pharmacol 2022; 451:116175. [PMID: 35901927 DOI: 10.1016/j.taap.2022.116175] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2022] [Revised: 07/15/2022] [Accepted: 07/20/2022] [Indexed: 02/05/2023]
Abstract
Oxidative stress causes cellular injury and damage in the heart primarily through apoptosis resulting in cardiac abnormalities such as heart failure and cardiomyopathy. During oxidative stress, stimulation of adenosine receptor (AR) has been shown to protect against oxidative damage due to their cytoprotective properties. However, the subtype specificity and signal transductions of adenosine A1 receptor (A1R) on cardiac protection during oxidative stress have remained elusive. In this study, we found that stimulation of A1Rs with N6-cyclopentyladenosine (CPA), a specific A1R agonist, attenuated the H2O2-induced intracellular and mitochondrial reactive oxygen species (ROS) production and apoptosis. In addition, A1R stimulation upregulated the synthesis of antioxidant enzymes (catalase and GPx-1), antiapoptotic proteins (Bcl-2 and Bcl-xL), and mitochondria-related markers (UCP2 and UCP3). Blockades of Gβγ subunit of heterotrimeric Gαi protein antagonized A1R-mediated antioxidant and antiapoptotic effects, confirming the potential role of Gβγ subunit-mediated A1R signaling. Additionally, cardioprotective effects of CPA mediated through PI3K/Akt- and ERK1/2-dependent signaling pathways. Thus, we propose that A1R represents a promising therapeutic target for prevention of oxidative injury in the heart.
Collapse
Affiliation(s)
- Supachoke Mangmool
- Department of Pharmacology, Faculty of Science, Mahidol University, Bangkok 10400, Thailand
| | - Ei Thet Htar Kyaw
- Department of Pharmacology, Faculty of Pharmacy, Mahidol University, Bangkok 10400, Thailand
| | - Narawat Nuamnaichati
- Department of Pharmacology, Faculty of Pharmacy, Mahidol University, Bangkok 10400, Thailand
| | - Sudhir Pandey
- Department of Pharmacology, Faculty of Pharmacy, Mahidol University, Bangkok 10400, Thailand
| | - Warisara Parichatikanond
- Department of Pharmacology, Faculty of Pharmacy, Mahidol University, Bangkok 10400, Thailand; Center of Biopharmaceutical Science for Healthy Ageing (BSHA), Faculty of Pharmacy, Mahidol University, Bangkok 10400, Thailand.
| |
Collapse
|
13
|
Wölkart G, Stessel H, Fassett E, Teschl E, Friedl K, Trummer M, Schrammel A, Kollau A, Mayer B, Fassett J. Adenosine kinase (ADK) inhibition with ABT-702 induces ADK protein degradation and a distinct form of sustained cardioprotection. Eur J Pharmacol 2022; 927:175050. [PMID: 35618039 DOI: 10.1016/j.ejphar.2022.175050] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2021] [Revised: 05/17/2022] [Accepted: 05/19/2022] [Indexed: 11/18/2022]
Abstract
Pharmacological inhibition of adenosine kinase (ADK), the major route of myocardial adenosine metabolism, can elicit acute cardioprotection against ischemia-reperfusion (IR) by increasing adenosine signaling. Here, we identified a novel, extended effect of the ADK inhibitor, ABT-702, on cardiac ADK protein longevity and investigated its impact on sustained adenosinergic cardioprotection. We found that ABT-702 treatment significantly reduced cardiac ADK protein content in mice 24-72 h after administration (IP or oral). ABT-702 did not alter ADK mRNA levels, but strongly diminished (ADK-L) isoform protein content through a proteasome-dependent mechanism. Langendorff perfusion experiments revealed that hearts from ABT-702-treated mice maintain higher adenosine release long after ABT-702 tissue elimination, accompanied by increased basal coronary flow (CF) and robust tolerance to IR. Sustained cardioprotection by ABT-702 did not involve increased nitric oxide synthase expression, but was completely dependent upon increased adenosine release in the delayed phase (24 h), as indicated by the loss of cardioprotection and CF increase upon perfusion of adenosine deaminase or adenosine receptor antagonist, 8-phenyltheophylline. Importantly, blocking adenosine receptor activity with theophylline during ABT-702 administration prevented ADK degradation, preserved late cardiac ADK activity, diminished CF increase and abolished delayed cardioprotection, indicating that early adenosine receptor signaling induces late ADK degradation to elicit sustained adenosine release. Together, these results indicate that ABT-702 induces a distinct form of delayed cardioprotection mediated by adenosine receptor-dependent, proteasomal degradation of cardiac ADK and enhanced adenosine signaling in the late phase. These findings suggest ADK protein stability may be pharmacologically targeted to achieve sustained adenosinergic cardioprotection.
Collapse
Affiliation(s)
- Gerald Wölkart
- Department of Pharmacology and Toxicology, University of Graz, 8010, Graz, Austria
| | - Heike Stessel
- Department of Pharmacology and Toxicology, University of Graz, 8010, Graz, Austria
| | - Erin Fassett
- Department of Pharmacology and Toxicology, University of Graz, 8010, Graz, Austria
| | - Eva Teschl
- Department of Pharmacology and Toxicology, University of Graz, 8010, Graz, Austria
| | - Katrin Friedl
- Department of Pharmacology and Toxicology, University of Graz, 8010, Graz, Austria
| | - Modesta Trummer
- Department of Pharmacology and Toxicology, University of Graz, 8010, Graz, Austria
| | - Astrid Schrammel
- Department of Pharmacology and Toxicology, University of Graz, 8010, Graz, Austria
| | - Alexander Kollau
- Department of Pharmacology and Toxicology, University of Graz, 8010, Graz, Austria
| | - Bernd Mayer
- Department of Pharmacology and Toxicology, University of Graz, 8010, Graz, Austria
| | - John Fassett
- Department of Pharmacology and Toxicology, University of Graz, 8010, Graz, Austria.
| |
Collapse
|
14
|
Mahmood A, Iqbal J. Purinergic receptors modulators: An emerging pharmacological tool for disease management. Med Res Rev 2022; 42:1661-1703. [PMID: 35561109 DOI: 10.1002/med.21888] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2021] [Revised: 03/16/2022] [Accepted: 05/04/2022] [Indexed: 11/10/2022]
Abstract
Purinergic signaling is mediated through extracellular nucleotides (adenosine 5'-triphosphate, uridine-5'-triphosphate, adenosine diphosphate, uridine-5'-diphosphate, and adenosine) that serve as signaling molecules. In the early 1990s, purines and pyrimidine receptors were cloned and characterized drawing the attention of scientists toward this aspect of cellular signaling. This signaling pathway is comprised of four subtypes of adenosine receptors (P1), eight subtypes of G-coupled protein receptors (P2YRs), and seven subtypes of ligand-gated ionotropic receptors (P2XRs). In current studies, the pathophysiology and therapeutic potentials of these receptors have been focused on. Various ligands, modulating the functions of purinergic receptors, are in current clinical practices for the treatment of various neurodegenerative disorders and cardiovascular diseases. Moreover, several purinergic receptors ligands are in advanced phases of clinical trials as a remedy for depression, epilepsy, autism, osteoporosis, atherosclerosis, myocardial infarction, diabetes, irritable bowel syndrome, and cancers. In the present study, agonists and antagonists of purinergic receptors have been summarized that may serve as pharmacological tools for drug design and development.
Collapse
Affiliation(s)
- Abid Mahmood
- Centre for Advanced Drug Research, COMSATS University Islamabad, Abbottabad, Pakistan
| | - Jamshed Iqbal
- Centre for Advanced Drug Research, COMSATS University Islamabad, Abbottabad, Pakistan
| |
Collapse
|
15
|
Penna C, Comità S, Tullio F, Alloatti G, Pagliaro P. Challenges facing the clinical translation of cardioprotection: 35 years after the discovery of ischemic preconditioning. Vascul Pharmacol 2022; 144:106995. [DOI: 10.1016/j.vph.2022.106995] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2022] [Revised: 03/17/2022] [Accepted: 04/16/2022] [Indexed: 12/19/2022]
|
16
|
Acute Coronary Syndromes (ACS)-Unravelling Biology to Identify New Therapies-The Microcirculation as a Frontier for New Therapies in ACS. Cells 2021; 10:cells10092188. [PMID: 34571836 PMCID: PMC8468909 DOI: 10.3390/cells10092188] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2021] [Revised: 08/18/2021] [Accepted: 08/23/2021] [Indexed: 12/12/2022] Open
Abstract
In acute coronary syndrome (ACS) patients, restoring epicardial culprit vessel patency and flow with percutaneous coronary intervention or coronary artery bypass grafting has been the mainstay of treatment for decades. However, there is an emerging understanding of the crucial role of coronary microcirculation in predicting infarct burden and subsequent left ventricular remodelling, and the prognostic significance of coronary microvascular obstruction (MVO) in mortality and morbidity. This review will elucidate the multifaceted and interconnected pathophysiological processes which underpin MVO in ACS, and the various diagnostic modalities as well as challenges, with a particular focus on the invasive but specific and reproducible index of microcirculatory resistance (IMR). Unfortunately, a multitude of purported therapeutic strategies to address this unmet need in cardiovascular care, outlined in this review, have so far been disappointing with conflicting results and a lack of hard clinical end-point benefit. There are however a number of exciting and novel future prospects in this field that will be evaluated over the coming years in large adequately powered clinical trials, and this review will briefly appraise these.
Collapse
|
17
|
Hepatic cell mobilization for protection against ischemic myocardial injury. Sci Rep 2021; 11:15830. [PMID: 34349157 PMCID: PMC8339068 DOI: 10.1038/s41598-021-94170-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2020] [Accepted: 06/29/2021] [Indexed: 11/29/2022] Open
Abstract
The heart is capable of activating protective mechanisms in response to ischemic injury to support myocardial survival and performance. These mechanisms have been recognized primarily in the ischemic heart, involving paracrine signaling processes. Here, we report a distant cardioprotective mechanism involving hepatic cell mobilization to the ischemic myocardium in response to experimental myocardial ischemia–reperfusion (MI-R) injury. A parabiotic mouse model was generated by surgical skin-union of two mice and used to induce bilateral MI-R injury with unilateral hepatectomy, establishing concurrent gain- and loss-of-hepatic cell mobilization conditions. Hepatic cells, identified based on the cell-specific expression of enhanced YFP, were found in the ischemic myocardium of parabiotic mice with intact liver (0.2 ± 0.1%, 1.1 ± 0.3%, 2.7 ± 0.6, and 0.7 ± 0.4% at 1, 3, 5, and 10 days, respectively, in reference to the total cell nuclei), but not significantly in the ischemic myocardium of parabiotic mice with hepatectomy (0 ± 0%, 0.1 ± 0.1%, 0.3 ± 0.2%, and 0.08 ± 0.08% at the same time points). The mobilized hepatic cells were able to express and release trefoil factor 3 (TFF3), a protein mitigating MI-R injury as demonstrated in TFF3−/− mice (myocardium infarcts 17.6 ± 2.3%, 20.7 ± 2.6%, and 15.3 ± 3.8% at 1, 5, and 10 days, respectively) in reference to wildtype mice (11.7 ± 1.9%, 13.8 ± 2.3%, and 11.0 ± 1.8% at the same time points). These observations suggest that MI-R injury can induce hepatic cell mobilization to support myocardial survival by releasing TFF3.
Collapse
|
18
|
Reducing Cardiac Injury during ST-Elevation Myocardial Infarction: A Reasoned Approach to a Multitarget Therapeutic Strategy. J Clin Med 2021; 10:jcm10132968. [PMID: 34279451 PMCID: PMC8268641 DOI: 10.3390/jcm10132968] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2021] [Revised: 06/22/2021] [Accepted: 06/27/2021] [Indexed: 02/06/2023] Open
Abstract
The significant reduction in ‘ischemic time’ through capillary diffusion of primary percutaneous intervention (pPCI) has rendered myocardial-ischemia reperfusion injury (MIRI) prevention a major issue in order to improve the prognosis of ST elevation myocardial infarction (STEMI) patients. In fact, while the ischemic damage increases with the severity and the duration of blood flow reduction, reperfusion injury reaches its maximum with a moderate amount of ischemic injury. MIRI leads to the development of post-STEMI left ventricular remodeling (post-STEMI LVR), thereby increasing the risk of arrhythmias and heart failure. Single pharmacological and mechanical interventions have shown some benefits, but have not satisfactorily reduced mortality. Therefore, a multitarget therapeutic strategy is needed, but no univocal indications have come from the clinical trials performed so far. On the basis of the results of the consistent clinical studies analyzed in this review, we try to design a randomized clinical trial aimed at evaluating the effects of a reasoned multitarget therapeutic strategy on the prevention of post-STEMI LVR. In fact, we believe that the correct timing of pharmacological and mechanical intervention application, according to their specific ability to interfere with survival pathways, may significantly reduce the incidence of post-STEMI LVR and thus improve patient prognosis.
