1
|
Triska J, Maitra N, Deshotels MR, Haddadin F, Angiolillo DJ, Vilahur G, Jneid H, Atar D, Birnbaum Y. A Comprehensive Review of the Pleiotropic Effects of Ticagrelor. Cardiovasc Drugs Ther 2024; 38:775-797. [PMID: 36001200 DOI: 10.1007/s10557-022-07373-5] [Citation(s) in RCA: 13] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 08/12/2022] [Indexed: 11/03/2022]
Abstract
AIMS This review summarizes the findings of preclinical studies evaluating the pleiotropic effects of ticagrelor. These include attenuation of ischemia-reperfusion injury (IRI), inflammation, adverse cardiac remodeling, and atherosclerosis. In doing so, it aims to provide novel insights into ticagrelor's mechanisms and benefits over other P2Y12 inhibitors. It also generates viable hypotheses for the results of seminal clinical trials assessing ticagrelor use in acute and chronic coronary syndromes. METHODS AND RESULTS A comprehensive review of the preclinical literature demonstrates that ticagrelor protects against IRI in the setting of both an acute myocardial infarction (MI), and when MI occurs while on chronic treatment. Maintenance therapy with ticagrelor also likely mitigates adverse inflammation, cardiac remodeling, and atherosclerosis, while improving stem cell recruitment. These effects are probably mediated by ticagrelor's ability to increase local interstitial adenosine levels which activate downstream cardio-protective molecules. Attenuation and augmentation of these pleiotropic effects by high-dose aspirin and caffeine, and statins respectively may help explain variable outcomes in PLATO and subsequent randomized controlled trials (RCTs). CONCLUSION Most RCTs and meta-analyses have not evaluated the pleiotropic effects of ticagrelor. We need further studies comparing cardiovascular outcomes in patients treated with ticagrelor versus other P2Y12 inhibitors that are mindful of the unique pleiotropic advantages afforded by ticagrelor, as well as possible interactions with other therapies (e.g., aspirin, statins, caffeine).
Collapse
Affiliation(s)
- Jeffrey Triska
- The Department of Medicine, Baylor College of Medicine, Houston, TX, USA.
| | - Neil Maitra
- The Department of Medicine, Baylor College of Medicine, Houston, TX, USA
| | | | - Faris Haddadin
- The Section of Cardiology, Baylor College of Medicine, Houston, TX, USA
| | - Dominick J Angiolillo
- Division of Cardiology, University of Florida College of Medicine, Jacksonville, FL, USA
| | - Gemma Vilahur
- Cardiovascular Program, Research Institute Hospital de La Santa Creu I Sant Pau, IIB-Sant Pau, Barcelona, Spain
- CiberCV, Institute Carlos III, Madrid, Spain
| | - Hani Jneid
- Department of Medicine, Section of Cardiology, University of Texas Medical Branch, Galveston, TX, USA
| | - Dan Atar
- The Department of Cardiology, Oslo University Hospital Ulleval, Oslo, Norway
- Institute of Clinical Medicine, University of Oslo, Oslo, Norway
| | - Yochai Birnbaum
- The Section of Cardiology, Baylor College of Medicine, Houston, TX, USA
| |
Collapse
|
2
|
Ravic M, Srejovic I, Novakovic J, Andjic M, Sretenovic J, Muric M, Nikolic M, Bolevich S, Alekseevich Kasabov K, Petrovich Fisenko V, Stojanovic A, Jakovljevic V. Effect of GLP-1 Receptor Agonist on Ischemia Reperfusion Injury in Rats with Metabolic Syndrome. Pharmaceuticals (Basel) 2024; 17:525. [PMID: 38675485 PMCID: PMC11053642 DOI: 10.3390/ph17040525] [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: 03/11/2024] [Revised: 04/14/2024] [Accepted: 04/15/2024] [Indexed: 04/28/2024] Open
Abstract
Metabolic syndrome (MetS) represents an important factor that increases the risk of myocardial infarction, and more severe complications. Glucagon Like Peptide-1 Receptor Agonists (GLP-1RAs) exhibit cardioprotective potential, but their efficacy in MetS-related myocardial dysfunction has not been fully explored. Therefore, we aimed to assess the effects of exenatide and dulaglutide on heart function and redox balance in MetS-induced rats. Twenty-four Wistar albino rats with induced MetS were divided into three groups: MetS, exenatide-treated (5 µg/kg), dulaglutide-treated (0.6 mg/kg). After 6 weeks of treatment, in vivo heart function was assessed via echocardiography, while ex vivo function was evaluated using a Langendorff apparatus to simulate ischemia-reperfusion injury. Heart tissue samples were analyzed histologically, and oxidative stress biomarkers were measured spectrophotometrically from the coronary venous effluent. Both exenatide and dulaglutide significantly improved the ejection fraction by 3% and 7%, respectively, compared to the MetS group. Histological analyses corroborated these findings, revealing a reduction in the cross-sectional area of cardiomyocytes by 11% in the exenatide and 18% in the dulaglutide group, indicating reduced myocardial damage in GLP-1RA-treated rats. Our findings suggest strong cardioprotective potential of GLP-1RAs in MetS, with dulaglutide showing a slight advantage. Thus, both exenatide and dulaglutide are potentially promising targets for cardioprotection and reducing mortality in MetS patients.
Collapse
Affiliation(s)
- Marko Ravic
- Department of Pharmacy, Faculty of Medical Sciences, University of Kragujevac, Svetozara Markovica 69, 34000 Kragujevac, Serbia; (M.R.); (J.N.); (M.A.)
- Center of Excellence for the Study of Redox Balance in Cardiovascular and Metabolic Disorders, University of Kragujevac, Svetozara Markovica 69, 34000 Kragujevac, Serbia; (I.S.); (J.S.); (M.M.); (M.N.); (V.J.)
| | - Ivan Srejovic
- Center of Excellence for the Study of Redox Balance in Cardiovascular and Metabolic Disorders, University of Kragujevac, Svetozara Markovica 69, 34000 Kragujevac, Serbia; (I.S.); (J.S.); (M.M.); (M.N.); (V.J.)
- Department of Physiology, Faculty of Medical Sciences, University of Kragujevac, Svetozara Markovica 69, 34000 Kragujevac, Serbia
- Department of Pharmacology, First Moscow State Medical University I.M. Sechenov, Trubetskaya Street 8, Str. 2, 119991 Moscow, Russia; (K.A.K.); (V.P.F.)
| | - Jovana Novakovic
- Department of Pharmacy, Faculty of Medical Sciences, University of Kragujevac, Svetozara Markovica 69, 34000 Kragujevac, Serbia; (M.R.); (J.N.); (M.A.)
- Center of Excellence for the Study of Redox Balance in Cardiovascular and Metabolic Disorders, University of Kragujevac, Svetozara Markovica 69, 34000 Kragujevac, Serbia; (I.S.); (J.S.); (M.M.); (M.N.); (V.J.)
| | - Marijana Andjic
- Department of Pharmacy, Faculty of Medical Sciences, University of Kragujevac, Svetozara Markovica 69, 34000 Kragujevac, Serbia; (M.R.); (J.N.); (M.A.)
- Center of Excellence for the Study of Redox Balance in Cardiovascular and Metabolic Disorders, University of Kragujevac, Svetozara Markovica 69, 34000 Kragujevac, Serbia; (I.S.); (J.S.); (M.M.); (M.N.); (V.J.)
| | - Jasmina Sretenovic
- Center of Excellence for the Study of Redox Balance in Cardiovascular and Metabolic Disorders, University of Kragujevac, Svetozara Markovica 69, 34000 Kragujevac, Serbia; (I.S.); (J.S.); (M.M.); (M.N.); (V.J.)
