1
|
Silvain J, Materne C, Zeitouni M, Procopi N, Guedeney P, Brugier D, Galier S, Lhomme M, Ponnaiah M, Guillas I, Kc P, Dahik VD, Frisdal E, Vicaut E, Lesnik P, Rahoual G, Le Goff W, Montalescot G, Kerneis M, Guerin M. Defective biological activities of high-density lipoprotein identify patients at highest risk of recurrent cardiovascular event. Eur J Prev Cardiol 2024:zwae356. [PMID: 39506545 DOI: 10.1093/eurjpc/zwae356] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/23/2024] [Revised: 10/07/2024] [Accepted: 11/05/2024] [Indexed: 11/08/2024]
Abstract
AIMS Low cholesterol efflux capacity and elevated levels of Interleukin-1ß (IL-1ß) are both associated with residual cardiovascular risk in patients with acute myocardial infarction (MI) and may be used as new biomarkers to identify patients at higher cardiovascular risk. METHODS We evaluated potential synergetic effect of cholesterol efflux capacity and IL-1ß on recurrent major adverse cardiovascular events (MACE) at one-year in 2012 patients with acute ST- segment elevation MI who underwent primary percutaneous coronary intervention. In addition, we evaluated the contribution to residual risk of HDL biological functions from 20 patients of the two extreme subgroups, focusing on cholesterol efflux capacity and anti-inflammatory properties. RESULTS Patients with MACE during the first year after the MI had significantly lower serum cholesterol efflux capacity as compared to those without recurrent events and higher level of IL-1ß, both associations were confirmed after multivariate analysis. We found an inverse relationship between CEC and circulating levels of the inflammatory markers IL-1ß, defining a very high risk (Low CEC/High IL-1ß) and a low risk (High CEC/Low IL-1ß) group of patients. Patients combining Low CEC/High IL-1ß exhibited the highest risk of recurrent MACE at one year showing an additive prognostic value of these biomarkers, regardless of all the other clinical or biological factors. In this very high-risk subgroup, patients exhibited reduced HDL-efflux capacity and defective ABCA1 and SR-BI with enhanced pro-inflammatory activity as a potential explanation for our clinical findings. CONCLUSION Impaired cholesterol efflux capacity and elevated IL-1β synergistically increase the residual cardiovascular risk in MI patients, which could be explained by reduced HDL-efflux capacity and enhanced HDL pro-inflammatory activity.
Collapse
Affiliation(s)
- Johanne Silvain
- ACTION Study Group, Institut de Cardiologie Hôpital Pitié-Salpêtrière (AP-HP), F-75013 Paris, France
- Sorbonne University, INSERM Unité de recherche sur les maladies cardiovasculaires, le métabolisme et la nutrition, UMR_S1166-ICAN F-75013 Paris, France
| | - Clément Materne
- Sorbonne University, INSERM Unité de recherche sur les maladies cardiovasculaires, le métabolisme et la nutrition, UMR_S1166-ICAN F-75013 Paris, France
| | - Michel Zeitouni
- ACTION Study Group, Institut de Cardiologie Hôpital Pitié-Salpêtrière (AP-HP), F-75013 Paris, France
- Sorbonne University, INSERM Unité de recherche sur les maladies cardiovasculaires, le métabolisme et la nutrition, UMR_S1166-ICAN F-75013 Paris, France
| | - Niki Procopi
- ACTION Study Group, Institut de Cardiologie Hôpital Pitié-Salpêtrière (AP-HP), F-75013 Paris, France
- Sorbonne University, INSERM Unité de recherche sur les maladies cardiovasculaires, le métabolisme et la nutrition, UMR_S1166-ICAN F-75013 Paris, France
| | - Paul Guedeney
- ACTION Study Group, Institut de Cardiologie Hôpital Pitié-Salpêtrière (AP-HP), F-75013 Paris, France
- Sorbonne University, INSERM Unité de recherche sur les maladies cardiovasculaires, le métabolisme et la nutrition, UMR_S1166-ICAN F-75013 Paris, France
| | - Delphine Brugier
- ACTION Study Group, Institut de Cardiologie Hôpital Pitié-Salpêtrière (AP-HP), F-75013 Paris, France
- Sorbonne University, INSERM Unité de recherche sur les maladies cardiovasculaires, le métabolisme et la nutrition, UMR_S1166-ICAN F-75013 Paris, France
| | - Sophie Galier
- Sorbonne University, INSERM Unité de recherche sur les maladies cardiovasculaires, le métabolisme et la nutrition, UMR_S1166-ICAN F-75013 Paris, France