Collapse
|
19
|
Aetesam-Ur-Rahman M, Giblett JP, Khialani B, Kyranis S, Clarke SJ, Zhao TX, Braganza DM, Clarke SC, West NEJ, Bennett MR, Hoole SP. GLP-1 vasodilatation in humans with coronary artery disease is not adenosine mediated. BMC Cardiovasc Disord 2021; 21:223. [PMID: 33932990 PMCID: PMC8088691 DOI: 10.1186/s12872-021-02030-5] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2020] [Accepted: 04/21/2021] [Indexed: 12/23/2022] Open
Abstract
BACKGROUND Incretin therapies appear to provide cardioprotection and improve cardiovascular outcomes in patients with diabetes, but the mechanism of this effect remains elusive. We have previously shown that glucagon-like peptide (GLP)-1 is a coronary vasodilator and we sought to investigate if this is an adenosine-mediated effect. METHODS We recruited 41 patients having percutaneous coronary intervention (PCI) for stable angina and allocated them into four groups administering a specific study-related infusion following successful PCI: GLP-1 infusion (Group G) (n = 10); Placebo, normal saline infusion (Group P) (n = 11); GLP-1 + Theophylline infusion (Group GT) (n = 10); and Theophylline infusion (Group T) (n = 10). A pressure wire assessment of coronary distal pressure and flow velocity (thermodilution transit time-Tmn) at rest and hyperaemia was performed after PCI and repeated following the study infusion to derive basal and index of microvascular resistance (BMR and IMR). RESULTS There were no significant differences in the demographics of patients recruited to our study. Most of the patients were not diabetic. GLP-1 caused significant reduction of resting Tmn that was not attenuated by theophylline: mean delta Tmn (SD) group G - 0.23 s (0.27) versus group GT - 0.18 s (0.37), p = 0.65. Theophylline alone (group T) did not significantly alter resting flow velocity compared to group GT: delta Tmn in group T 0.04 s (0.15), p = 0.30. The resulting decrease in BMR observed in group G persisted in group GT: - 20.83 mmHg s (24.54 vs. - 21.20 mmHg s (30.41), p = 0.97. GLP-1 did not increase circulating adenosine levels in group GT more than group T: delta median adenosine - 2.0 ng/ml (- 117.1, 14.8) versus - 0.5 ng/ml (- 19.6, 9.4); p = 0.60. CONCLUSION The vasodilatory effect of GLP-1 is not abolished by theophylline and GLP-1 does not increase adenosine levels, indicating an adenosine-independent mechanism of GLP-1 coronary vasodilatation. TRIAL REGISTRATION The local research ethics committee approved the study (National Research Ethics Service-NRES Committee, East of England): REC reference 14/EE/0018. The study was performed according to institutional guidelines, was registered on http://www.clinicaltrials.gov (unique identifier: NCT03502083) and the study conformed to the principles outlined in the Declaration of Helsinki.
Collapse
Affiliation(s)
- Muhammad Aetesam-Ur-Rahman
- Department of Interventional Cardiology, Royal Papworth Hospital, Papworth Road, Cambridge Biomedical Campus, Cambridge, CB2 0AY, UK
- Division of Cardiovascular Medicine, University of Cambridge, Cambridge, UK
| | - Joel P Giblett
- Department of Interventional Cardiology, Royal Papworth Hospital, Papworth Road, Cambridge Biomedical Campus, Cambridge, CB2 0AY, UK
| | - Bharat Khialani
- Department of Interventional Cardiology, Royal Papworth Hospital, Papworth Road, Cambridge Biomedical Campus, Cambridge, CB2 0AY, UK
| | - Stephen Kyranis
- Department of Interventional Cardiology, Royal Papworth Hospital, Papworth Road, Cambridge Biomedical Campus, Cambridge, CB2 0AY, UK
| | - Sophie J Clarke
- Department of Interventional Cardiology, Royal Papworth Hospital, Papworth Road, Cambridge Biomedical Campus, Cambridge, CB2 0AY, UK
| | - Tian X Zhao
- Department of Interventional Cardiology, Royal Papworth Hospital, Papworth Road, Cambridge Biomedical Campus, Cambridge, CB2 0AY, UK
- Division of Cardiovascular Medicine, University of Cambridge, Cambridge, UK
| | - Denise M Braganza
- Department of Interventional Cardiology, Royal Papworth Hospital, Papworth Road, Cambridge Biomedical Campus, Cambridge, CB2 0AY, UK
| | - Sarah C Clarke
- Department of Interventional Cardiology, Royal Papworth Hospital, Papworth Road, Cambridge Biomedical Campus, Cambridge, CB2 0AY, UK
| | - Nick E J West
- Department of Interventional Cardiology, Royal Papworth Hospital, Papworth Road, Cambridge Biomedical Campus, Cambridge, CB2 0AY, UK
| | - Martin R Bennett
- Division of Cardiovascular Medicine, University of Cambridge, Cambridge, UK
| | - Stephen P Hoole
- Department of Interventional Cardiology, Royal Papworth Hospital, Papworth Road, Cambridge Biomedical Campus, Cambridge, CB2 0AY, UK.
| |
Collapse
|
20
|
Woo SH, Trinh TN. P2 Receptors in Cardiac Myocyte Pathophysiology and Mechanotransduction. Int J Mol Sci 2020; 22:ijms22010251. [PMID: 33383710 PMCID: PMC7794727 DOI: 10.3390/ijms22010251] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2020] [Revised: 12/22/2020] [Accepted: 12/22/2020] [Indexed: 12/30/2022] Open
Abstract
ATP is a major energy source in the mammalian cells, but it is an extracellular chemical messenger acting on P2 purinergic receptors. A line of evidence has shown that ATP is released from many different types of cells including neurons, endothelial cells, and muscle cells. In this review, we described the distribution of P2 receptor subtypes in the cardiac cells and their physiological and pathological roles in the heart. So far, the effects of external application of ATP or its analogues, and those of UTP on cardiac contractility and rhythm have been reported. In addition, specific genetic alterations and pharmacological agonists and antagonists have been adopted to discover specific roles of P2 receptor subtypes including P2X4-, P2X7-, P2Y2- and P2Y6-receptors in cardiac cells under physiological and pathological conditions. Accumulated data suggest that P2X4 receptors may play a beneficial role in cardiac muscle function, and that P2Y2- and P2Y6-receptors can induce cardiac fibrosis. Recent evidence further demonstrates P2Y1 receptor and P2X4 receptor as important mechanical signaling molecules to alter membrane potential and Ca2+ signaling in atrial myocytes and their uneven expression profile between right and left atrium.
Collapse
|
21
|
Oladimeji O, Akinyelu J, Singh M. Nanomedicines for Subcellular Targeting: The Mitochondrial Perspective. Curr Med Chem 2020; 27:5480-5509. [PMID: 31763965 DOI: 10.2174/0929867326666191125092111] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2019] [Revised: 10/28/2019] [Accepted: 10/30/2019] [Indexed: 01/01/2023]
Abstract
BACKGROUND Over the past decade, there has been a surge in the number of mitochondrialactive therapeutics for conditions ranging from cancer to aging. Subcellular targeting interventions can modulate adverse intracellular processes unique to the compartments within the cell. However, there is a dearth of reviews focusing on mitochondrial nano-delivery, and this review seeks to fill this gap with regards to nanotherapeutics of the mitochondria. METHODS Besides its potential for a higher therapeutic index than targeting at the tissue and cell levels, subcellular targeting takes into account the limitations of systemic drug administration and significantly improves pharmacokinetics. Hence, an extensive literature review was undertaken and salient information was compiled in this review. RESULTS From literature, it was evident that nanoparticles with their tunable physicochemical properties have shown potential for efficient therapeutic delivery, with several nanomedicines already approved by the FDA and others in clinical trials. However, strategies for the development of nanomedicines for subcellular targeting are still emerging, with an increased understanding of dysfunctional molecular processes advancing the development of treatment modules. For optimal delivery, the design of an ideal carrier for subcellular delivery must consider the features of the diseased microenvironment. The functional and structural features of the mitochondria in the diseased state are highlighted and potential nano-delivery interventions for treatment and diagnosis are discussed. CONCLUSION This review provides an insight into recent advances in subcellular targeting, with a focus on en route barriers to subcellular targeting. The impact of mitochondrial dysfunction in the aetiology of certain diseases is highlighted, and potential therapeutic sites are identified.
Collapse
Affiliation(s)
- Olakunle Oladimeji
- Nano-Gene and Drug Delivery Group, Discipline of Biochemistry, School of Life Sciences, University of Kwa-Zulu Natal, Private Bag X54001, Durban, South Africa
| | - Jude Akinyelu
- Nano-Gene and Drug Delivery Group, Discipline of Biochemistry, School of Life Sciences, University of Kwa-Zulu Natal, Private Bag X54001, Durban, South Africa
| | - Moganavelli Singh
- Nano-Gene and Drug Delivery Group, Discipline of Biochemistry, School of Life Sciences, University of Kwa-Zulu Natal, Private Bag X54001, Durban, South Africa
| |
Collapse
|
22
|
Wang WL, Ge TY, Chen X, Mao Y, Zhu YZ. Advances in the Protective Mechanism of NO, H 2S, and H 2 in Myocardial Ischemic Injury. Front Cardiovasc Med 2020; 7:588206. [PMID: 33195476 PMCID: PMC7661694 DOI: 10.3389/fcvm.2020.588206] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2020] [Accepted: 09/28/2020] [Indexed: 12/30/2022] Open
Abstract
Myocardial ischemic injury is among the top 10 leading causes of death from cardiovascular diseases worldwide. Myocardial ischemia is caused mainly by coronary artery occlusion or obstruction. It usually occurs when the heart is insufficiently perfused, oxygen supply to the myocardium is reduced, and energy metabolism in the myocardium is abnormal. Pathologically, myocardial ischemic injury generates a large number of inflammatory cells, thus inducing a state of oxidative stress. This sharp reduction in the number of normal cells as a result of apoptosis leads to organ and tissue damage, which can be life-threatening. Therefore, effective methods for the treatment of myocardial ischemic injury and clarification of the underlying mechanisms are urgently required. Gaseous signaling molecules, such as NO, H2S, H2, and combined gas donors, have gradually become a focus of research. Gaseous signaling molecules have shown anti-apoptotic, anti-oxidative and anti-inflammatory effects as potential therapeutic agents for myocardial ischemic injury in a large number of studies. In this review, we summarize and discuss the mechanism underlying the protective effect of gaseous signaling molecules on myocardial ischemic injury.
Collapse
Affiliation(s)
| | | | - Xu Chen
- Guilin Medical College, Guilin, China
| | - Yicheng Mao
- Shanghai Key Laboratory of Bioactive Small Molecules, Department of Pharmacology, School of Pharmacy, Fudan University, Shanghai, China
| | - Yi-Zhun Zhu
- Guilin Medical College, Guilin, China.,Shanghai Key Laboratory of Bioactive Small Molecules, Department of Pharmacology, School of Pharmacy, Fudan University, Shanghai, China.,State Key Laboratory of Quality Research in Chinese Medicine and School of Pharmacy, Macau University of Science and Technology, Macau, China
| |
Collapse
|
23
|
Yeung PK, Mohammadizadeh S, Akhoundi F, Mann K, Agu RU, Pulinilkunnil T. Hemodynamic Assessment and in vivo Catabolism of Adenosine 5'- Triphosphate in Doxorubicin or Isoproterenol-induced Cardiovascular Toxicity. Drug Metab Lett 2020; 14:80-88. [PMID: 33092518 DOI: 10.2174/1872312814666201022103802] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2020] [Revised: 08/18/2020] [Accepted: 08/30/2020] [Indexed: 11/22/2022]
Abstract
PURPOSE Previous studies have shown catabolism of adenosine 5'-triphosphate (ATP) in systemic blood is a potential surrogate biomarker for cardiovascular toxicity. We compared the acute toxicity of high doses of doxorubicin (DOX) and isoproterenol (ISO) on hemodynamics and ATP catabolism in systemic circulation. METHODS Sprague Dawley (SD) rats (n = 8 - 11) were each given either a single dose of 30 mg/kg ISO, or twice-daily dose of 10 mg/kg of DOX or normal saline (control) for 4 doses by subcutaneous injection. Blood samples were collected up to 6 hours for measuring concentrations of ATP and its catabolites. Hemodynmics was recorded continuously. Difference was considered significant at p < 0.05 (ANOVA). RESULTS AND DISCUSSION Mortality was 1/8, 5/11 and 0/11 for the DOX, ISO and control groups, respectively. Systolic blood pressure was significantly lower in the DOX and ISO treated rats than in the control measured at the last recorded time (76 ± 9 for DOX vs 42 ± 8 for ISO vs 103 ± 5 mmHg for Control, p < 0.05 for all). Blood pressure fell gradually after the final injection for both DOX and control groups, but abruptly after ISO followed by a rebound and then gradual decline till the end of the experiment. Heart rate was significantly higher after ISO, but no difference between the DOX and control rats (p > 0.05). RBC concentrations of ADP and AMP, and plasma concentrations of adenosine and uric acid were significantly higher in the ISO group. In contrast, hypoxanthine concentrations were significantly higher in the DOX treated group (p < 0.05). CONCLUSION Acute cardiovascular toxicity induced by DOX and ISO may be measured by changes in hemodynamics and breakdown of ATP and adenosine in the systemic circulation, albeit a notable qualitative and quantitative difference was observed.