- Department of Physiology, Faculty of Medical Sciences, University of Kragujevac, Svetozara Markovica 69, 34000 Kragujevac, Serbia
| | - Maja Muric
- Center of Excellence for the Study of Redox Balance in Cardiovascular and Metabolic Disorders, University of Kragujevac, Svetozara Markovica 69, 34000 Kragujevac, Serbia; (I.S.); (J.S.); (M.M.); (M.N.); (V.J.)
- Department of Physiology, Faculty of Medical Sciences, University of Kragujevac, Svetozara Markovica 69, 34000 Kragujevac, Serbia
| | - Marina Nikolic
- Center of Excellence for the Study of Redox Balance in Cardiovascular and Metabolic Disorders, University of Kragujevac, Svetozara Markovica 69, 34000 Kragujevac, Serbia; (I.S.); (J.S.); (M.M.); (M.N.); (V.J.)
- Department of Physiology, Faculty of Medical Sciences, University of Kragujevac, Svetozara Markovica 69, 34000 Kragujevac, Serbia
| | - Sergey Bolevich
- Department of Human Pathology, First Moscow State Medical University I.M. Sechenov, Trubetskaya Street 8, Str. 2, 119991 Moscow, Russia;
| | - Kirill Alekseevich Kasabov
- Department of Pharmacology, First Moscow State Medical University I.M. Sechenov, Trubetskaya Street 8, Str. 2, 119991 Moscow, Russia; (K.A.K.); (V.P.F.)
| | - Vladimir Petrovich Fisenko
- Department of Pharmacology, First Moscow State Medical University I.M. Sechenov, Trubetskaya Street 8, Str. 2, 119991 Moscow, Russia; (K.A.K.); (V.P.F.)
| | - Aleksandra Stojanovic
- Department of Pharmacy, Faculty of Medical Sciences, University of Kragujevac, Svetozara Markovica 69, 34000 Kragujevac, Serbia; (M.R.); (J.N.); (M.A.)
- Center of Excellence for the Study of Redox Balance in Cardiovascular and Metabolic Disorders, University of Kragujevac, Svetozara Markovica 69, 34000 Kragujevac, Serbia; (I.S.); (J.S.); (M.M.); (M.N.); (V.J.)
| | - Vladimir Jakovljevic
- Center of Excellence for the Study of Redox Balance in Cardiovascular and Metabolic Disorders, University of Kragujevac, Svetozara Markovica 69, 34000 Kragujevac, Serbia; (I.S.); (J.S.); (M.M.); (M.N.); (V.J.)
- Department of Physiology, Faculty of Medical Sciences, University of Kragujevac, Svetozara Markovica 69, 34000 Kragujevac, Serbia
- Department of Human Pathology, First Moscow State Medical University I.M. Sechenov, Trubetskaya Street 8, Str. 2, 119991 Moscow, Russia;
| |
Collapse
|
3
|
Czigle S, Nagy M, Mladěnka P, Tóth J. Pharmacokinetic and pharmacodynamic herb-drug interactions-part I. Herbal medicines of the central nervous system. PeerJ 2023; 11:e16149. [PMID: 38025741 PMCID: PMC10656908 DOI: 10.7717/peerj.16149] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2023] [Accepted: 08/30/2023] [Indexed: 12/01/2023] Open
Abstract
Unlike conventional drug substances, herbal medicines are composed of a complex of biologically active compounds. Therefore, the potential occurrence of herb-drug interactions is even more probable than for drug-drug interactions. Interactions can occur on both the pharmacokinetic and pharmacodynamic level. Herbal medicines may affect the resulting efficacy of the concomitantly used (synthetic) drugs, mainly on the pharmacokinetic level, by changing their absorption, distribution, metabolism, and excretion. Studies on the pharmacodynamic interactions of herbal medicines and conventional drugs are still very limited. This interaction level is related to the mechanism of action of different plant constituents. Herb-drug interactions can cause changes in drug levels and activities and lead to therapeutic failure and/or side effects (sometimes toxicities, even fatal). This review aims to provide a summary of recent information on the potential drug interactions involving commonly used herbal medicines that affect the central nervous system (Camellia, Valeriana, Ginkgo, Hypericum, Humulus, Cannabis) and conventional drugs. The survey databases were used to identify primary scientific publications, case reports, and secondary databases on interactions were used later on as well. Search keywords were based on plant names (botanical genera), officinal herbal drugs, herbal drug preparations, herbal drug extracts.
Collapse
Affiliation(s)
- Szilvia Czigle
- Department of Pharmacognosy and Botany, Faculty of Pharmacy, Comenius University Bratislava, Bratislava, Slovak Republic
| | - Milan Nagy
- Department of Pharmacognosy and Botany, Faculty of Pharmacy, Comenius University Bratislava, Bratislava, Slovak Republic
| | - Přemysl Mladěnka
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Charles University, Hradec Králové, Czech Republic
| | - Jaroslav Tóth
- Department of Pharmacognosy and Botany, Faculty of Pharmacy, Comenius University Bratislava, Bratislava, Slovak Republic
| | - the OEMONOM.
- Department of Pharmacognosy and Botany, Faculty of Pharmacy, Comenius University Bratislava, Bratislava, Slovak Republic
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Charles University, Hradec Králové, Czech Republic
| |
Collapse
|
4
|
Zhao ZZ, Li E, Li XJ, Guo Q, Shi QB, Li MW. Effects of remote ischemic preconditioning on coronary blood flow and microcirculation. BMC Cardiovasc Disord 2023; 23:404. [PMID: 37592218 PMCID: PMC10433538 DOI: 10.1186/s12872-023-03419-0] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2023] [Accepted: 07/26/2023] [Indexed: 08/19/2023] Open
Abstract
This study aimed to determine the effect of short-term remote ischemic preconditioning (RIPC) on coronary blood flow and microcirculation function using the quantitative flow ratio (QFR) and index of microcirculatory resistance (IMR). We randomly divided 129 patients undergoing coronary angiography (CAG) into RIPC and control groups. Following the first CAG, we randomly divided the patients further into the unilateral upper limb and lower limb groups for four cycles of ischemia/reperfusion circulation; subsequently, we performed the second CAG. During each CAG, contrast-flow QFR (cQFR), fixed-flow QFR (fQFR), and IMR (in patients with cardiac syndrome X) were calculated and compared. We measured 253 coronary arteries in 129 patients. Compared to the control group, the average cQFR of the RIPC group increased significantly after RIPC. Additionally, 23 patients with cardiac syndrome X (IMR > 30) were included in this study. Compared to the control group, IMR and the difference between cQFR and fQFR (cQFR-fQFR) both decreased significantly after receiving RIPC. The application of RIPC can increase coronary blood flow and improve coronary microcirculation function.