| | - Marie Lhomme
- Foundation for Innovation in Cardiometabolism and Nutrition (IHU ICAN, ICAN OMICS and ICAN I/O), F-75013 Paris, France
| | - Maharajah Ponnaiah
- Foundation for Innovation in Cardiometabolism and Nutrition (IHU ICAN, ICAN OMICS and ICAN I/O), F-75013 Paris, France
| | - Isabelle Guillas
- Sorbonne University, INSERM Unité de recherche sur les maladies cardiovasculaires, le métabolisme et la nutrition, UMR_S1166-ICAN F-75013 Paris, France
| | - Pukar Kc
- Sorbonne University, INSERM Unité de recherche sur les maladies cardiovasculaires, le métabolisme et la nutrition, UMR_S1166-ICAN F-75013 Paris, France
| | - Veronica D Dahik
- Sorbonne University, INSERM Unité de recherche sur les maladies cardiovasculaires, le métabolisme et la nutrition, UMR_S1166-ICAN F-75013 Paris, France
| | - Eric Frisdal
- Sorbonne University, INSERM Unité de recherche sur les maladies cardiovasculaires, le métabolisme et la nutrition, UMR_S1166-ICAN F-75013 Paris, France
| | - Eric Vicaut
- ACTION Study Group, Institut de Cardiologie Hôpital Pitié-Salpêtrière (AP-HP), F-75013 Paris, France
- Unité de Recherche Clinique, Hôpital Fernand Widal (AP-HP), Paris, France. SAMM - Statistique, Analyse et Modélisation Multidisciplinaire EA 4543, Université Paris 1 Panthéon Sorbonne, France
| | - Philippe Lesnik
- Sorbonne University, INSERM Unité de recherche sur les maladies cardiovasculaires, le métabolisme et la nutrition, UMR_S1166-ICAN F-75013 Paris, France
| | - Ghilas Rahoual
- ACTION Study Group, Institut de Cardiologie Hôpital Pitié-Salpêtrière (AP-HP), F-75013 Paris, France
| | - Wilfried Le Goff
- Sorbonne University, INSERM Unité de recherche sur les maladies cardiovasculaires, le métabolisme et la nutrition, UMR_S1166-ICAN F-75013 Paris, France
| | - Gilles Montalescot
- ACTION Study Group, Institut de Cardiologie Hôpital Pitié-Salpêtrière (AP-HP), F-75013 Paris, France
- Sorbonne University, INSERM Unité de recherche sur les maladies cardiovasculaires, le métabolisme et la nutrition, UMR_S1166-ICAN F-75013 Paris, France
| | - Mathieu Kerneis
- ACTION Study Group, Institut de Cardiologie Hôpital Pitié-Salpêtrière (AP-HP), F-75013 Paris, France
- Sorbonne University, INSERM Unité de recherche sur les maladies cardiovasculaires, le métabolisme et la nutrition, UMR_S1166-ICAN F-75013 Paris, France
| | - Maryse Guerin
- Sorbonne University, INSERM Unité de recherche sur les maladies cardiovasculaires, le métabolisme et la nutrition, UMR_S1166-ICAN F-75013 Paris, France
| |
Collapse
|
2
|
Zormpas G, Boulmpou A, Potoupni V, Siskos F, Chatzipapa N, Fragakis N, Doumas M, Kassimis G, Vassilikos V, Papadopoulos CE. Identifying the Role of Flow-Mediated Dilatation Assessment in Acute Coronary Syndromes: A Systematic Review. Cardiol Rev 2024:00045415-990000000-00323. [PMID: 39254543 DOI: 10.1097/crd.0000000000000768] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 09/11/2024]
Abstract
In the context of the global burden of cardiovascular disease, the development of novel, patient-targeted diagnostic and therapeutic strategies is of paramount importance. Acute coronary syndromes (ACS) comprise a subset of cardiovascular disease, with constantly increasing prevalence requiring urgent attention. Flow-mediated dilatation (FMD), a noninvasive method for the evaluation of endothelial function, has been previously implemented in patients with ACS. A systematic review following Preferred Reporting Items for Systematic Reviews and Meta-Analyses guidelines was conducted in order to identify all relevant studies assessing the implementation of FMD among patients with ACS. Our review reflects an effort to present all available data regarding the role of FMD to date, a valuable noninvasive and easy accessible diagnostic tool, in the prognosis of patients with ACS. FMD evaluation in patients with ACS reveals a decline in values, indicative of the presence of endothelial function among this distinct patient group. FMD has also been used to assess the response to various treatments, as well as to predict major adverse cardiovascular events. Dynamic responses to interventions highlights its potential in the evolving field of interventional cardiology.