Collapse
Affiliation(s)
- Pollen K Yeung
- Pharmacokinetics and Metabolism Laboratory, College of Pharmacy and Department of Medicine, Faculties of Health and Medicine, Dalhousie University, Halifax, NS. Canada
| | - Sheyda Mohammadizadeh
- Pharmacokinetics and Metabolism Laboratory, College of Pharmacy and Department of Medicine, Faculties of Health and Medicine, Dalhousie University, Halifax, NS. Canada
| | - Fatemeh Akhoundi
- Pharmacokinetics and Metabolism Laboratory, College of Pharmacy and Department of Medicine, Faculties of Health and Medicine, Dalhousie University, Halifax, NS. Canada
| | - Kelsey Mann
- Pharmacokinetics and Metabolism Laboratory, College of Pharmacy and Department of Medicine, Faculties of Health and Medicine, Dalhousie University, Halifax, NS. Canada
| | - Remigius U Agu
- Biopharmaceutics and Drug Delivery Laboratory, College of Pharmacy, Faculty of Health, Dalhousie University, Halifax, NS. Canada
| | - Thomas Pulinilkunnil
- Faculty of Medicine, Department of Biochemistry and Molecular Biology, Dalhousie University, Dalhousie Medicine New Brunswick, Saint John, NB. Canada
| |
Collapse
|
24
|
Paganelli F, Gaudry M, Ruf J, Guieu R. Recent advances in the role of the adenosinergic system in coronary artery disease. Cardiovasc Res 2020; 117:1284-1294. [PMID: 32991685 DOI: 10.1093/cvr/cvaa275] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/06/2020] [Revised: 08/14/2020] [Accepted: 09/15/2020] [Indexed: 12/18/2022] Open
Abstract
Adenosine is an endogenous nucleoside that plays a major role in the physiology and physiopathology of the coronary artery system, mainly by activating its A2A receptors (A2AR). Adenosine is released by myocardial, endothelial, and immune cells during hypoxia, ischaemia, or inflammation, each condition being present in coronary artery disease (CAD). While activation of A2AR improves coronary blood circulation and leads to anti-inflammatory effects, down-regulation of A2AR has many deleterious effects during CAD. A decrease in the level and/or activity of A2AR leads to: (i) lack of vasodilation, which decreases blood flow, leading to a decrease in myocardial oxygenation and tissue hypoxia; (ii) an increase in the immune response, favouring inflammation; and (iii) platelet aggregation, which therefore participates, in part, in the formation of a fibrin-platelet thrombus after the rupture or erosion of the plaque, leading to the occurrence of acute coronary syndrome. Inflammation contributes to the development of atherosclerosis, leading to myocardial ischaemia, which in turn leads to tissue hypoxia. Therefore, a vicious circle is created that maintains and aggravates CAD. In some cases, studying the adenosinergic profile can help assess the severity of CAD. In fact, inducible ischaemia in CAD patients, as assessed by exercise stress test or fractional flow reserve, is associated with the presence of a reserve of A2AR called spare receptors. The purpose of this review is to present emerging experimental evidence supporting the existence of this adaptive adenosinergic response to ischaemia or inflammation in CAD. We believe that we have achieved a breakthrough in the understanding and modelling of spare A2AR, based upon a new concept allowing for a new and non-invasive CAD management.
Collapse
Affiliation(s)
- Franck Paganelli
- C2VN, INSERM, INRAE, Aix-Marseille University, Campus Santé Timone, Faculté de Pharmacie, 27 Bd Jean Moulin, F-13005 Marseille, France.,Department of Cardiology, North Hospital, Chemin des Bourrely, F-13015 Marseille, France
| | - Marine Gaudry
- Department of Vascular Surgery, Timone Hospital, 278 Rue Saint Pierre, F-13005 Marseille, France
| | - Jean Ruf
- C2VN, INSERM, INRAE, Aix-Marseille University, Campus Santé Timone, Faculté de Pharmacie, 27 Bd Jean Moulin, F-13005 Marseille, France
| | - Régis Guieu
- C2VN, INSERM, INRAE, Aix-Marseille University, Campus Santé Timone, Faculté de Pharmacie, 27 Bd Jean Moulin, F-13005 Marseille, France.,Laboratory of Biochemistry, Timone Hospital, 278 Rue Saint Pierre, F-13005 Marseille, France
| |
Collapse
|
25
|
Gaudry M, Vairo D, Marlinge M, Gaubert M, Guiol C, Mottola G, Gariboldi V, Deharo P, Sadrin S, Maixent JM, Fenouillet E, Ruf J, Guieu R, Paganelli F. Adenosine and Its Receptors: An Expected Tool for the Diagnosis and Treatment of Coronary Artery and Ischemic Heart Diseases. Int J Mol Sci 2020; 21:ijms21155321. [PMID: 32727116 PMCID: PMC7432452 DOI: 10.3390/ijms21155321] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2020] [Revised: 07/15/2020] [Accepted: 07/21/2020] [Indexed: 12/14/2022] Open
Abstract
Adenosine is an endogenous nucleoside which strongly impacts the cardiovascular system. Adenosine is released mostly by endothelial cells and myocytes during ischemia or hypoxia and greatly regulates the cardiovascular system via four specific G-protein-coupled receptors named A1R, A2AR, A2BR, and A3R. Among them, A2 subtypes are strongly expressed in coronary tissues, and their activation increases coronary blood flow via the production of cAMP in smooth muscle cells. A2A receptor modulators are an opportunity for intense research by the pharmaceutical industry to develop new cardiovascular therapies. Most innovative therapies are mediated by the modulation of adenosine release and/or the activation of the A2A receptor subtypes. This review aims to focus on the specific exploration of the adenosine plasma level and its relationship with the A2A receptor, which seems a promising biomarker for a diagnostic and/or a therapeutic tool for the screening and management of coronary artery disease. Finally, a recent class of selective adenosine receptor ligands has emerged, and A2A receptor agonists/antagonists are useful tools to improve the management of patients suffering from coronary artery disease.
Collapse
Affiliation(s)
- Marine Gaudry
- Department of Vascular Surgery, Timone Hospital, F-13008 Marseille, France;
| | - Donato Vairo
- C2VN, INSERM, INRA, Aix-Marseille University, F-13015 Marseille, France; (D.V.); (M.M.); (M.G.); (C.G.); (G.M.); (V.G.); (P.D.); (E.F.); (J.R.); (R.G.)
| | - Marion Marlinge
- C2VN, INSERM, INRA, Aix-Marseille University, F-13015 Marseille, France; (D.V.); (M.M.); (M.G.); (C.G.); (G.M.); (V.G.); (P.D.); (E.F.); (J.R.); (R.G.)
- Laboratory of Biochemistry, Timone Hospital, F-13008 Marseille, France
| | - Melanie Gaubert
- C2VN, INSERM, INRA, Aix-Marseille University, F-13015 Marseille, France; (D.V.); (M.M.); (M.G.); (C.G.); (G.M.); (V.G.); (P.D.); (E.F.); (J.R.); (R.G.)
| | - Claire Guiol
- C2VN, INSERM, INRA, Aix-Marseille University, F-13015 Marseille, France; (D.V.); (M.M.); (M.G.); (C.G.); (G.M.); (V.G.); (P.D.); (E.F.); (J.R.); (R.G.)
| | - Giovanna Mottola
- C2VN, INSERM, INRA, Aix-Marseille University, F-13015 Marseille, France; (D.V.); (M.M.); (M.G.); (C.G.); (G.M.); (V.G.); (P.D.); (E.F.); (J.R.); (R.G.)
- Laboratory of Biochemistry, Timone Hospital, F-13008 Marseille, France
| | - Vlad Gariboldi
- C2VN, INSERM, INRA, Aix-Marseille University, F-13015 Marseille, France; (D.V.); (M.M.); (M.G.); (C.G.); (G.M.); (V.G.); (P.D.); (E.F.); (J.R.); (R.G.)
- Department of Cardiac Surgery, Timone Hospital, F-13008 Marseille, France
| | - Pierre Deharo
- C2VN, INSERM, INRA, Aix-Marseille University, F-13015 Marseille, France; (D.V.); (M.M.); (M.G.); (C.G.); (G.M.); (V.G.); (P.D.); (E.F.); (J.R.); (R.G.)
- Department of Cardiology, Timone Hospital, F-13008 Marseille, France
| | | | - Jean Michel Maixent
- Unité de Recherche Clinique Pierre Deniker (URC C.S. 10587) Centre Hospitalier Henri Laborit, 86000 Poitiers, France
- I.A.P.S. Equipe Emergeante, Université de Toulon, 83957 Toulon-La Garde, UFR S.F.A., F-86073 Poitiers, France
- Correspondence: (J.M.M.); (F.P.)
| | - Emmanuel Fenouillet
- C2VN, INSERM, INRA, Aix-Marseille University, F-13015 Marseille, France; (D.V.); (M.M.); (M.G.); (C.G.); (G.M.); (V.G.); (P.D.); (E.F.); (J.R.); (R.G.)
| | - Jean Ruf
- C2VN, INSERM, INRA, Aix-Marseille University, F-13015 Marseille, France; (D.V.); (M.M.); (M.G.); (C.G.); (G.M.); (V.G.); (P.D.); (E.F.); (J.R.); (R.G.)
| | - Regis Guieu
- C2VN, INSERM, INRA, Aix-Marseille University, F-13015 Marseille, France; (D.V.); (M.M.); (M.G.); (C.G.); (G.M.); (V.G.); (P.D.); (E.F.); (J.R.); (R.G.)
- Laboratory of Biochemistry, Timone Hospital, F-13008 Marseille, France
| | - Franck Paganelli
- C2VN, INSERM, INRA, Aix-Marseille University, F-13015 Marseille, France; (D.V.); (M.M.); (M.G.); (C.G.); (G.M.); (V.G.); (P.D.); (E.F.); (J.R.); (R.G.)
- Department of Cardiology, Nord Hospital, ARCHANTEC, F-13015 Marseille, France
- Correspondence: (J.M.M.); (F.P.)
| |
Collapse
|
26
|
Abstract
Despite the increasing use and success of interventional coronary reperfusion strategies, morbidity and mortality from acute myocardial infarction are still substantial. Myocardial infarct size is a major determinant of prognosis in these patients. Therefore, cardioprotective strategies aim to reduce infarct size. However, a perplexing gap exists between the many preclinical studies reporting infarct size reduction with mechanical and pharmacological interventions and the poor translation into better clinical outcomes in patients. This Review revisits the pathophysiology of myocardial ischaemia-reperfusion injury, including the role of autophagy and forms of cell death such as necrosis, apoptosis, necroptosis and pyroptosis. Other cellular compartments in addition to cardiomyocytes are addressed, notably the coronary microcirculation. Preclinical and clinical research developments in mechanical and pharmacological approaches to induce cardioprotection, and their signal transduction pathways, are discussed. Additive cardioprotective interventions are advocated. For clinical translation into treatments for patients with acute myocardial infarction, who typically are of advanced age, have comorbidities and are receiving several medications, not only infarct size reduction but also attenuation of coronary microvascular obstruction, as well as longer-term targets including infarct repair and reverse remodelling, must be considered to improve patient outcomes. Future clinical trials must focus on patients who really need adjunct cardioprotection, that is, those with severe haemodynamic alterations.