Collapse
Affiliation(s)
- Zhen-Zhou Zhao
- Heart Center of Henan Provincial People's Hospital, Fuwai Central China Cardiovascular Hospital, Henan Province, Zhengzhou, 450003, Henan Province, China
| | - En Li
- Heart Center of Henan Provincial People's Hospital, Fuwai Central China Cardiovascular Hospital, Henan Province, Zhengzhou, 450003, Henan Province, China
| | - Xue-Jie Li
- Heart Center of Henan Provincial People's Hospital, Fuwai Central China Cardiovascular Hospital, Henan Province, Zhengzhou, 450003, Henan Province, China
| | - Quan Guo
- Heart Center of Henan Provincial People's Hospital, Fuwai Central China Cardiovascular Hospital, Henan Province, Zhengzhou, 450003, Henan Province, China
| | - Qing-Bo Shi
- Heart Center of Henan Provincial People's Hospital, Fuwai Central China Cardiovascular Hospital, Henan Province, Zhengzhou, 450003, Henan Province, China
| | - Mu-Wei Li
- Heart Center of Henan Provincial People's Hospital, Fuwai Central China Cardiovascular Hospital, Henan Province, Zhengzhou, 450003, Henan Province, China.
| |
Collapse
|
5
|
Ye R, Jneid H, Alam M, Uretsky BF, Atar D, Kitakaze M, Davidson SM, Yellon DM, Birnbaum Y. Do We Really Need Aspirin Loading for STEMI? Cardiovasc Drugs Ther 2022; 36:1221-1238. [PMID: 35171384 DOI: 10.1007/s10557-022-07327-x] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 02/11/2022] [Indexed: 12/12/2022]
Abstract
Aspirin loading (chewable or intravenous) as soon as possible after presentation is a class I recommendation by current ST elevation myocardial infarction (STEMI) guidelines. Earlier achievement of therapeutic antiplatelet effects by aspirin loading has long been considered the standard of care. However, the effects of the loading dose of aspirin (alone or in addition to a chronic maintenance oral dose) have not been studied. A large proportion of myocardial cell death occurs upon and after reperfusion (reperfusion injury). Numerous agents and interventions have been shown to limit infarct size in animal models when administered before or immediately after reperfusion. However, these interventions have predominantly failed to show significant protection in clinical studies. In the current review, we raise the hypothesis that aspirin loading may be the culprit. Data obtained from animal models consistently show that statins, ticagrelor, opiates, and ischemic postconditioning limit myocardial infarct size. In most of these studies, aspirin was not administered. However, when aspirin was administered before reperfusion (as is the case in the majority of studies enrolling STEMI patients), the protective effects of statin, ticagrelor, morphine, and ischemic postconditioning were attenuated, which can be plausibly attributable to aspirin loading. We therefore suggest studying the effects of aspirin loading before reperfusion on the infarct size limiting effects of statins, ticagrelor, morphine, and/ or postconditioning in large animal models using long reperfusion periods (at least 24 h). If indeed aspirin attenuates the protective effects, clinical trials should be conducted comparing aspirin loading to alternative antiplatelet regimens without aspirin loading in patients with STEMI undergoing primary percutaneous coronary intervention.
Collapse
Affiliation(s)
- Regina Ye
- University of Texas at Austin, Austin, TX, USA
| | - Hani Jneid
- Department of Medicine Baylor College of Medicine, 7200 Cambridge Street Houston, Texas, 77030, USA
| | - Mahboob Alam
- Department of Medicine Baylor College of Medicine, 7200 Cambridge Street Houston, Texas, 77030, USA
| | - Barry F Uretsky
- University of Arkansas for Medical Sciences, Central Arkansas Veterans Health System, Little Rock, AR, USA
| | - Dan Atar
- Department of Cardiology, Oslo University Hospital Ulleval, Oslo, Norway, and Institute of Clinical Medicine, University of Oslo, Oslo, Norway
| | - Masafumi Kitakaze
- Center of Medical Innovation and Translational Research, Department of Medical Data Science, Osaka University Graduate School of Medicine, Osaka, Japan
| | - Sean M Davidson
- The Hatter Cardiovascular Institute, University College London, 67 Chenies Mews, London, WC1E 6HX, UK
| | - Derek M Yellon
- The Hatter Cardiovascular Institute, University College London, 67 Chenies Mews, London, WC1E 6HX, UK
| | - Yochai Birnbaum
- Department of Medicine Baylor College of Medicine, 7200 Cambridge Street Houston, Texas, 77030, USA.
| |
Collapse
|
6
|
Kherallah RY, Khawaja M, Olson M, Angiolillo D, Birnbaum Y. Cilostazol: a Review of Basic Mechanisms and Clinical Uses. Cardiovasc Drugs Ther 2021; 36:777-792. [PMID: 33860901 DOI: 10.1007/s10557-021-07187-x] [Citation(s) in RCA: 42] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 04/05/2021] [Indexed: 12/24/2022]
Abstract
Primarily used in the treatment of intermittent claudication, cilostazol is a 2-oxyquinolone derivative that works through the inhibition of phosphodiesterase III and related increases in cyclic adenosine monophosphate (cAMP) levels. However, cilostazol has been implicated in a number of other basic pathways including the inhibition of adenosine reuptake, the inhibition of multidrug resistance protein 4, among others. It has been observed to exhibit antiplatelet, antiproliferative, vasodilatory, and ischemic-reperfusion protective properties. As such, cilostazol has been investigated for clinical use in a variety of settings including intermittent claudication, as an adjunctive for reduction of restenosis after coronary and peripheral endovascular interventions, and in the prevention of secondary stroke, although its widespread implementation for indications other than intermittent claudication has been limited by relatively modest effect sizes and lack of studies in western populations. In this review, we highlight the pleiotropic effects of cilostazol and the evidence for its clinical use.
Collapse
Affiliation(s)
- Riyad Y Kherallah
- Department of Medicine, Baylor College of Medicine, Houston, TX, USA
| | - Muzamil Khawaja
- Department of Medicine, Baylor College of Medicine, Houston, TX, USA
| | - Michael Olson
- Department of Medicine, Baylor College of Medicine, Houston, TX, USA
| | - Dominick Angiolillo
- Division of Cardiology, University of Florida College of Medicine, Jacksonville, FL, USA
| | - Yochai Birnbaum
- Section of Cardiology, Department of Medicine, Baylor College of Medicine, 7200 Cambridge Street, Houston, TX, USA.
| |
Collapse
|
7
|
Stiermaier T, Birnbaum Y, Eitel I. Is there a Future for Remote Ischemic Conditioning in Acute Myocardial Infarction? Cardiovasc Drugs Ther 2021; 36:197-199. [PMID: 33666821 PMCID: PMC8770378 DOI: 10.1007/s10557-020-07074-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 09/02/2020] [Indexed: 11/29/2022]
Affiliation(s)
- Thomas Stiermaier
- University Heart Center Lübeck, Medical Clinic II, University Hospital Schleswig-Holstein, Ratzeburger Allee 160, Lübeck, 23538, Germany.,German Center for Cardiovascular Research (DZHK), Partner site Hamburg/Kiel/Lübeck, Lübeck, Germany
| | - Yochai Birnbaum
- The Department of Medicine, The Section of Cardiology, Baylor College of Medicine, Houston, TX, USA
| | - Ingo Eitel
- University Heart Center Lübeck, Medical Clinic II, University Hospital Schleswig-Holstein, Ratzeburger Allee 160, Lübeck, 23538, Germany. .,German Center for Cardiovascular Research (DZHK), Partner site Hamburg/Kiel/Lübeck, Lübeck, Germany.