Collapse
Affiliation(s)
- Georgios Zormpas
- From the Second Department of Cardiology, Aristotle University of Thessaloniki, Ippokratio General Hospital of Thessaloniki, Greece
| | - Aristi Boulmpou
- Third Department of Cardiology, Aristotle University of Thessaloniki, Ippokratio General Hospital of Thessaloniki, Greece
| | - Victoria Potoupni
- Third Department of Cardiology, Aristotle University of Thessaloniki, Ippokratio General Hospital of Thessaloniki, Greece
| | - Fotios Siskos
- Second Propaedeutic Department of Internal Medicine, Aristotle University of Thessaloniki, Ippokratio General Hospital of Thessaloniki, Greece
| | - Nikoleta Chatzipapa
- Second Propaedeutic Department of Internal Medicine, Aristotle University of Thessaloniki, Ippokratio General Hospital of Thessaloniki, Greece
- Laboratory of Chemical and Environmental Technology, Department of Chemistry, Aristotle University of Thessaloniki, Greece
| | - Nikolaos Fragakis
- From the Second Department of Cardiology, Aristotle University of Thessaloniki, Ippokratio General Hospital of Thessaloniki, Greece
| | - Michael Doumas
- Second Propaedeutic Department of Internal Medicine, Aristotle University of Thessaloniki, Ippokratio General Hospital of Thessaloniki, Greece
| | - George Kassimis
- From the Second Department of Cardiology, Aristotle University of Thessaloniki, Ippokratio General Hospital of Thessaloniki, Greece
| | - Vassilios Vassilikos
- Third Department of Cardiology, Aristotle University of Thessaloniki, Ippokratio General Hospital of Thessaloniki, Greece
| | - Christodoulos E Papadopoulos
- Third Department of Cardiology, Aristotle University of Thessaloniki, Ippokratio General Hospital of Thessaloniki, Greece
| |
Collapse
|
3
|
Rexhaj E, Bär S, Soria R, Ueki Y, Häner JD, Otsuka T, Kavaliauskaite R, Siontis GC, Stortecky S, Shibutani H, Spirk D, Engstrøm T, Lang I, Morf L, Ambühl M, Windecker S, Losdat S, Koskinas KC, Räber L. Effects of alirocumab on endothelial function and coronary atherosclerosis in myocardial infarction: A PACMAN-AMI randomized clinical trial substudy. Atherosclerosis 2024; 392:117504. [PMID: 38513436 DOI: 10.1016/j.atherosclerosis.2024.117504] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/01/2023] [Revised: 02/22/2024] [Accepted: 02/27/2024] [Indexed: 03/23/2024]
Abstract
BACKGROUND AND AIMS The effects of protein convertase subtilisin/kexin type 9 (PCSK9) inhibitors on endothelial function as assessed by flow-mediated dilation (FMD) in patients with acute myocardial infarction (AMI) are unknown. Therefore, we aimed to investigate the effects of the PCSK9 inhibitor alirocumab added to high-intensity statin on FMD, and its association with coronary atherosclerosis in non-infarct related arteries using intracoronary intravascular ultrasound (IVUS), near-infrared spectroscopy (NIRS), and optical coherence tomography (OCT). METHODS This was a pre-specified substudy among patients recruited at Bern University Hospital, Switzerland, for the randomized-controlled, double-blind, PACMAN-AMI trial, which compared the effects of biweekly alirocumab 150 mg vs. placebo added to rosuvastatin. Brachial artery FMD was measured at 4 and 52 weeks, and intracoronary imaging at baseline and 52 weeks. RESULTS 139/173 patients completed the substudy. There was no difference in FMD at 52 weeks in the alirocumab (n = 68, 5.44 ± 2.24%) versus placebo (n = 71, 5.45 ± 2.19%) group (difference = -0.21%, 95% CI -0.77 to 0.35, p = 0.47). FMD improved throughout 52 weeks in both groups similarly (p < 0.001). There was a significant association between 4 weeks FMD and baseline plaque burden (IVUS) (n = 139, slope = -1.00, p = 0.006), but not with lipid pool (NIRS) (n = 139, slope = -7.36, p = 0.32), or fibrous cap thickness (OCT) (n = 81, slope = -1.57, p = 0.62). CONCLUSIONS Among patients with AMI, the addition of alirocumab did not result in further improvement of FMD as compared to 52 weeks secondary preventative medical therapy including high-intensity statin therapy. FMD was significantly associated with coronary plaque burden at baseline, but not with lipid pool or fibrous cap thickness.