Collapse
|
27
|
Feliu C, Peyret H, Vautier D, Djerada Z. Simultaneous quantification of 8 nucleotides and adenosine in cells and their medium using UHPLC-HRMS. J Chromatogr B Analyt Technol Biomed Life Sci 2020; 1148:122156. [PMID: 32446186 DOI: 10.1016/j.jchromb.2020.122156] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2020] [Revised: 05/10/2020] [Accepted: 05/11/2020] [Indexed: 10/24/2022]
Abstract
Purinergic signalling is involved in physiological processes, particularly during ischemia-reperfusion injuries for which it has a protective effect. The purpose of this work was to develop a method for simultaneous quantification of eight nucleotides and adenosine in biological matrices by liquid chromatography coupled with high-resolution mass spectrometry. A method was developed that was sufficiently robust to quantify the targeted analytes in 20 min with good sensitivity. Analysis of extracellular media from cultured endothelial cells detected the release of nucleotides and adenosine during 2 h of hypoxia. The quantification of cylic adenosine monophosphate (cAMP) allowed to establish a dose-response curve after receptor stimulation. Therefore, our method allows us to study the involvement of nucleotides in various processes in both the intracellular and extracellular compartment.
Collapse
Affiliation(s)
- Catherine Feliu
- Department of Pharmacology, E.A.3801, SFR CAP-santé, Reims University Hospital, 51, rue Cognacq-Jay, 51095 Reims Cedex, France
| | - Hélène Peyret
- Department of Pharmacology, E.A.3801, SFR CAP-santé, Reims University Hospital, 51, rue Cognacq-Jay, 51095 Reims Cedex, France
| | - Damien Vautier
- Department of Pharmacology, E.A.3801, SFR CAP-santé, Reims University Hospital, 51, rue Cognacq-Jay, 51095 Reims Cedex, France
| | - Zoubir Djerada
- Department of Pharmacology, E.A.3801, SFR CAP-santé, Reims University Hospital, 51, rue Cognacq-Jay, 51095 Reims Cedex, France.
| |
Collapse
|
28
|
Adenosine and the Cardiovascular System: The Good and the Bad. J Clin Med 2020; 9:jcm9051366. [PMID: 32384746 PMCID: PMC7290927 DOI: 10.3390/jcm9051366] [Citation(s) in RCA: 56] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2020] [Revised: 04/27/2020] [Accepted: 04/29/2020] [Indexed: 12/18/2022] Open
Abstract
Adenosine is a nucleoside that impacts the cardiovascular system via the activation of its membrane receptors, named A1R, A2AR, A2BR and A3R. Adenosine is released during hypoxia, ischemia, beta-adrenergic stimulation or inflammation and impacts heart rhythm and produces strong vasodilation in the systemic, coronary or pulmonary vascular system. This review summarizes the main role of adenosine on the cardiovascular system in several diseases and conditions. Adenosine release participates directly in the pathophysiology of atrial fibrillation and neurohumoral syncope. Adenosine has a key role in the adaptive response in pulmonary hypertension and heart failure, with the most relevant effects being slowing of heart rhythm, coronary vasodilation and decreasing blood pressure. In other conditions, such as altitude or apnea-induced hypoxia, obstructive sleep apnea, or systemic hypertension, the adenosinergic system activation appears in a context of an adaptive response. Due to its short half-life, adenosine allows very rapid adaptation of the cardiovascular system. Finally, the effects of adenosine on the cardiovascular system are sometimes beneficial and other times harmful. Future research should aim to develop modulating agents of adenosine receptors to slow down or conversely amplify the adenosinergic response according to the occurrence of different pathologic conditions.
Collapse
|
29
|
Diez ER, Sánchez JA, Prado NJ, Ponce Zumino AZ, García-Dorado D, Miatello RM, Rodríguez-Sinovas A. Ischemic Postconditioning Reduces Reperfusion Arrhythmias by Adenosine Receptors and Protein Kinase C Activation but Is Independent of K ATP Channels or Connexin 43. Int J Mol Sci 2019; 20:E5927. [PMID: 31775376 PMCID: PMC6928819 DOI: 10.3390/ijms20235927] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2019] [Revised: 11/20/2019] [Accepted: 11/22/2019] [Indexed: 12/18/2022] Open
Abstract
Ischemic postconditioning (IPoC) reduces reperfusion arrhythmias but the antiarrhythmic mechanisms remain unknown. The aim of this study was to analyze IPoC electrophysiological effects and the role played by adenosine A1, A2A and A3 receptors, protein kinase C, ATP-dependent potassium (KATP) channels, and connexin 43. IPoC reduced reperfusion arrhythmias (mainly sustained ventricular fibrillation) in isolated rat hearts, an effect associated with a transient delay in epicardial electrical activation, and with action potential shortening. Electrical impedance measurements and Lucifer-Yellow diffusion assays agreed with such activation delay. However, this delay persisted during IPoC in isolated mouse hearts in which connexin 43 was replaced by connexin 32 and in mice with conditional deletion of connexin 43. Adenosine A1, A2A and A3 receptor blockade antagonized the antiarrhythmic effect of IPoC and the associated action potential shortening, whereas exogenous adenosine reduced reperfusion arrhythmias and shortened action potential duration. Protein kinase C inhibition by chelerythrine abolished the protective effect of IPoC but did not modify the effects on action potential duration. On the other hand, glibenclamide, a KATP inhibitor, antagonized the action potential shortening but did not interfere with the antiarrhythmic effect. The antiarrhythmic mechanisms of IPoC involve adenosine receptor activation and are associated with action potential shortening. However, this action potential shortening is not essential for protection, as it persisted during protein kinase C inhibition, a maneuver that abolished IPoC protection. Furthermore, glibenclamide induced the opposite effects. In addition, IPoC delays electrical activation and electrical impedance recovery during reperfusion, but these effects are independent of connexin 43.
Collapse
Affiliation(s)
- Emiliano Raúl Diez
- Facultad de Ciencias Médicas, Universidad Nacional de Cuyo, Mendoza 5500, Argentina; (E.R.D.); (N.J.P.); (A.Z.P.Z.); (R.M.M.)
- Institute of Medical and Experimental Biology of Cuyo, CONICET, Mendoza 5500, Argentina
| | - Jose Antonio Sánchez
- Cardiovascular Diseases Research Group, Department of Cardiology, Vall d’Hebron University Hospital and Research Institute, Universitat Autònoma de Barcelona, Departament de Medicina, Pg. Vall d’Hebron 119-129, 08035 Barcelona, Spain; (J.A.S.); (D.G.-D.)
- Centro de Investigación Biomédica en Red sobre Enfermedades Cardiovasculares (CIBERCV), 28029 Madrid, Spain
| | - Natalia Jorgelina Prado
- Facultad de Ciencias Médicas, Universidad Nacional de Cuyo, Mendoza 5500, Argentina; (E.R.D.); (N.J.P.); (A.Z.P.Z.); (R.M.M.)
- Institute of Medical and Experimental Biology of Cuyo, CONICET, Mendoza 5500, Argentina
| | - Amira Zulma Ponce Zumino
- Facultad de Ciencias Médicas, Universidad Nacional de Cuyo, Mendoza 5500, Argentina; (E.R.D.); (N.J.P.); (A.Z.P.Z.); (R.M.M.)
- Institute of Medical and Experimental Biology of Cuyo, CONICET, Mendoza 5500, Argentina
| | - David García-Dorado
- Cardiovascular Diseases Research Group, Department of Cardiology, Vall d’Hebron University Hospital and Research Institute, Universitat Autònoma de Barcelona, Departament de Medicina, Pg. Vall d’Hebron 119-129, 08035 Barcelona, Spain; (J.A.S.); (D.G.-D.)
- Centro de Investigación Biomédica en Red sobre Enfermedades Cardiovasculares (CIBERCV), 28029 Madrid, Spain
| | - Roberto Miguel Miatello
- Facultad de Ciencias Médicas, Universidad Nacional de Cuyo, Mendoza 5500, Argentina; (E.R.D.); (N.J.P.); (A.Z.P.Z.); (R.M.M.)
- Institute of Medical and Experimental Biology of Cuyo, CONICET, Mendoza 5500, Argentina
| | - Antonio Rodríguez-Sinovas
- Cardiovascular Diseases Research Group, Department of Cardiology, Vall d’Hebron University Hospital and Research Institute, Universitat Autònoma de Barcelona, Departament de Medicina, Pg. Vall d’Hebron 119-129, 08035 Barcelona, Spain; (J.A.S.); (D.G.-D.)
- Centro de Investigación Biomédica en Red sobre Enfermedades Cardiovasculares (CIBERCV), 28029 Madrid, Spain
| |
Collapse
|
30
|
Gorain B, Choudhury H, Yee GS, Bhattamisra SK. Adenosine Receptors as Novel Targets for the Treatment of Various Cancers. Curr Pharm Des 2019; 25:2828-2841. [DOI: 10.2174/1381612825666190716102037] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2019] [Accepted: 07/01/2019] [Indexed: 01/28/2023]
Abstract
Adenosine is a ubiquitous signaling nucleoside molecule, released from different cells within the body
to act on vasculature and immunoescape. The physiological action on the proliferation of tumour cell has been
reported by the presence of high concentration of adenosine within the tumour microenvironment, which results
in the progression of the tumour, even leading to metastases. The activity of adenosine exclusively depends upon
the interaction with four subtypes of heterodimeric G-protein-coupled adenosine receptors (AR), A1, A2A, A2B,
and A3-ARs on the cell surface. Research evidence supports that the activation of those receptors via specific
agonist or antagonist can modulate the proliferation of tumour cells. The first category of AR, A1 is known to play
an antitumour activity via tumour-associated microglial cells to prevent the development of glioblastomas.
A2AAR are found in melanoma, lung, and breast cancer cells, where tumour proliferation is stimulated due to
inhibition of the immune response via inhibition of natural killer cells cytotoxicity, T cell activity, and tumourspecific
CD4+/CD8+ activity. Alternatively, A2BAR helps in the development of tumour upon activation via
upregulation of angiogenin factor in the microvascular endothelial cells, inhibition of MAPK and ERK 1/2 phosphorylation
activity. Lastly, A3AR is expressed in low levels in normal cells whereas the expression is upregulated
in tumour cells, however, agonists to this receptor inhibit tumour proliferation through modulation of Wnt
and NF-κB signaling pathways. Several researchers are in search for potential agents to modulate the overexpressed
ARs to control cancer. Active components of A2AAR antagonists and A3AR agonists have already entered
in Phase-I clinical research to prove their safety in human. This review focused on novel research targets towards
the prevention of cancer progression through stimulation of the overexpressed ARs with the hope to protect lives
and advance human health.
Collapse
Affiliation(s)
- Bapi Gorain
- School of Pharmacy, Faculty of Health and Medical Science, Taylor’s University, Subang Jaya, Selangor, Malaysia
| | - Hira Choudhury
- Department of Pharmaceutical Technology, School of Pharmacy, International Medical University, Bukit Jalil 57000, Kuala Lumpur, Malaysia
| | - Gan Sook Yee
- Department of Life Sciences, School of Pharmacy, International Medical University, Bukit Jalil 57000, Kuala Lumpur, Malaysia
| | - Subrat Kumar Bhattamisra
- Department of Life Sciences, School of Pharmacy, International Medical University, Bukit Jalil 57000, Kuala Lumpur, Malaysia
| |
Collapse
|
31
|
Niccoli G, Montone RA, Ibanez B, Thiele H, Crea F, Heusch G, Bulluck H, Hausenloy DJ, Berry C, Stiermaier T, Camici PG, Eitel I. Optimized Treatment of ST-Elevation Myocardial Infarction. Circ Res 2019; 125:245-258. [PMID: 31268854 DOI: 10.1161/circresaha.119.315344] [Citation(s) in RCA: 132] [Impact Index Per Article: 26.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Primary percutaneous coronary intervention is nowadays the preferred reperfusion strategy for patients with acute ST-segment-elevation myocardial infarction, aiming at restoring epicardial infarct-related artery patency and achieving microvascular reperfusion as early as possible, thus limiting the extent of irreversibly injured myocardium. Yet, in a sizeable proportion of patients, primary percutaneous coronary intervention does not achieve effective myocardial reperfusion due to the occurrence of coronary microvascular obstruction (MVO). The amount of infarcted myocardium, the so-called infarct size, has long been known to be an independent predictor for major adverse cardiovascular events and adverse left ventricular remodeling after myocardial infarction. Previous cardioprotection studies were mainly aimed at protecting cardiomyocytes and reducing infarct size. However, several clinical and preclinical studies have reported that the presence and extent of MVO represent another important independent predictor of adverse left ventricular remodeling, and recent evidences support the notion that MVO may be more predictive of major adverse cardiovascular events than infarct size itself. Although timely and complete reperfusion is the most effective way of limiting myocardial injury and subsequent ventricular remodeling, the translation of effective therapeutic strategies into improved clinical outcomes has been largely disappointing. Of importance, despite the presence of a large number of studies focused on infarct size, only few cardioprotection studies addressed MVO as a therapeutic target. In this review, we provide a detailed summary of MVO including underlying causes, diagnostic techniques, and current therapeutic approaches. Furthermore, we discuss the hypothesis that simultaneously addressing infarct size and MVO may help to translate cardioprotective strategies into improved clinical outcome following ST-segment-elevation myocardial infarction.