| |
Collapse
|
8
|
Mendieta G, Ben-Aicha S, Gutiérrez M, Casani L, Aržanauskaitė M, Carreras F, Sabate M, Badimon L, Vilahur G. Intravenous Statin Administration During Myocardial Infarction Compared With Oral Post-Infarct Administration. J Am Coll Cardiol 2020; 75:1386-1402. [PMID: 32216907 DOI: 10.1016/j.jacc.2020.01.042] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/22/2019] [Revised: 01/03/2020] [Accepted: 01/23/2020] [Indexed: 12/20/2022]
Abstract
BACKGROUND Beyond lipid-lowering, statins exert cardioprotective effects. High-dose statin treatment seems to reduce cardiovascular complications in high-risk patients. The ideal timing and administration regime remain unknown. OBJECTIVES This study compared the cardioprotective effects of intravenous statin administration during myocardial infarction (MI) with oral administration immediately post-MI. METHODS Hypercholesterolemic pigs underwent MI induction (90 min of ischemia) and were kept for 42 days. Animals were distributed in 3 arms (A): A1 received an intravenous bolus of atorvastatin during MI; A2 received an intravenous bolus of vehicle during MI; and A3 received oral atorvastatin within 2 h post-MI. A1 and A3 remained on daily oral atorvastatin for the following 42 days. Cardiac magnetic resonance analysis (days 3 and 42 post-MI) and molecular/histological studies were performed. RESULTS At day 3, A1 showed a 10% reduction in infarct size compared with A3 and A2 and a 50% increase in myocardial salvage. At day 42, both A1 and A3 showed a significant decrease in scar size versus A2; however, A1 showed a further 24% reduction versus A3. Functional analyses revealed improved systolic performance in A1 compared with A2 and less wall motion abnormalities in the jeopardized myocardium versus both groups at day 42. A1 showed enhanced collagen content and AMP-activated protein kinase activation in the scar, increased vessel density in the penumbra, higher tumor necrosis factor α plasma levels and lower peripheral blood mononuclear cell activation versus both groups. CONCLUSIONS Intravenous administration of atorvastatin during MI limits cardiac damage, improves cardiac function, and mitigates remodeling to a larger extent than when administered orally shortly after reperfusion. This therapeutic approach deserves to be investigated in ST-segment elevation MI patients.
Collapse
Affiliation(s)
- Guiomar Mendieta
- Cardiovascular Research Center-ICCC, Hospital de la Santa Creu i Sant Pau, IIB-Sant Pau, Barcelona, Spain; Department of Cardiology, Clinic Hospital, Barcelona, Spain
| | - Soumaya Ben-Aicha
- Cardiovascular Research Center-ICCC, Hospital de la Santa Creu i Sant Pau, IIB-Sant Pau, Barcelona, Spain
| | - Manuel Gutiérrez
- Radiology, Hospital de la Santa Creu i Sant Pau, IIB-Sant Pau, Barcelona, Spain
| | - Laura Casani
- Cardiovascular Research Center-ICCC, Hospital de la Santa Creu i Sant Pau, IIB-Sant Pau, Barcelona, Spain; Radiology, Hospital de la Santa Creu i Sant Pau, IIB-Sant Pau, Barcelona, Spain
| | - Monika Aržanauskaitė
- Cardiovascular Research Center-ICCC, Hospital de la Santa Creu i Sant Pau, IIB-Sant Pau, Barcelona, Spain
| | - Francesc Carreras
- Cardiology, Hospital de la Santa Creu i Sant Pau, IIB-Sant Pau, Barcelona, Spain
| | - Manel Sabate
- Department of Cardiology, Clinic Hospital, Barcelona, Spain
| | - Lina Badimon
- Cardiovascular Research Center-ICCC, Hospital de la Santa Creu i Sant Pau, IIB-Sant Pau, Barcelona, Spain; CiberCV, Institute Carlos III, Barcelona, Spain
| | - Gemma Vilahur
- Cardiovascular Research Center-ICCC, Hospital de la Santa Creu i Sant Pau, IIB-Sant Pau, Barcelona, Spain; CiberCV, Institute Carlos III, Barcelona, Spain.
| |
Collapse
|
9
|
Kleinbongard P, Bøtker HE, Ovize M, Hausenloy DJ, Heusch G. Co-morbidities and co-medications as confounders of cardioprotection-Does it matter in the clinical setting? Br J Pharmacol 2020; 177:5252-5269. [PMID: 31430831 PMCID: PMC7680006 DOI: 10.1111/bph.14839] [Citation(s) in RCA: 100] [Impact Index Per Article: 20.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2019] [Revised: 07/26/2019] [Accepted: 08/15/2019] [Indexed: 02/06/2023] Open
Abstract
The translation of cardioprotection from robust experimental evidence to beneficial clinical outcome for patients suffering acute myocardial infarction or undergoing cardiovascular surgery has been largely disappointing. The present review attempts to critically analyse the evidence for confounders of cardioprotection in patients with acute myocardial infarction and in patients undergoing cardiovascular surgery. One reason that has been proposed to be responsible for such lack of translation is the confounding of cardioprotection by co-morbidities and co-medications. Whereas there is solid experimental evidence for such confounding of cardioprotection by single co-morbidities and co-medications, the clinical evidence from retrospective analyses of the limited number of clinical data is less robust. The best evidence for interference of co-medications is that for platelet inhibitors to recruit cardioprotection per se and thus limit the potential for further protection from myocardial infarction and for propofol anaesthesia to negate the protection from remote ischaemic conditioning in cardiovascular surgery. LINKED ARTICLES: This article is part of a themed issue on Risk factors, comorbidities, and comedications in cardioprotection. To view the other articles in this section visit http://onlinelibrary.wiley.com/doi/10.1111/bph.v177.23/issuetoc.
Collapse
Affiliation(s)
- Petra Kleinbongard
- Institute for Pathophysiology, West German Heart and Vascular CenterUniversity of Essen Medical SchoolEssenGermany
| | - Hans Erik Bøtker
- Department of CardiologyAarhus University Hospital SkejbyAarhusDenmark
| | - Michel Ovize
- INSERM U1060, CarMeN Laboratory, Université de Lyon and Explorations Fonctionnelles Cardiovasculaires, Hôpital Louis Pradel, Hospices Civils de LyonLyonFrance
| | - Derek J. Hausenloy
- Cardiovascular and Metabolic Disorders ProgramDuke‐National University of Singapore Medical SchoolSingapore
- National Heart Research Institute SingaporeNational Heart CentreSingapore
- Yong Loo Lin School of MedicineNational University SingaporeSingapore
- The Hatter Cardiovascular InstituteUniversity College LondonLondonUK
- Research and DevelopmentThe National Institute of Health Research University College London Hospitals Biomedical Research CentreLondonUK
- Tecnologico de MonterreyCentro de Biotecnologia‐FEMSAMonterreyNuevo LeonMexico
| | - Gerd Heusch
- Institute for Pathophysiology, West German Heart and Vascular CenterUniversity of Essen Medical SchoolEssenGermany
| |
Collapse
|
10
|
Klinnikova MG, Koldysheva EV, Tursunova NV, Semenov DE, Lushnikova EL. Features of Myocardial Remodeling and Changes in the Blood Lipid Spectrum in Experimental Doxorubicin-Induced Cardiomyopathy and Atorvastatin Administration. Bull Exp Biol Med 2020; 170:24-29. [PMID: 33222079 DOI: 10.1007/s10517-020-04997-1] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2020] [Indexed: 11/30/2022]
Abstract
Structural myocardial reorganization and changes in the blood lipid spectrum in rats were studied after administration of a single sublethal dose of doxorubicin (15 mg/kg) alone and in combination with atorvastatin (20 mg/kg/day over 7 days). It was established that doxorubicin induced the development of dyslipidemia in experimental animals (the concentrations of total cholesterol, triglycerides, and VLDL increased by 2.2, 2.0, and 1.96 times, respectively; the atherogenic coefficient increased by 3.4 times by day 7 of the experiment). In animals with experimental anthracycline cardiomyopathy treated with atorvastatin, the concentrations of the main components of the blood lipid spectrum increased less markedly. Atorvastatin alone induces moderate myocardial remodeling in comparison with more pronounced changes in the structural organization of the myocardium in rats treated with doxorubicin alone. Course treatment with atorvastatin under conditions of doxorubicin-induced cardiomyopathy reduced the severity of myocardial remodeling: the decrease in the volume density of cardiomyocytes and the increase in the volume density of the connective tissue were less pronounced in the dynamics of the experiment.