Collapse
MESH Headings
- Humans
- Male
- Female
- Antibodies, Monoclonal, Humanized/therapeutic use
- Antibodies, Monoclonal, Humanized/pharmacology
- Middle Aged
- Coronary Artery Disease/drug therapy
- Coronary Artery Disease/diagnostic imaging
- Coronary Artery Disease/complications
- Endothelium, Vascular/drug effects
- Endothelium, Vascular/physiopathology
- Double-Blind Method
- Aged
- PCSK9 Inhibitors
- Myocardial Infarction/drug therapy
- Myocardial Infarction/complications
- Myocardial Infarction/diagnostic imaging
- Myocardial Infarction/physiopathology
- Ultrasonography, Interventional
- Rosuvastatin Calcium/therapeutic use
- Treatment Outcome
- Hydroxymethylglutaryl-CoA Reductase Inhibitors/therapeutic use
- Tomography, Optical Coherence
- Vasodilation/drug effects
- Drug Therapy, Combination
- Spectroscopy, Near-Infrared
- Plaque, Atherosclerotic/drug therapy
- Coronary Vessels/diagnostic imaging
- Coronary Vessels/drug effects
- Coronary Vessels/physiopathology
- Brachial Artery/drug effects
- Brachial Artery/physiopathology
- Brachial Artery/diagnostic imaging
- Time Factors
- Proprotein Convertase 9
Collapse
Affiliation(s)
- Emrush Rexhaj
- Department of Cardiology, Bern University Hospital Inselspital, Freiburgstrasse 18, 3010, Bern, Switzerland
| | - Sarah Bär
- Department of Cardiology, Bern University Hospital Inselspital, Freiburgstrasse 18, 3010, Bern, Switzerland
| | - Rodrigo Soria
- Department of Cardiology, Bern University Hospital Inselspital, Freiburgstrasse 18, 3010, Bern, Switzerland
| | - Yasushi Ueki
- Department of Cardiology, Bern University Hospital Inselspital, Freiburgstrasse 18, 3010, Bern, Switzerland
| | - Jonas D Häner
- Department of Cardiology, Bern University Hospital Inselspital, Freiburgstrasse 18, 3010, Bern, Switzerland
| | - Tatsuhiko Otsuka
- Department of Cardiology, Bern University Hospital Inselspital, Freiburgstrasse 18, 3010, Bern, Switzerland
| | - Raminta Kavaliauskaite
- Department of Cardiology, Bern University Hospital Inselspital, Freiburgstrasse 18, 3010, Bern, Switzerland
| | - George Cm Siontis
- Department of Cardiology, Bern University Hospital Inselspital, Freiburgstrasse 18, 3010, Bern, Switzerland
| | - Stefan Stortecky
- Department of Cardiology, Bern University Hospital Inselspital, Freiburgstrasse 18, 3010, Bern, Switzerland
| | - Hiroki Shibutani
- Department of Cardiology, Bern University Hospital Inselspital, Freiburgstrasse 18, 3010, Bern, Switzerland
| | - David Spirk
- Institute of Pharmacology, Bern University Hospital and University of Bern, Freiburgstrasse 18, 3010, Bern, Switzerland; Sanofi, Suurstofi 2, 6343, Risch-Rotkreuz, Switzerland
| | - Thomas Engstrøm
- Department of Cardiology, Rigshospitalet, Copenhagen University Hospital, Blegdamsvej 9, 20100, Copenhagen, Denmark
| | - Irene Lang
- Department of Cardiology, Medical University of Vienna, Währinger Gürtel 18-20, 1090, Vienna, Austria
| | - Laura Morf
- Department of Cardiology, Bern University Hospital Inselspital, Freiburgstrasse 18, 3010, Bern, Switzerland
| | - Maria Ambühl
- Department of Cardiology, Bern University Hospital Inselspital, Freiburgstrasse 18, 3010, Bern, Switzerland
| | - Stephan Windecker
- Department of Cardiology, Bern University Hospital Inselspital, Freiburgstrasse 18, 3010, Bern, Switzerland
| | - Sylvain Losdat
- CTU Bern, University of Bern, Mittelstrasse 43, 3012, Bern, Switzerland
| | - Konstantinos C Koskinas
- Department of Cardiology, Bern University Hospital Inselspital, Freiburgstrasse 18, 3010, Bern, Switzerland
| | - Lorenz Räber
- Department of Cardiology, Bern University Hospital Inselspital, Freiburgstrasse 18, 3010, Bern, Switzerland.