Collapse
Affiliation(s)
- Giampaolo Niccoli
- From the Department of Cardiovascular and Thoracic Sciences, Fondazione Policlinico Universitario A. Gemelli IRCCS, Rome, Italy (G.N., R.A.M., F.C.).,Institute of Cardiology, Catholic University of the Sacred Heart, Rome, Italy (G.N., F.C.)
| | - Rocco A Montone
- From the Department of Cardiovascular and Thoracic Sciences, Fondazione Policlinico Universitario A. Gemelli IRCCS, Rome, Italy (G.N., R.A.M., F.C.)
| | - Borja Ibanez
- Centro Nacional de Investigaciones Cardiovasculares Carlos III (CNIC), Madrid, Spain (B.I.).,Cardiology Department, IIS-Fundación Jiménez Díaz University Hospital, Madrid, Spain(B.I.).,CIBER de enfermedades CardioVasculares (CIBERCV), Madrid, Spain (B.I.)
| | - Holger Thiele
- Department of Internal Medicine/Cardiology, Heart Center Leipzig at University of Leipzig and Leipzig Heart Institute, Germany (H.T.)
| | - Filippo Crea
- From the Department of Cardiovascular and Thoracic Sciences, Fondazione Policlinico Universitario A. Gemelli IRCCS, Rome, Italy (G.N., R.A.M., F.C.).,Institute of Cardiology, Catholic University of the Sacred Heart, Rome, Italy (G.N., F.C.)
| | - Gerd Heusch
- Institute for Pathophysiology, West German Heart and Vascular Center, University of Essen Medical School, Germany (G.H.)
| | - Heerajnarain Bulluck
- The Hatter Cardiovascular Institute, Institute of Cardiovascular Science, University College London, United Kingdom (H.B., D.J.H.)
| | - Derek J Hausenloy
- The Hatter Cardiovascular Institute, Institute of Cardiovascular Science, University College London, United Kingdom (H.B., D.J.H.).,Cardiovascular & Metabolic Disorders Program, Duke-National University of Singapore Medical School (D.J.H.).,National Heart Research Institute Singapore, National Heart Centre (D.J.H.).,Yong Loo Lin School of Medicine, National University Singapore (D.J.H.).,The Hatter Cardiovascular Institute, University College London, United Kingdom (D.J.H.).,The National Institute of Health Research University College London Hospitals Biomedical Research Centre, Research and Development, United Kingdom (D.J.H.).,Department of Cardiology, Tecnologico de Monterrey, Centro de Biotecnologia-FEMSA, Nuevo Leon, Mexico (D.J.H.)
| | - Colin Berry
- West of Scotland Heart and Lung Centre, Golden Jubilee National Hospital, United Kingdom (C.B.).,British Heart Foundation Glasgow Cardiovascular Research Centre, Institute of Cardiovascular and Medical Sciences, University of Glasgow, United Kingdom (C.B.)
| | - Thomas Stiermaier
- University Heart Center Lübeck, Medical Clinic II (Cardiology/Angiology/Intensive Care Medicine) and German Center for Cardiovascular Research (DZHK), partner site Hamburg/Kiel/Lübeck, Germany (T.S., I.E.)
| | - Paolo G Camici
- Vita-Salute University and San Raffaele Hospital, Milan, Italy (P.G.C.)
| | - Ingo Eitel
- University Heart Center Lübeck, Medical Clinic II (Cardiology/Angiology/Intensive Care Medicine) and German Center for Cardiovascular Research (DZHK), partner site Hamburg/Kiel/Lübeck, Germany (T.S., I.E.)
| |
Collapse
|
32
|
Jackson EK, Mi Z, Janesko-Feldman K, Jackson TC, Kochanek PM. 2',3'-cGMP exists in vivo and comprises a 2',3'-cGMP-guanosine pathway. Am J Physiol Regul Integr Comp Physiol 2019; 316:R783-R790. [PMID: 30789788 PMCID: PMC6620655 DOI: 10.1152/ajpregu.00401.2018] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2018] [Revised: 02/11/2019] [Accepted: 02/19/2019] [Indexed: 01/10/2023]
Abstract
The discovery in 2009 that 2',3'-cAMP exists in biological systems was rapidly followed by identification of 2',3'-cGMP in cell and tissue extracts. To determine whether 2',3'-cGMP exists in mammals under physiological conditions, we used ultraperformance LC-MS/MS to measure 2',3'-cAMP and 2',3'-cGMP in timed urine collections (via direct bladder cannulation) from 25 anesthetized mice. Urinary excretion rates (means ± SE) of 2',3'-cAMP (15.5 ± 1.8 ng/30 min) and 2',3'-cGMP (17.9 ± 1.9 ng/30 min) were similar. Mice also excreted 2'-AMP (3.6 ± 1.1 ng/20 min) and 3'-AMP (9.5 ± 1.2 ng/min), hydrolysis products of 2',3'-cAMP, and 2'-GMP (4.7 ± 1.7 ng/30 min) and 3'-GMP (12.5 ± 1.8 ng/30 min), hydrolysis products of 2',3'-cGMP. To validate that the chromatographic signals were from these endogenous noncanonical nucleotides, we repeated these experiments in mice (n = 18) lacking 2',3'-cyclic nucleotide 3'-phosphodiesterase (CNPase), an enzyme known to convert 2',3'-cyclic nucleotides to their corresponding 2'-nucleotides. In CNPase-knockout mice, urinary excretions of 2',3'-cAMP, 3'-AMP, 2',3'-cGMP, and 3'-GMP were increased, while urinary excretions of 2'-AMP and 2'-GMP were decreased. Infusions of exogenous 2',3'-cAMP increased urinary excretion of 2',3'-cAMP, 2'-AMP, 3'-AMP, and adenosine, whereas infusions of exogenous 2',3'-cGMP increased excretion of 2',3'-cGMP, 2'-GMP, 3'-GMP, and guanosine. Together, these data suggest the endogenous existence of not only a 2',3'-cAMP-adenosine pathway (2',3'-cAMP → 2'-AMP/3'-AMP → adenosine), which was previously identified, but also a 2',3'-cGMP-guanosine pathway (2',3'-cGMP → 2'-GMP/3'-GMP → guanosine), observed here for the first time. Because it is well known that adenosine and guanosine protect tissues from injury, our data support the concept that both pathways may work together to protect tissues from injury.
Collapse
Affiliation(s)
- Edwin K Jackson
- Department of Pharmacology and Chemical Biology, University of Pittsburgh School of Medicine , Pittsburgh, Pennsylvania
| | - Zaichuan Mi
- Department of Pharmacology and Chemical Biology, University of Pittsburgh School of Medicine , Pittsburgh, Pennsylvania
| | - Keri Janesko-Feldman
- Safar Center for Resuscitation Research, University of Pittsburgh School of Medicine , Pittsburgh, Pennsylvania
- Department of Critical Care Medicine, University of Pittsburgh School of Medicine , Pittsburgh, Pennsylvania
| | - Travis C Jackson
- Safar Center for Resuscitation Research, University of Pittsburgh School of Medicine , Pittsburgh, Pennsylvania
- Department of Critical Care Medicine, University of Pittsburgh School of Medicine , Pittsburgh, Pennsylvania
| | - Patrick M Kochanek
- Safar Center for Resuscitation Research, University of Pittsburgh School of Medicine , Pittsburgh, Pennsylvania
- Department of Critical Care Medicine, University of Pittsburgh School of Medicine , Pittsburgh, Pennsylvania
| |
Collapse
|
33
|
Feliu C, Peyret H, Poitevin G, Cazaubon Y, Oszust F, Nguyen P, Millart H, Djerada Z. Complementary Role of P2 and Adenosine Receptors in ATP Induced-Anti-Apoptotic Effects Against Hypoxic Injury of HUVECs. Int J Mol Sci 2019; 20:ijms20061446. [PMID: 30909368 PMCID: PMC6470483 DOI: 10.3390/ijms20061446] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2019] [Revised: 03/16/2019] [Accepted: 03/20/2019] [Indexed: 01/08/2023] Open
Abstract
BACKGROUND Vascular endothelial injury during ischemia generates apoptotic cell death and precedes apoptosis of underlying tissues. We aimed at studying the role of extracellular adenosine triphosphate (ATP) on endothelial cells protection against hypoxia injury. METHODS In a hypoxic model on endothelial cells, we quantified the extracellular concentration of ATP and adenosine. The expression of mRNA (ectonucleotidases, adenosine, and P2 receptors) was measured. Apoptosis was evaluated by the expression of cleaved caspase 3. The involvement of P2 and adenosine receptors and signaling pathways was investigated using selective inhibitors. RESULTS Hypoxic stress induced a significant increase in extracellular ATP and adenosine. After a 2-h hypoxic injury, an increase of cleaved caspase 3 was observed. ATP anti-apoptotic effect was prevented by suramin, pyridoxalphosphate-6-azophenyl-2',4'-disulfonic acid (PPADS), and CGS15943, as well as by selective A2A, A2B, and A3 receptor antagonists. P2 receptor-mediated anti-apoptotic effect of ATP involved phosphoinositide 3-kinase (PI3K), extracellular signal-regulated kinases (ERK1/2), mitoKATP, and nitric oxide synthase (NOS) pathways whereas adenosine receptor-mediated anti-apoptotic effect involved ERK1/2, protein kinase A (PKA), and NOS. CONCLUSIONS These results suggest a complementary role of P2 and adenosine receptors in ATP-induced protective effects against hypoxia injury of endothelial. This could be considered therapeutic targets to limit the development of ischemic injury of organs such as heart, brain, and kidney.