Collapse
Affiliation(s)
- M G Klinnikova
- Institute of Molecular Pathology and Pathomorphology, Federal Research Center of Fundamental and Translational Medicine, Novosibirsk, Russia.
| | - E V Koldysheva
- Institute of Molecular Pathology and Pathomorphology, Federal Research Center of Fundamental and Translational Medicine, Novosibirsk, Russia
| | - N V Tursunova
- Institute of Molecular Pathology and Pathomorphology, Federal Research Center of Fundamental and Translational Medicine, Novosibirsk, Russia
| | - D E Semenov
- Institute of Molecular Pathology and Pathomorphology, Federal Research Center of Fundamental and Translational Medicine, Novosibirsk, Russia
| | - E L Lushnikova
- Institute of Molecular Pathology and Pathomorphology, Federal Research Center of Fundamental and Translational Medicine, Novosibirsk, Russia
| |
Collapse
|
11
|
Fuernau G, Beck J, Desch S, Eitel I, Jung C, Erbs S, Mangner N, Lurz P, Fengler K, Jobs A, Vonthein R, de Waha-Thiele S, Sandri M, Schuler G, Thiele H. Mild Hypothermia in Cardiogenic Shock Complicating Myocardial Infarction. Circulation 2019; 139:448-457. [DOI: 10.1161/circulationaha.117.032722] [Citation(s) in RCA: 33] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Affiliation(s)
- Georg Fuernau
- Medical Clinic II (Cardiology/Angiology/Intensive Care Medicine), University Heart Center Luebeck, University Hospital Schleswig-Holstein (G.F., S.D., I.E., A.J., S.d.W.-T), University of Luebeck, Germany
- German Center for Cardiovascular Research (Deutsches Zentrum für Herz-Kreislauf-Forschung [DZHK]), Partner Site Hamburg/Kiel/Lübeck, Luebeck, Germany (G.F., S.D., I.E., A.J., S.d.W.-T)
- Department of Internal Medicine/Cardiology, Heart Center Leipzig–University Hospital, Germany (G.F., J.B., S.D., I.E., S.E., N.M., P.L., K.F., S.d.W.-T, M.S., G.S., H.T.)
| | - Johannes Beck
- Department of Internal Medicine/Cardiology, Heart Center Leipzig–University Hospital, Germany (G.F., J.B., S.D., I.E., S.E., N.M., P.L., K.F., S.d.W.-T, M.S., G.S., H.T.)
| | - Steffen Desch
- Medical Clinic II (Cardiology/Angiology/Intensive Care Medicine), University Heart Center Luebeck, University Hospital Schleswig-Holstein (G.F., S.D., I.E., A.J., S.d.W.-T), University of Luebeck, Germany
- German Center for Cardiovascular Research (Deutsches Zentrum für Herz-Kreislauf-Forschung [DZHK]), Partner Site Hamburg/Kiel/Lübeck, Luebeck, Germany (G.F., S.D., I.E., A.J., S.d.W.-T)
- Department of Internal Medicine/Cardiology, Heart Center Leipzig–University Hospital, Germany (G.F., J.B., S.D., I.E., S.E., N.M., P.L., K.F., S.d.W.-T, M.S., G.S., H.T.)
| | - Ingo Eitel
- Medical Clinic II (Cardiology/Angiology/Intensive Care Medicine), University Heart Center Luebeck, University Hospital Schleswig-Holstein (G.F., S.D., I.E., A.J., S.d.W.-T), University of Luebeck, Germany
- German Center for Cardiovascular Research (Deutsches Zentrum für Herz-Kreislauf-Forschung [DZHK]), Partner Site Hamburg/Kiel/Lübeck, Luebeck, Germany (G.F., S.D., I.E., A.J., S.d.W.-T)
- Department of Internal Medicine/Cardiology, Heart Center Leipzig–University Hospital, Germany (G.F., J.B., S.D., I.E., S.E., N.M., P.L., K.F., S.d.W.-T, M.S., G.S., H.T.)
| | - Christian Jung
- Medical Faculty, Division of Cardiology, Pulmonology and Vascular Medicine, University Hospital Düsseldorf, Heinrich-Heine-University Düsseldorf, Germany (C.J.)
| | - Sandra Erbs
- Department of Internal Medicine/Cardiology, Heart Center Leipzig–University Hospital, Germany (G.F., J.B., S.D., I.E., S.E., N.M., P.L., K.F., S.d.W.-T, M.S., G.S., H.T.)
| | - Norman Mangner
- Department of Internal Medicine/Cardiology, Heart Center Leipzig–University Hospital, Germany (G.F., J.B., S.D., I.E., S.E., N.M., P.L., K.F., S.d.W.-T, M.S., G.S., H.T.)
| | - Philipp Lurz
- Department of Internal Medicine/Cardiology, Heart Center Leipzig–University Hospital, Germany (G.F., J.B., S.D., I.E., S.E., N.M., P.L., K.F., S.d.W.-T, M.S., G.S., H.T.)
| | - Karl Fengler
- Department of Internal Medicine/Cardiology, Heart Center Leipzig–University Hospital, Germany (G.F., J.B., S.D., I.E., S.E., N.M., P.L., K.F., S.d.W.-T, M.S., G.S., H.T.)
| | - Alexander Jobs
- Medical Clinic II (Cardiology/Angiology/Intensive Care Medicine), University Heart Center Luebeck, University Hospital Schleswig-Holstein (G.F., S.D., I.E., A.J., S.d.W.-T), University of Luebeck, Germany
- German Center for Cardiovascular Research (Deutsches Zentrum für Herz-Kreislauf-Forschung [DZHK]), Partner Site Hamburg/Kiel/Lübeck, Luebeck, Germany (G.F., S.D., I.E., A.J., S.d.W.-T)
| | - Reinhard Vonthein
- Institute of Medical Biometry and Statistics and Center for Clinical Trials (R.V.), University of Luebeck, Germany
| | - Suzanne de Waha-Thiele
- Medical Clinic II (Cardiology/Angiology/Intensive Care Medicine), University Heart Center Luebeck, University Hospital Schleswig-Holstein (G.F., S.D., I.E., A.J., S.d.W.-T), University of Luebeck, Germany
- German Center for Cardiovascular Research (Deutsches Zentrum für Herz-Kreislauf-Forschung [DZHK]), Partner Site Hamburg/Kiel/Lübeck, Luebeck, Germany (G.F., S.D., I.E., A.J., S.d.W.-T)
- Department of Internal Medicine/Cardiology, Heart Center Leipzig–University Hospital, Germany (G.F., J.B., S.D., I.E., S.E., N.M., P.L., K.F., S.d.W.-T, M.S., G.S., H.T.)
| | - Marcus Sandri
- Department of Internal Medicine/Cardiology, Heart Center Leipzig–University Hospital, Germany (G.F., J.B., S.D., I.E., S.E., N.M., P.L., K.F., S.d.W.-T, M.S., G.S., H.T.)