Collapse
Affiliation(s)
- Catherine Feliu
- Department of Pharmacology, E.A.3801, SFR CAP-santé, Reims University Hospital, 51, rue Cognacq-Jay, 51095 Reims CEDEX, France.
| | - Hélène Peyret
- Department of Pharmacology, E.A.3801, SFR CAP-santé, Reims University Hospital, 51, rue Cognacq-Jay, 51095 Reims CEDEX, France.
| | - Gael Poitevin
- Laboratory of Hematology, E.A.3801, SFR CAP-santé, Reims University Hospital, 51, rue Cognacq-Jay, 51095 Reims CEDEX, France.
| | - Yoann Cazaubon
- Department of Pharmacology, E.A.3801, SFR CAP-santé, Reims University Hospital, 51, rue Cognacq-Jay, 51095 Reims CEDEX, France.
| | - Floriane Oszust
- Department of Pharmacology, E.A.3801, SFR CAP-santé, Reims University Hospital, 51, rue Cognacq-Jay, 51095 Reims CEDEX, France.
| | - Philippe Nguyen
- Laboratory of Hematology, E.A.3801, SFR CAP-santé, Reims University Hospital, 51, rue Cognacq-Jay, 51095 Reims CEDEX, France.
| | - Hervé Millart
- Department of Pharmacology, E.A.3801, SFR CAP-santé, Reims University Hospital, 51, rue Cognacq-Jay, 51095 Reims CEDEX, France.
| | - Zoubir Djerada
- Department of Pharmacology, E.A.3801, SFR CAP-santé, Reims University Hospital, 51, rue Cognacq-Jay, 51095 Reims CEDEX, France.
| |
Collapse
|
34
|
Nakamura TY, Nakao S, Wakabayashi S. Emerging Roles of Neuronal Ca 2+ Sensor-1 in Cardiac and Neuronal Tissues: A Mini Review. Front Mol Neurosci 2019; 12:56. [PMID: 30886571 PMCID: PMC6409499 DOI: 10.3389/fnmol.2019.00056] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2018] [Accepted: 02/15/2019] [Indexed: 11/13/2022] Open
Abstract
The EF-hand calcium (Ca2+)-binding protein, neuronal Ca2+ sensor-1 (NCS-1/frequenin), is predominantly expressed in neuronal tissues and plays a crucial role in neuronal functions, including synaptic transmission and plasticity. NCS-1 has diverse functional roles, as elucidated in the past 15 years, which include the regulation of phosphatidylinositol 4-kinase IIIβ (PI-4K-β) and several ion channels such as voltage-gated K+ and Ca2+ channels, the D2 dopamine receptors, and inositol 1,4,5-trisphosphate receptors (InsP3Rs). Functional analyses demonstrated that NCS-1 enhances exocytosis and neuronal survival after injury, as well as promotes learning and memory in mice. NCS-1 is also expressed in the heart including the Purkinje fibers (PFs) of the conduction system. NCS-1 interacts with KV4 K+ channels together with dipeptidyl peptidase-like protein-6 (DPP-6), and this macromolecule then composes the transient outward current in PFs and contributes to the repolarization of PF action potential, thus being responsible for idiopathic arrhythmia. Moreover, NCS-1 expression was reported to be significantly high at the immature stage and at hypertrophy in adults. That report demonstrated that NCS-1 positively regulates cardiac contraction in immature hearts by increasing intracellular Ca2+ signals through interaction with InsP3Rs. With the related signals, NCS-1 activates nuclear Ca2+ signals, which would be a mechanism underlying hormone-induced cardiac hypertrophy. Furthermore, NCS-1 contributes to stress tolerance in cardiomyocytes by activating mitochondrial detoxification pathways, with a key role in Ca2+-dependent pathways. In this review, we will discuss recent findings supporting the functional significance of NCS-1 in the brain and heart and will address possible underlying molecular mechanisms.
Collapse
Affiliation(s)
- Tomoe Y Nakamura
- Department of Molecular Physiology, National Cerebral and Cardiovascular Center Research Institute, Suita, Japan
| | - Shu Nakao
- Department of Molecular Physiology, National Cerebral and Cardiovascular Center Research Institute, Suita, Japan.,Department of Biomedical Sciences, College of Life Sciences, Ritsumeikan University, Kusatsu, Japan
| | - Shigeo Wakabayashi
- Department of Molecular Physiology, National Cerebral and Cardiovascular Center Research Institute, Suita, Japan.,Department of Pharmacology, Osaka Medical College, Takatsuki, Japan
| |
Collapse
|
35
|
CD39 and CD73 in the aortic valve-biochemical and immunohistochemical analysis in valve cell populations and its changes in valve mineralization. Cardiovasc Pathol 2018; 36:53-63. [PMID: 30056298 DOI: 10.1016/j.carpath.2018.05.008] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/06/2018] [Revised: 04/21/2018] [Accepted: 05/30/2018] [Indexed: 01/09/2023] Open
Abstract
BACKGROUND The calcific aortic valve disease (CAVD) is a common heart pathology that involves inflammation, fibrosis, and calcification of aortic valve leaflets. All these processes could be affected by changes in the extracellular purinergic signaling that depend on the activity of ectonucleotidases, mainly ectonucleoside triphosphate diphosphohydrolase 1 (CD39, eNTPD1) and ecto-5'nucleotidase (CD73, e5NT). OBJECTIVE AND METHODS We investigated the localization of CD39 and CD73 proteins in human noncalcified and calcified aortic valves using immunohistochemistry together with analysis of NTPDases and e5NT activities in aortic valve homogenates by analysis of substrate into product conversion by high-performance liquid chromatography. We also measured the rates of extracellular nucleotide catabolism on the surface of isolated cultured aortic valve endothelial (hAVECs) and interstitial cells (hAVICs) as well as characterized cellular CD39 and CD73 distribution. RESULTS In noncalcified valves, CD39 and CD73 were expressed in both endothelial and interstitial cells, while in calcified valves, the expressions of CD39 and CD73 were significantly down-regulated with the exception of calcified regions where the expression of CD73 was maintained. This correlated with activities in valve homogenates. NTPDase was reduced by 35% and e5NT activity by 50% in calcified vs. noncalcified valve. CD39 and CD73 were present mainly in the cell membrane of hAVECs, but in hAVICs, these proteins were also present intracellularly. The rates of extracellular adenosine triphosphate and adenosine monophosphate hydrolysis in isolated hAVECs and hAVICs were comparable. CONCLUSION The presence of ectonucleotidases in valves and especially in aortic valve interstitial cells highlights important local role of purinergic signaling and metabolism. Changes in the local expression and hence the activity of CD39 and CD73 in calcified valves suggest their potential role in CAVD.
Collapse
|
36
|
Yeung PK, Kolathuru SS, Mohammadizadeh S, Akhoundi F, Linderfield B. Adenosine 5'-Triphosphate Metabolism in Red Blood Cells as a Potential Biomarker for Post-Exercise Hypotension and a Drug Target for Cardiovascular Protection. Metabolites 2018; 8:metabo8020030. [PMID: 29724022 PMCID: PMC6027528 DOI: 10.3390/metabo8020030] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2018] [Revised: 04/27/2018] [Accepted: 04/29/2018] [Indexed: 12/22/2022] Open
Abstract
The importance of adenosine and ATP in regulating many biological functions has long been recognized, especially for their effects on the cardiovascular system, which may be used for management of hypertension and cardiometabolic diseases. In response to ischemia and cardiovascular injury, ATP is broken down to release adenosine. The effect of adenosine is very short lived because it is rapidly taken up by erythrocytes (RBCs), myocardial and endothelial cells, and also rapidly catabolized to oxypurine metabolites. Intracellular adenosine is phosphorylated back to adenine nucleotides via a salvage pathway. Extracellular and intracellular ATP is broken down rapidly to ADP and AMP, and finally to adenosine by 5′-nucleotidase. These metabolic events are known to occur in the myocardium, endothelium as well as in RBCs. Exercise has been shown to increase metabolism of ATP in RBCs, which may be an important mechanism for post-exercise hypotension and cardiovascular protection. The post-exercise effect was greater in hypertensive than in normotensive rats. The review summarizes current evidence in support of ATP metabolism in the RBC as a potential surrogate biomarker for cardiovascular protection and toxicities. It also discusses the opportunities, challenges, and obstacles of exploiting ATP metabolism in RBCs as a target for drug development and precision medicine.
Collapse
Affiliation(s)
- Pollen K Yeung
- Pharmacokinetics and Metabolism Laboratory, College of Pharmacy and Department of Medicine, Dalhousie University, Halifax, NS B3H 4R2, Canada.
| | - Shyam Sundar Kolathuru
- Pharmacokinetics and Metabolism Laboratory, College of Pharmacy and Department of Medicine, Dalhousie University, Halifax, NS B3H 4R2, Canada.
| | - Sheyda Mohammadizadeh
- Pharmacokinetics and Metabolism Laboratory, College of Pharmacy and Department of Medicine, Dalhousie University, Halifax, NS B3H 4R2, Canada.
| | - Fatemeh Akhoundi
- Pharmacokinetics and Metabolism Laboratory, College of Pharmacy and Department of Medicine, Dalhousie University, Halifax, NS B3H 4R2, Canada.
| | - Brett Linderfield
- Pharmacokinetics and Metabolism Laboratory, College of Pharmacy and Department of Medicine, Dalhousie University, Halifax, NS B3H 4R2, Canada.
| |
Collapse
|
37
|
Gao ZG, Inoue A, Jacobson KA. On the G protein-coupling selectivity of the native A 2B adenosine receptor. Biochem Pharmacol 2018; 151:201-213. [PMID: 29225130 PMCID: PMC5899946 DOI: 10.1016/j.bcp.2017.12.003] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2017] [Accepted: 12/05/2017] [Indexed: 12/14/2022]
Abstract
A2B adenosine receptor (A2BAR) activation induces Gs-dependent cyclic AMP accumulation. However, A2BAR G protein-coupling to other signaling events, e.g. ERK1/2 and calcium, is not well documented. We explored Gi, Gq/11 and Gs coupling in 1321 N1 astrocytoma, HEK293, and T24 bladder cancer cells endogenously expressing human A2BAR, using NECA or nonnucleoside BAY60-6583 as agonist, selective Gi, Gs and Gq/11 blockers, and CRISPR/Cas9-based Gq- and Gs-null HEK293 cells. In HEK293 cells, A2BAR-mediated ERK1/2 activity occurred via both Gi and Gs, but not Gq/11. However, HEK293 cell calcium mobilization was completely blocked by Gq/11 inhibitor UBO-QIC and by Gq/11 knockout. In T24 cells, Gi was solely responsible for A2BAR-mediated ERK1/2 stimulation, and Gs suppressed ERK1/2 activity. A2BAR-mediated intracellular calcium mobilization in T24 cells was mainly via Gi, although Gs may also play a role, but Gq/11 is not involved. In 1321 N1 astrocytoma cells A2BAR activation suppressed rather than stimulated ERK1/2 activity. The ERK1/2 activity decrease was reversed by Gs downregulation using cholera toxin, but potentiated by Gi inhibitor pertussis toxin, and UBO-QIC had no effect. EPACs played an important role in A2BAR-mediated ERK1/2 signaling in all three cells. Thus, A2BAR may: couple to the same downstream pathway via different G proteins in different cell types; activate different downstream events via different G proteins in the same cell type; activate Gi and Gs, which have opposing or synergistic roles in different cell types/signaling pathways. The findings, relevant to drug discovery, address some reported controversial roles of A2BAR and could apply to signaling mechanisms in other GPCRs.
Collapse
Affiliation(s)
- Zhan-Guo Gao
- Molecular Recognition Section, Laboratory of Bioorganic Chemistry, NIDDK, National Institutes of Health, 9000 Rockville Pike, Bethesda, MD 20892, USA.
| | - Asuka Inoue
- Graduate School of Pharmaceutical Sciences, Tohoku University, Sendai, Miyagi 980-8578, Japan
| | - Kenneth A Jacobson
- Molecular Recognition Section, Laboratory of Bioorganic Chemistry, NIDDK, National Institutes of Health, 9000 Rockville Pike, Bethesda, MD 20892, USA.
| |
Collapse
|
38
|
Almohanna AM, Wray S. Hypoxic conditioning in blood vessels and smooth muscle tissues: effects on function, mechanisms, and unknowns. Am J Physiol Heart Circ Physiol 2018; 315:H756-H770. [PMID: 29702009 DOI: 10.1152/ajpheart.00725.2017] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Hypoxic preconditioning, the protective effect of brief, intermittent hypoxic or ischemic episodes on subsequent more severe hypoxic episodes, has been known for 30 yr from studies on cardiac muscle. The concept of hypoxic preconditioning has expanded; excitingly, organs beyond the heart, including the brain, liver, and kidney, also benefit. Preconditioning of vascular and visceral smooth muscles has received less attention despite their obvious importance to health. In addition, there has been no attempt to synthesize the literature in this field. Therefore, in addition to overviewing the current understanding of hypoxic conditioning, in the present review, we consider the role of blood vessels in conditioning and explore evidence for conditioning in other smooth muscles. Where possible, we have distinguished effects on myocytes from other cell types in the visceral organs. We found evidence of a pivotal role for blood vessels in conditioning and for conditioning in other smooth muscle, including the bladder, vascular myocytes, and gastrointestinal tract, and a novel response in the uterus of a hypoxic-induced force increase, which helps maintain contractions during labor. To date, however, there are insufficient data to provide a comprehensive or unifying mechanism for smooth muscles or visceral organs and the effects of conditioning on their function. This also means that no firm conclusions can be drawn as to how differences between smooth muscles in metabolic and contractile activity may contribute to conditioning. Therefore, we have suggested what may be general mechanisms of conditioning occurring in all smooth muscles and tabulated tissue-specific mechanistic findings and suggested ideas for further progress.
Collapse
Affiliation(s)
- Asmaa M Almohanna
- Department of Molecular and Cellular Physiology, Institute of Translational Medicine University of Liverpool , Liverpool , United Kingdom.,Princess Nourah bint Abdulrahman University , Riyadh , Saudi Arabia
| | - Susan Wray
- Department of Molecular and Cellular Physiology, Institute of Translational Medicine University of Liverpool , Liverpool , United Kingdom
| |
Collapse
|
39
|
Lambertucci C, Marucci G, Dal Ben D, Buccioni M, Spinaci A, Kachler S, Klotz KN, Volpini R. New potent and selective A 1 adenosine receptor antagonists as potential tools for the treatment of gastrointestinal diseases. Eur J Med Chem 2018; 151:199-213. [PMID: 29614417 DOI: 10.1016/j.ejmech.2018.03.067] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2018] [Revised: 03/21/2018] [Accepted: 03/22/2018] [Indexed: 01/31/2023]
Abstract
The synthesis of 9-alkyl substituted adenine derivatives presenting aromatic groups and cycloalkyl rings in 8- and N6-position, respectively, is reported. The compounds were tested with radioligand binding studies showing, in some cases, a low nanomolar A1 adenosine receptor affinity and a very good selectivity versus the other adenosine receptor subtypes. Functional assays at human adenosine receptors and at a mouse ileum tissue preparation clearly demonstrate the antagonist profile of these molecules, with inhibitory potency at nanomolar level. A molecular modeling study, consisting in docking analysis at the recently reported A1 adenosine receptor crystal structure, was performed for the interpretation of the obtained pharmacological results. The N6-cyclopentyl-9-methyl-8-phenyladenine (17), resulting the most active derivative of the series (Ki = 2.8 nM and IC50 = 14 nM), was also very efficacious in counteracting the effect of the agonist CCPA on mouse ileum contractility. This new compound represents a tool for the development of new agents for the treatment of intestinal diseases as constipation and postoperative ileus.
Collapse
Affiliation(s)
- Catia Lambertucci
- School of Pharmacy, Medicinal Chemistry Unit, University of Camerino, Via S. Agostino 1, 62032 Camerino, MC, Italy
| | - Gabriella Marucci
- School of Pharmacy, Medicinal Chemistry Unit, University of Camerino, Via S. Agostino 1, 62032 Camerino, MC, Italy
| | - Diego Dal Ben
- School of Pharmacy, Medicinal Chemistry Unit, University of Camerino, Via S. Agostino 1, 62032 Camerino, MC, Italy
| | - Michela Buccioni
- School of Pharmacy, Medicinal Chemistry Unit, University of Camerino, Via S. Agostino 1, 62032 Camerino, MC, Italy
| | - Andrea Spinaci
- School of Pharmacy, Medicinal Chemistry Unit, University of Camerino, Via S. Agostino 1, 62032 Camerino, MC, Italy
| | - Sonja Kachler
- Institut für Pharmakologie und Toxikologie, Universität Würzburg, Versbacher Str. 9, 97078 Würzburg, Germany
| | - Karl-Norbert Klotz
- Institut für Pharmakologie und Toxikologie, Universität Würzburg, Versbacher Str. 9, 97078 Würzburg, Germany
| | - Rosaria Volpini
- School of Pharmacy, Medicinal Chemistry Unit, University of Camerino, Via S. Agostino 1, 62032 Camerino, MC, Italy.
| |
Collapse
|
40
|
Jung H. Kidney transplantation and ischemic conditioning: past, present and future perspectives. Anesth Pain Med (Seoul) 2018. [DOI: 10.17085/apm.2018.13.1.10] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
Affiliation(s)
- Hoon Jung
- Department of Anesthesiology and Pain Medicine, School of Medicine, Kyungpook National University, Daegu, Korea
| |
Collapse
|
41
|
Phosri S, Bunrukchai K, Parichatikanond W, Sato VH, Mangmool S. Epac is required for exogenous and endogenous stimulation of adenosine A 2B receptor for inhibition of angiotensin II-induced collagen synthesis and myofibroblast differentiation. Purinergic Signal 2018; 14:141-156. [PMID: 29322373 DOI: 10.1007/s11302-017-9600-5] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2017] [Accepted: 12/25/2017] [Indexed: 12/31/2022] Open
Abstract
Angiotensin II (Ang II) plays an important role on the pathogenesis of cardiac fibrosis. Prolong and overstimulation of angiotensin II type 1 receptor with Ang II-induced collagen synthesis and myofibroblast differentiation in cardiac fibroblasts, leading to cardiac fibrosis. Although adenosine and its analogues are known to have cardioprotective effects, the mechanistic by which adenosine A2 receptors (A2Rs) inhibit Ang II-induced cardiac fibrosis is not clearly understood. In the present study, we examined the effects of exogenous adenosine and endogenous adenosine on Ang II-induced collagen and myofibroblast differentiation determined by α-smooth muscle action (α-SMA) overexpression and their underlying signal transduction. Elevation of endogenous adenosine levels resulted in the inhibition of Ang II-induced collagen type I and III and α-SMA synthesis in cardiac fibroblasts. Moreover, treatment with exogenous adenosine which selectively stimulated A2Rs also suppressed Ang II-induced collagen synthesis and α-SMA production. These antifibrotic effects of both endogenous and exogenous adenosines are mediated through the A2B receptor (A2BR) subtype. Stimulation of A2BR exhibited antifibrotic effects via the cAMP-dependent and Epac-dependent pathways. Our results provide new mechanistic insights regarding the role for cAMP and Epac on A2BR-mediated antifibrotic effects. Thus, A2BR is one of the potential therapeutic targets against cardiac fibrosis.
Collapse
Affiliation(s)
- Sarawuth Phosri
- Department of Pharmacology, Faculty of Pharmacy, Mahidol University, Bangkok, 10400, Thailand
| | - Kwanchai Bunrukchai
- Department of Pharmacology, Faculty of Pharmacy, Mahidol University, Bangkok, 10400, Thailand
| | | | - Vilasinee H Sato
- Department of Pharmacology, Faculty of Pharmacy, Mahidol University, Bangkok, 10400, Thailand
| | - Supachoke Mangmool
- Department of Pharmacology, Faculty of Pharmacy, Mahidol University, Bangkok, 10400, Thailand.
| |
Collapse
|
42
|
Ammar A, Mahmoud K, Elkersh A, Kasemy Z. A Randomized Controlled Trial of Intra-Aortic Adenosine Infusion Before Release of the Aortic Cross-Clamp During Coronary Artery Bypass Surgery. J Cardiothorac Vasc Anesth 2017; 32:2520-2527. [PMID: 29225152 DOI: 10.1053/j.jvca.2017.10.041] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/14/2017] [Indexed: 11/11/2022]
Abstract
OBJECTIVES To assess the feasibility, safety, and potential useful effect of adenosine as a postconditioning agent in patients undergoing coronary artery bypass grafting surgeries. DESIGN Prospective randomized controlled study. SETTING University hospital. PARTICIPANTS The study comprised 60 patients scheduled for coronary artery bypass grafting surgery. INTERVENTIONS Adenosine (postconditioning group) or placebo (control group). Adenosine infusion (150 µg/kg/min) for 10 minutes via a cardioplegia needle into the aortic root was started 10 minutes before aortic cross-clamp removal. MEASUREMENTS AND MAIN RESULTS Compared with the control group, ejection fraction, fractional shortening, cardiac index (2.9 ± 0.3 v 2.2 ± 0.3 L/min/m2, p = 0.032 at 60 min postbypass) and diastolic function indices were significantly better in the postconditioning group at most time points in the postbypass period. Cardiac troponin I and creatine kinase-MB release and the inotropic score were significantly lower in the postconditioning group at most time points in the postoperative period. The need for intra-aortic balloon and epicardial pacing were comparable in both groups, whereas incidence of arrhythmia, duration of postoperative mechanical ventilation, and intensive care unit and total hospital stays were significantly lower in the postconditioning group. CONCLUSIONS Adenosine postconditioning provided cardiac protection as evidenced by a favorable outcome on systolic and diastolic function indices, less cardiac troponin I and creatine kinase-MB release, lower incidence of arrhythmia, lower inotropic score, and shorter duration of postoperative mechanical ventilation and intensive care unit stay.
Collapse
Affiliation(s)
- Amany Ammar
- Faculty of Medicine, Minoufiya University, Minoufiya, Egypt
| | - Khaled Mahmoud
- Faculty of Medicine, Minoufiya University, Minoufiya, Egypt.
| | - Ahmed Elkersh
- Faculty of Medicine, Minoufiya University, Minoufiya, Egypt
| | - Zeinab Kasemy
- Faculty of Medicine, Minoufiya University, Minoufiya, Egypt
| |
Collapse
|
43
|
Tullio F, Penna C, Cabiale K, Femminò S, Galloni M, Pagliaro P. Cardioprotective effects of calcitonin gene-related peptide in isolated rat heart and in H9c2 cells via redox signaling. Biomed Pharmacother 2017; 90:194-202. [PMID: 28364596 DOI: 10.1016/j.biopha.2017.03.043] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2017] [Revised: 03/10/2017] [Accepted: 03/14/2017] [Indexed: 02/02/2023] Open
Abstract
The calcitonin-gene-related-peptide (CGRP) release is coupled to the signaling of Angeli's salt in determining vasodilator effects. However, it is unknown whether CGRP is involved in Angeli's salt cardioprotective effects and which are the mechanisms of protection. We aimed to determine whether CGRP is involved in myocardial protection induced by Angeli's salt. We also analyzed the intracellular signaling pathway activated by CGRP. Isolated rat hearts were pre-treated with Angeli's salt or Angeli's salt plus CGRP8-37, a specific CGRP-receptor antagonist, and subjected to ischemia (30-min) and reperfusion (120-min). Moreover, we studied CGRP-induced protection during oxidative stress (H2O2) and hypoxia/reoxygenation protocols in H9c2 cardiomyocytes. Cell vitality and mitochondrial membrane potential (ΔYm, MMP) were measured using MTT and JC-1 dyes. Angeli's salt reduced infarct size and ameliorated post-ischemic cardiac function via a CGRP-receptor-dependent mechanism. Pre-treatment with CGRP increased H9c2 survival upon challenging with either H2O2 (redox stress) or hypoxia/reoxygenation (H/R stress). Under these stress conditions, reduction in MMP and cell death were partly prevented by CGRP. These CGRP beneficial effects were blocked by CGRP8-37. During H/R stress, pre-treatment with either CGRP-receptor, protein kinase C (PKC) or mitochondrial KATP channel antagonists, and pre-treatment with an antioxidant (2-mercaptopropionylglycine) blocked the protection mediated by CGRP. In conclusion, CGRP is involved in the cardioprotective effects of Angeli's salt. In H9c2 cardiomyocytes, CGRP elicits PKC-dependent and mitochondrial-KATP-redox-dependent mechanisms. Hence, CGRP is an important factor in the redox-sensible cardioprotective effects of Angeli's salt.
Collapse
Affiliation(s)
- Francesca Tullio
- Department of Clinical and Biological Sciences, University of Turin, Italy
| | - Claudia Penna
- Department of Clinical and Biological Sciences, University of Turin, Italy.
| | - Karine Cabiale
- Department of Clinical and Biological Sciences, University of Turin, Italy; Department of Veterinary Science, University of Torino, Italy
| | - Saveria Femminò
- Department of Clinical and Biological Sciences, University of Turin, Italy
| | - Marco Galloni
- Department of Veterinary Science, University of Torino, Italy
| | - Pasquale Pagliaro
- Department of Clinical and Biological Sciences, University of Turin, Italy.
| |
Collapse
|
44
|
Na+/Ca2+ exchanger 1 inhibition abolishes ischemic tolerance induced by ischemic preconditioning in different cardiac models. Eur J Pharmacol 2017; 794:246-256. [DOI: 10.1016/j.ejphar.2016.11.045] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2016] [Revised: 11/22/2016] [Accepted: 11/24/2016] [Indexed: 01/22/2023]
|
45
|
Kang C, Qin J, Osei W, Hu K. Regulation of protein kinase C-epsilon and its age-dependence. Biochem Biophys Res Commun 2016; 482:1201-1206. [PMID: 27919679 DOI: 10.1016/j.bbrc.2016.12.012] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2016] [Accepted: 12/02/2016] [Indexed: 12/20/2022]
Abstract
Protein kinase C (PKC) is an important mediator in the cardioprotection of ischemic preconditioning and has been shown to translocate to mitochondria upon activation. However, little is known about the cellular signaling underlying the translocation of PKC isoforms to mitochondria and its age-dependence. The present study aimed to explore whether adenosine-induced translocation of PKCε to mitochondria is mediated by caveolin-3 and/or adenosine A2B receptor/PI3 kinase mediated signaling, and whether the mitochondrial targeting of PKCε is age-related. Immunofluorescence imaging of isolated mitochondria from cardiomyocytes and H9c2 cells showed that while adenosine-induced increase in mitochondrial PKCε was inhibited by adenosine A1 receptor blocker, pretreatment with adenosine A2B receptor specific inhibitor MRS 1754 or PI3K inhibitor Wortmannin did not significantly reduce adenosine-mediated increase in mitochondrial PKCε. Interestingly, adenosine-induced increase in mitochondrial translocation of PKCε was significantly blocked by suppressing caveolin-3 expression with specific siRNA. When compared to that in young adult rat hearts, the level of mitochondrial PKCε in middle-aged rat hearts was significantly lower at the basal condition and in response to adenosine treatment, along with largely decreased mitochondrial HSP90 and TOM70 protein expression. We demonstrate that adenosine-induced translocation of PKCε to mitochondria is mediated by a caveolin-3-dependent mechanism and this process is age-related, possibly in part, through regulation of HSP90 and TOM70 expression. These results point out a novel mechanism in regulating PKC function in mitochondria.