| | - Gerhard Schuler
- Department of Internal Medicine/Cardiology, Heart Center Leipzig–University Hospital, Germany (G.F., J.B., S.D., I.E., S.E., N.M., P.L., K.F., S.d.W.-T, M.S., G.S., H.T.)
| | - Holger Thiele
- Department of Internal Medicine/Cardiology, Heart Center Leipzig–University Hospital, Germany (G.F., J.B., S.D., I.E., S.E., N.M., P.L., K.F., S.d.W.-T, M.S., G.S., H.T.)
| |
Collapse
|
12
|
Unraveling the Interaction of Aspirin, Ticagrelor, and Rosuvastatin on the Progression of Atherosclerosis and Inflammation in Diabetic Mice. Cardiovasc Drugs Ther 2018; 31:489-500. [PMID: 29185103 DOI: 10.1007/s10557-017-6763-9] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/18/2023]
Abstract
PURPOSE We explored the effects of rosuvastatin, aspirin, ticagrelor, and clopidogrel, alone or in combinations on the progression of atherosclerosis and inflammasome activation in diabetic mice. Statins and ticagrelor increase the production of 15-epi-lipoxin A4 via cyclooxygenase-2. Aspirin alone increases 15-epi-lipoxin A4, but when combined with statins, cyclooxygenase-2 is completely blocked. METHODS ApoE-/-/db+/db+ double-knockout mice received rosuvastatin (5 mg/kg/day), aspirin (25 mg/kg/day), ticagrelor (300 mg/kg/day), clopidogrel (75 mg/kg/day), or their combination for 14 weeks. Serum 15-epi-lipoxin A4 levels and aortic wall cholesterol content, IL-1β, IL-6, and TNF-α levels, and plaque area were assessed. RESULTS Aspirin, ticagrelor, and rosuvastatin increased 15-epi-lipoxin A4 levels. The combination of rosuvastatin + ticagrelor provided an additive effect. Aspirin attenuated the effect of both ticagrelor and rosuvastatin. Aspirin, ticagrelor, and rosuvastatin reduced the area of the atherosclerotic plaque. The combination of ticagrelor + rosuvastatin provided additive effects. There was a negative interaction when aspirin was combined with ticagrelor or rosuvastatin. Aspirin, ticagrelor, and rosuvastatin decreased serum IL-1β and IL-6 levels. There was no interaction between aspirin and ticagrelor or aspirin and rosuvastatin, whereas combining rosuvastatin and ticagrelor provided an additive effect. Aspirin, ticagrelor, and rosuvastatin all decreased TNF-α levels. Aspirin attenuated the effect of both ticagrelor and rosuvastatin, and there was no additive effect of combining ticagrelor + rosuvastatin. CONCLUSIONS We found an intricate interaction between aspirin, ticagrelor, and rosuvastatin, as aspirin reduced both ticagrelor and rosuvastatin ability to ameliorate inflammation and atherosclerosis. In contrast, we found additive effects when ticagrelor and rosuvastatin were combined.
Collapse
|
13
|
Weisshaar S, Litschauer B, Kerbel T, Wolzt M. Atorvastatin combined with ticagrelor prevent ischemia-reperfusion induced vascular endothelial dysfunction in healthy young males – A randomized, placebo-controlled, double-blinded study. Int J Cardiol 2018; 255:1-7. [DOI: 10.1016/j.ijcard.2017.12.067] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/19/2017] [Revised: 11/28/2017] [Accepted: 12/16/2017] [Indexed: 10/18/2022]
|
14
|
Venturi E, Lindsay C, Lotteau S, Yang Z, Steer E, Witschas K, Wilson AD, Wickens JR, Russell AJ, Steele D, Calaghan S, Sitsapesan R. Simvastatin activates single skeletal RyR1 channels but exerts more complex regulation of the cardiac RyR2 isoform. Br J Pharmacol 2018; 175:938-952. [PMID: 29278865 PMCID: PMC5825303 DOI: 10.1111/bph.14136] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2017] [Revised: 11/23/2017] [Accepted: 12/13/2017] [Indexed: 12/18/2022] Open
Abstract
BACKGROUND AND PURPOSE Statins are amongst the most widely prescribed drugs for those at risk of cardiovascular disease, lowering cholesterol levels by inhibiting 3-hydroxy-3-methylglutaryl (HMG)-CoA reductase. Although effective at preventing cardiovascular disease, statin use is associated with muscle weakness, myopathies and, occasionally, fatal rhabdomyolysis. As simvastatin, a commonly prescribed statin, promotes Ca2+ release from sarcoplasmic reticulum (SR) vesicles, we investigated if simvastatin directly activates skeletal (RyR1) and cardiac (RyR2) ryanodine receptors. EXPERIMENTAL APPROACH RyR1 and RyR2 single-channel behaviour was investigated after incorporation of sheep cardiac or mouse skeletal SR into planar phospholipid bilayers under voltage-clamp conditions. LC-MS was used to monitor the kinetics of interconversion of simvastatin between hydroxy-acid and lactone forms during these experiments. Cardiac and skeletal myocytes were permeabilised to examine simvastatin modulation of SR Ca2+ release. KEY RESULTS Hydroxy acid simvastatin (active at HMG-CoA reductase) significantly and reversibly increased RyR1 open probability (Po) and shifted the distribution of Ca2+ spark frequency towards higher values in skeletal fibres. In contrast, simvastatin reduced RyR2 Po and shifted the distribution of spark frequency towards lower values in ventricular cardiomyocytes. The lactone pro-drug form of simvastatin (inactive at HMG-CoA reductase) also activated RyR1, suggesting that the HMG-CoA inhibitor pharmacophore was not responsible for RyR1 activation. CONCLUSION AND IMPLICATIONS Simvastatin interacts with RyR1 to increase SR Ca2+ release and thus may contribute to its reported adverse effects on skeletal muscle. The ability of low concentrations of simvastatin to reduce RyR2 Po may also protect against Ca2+ -dependent arrhythmias and sudden cardiac death.