Collapse
Affiliation(s)
- Chen Kang
- Division of Pharmacology, College of Pharmacy, The Ohio State University, Columbus, OH 43210, USA
| | - Jingping Qin
- Division of Pharmacology, College of Pharmacy, The Ohio State University, Columbus, OH 43210, USA
| | - Wil Osei
- Division of Pharmacology, College of Pharmacy, The Ohio State University, Columbus, OH 43210, USA
| | - Keli Hu
- Division of Pharmacology, College of Pharmacy, The Ohio State University, Columbus, OH 43210, USA.
| |
Collapse
|
46
|
Yetgin T, Uitterdijk A, Te Lintel Hekkert M, Merkus D, Krabbendam-Peters I, van Beusekom HMM, Falotico R, Serruys PW, Manintveld OC, van Geuns RJM, Zijlstra F, Duncker DJ. Limitation of Infarct Size and No-Reflow by Intracoronary Adenosine Depends Critically on Dose and Duration. JACC Cardiovasc Interv 2016; 8:1990-1999. [PMID: 26738671 DOI: 10.1016/j.jcin.2015.08.033] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/20/2015] [Accepted: 08/14/2015] [Indexed: 11/19/2022]
Abstract
OBJECTIVES In the absence of effective clinical pharmacotherapy for prevention of reperfusion-mediated injury, this study re-evaluated the effects of intracoronary adenosine on infarct size and no-reflow in a porcine model of acute myocardial infarction using clinical bolus and experimental high-dose infusion regimens. BACKGROUND Despite the clear cardioprotective effects of adenosine, when administered prior to ischemia, studies on cardioprotection by adenosine when administered at reperfusion have yielded contradictory results in both pre-clinical and clinical settings. METHODS Swine (54 ± 1 kg) were subjected to a 45-min mid-left anterior descending artery occlusion followed by 2 h of reperfusion. In protocol A, an intracoronary bolus of 3 mg adenosine injected over 1 min (n = 5) or saline (n = 10) was administered at reperfusion. In protocol B, an intracoronary infusion of 50 μg/kg/min adenosine (n = 15) or saline (n = 21) was administered starting 5 min prior to reperfusion and continued throughout the 2-h reperfusion period. RESULTS In protocol A, area-at-risk, infarct size, and no-reflow were similar between groups. In protocol B, risk zones were similar, but administration of adenosine resulted in significant reductions in infarct size from 59 ± 3% of the area-at-risk in control swine to 46 ± 4% (p = 0.02), and no-reflow from 49 ± 6% of the infarct area to 26 ± 6% (p = 0.03). CONCLUSIONS During reperfusion, intracoronary adenosine can limit infarct size and no-reflow in a porcine model of acute myocardial infarction. However, protection was only observed when adenosine was administered via prolonged high-dose infusion, and not via short-acting bolus injection. These findings warrant reconsideration of adenosine as an adjuvant therapy during early reperfusion.
Collapse
Affiliation(s)
- Tuncay Yetgin
- Department of Cardiology, Thoraxcenter, Erasmus University Medical Center, Rotterdam, the Netherlands; Interuniversity Cardiology Institute of the Netherlands, Utrecht, the Netherlands
| | - André Uitterdijk
- Department of Cardiology, Thoraxcenter, Erasmus University Medical Center, Rotterdam, the Netherlands
| | - Maaike Te Lintel Hekkert
- Department of Cardiology, Thoraxcenter, Erasmus University Medical Center, Rotterdam, the Netherlands
| | - Daphne Merkus
- Department of Cardiology, Thoraxcenter, Erasmus University Medical Center, Rotterdam, the Netherlands
| | - Ilona Krabbendam-Peters
- Department of Cardiology, Thoraxcenter, Erasmus University Medical Center, Rotterdam, the Netherlands
| | - Heleen M M van Beusekom
- Department of Cardiology, Thoraxcenter, Erasmus University Medical Center, Rotterdam, the Netherlands
| | | | - Patrick W Serruys
- Department of Cardiology, Thoraxcenter, Erasmus University Medical Center, Rotterdam, the Netherlands
| | - Olivier C Manintveld
- Department of Cardiology, Thoraxcenter, Erasmus University Medical Center, Rotterdam, the Netherlands
| | - Robert-Jan M van Geuns
- Department of Cardiology, Thoraxcenter, Erasmus University Medical Center, Rotterdam, the Netherlands
| | - Felix Zijlstra
- Department of Cardiology, Thoraxcenter, Erasmus University Medical Center, Rotterdam, the Netherlands; Interuniversity Cardiology Institute of the Netherlands, Utrecht, the Netherlands
| | - Dirk J Duncker
- Department of Cardiology, Thoraxcenter, Erasmus University Medical Center, Rotterdam, the Netherlands; Interuniversity Cardiology Institute of the Netherlands, Utrecht, the Netherlands.
| |
Collapse
|
47
|
See Hoe LE, May LT, Headrick JP, Peart JN. Sarcolemmal dependence of cardiac protection and stress-resistance: roles in aged or diseased hearts. Br J Pharmacol 2016; 173:2966-91. [PMID: 27439627 DOI: 10.1111/bph.13552] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2015] [Revised: 06/27/2016] [Accepted: 06/28/2016] [Indexed: 12/25/2022] Open
Abstract
Disruption of the sarcolemmal membrane is a defining feature of oncotic death in cardiac ischaemia-reperfusion (I-R), and its molecular makeup not only fundamentally governs this process but also affects multiple determinants of both myocardial I-R injury and responsiveness to cardioprotective stimuli. Beyond the influences of membrane lipids on the cytoprotective (and death) receptors intimately embedded within this bilayer, myocardial ionic homeostasis, substrate metabolism, intercellular communication and electrical conduction are all sensitive to sarcolemmal makeup, and critical to outcomes from I-R. As will be outlined in this review, these crucial sarcolemmal dependencies may underlie not only the negative effects of age and common co-morbidities on myocardial ischaemic tolerance but also the on-going challenge of implementing efficacious cardioprotection in patients suffering accidental or surgically induced I-R. We review evidence for the involvement of sarcolemmal makeup changes in the impairment of stress-resistance and cardioprotection observed with ageing and highly prevalent co-morbid conditions including diabetes and hypercholesterolaemia. A greater understanding of membrane changes with age/disease, and the inter-dependences of ischaemic tolerance and cardioprotection on sarcolemmal makeup, can facilitate the development of strategies to preserve membrane integrity and cell viability, and advance the challenging goal of implementing efficacious 'cardioprotection' in clinically relevant patient cohorts. Linked Articles This article is part of a themed section on Molecular Pharmacology of G Protein-Coupled Receptors. To view the other articles in this section visit http://onlinelibrary.wiley.com/doi/10.1111/bph.v173.20/issuetoc.
Collapse
Affiliation(s)
- Louise E See Hoe
- Menzies Health Institute Queensland, Griffith University, Gold Coast, Australia.,Critical Care Research Group, The Prince Charles Hospital and The University of Queensland, Chermside, Queensland, Australia
| | - Lauren T May
- Monash Institute of Pharmaceutical Sciences, Monash University, Clayton, VIC, Australia
| | - John P Headrick
- Menzies Health Institute Queensland, Griffith University, Gold Coast, Australia
| | - Jason N Peart
- Menzies Health Institute Queensland, Griffith University, Gold Coast, Australia.
| |
Collapse
|
48
|
Lai LN, Zhang XJ, Zhang XY, Song LH, Guo CH, Lei JW, Song XL. Lazaroid U83836E protects the heart against ischemia reperfusion injury via inhibition of oxidative stress and activation of PKC. Mol Med Rep 2016; 13:3993-4000. [PMID: 27035121 DOI: 10.3892/mmr.2016.5030] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2015] [Accepted: 03/07/2016] [Indexed: 11/05/2022] Open
Abstract
Oxidative stress has been demonstrated to be important during myocardial ischemia/reperfusion injury (MIRI). The lazaroid U83836E, which combines the amino functionalities of the 21‑aminosteroids with the antioxidant ring portion of vitamin E, is a reactive oxygen species scavenger. The aim of the current study was to investigate the effect of U83836E on MIRI and its mechanisms of action. Rat hearts were subjected to 30 min ligation of the left anterior descending coronary artery, followed by 2 h reperfusion. The results demonstrated that at 5 mg/kg, U83836E markedly protected cardiac function in ischemia/reperfusion rat models, decreased the malondialdehyde content and creatinine kinase activity, while increasing superoxide dismutase and glutathione peroxidase activity. Additionally, U83836E significantly decreased the histological damage to the myocardium, reduced the area of myocardial infarction in the left ventricle and modified the mitochondrial dysfunction. Furthermore, U83836E enhanced the translocation of protein kinase Cε (PKCε) from the cytoplasm to the membrane. However, the cardioprotective effects of U83836E were reduced in the presence of the PKC inhibitor, chelerythrine (1 mg/kg). Therefore, the results of the present study suggest that U83836E has a potent protective effect against MIRI in rat models through the direct anti‑oxidative stress mechanisms and the activation of PKC signaling.
Collapse
Affiliation(s)
- Li-Na Lai
- Department of Pharmacology, Changzhi Medical College, Changzhi, Shanxi 046000, P.R. China
| | - Xiao-Jing Zhang
- Department of Pharmacology, Changzhi Medical College, Changzhi, Shanxi 046000, P.R. China
| | - Xiao-Yi Zhang
- Department of Pharmacology, Changzhi Medical College, Changzhi, Shanxi 046000, P.R. China
| | - Li-Hua Song
- Department of Pharmacology, Changzhi Medical College, Changzhi, Shanxi 046000, P.R. China
| | - Chun-Hua Guo
- Department of Pharmacology, Changzhi Medical College, Changzhi, Shanxi 046000, P.R. China
| | - Jing-Wen Lei
- Department of Pharmacology, Changzhi Medical College, Changzhi, Shanxi 046000, P.R. China
| | - Xiao-Liang Song
- Department of Pharmacology, Changzhi Medical College, Changzhi, Shanxi 046000, P.R. China
| |
Collapse
|
49
|
Abstract
We showed that exercise induces early and late myocardial preconditioning in dogs and that these effects are mediated through nicotinamide adenine dinucleotide phosphate reduced form (NADPH) oxidase activation. As the intracoronary administration of calcium induces preconditioning and exercise enhances the calcium inflow to the cell, we studied if this effect of exercise triggers exercise preconditioning independently of its hemodynamic effects. We analyzed in 81 dogs the effect of blocking sarcolemmal L-type Ca channels with a low dose of verapamil on early and late preconditioning by exercise, and in other 50 dogs, we studied the effect of verapamil on NADPH oxidase activation in early exercise preconditioning. Exercise reduced myocardial infarct size by 76% and 52% (early and late windows respectively; P < 0.001 both), and these effects were abolished by a single low dose of verapamil given before exercise. This dose of verapamil did not modify the effect of exercise on metabolic and hemodynamic parameters. In addition, verapamil blocked the activation of NADPH oxidase during early preconditioning. The protective effect of exercise preconditioning on myocardial infarct size is triggered, at least in part, by calcium inflow increase to the cell during exercise and, during the early window, is mediated by NADPH oxidase activation.
Collapse
|
50
|
Caru M, Lalonde F, Gravel H, Daigle C, Tournoux F, Jacquemet V, Curnier D. Remote ischaemic preconditioning shortens QT intervals during exercise in healthy subjects. Eur J Sport Sci 2016; 16:1005-13. [DOI: 10.1080/17461391.2016.1156161] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
|