Collapse
Affiliation(s)
- Elisa Venturi
- Department of PharmacologyUniversity of OxfordOxfordUK
| | - Chris Lindsay
- Department of PharmacologyUniversity of OxfordOxfordUK
- Department of Chemistry, Chemistry Research LaboratoryUniversity of OxfordOxfordUK
| | | | - Zhaokang Yang
- School of Biomedical SciencesUniversity of LeedsLeedsUK
| | - Emma Steer
- School of Biomedical SciencesUniversity of LeedsLeedsUK
| | | | | | - James R Wickens
- Department of Chemistry, Chemistry Research LaboratoryUniversity of OxfordOxfordUK
| | - Angela J Russell
- Department of PharmacologyUniversity of OxfordOxfordUK
- Department of Chemistry, Chemistry Research LaboratoryUniversity of OxfordOxfordUK
| | - Derek Steele
- School of Biomedical SciencesUniversity of LeedsLeedsUK
| | | | | |
Collapse
|
15
|
Kubisa MJ, Jezewski MP, Gasecka A, Siller-Matula JM, Postuła M. Ticagrelor - toward more efficient platelet inhibition and beyond. Ther Clin Risk Manag 2018; 14:129-140. [PMID: 29398917 PMCID: PMC5775739 DOI: 10.2147/tcrm.s152369] [Citation(s) in RCA: 47] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
Novel antiplatelet drugs, including ticagrelor, are being successively introduced into the therapy of atherothrombotic conditions due to their superiority over a standard combination of clopidogrel with acetylsalicylic acid in patients with acute coronary syndromes (ACS). A P2Y12 receptor antagonist, ticagrelor, is unique among antiplatelet drugs, because ticagrelor inhibits the platelet P2Y12 receptor in a reversible manner, and because it demonstrates a wide palette of advantageous pleiotropic effects associated with the increased concentration of adenosine. The pleiotropic effects of ticagrelor comprise cardioprotection, restoration of the myocardium after an ischemic event, promotion of the release of anticoagulative factors and, eventually, anti-inflammatory effects. Beyond the advantageous effects, the increased concentration of adenosine is responsible for some of ticagrelor's adverse effects, including dyspnea and bradycardia. Large-scale clinical trials demonstrated that both standard 12-month therapy and long-term use of ticagrelor reduce the risk of cardiovascular events in patients with ACS, but at the expense of a higher risk of major bleeding. Further trials focused on the use of ticagrelor in conditions other than ACS, including ischemic stroke, peripheral artery disease and status after coronary artery bypass grafting. The results of these trials suggest comparable efficacy and safety of ticagrelor and clopidogrel in extra-coronary indications, but firm conclusions are anticipated from currently ongoing studies. Here, we summarize current evidence on the superiority of ticagrelor over other P2Y12 antagonists in ACS, discuss the mechanism underlying the drug-drug interactions and pleiotropic effects of ticagrelor, and present future perspectives of non-coronary indications for ticagrelor.
Collapse
Affiliation(s)
- Michał J Kubisa
- Department of Experimental and Clinical Pharmacology, Centre for Preclinical Research and Technology (CEPT)
| | - Mateusz P Jezewski
- Department of Experimental and Clinical Pharmacology, Centre for Preclinical Research and Technology (CEPT)
| | - Aleksandra Gasecka
- 1st Chair and Department of Cardiology, Medical University of Warsaw, Warsaw, Poland
- Vesicle Observation Centre, Laboratory of Experimental Clinical Chemistry, Academic Medical Centre, University of Amsterdam, the Netherlands
| | | | - Marek Postuła
- Department of Experimental and Clinical Pharmacology, Centre for Preclinical Research and Technology (CEPT)
| |
Collapse
|
16
|
Biochemical targets of drugs mitigating oxidative stress via redox-independent mechanisms. Biochem Soc Trans 2017; 45:1225-1252. [PMID: 29101309 DOI: 10.1042/bst20160473] [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: 06/14/2017] [Revised: 09/24/2017] [Accepted: 09/26/2017] [Indexed: 12/13/2022]
Abstract
Acute or chronic oxidative stress plays an important role in many pathologies. Two opposite approaches are typically used to prevent the damage induced by reactive oxygen and nitrogen species (RONS), namely treatment either with antioxidants or with weak oxidants that up-regulate endogenous antioxidant mechanisms. This review discusses options for the third pharmacological approach, namely amelioration of oxidative stress by 'redox-inert' compounds, which do not inactivate RONS but either inhibit the basic mechanisms leading to their formation (i.e. inflammation) or help cells to cope with their toxic action. The present study describes biochemical targets of many drugs mitigating acute oxidative stress in animal models of ischemia-reperfusion injury or N-acetyl-p-aminophenol overdose. In addition to the pro-inflammatory molecules, the targets of mitigating drugs include protein kinases and transcription factors involved in regulation of energy metabolism and cell life/death balance, proteins regulating mitochondrial permeability transition, proteins involved in the endoplasmic reticulum stress and unfolded protein response, nuclear receptors such as peroxisome proliferator-activated receptors, and isoprenoid synthesis. The data may help in identification of oxidative stress mitigators that will be effective in human disease on top of the current standard of care.
Collapse
|
17
|
Birnbaum Y, Birnbaum GD, Birnbaum I, Nylander S, Ye Y. Ticagrelor and Rosuvastatin Have Additive Cardioprotective Effects via Adenosine. Cardiovasc Drugs Ther 2017; 30:539-550. [PMID: 27830382 DOI: 10.1007/s10557-016-6701-2] [Citation(s) in RCA: 46] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
BACKGROUND Ticagrelor inhibits the equilibrative-nucleoside-transporter-1 and thereby, adenosine cell re-uptake. Ticagrelor limits infarct size (IS) in non-diabetic rats and the effect is adenosine-dependent. Statins, via ecto-5'-nucleotidase activation, also increase adenosine levels and limit IS. HYPOTHESIS Ticagrelor and rosuvastatin have additive effects on myocardial adenosine levels, and therefore, on IS and post-reperfusion activation of the NLRP3-inflammasome. METHODS Diabetic ZDF rats received via oral gavage; water (control), ticagrelor (150 mg/kg/d), prasugrel (7.5 mg/kg/d), rosuvastatin (5 mg/kg/d), ticagrelor + rosuvastatin and prasugrel + rosuvastatin for 3d. On day 4, rats underwent 30 min coronary artery occlusion and 24 h of reperfusion. Two additional groups received, ticagrelor + rosuvastatin or water in combination with CGS15943 (CGS, an adenosine receptor antagonist, 10 mg/kg i.p. 1 h before ischemia). RESULTS Both ticagrelor and rosuvastatin increased myocardial adenosine levels with an additive effect of the combination whereas prasugrel had no effect. Similarly, both ticagrelor and rosuvastatin significantly reduced IS with an additive effect of the combination whereas prasugrel had no effect. The effect on IS was adenosine dependent as CGS15943 reversed the effect of ticagrelor + rosuvastatin. The ischemia-reperfusion injury increased myocardial mRNA levels of NLRP3, ASC, IL-1β and IL-6. Ticagrelor and rosuvastatin, but not prasugrel, significantly decreased these pro-inflammatory mediators with a trend to an additive effect of the combination. The combination also increased the levels of anti-inflammatory 15-epilipoxin A4. CONCLUSIONS Ticagrelor and rosuvastatin when given in combination have an additive effect on local myocardial adenosine levels in the setting of ischemia reperfusion. This translates into an additive cardioprotective effect mediated by adenosine-induced effects including downregulation of pro- but upregulation of anti-inflammatory mediators.
Collapse
Affiliation(s)
- Yochai Birnbaum
- The Department of Biochemistry and Molecular Biology, University of Texas Medical Branch, 301 University Blvd., Galveston, TX, 77555, USA.,The section of Cardiology, Baylor College of Medicine, Houston, TX, 77030, USA
| | - Gilad D Birnbaum
- The Department of Medicine, Baylor College of Medicine, Houston, TX, 77030, USA
| | - Itamar Birnbaum
- The section of Cardiology, Baylor College of Medicine, Houston, TX, 77030, USA
| | | | - Yumei Ye
- The Department of Biochemistry and Molecular Biology, University of Texas Medical Branch, 301 University Blvd., Galveston, TX, 77555, USA.
| |
Collapse
|
18
|
Chen Y, Chen H, Birnbaum Y, Nanhwan MK, Bajaj M, Ye Y, Qian J. Aleglitazar, a dual peroxisome proliferator-activated receptor-α and -γ agonist, protects cardiomyocytes against the adverse effects of hyperglycaemia. Diab Vasc Dis Res 2017; 14:152-162. [PMID: 28111985 PMCID: PMC5305042 DOI: 10.1177/1479164116679081] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/17/2023] Open
Abstract
PURPOSE To assess the effects of Aleglitazar on hyperglycaemia-induced apoptosis. METHODS We incubated human cardiomyocytes, cardiomyocytes from cardiac-specific peroxisome proliferator-activated receptor-γ knockout or wild-type mice in normoglycaemic or hyperglycaemic conditions (glucose 25 mM). Cells were treated with different concentrations of Aleglitazar for 48 h. We measured viability, apoptosis, caspase-3 activity, cytochrome-C release, total antioxidant capacity and reactive oxygen species formation in the treated cardiomyocytes. Human cardiomyocytes were transfected with short interfering RNA against peroxisome proliferator-activated receptor-α or peroxisome proliferator-activated receptor-γ. RESULTS Aleglitazar attenuated hyperglycaemia-induced apoptosis, caspase-3 activity and cytochrome-C release and increased viability in human cardiomyocyte, cardiomyocytes from cardiac-specific peroxisome proliferator-activated receptor-γ knockout and wild-type mice. Hyperglycaemia reduced the antioxidant capacity and Aleglitazar significantly blunted this effect. Hyperglycaemia-induced reactive oxygen species production was attenuated by Aleglitazar in both human cardiomyocyte and wild-type mice cardiomyocytes. Aleglitazar improved cell viability in cells exposed to hyperglycaemia. The protective effect was partially blocked by short interfering RNA against peroxisome proliferator-activated receptor-α alone and short interfering RNA against peroxisome proliferator-activated receptor-γ alone and completely blocked by short interfering RNA to both peroxisome proliferator-activated receptor-α and peroxisome proliferator-activated receptor-γ. CONCLUSION Aleglitazar protects cardiomyocytes against hyperglycaemia-induced apoptosis by combined activation of both peroxisome proliferator-activated receptor-α and peroxisome proliferator-activated receptor-γ in a short-term vitro model.
Collapse
Affiliation(s)
- Yan Chen
- Department of Anesthesiology, The First Affiliated Hospital of Kunming Medical University, Kunming, China
| | - Hongmei Chen
- Department of Anesthesiology, Kunming Tongren Hospital, Kunming, China
| | - Yochai Birnbaum
- Section of Cardiology, Department of Medicine, Baylor College of Medicine, Houston, TX, USA
| | - Manjyot K Nanhwan
- Department of Biochemistry and Molecular Biology, The University of Texas Medical Branch at Galveston, Galveston, TX, USA
| | - Mandeep Bajaj
- Section of Endocrinology, Department of Medicine, Baylor College of Medicine, Houston, TX, USA
| | - Yumei Ye
- Department of Biochemistry and Molecular Biology, The University of Texas Medical Branch at Galveston, Galveston, TX, USA
| | - Jinqiao Qian
- Department of Anesthesiology, The First Affiliated Hospital of Kunming Medical University, Kunming, China
- Jinqiao Qian, Department of Anesthesiology, The First Affiliated Hospital of Kunming Medical University, #295 Xichang Road, Kunming 650032, Yunnan Province, China.
| |
Collapse
|
19
|
Qian J, Chen H, Birnbaum Y, Nanhwan MK, Bajaj M, Ye Y. Aleglitazar, a Balanced Dual PPARα and -γ Agonist, Protects the Heart Against Ischemia-Reperfusion Injury. Cardiovasc Drugs Ther 2017; 30:129-41. [PMID: 26861490 DOI: 10.1007/s10557-016-6650-9] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
Abstract
PURPOSE To evaluate whether aleglitazar (Ale), a dual PPARα/γ agonist, has additive effects on myocardial protection against ischemia-reperfusion injury. METHODS Human cardiomyocytes (HCMs), cardiomyocytes from cardiac-specific PPARγ knockout (MCM-PPARγ (CKO) ) or wild type (MCM-WT) mice were incubated with different concentrations of Ale, and subjected to simulated ischemia-reperfusion (SIR) or normoxic conditions (NSIR). Cell viability, apoptosis and caspase-3 activity were determined. HCMs were transfected with siRNA against PPARα (siPPARα) or PPARγ (siPPARγ) followed by incubation with Ale. PPARα/γ DNA binding capacity was measured. Cell viability, apoptosis and levels of P-AKT and P-eNOS were assessed. Infarct size following 30 min coronary artery occlusion and 24 h reperfusion were assessed in WT and db/db diabetic mice following 3-day pretreatment with vehicle, Ale or glimeperide. RESULTS Ale (at concentrations of 150-600 nM) increased cell viability and reduced apoptosis in HCMs, MCM-WT and MCM-PPAR (CKO) exposed to SIR. In HCM, the protective effect was partially blocked by siPPARα alone or siPPARγ alone, and completely blocked by siPPARα+siPPARγ. Ale increased P-Akt/P-eNOS in HCMs. P-Akt or P-eNOS levels were decreased when PPARα alone, PPARγ alone and especially when both were knocked down. Peritoneal GTTs revealed that db/db mice had developed impaired glucose tolerance and insulin sensitivity, which were normalized by Ale or glimepiride treatment. Ale, but not glimepiride, limited infarct size in both WT and diabetic mice after ischemia-reperfusion. CONCLUSIONS Ale protects against myocardial apoptosis caused by hypoxia-reoxygenation in vitro and reduces infarct size in vivo.
Collapse
Affiliation(s)
- Jinqiao Qian
- Department of Anesthesiology, the First Affiliated Hospital of Kunming Medical University, Kunming, Yunnan Province, China.
| | - Hongmei Chen
- Department of Anesthesiology, Kunming Tongren Hospital, Kunming, Yunnan Province, China
| | - Yochai Birnbaum
- Department of Medicine, Section of Cardiology, Baylor College of Medicine, One Baylor Plaza, MS BCM620, Houston, TX, USA.,Department of Biochemistry and Molecular Biology, University of Texas Medical Branch, Galveston, TX, USA
| | - Manjyot K Nanhwan
- Department of Biochemistry and Molecular Biology, University of Texas Medical Branch, Galveston, TX, USA
| | - Mandeep Bajaj
- Department of Medicine, Section of Endocrinology, Baylor College of Medicine, One Baylor Plaza, Houston, TX, USA
| | - Yumei Ye
- Department of Biochemistry and Molecular Biology, University of Texas Medical Branch, Galveston, TX, USA
| |
Collapse
|
20
|
Birnbaum Y, Ye Y, Bajaj M. Type 2 diabetes and cardiovascular disease: A metabolic overview of recent clinical trials. J Diabetes Complications 2017; 31:291-294. [PMID: 27780670 DOI: 10.1016/j.jdiacomp.2016.09.007] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/20/2016] [Revised: 09/14/2016] [Accepted: 09/16/2016] [Indexed: 12/22/2022]
Affiliation(s)
- Yochai Birnbaum
- The Department of Biochemistry and Molecular Biology, University of Texas Medical Branch, Galveston, TX, USA; The Section of Cardiology, Department of Medicine, Baylor College of Medicine, Houston, TX, USA
| | - Yumei Ye
- The Department of Biochemistry and Molecular Biology, University of Texas Medical Branch, Galveston, TX, USA
| | - Mandeep Bajaj
- Endocrinology and Diabetes Division, Department of Medicine, Baylor College of Medicine, Houston, TX, USA.
| |
Collapse
|
21
|
Rossello X, Yellon DM. A critical review on the translational journey of cardioprotective therapies! Int J Cardiol 2016; 220:176-84. [DOI: 10.1016/j.ijcard.2016.06.131] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/05/2016] [Revised: 06/03/2016] [Accepted: 06/23/2016] [Indexed: 01/08/2023]
|