| |
Collapse
|
4
|
Carmo HRP, Bonilha I, Barreto J, Tognolini M, Zanotti I, Sposito AC. High-Density Lipoproteins at the Interface between the NLRP3 Inflammasome and Myocardial Infarction. Int J Mol Sci 2024; 25:1290. [PMID: 38279290 PMCID: PMC10816227 DOI: 10.3390/ijms25021290] [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: 12/30/2023] [Revised: 01/10/2024] [Accepted: 01/18/2024] [Indexed: 01/28/2024] Open
Abstract
Despite significant therapeutic advancements, morbidity and mortality following myocardial infarction (MI) remain unacceptably high. This clinical challenge is primarily attributed to two significant factors: delayed reperfusion and the myocardial injury resulting from coronary reperfusion. Following reperfusion, there is a rapid intracellular pH shift, disruption of ionic balance, heightened oxidative stress, increased activity of proteolytic enzymes, initiation of inflammatory responses, and activation of several cell death pathways, encompassing apoptosis, necroptosis, and pyroptosis. The inflammatory cell death or pyroptosis encompasses the activation of the intracellular multiprotein complex known as the NLRP3 inflammasome. High-density lipoproteins (HDL) are endogenous particles whose components can either promote or mitigate the activation of the NLRP3 inflammasome. In this comprehensive review, we explore the role of inflammasome activation in the context of MI and provide a detailed analysis of how HDL can modulate this process.
Collapse
Affiliation(s)
- Helison R. P. Carmo
- Atherosclerosis and Vascular Biology Laboratory (Aterolab), Division of Cardiology, State University of Campinas (UNICAMP), Campinas 13084-971, SP, Brazil; (H.R.P.C.); (I.B.); (J.B.); (A.C.S.)
| | - Isabella Bonilha
- Atherosclerosis and Vascular Biology Laboratory (Aterolab), Division of Cardiology, State University of Campinas (UNICAMP), Campinas 13084-971, SP, Brazil; (H.R.P.C.); (I.B.); (J.B.); (A.C.S.)
| | - Joaquim Barreto
- Atherosclerosis and Vascular Biology Laboratory (Aterolab), Division of Cardiology, State University of Campinas (UNICAMP), Campinas 13084-971, SP, Brazil; (H.R.P.C.); (I.B.); (J.B.); (A.C.S.)
| | | | - Ilaria Zanotti
- Department of Food and Drug, University of Parma, 43124 Parma, Italy;
| | - Andrei C. Sposito
- Atherosclerosis and Vascular Biology Laboratory (Aterolab), Division of Cardiology, State University of Campinas (UNICAMP), Campinas 13084-971, SP, Brazil; (H.R.P.C.); (I.B.); (J.B.); (A.C.S.)
| |
Collapse
|
5
|
Klobučar I, Stadler JT, Klobučar L, Lechleitner M, Trbušić M, Pregartner G, Berghold A, Habisch H, Madl T, Marsche G, Frank S, Degoricija V. Associations between Endothelial Lipase, High-Density Lipoprotein, and Endothelial Function Differ in Healthy Volunteers and Metabolic Syndrome Patients. Int J Mol Sci 2023; 24:2073. [PMID: 36768410 PMCID: PMC9916974 DOI: 10.3390/ijms24032073] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2022] [Revised: 01/13/2023] [Accepted: 01/17/2023] [Indexed: 01/21/2023] Open
Abstract
Metabolic syndrome (MS) is characterized by endothelial- and high-density lipoprotein (HDL) dysfunction and increased endothelial lipase (EL) serum levels. We examined the associations between EL serum levels, HDL (serum levels, lipid content, and function), and endothelial function in healthy volunteers (HV) and MS patients. Flow-mediated dilation (FMD), nitroglycerin-mediated dilation (NMD), serum levels of HDL subclasses (measured by nuclear magnetic resonance (NMR) spectroscopy), and EL serum levels differed significantly between HV and MS patients. The serum levels of triglycerides in large HDL particles were significantly positively correlated with FMD and NMD in HV, but not in MS patients. Cholesterol (C) and phospholipid (PL) contents of large HDL particles, calculated as HDL1-C/HDL1-apoA-I and HDL1-PL/HDL1-apoA-I, respectively, were significantly negatively correlated with FMD in HV, but not in MS patients. Cholesterol efflux capacity and arylesterase activity of HDL, as well as EL, were correlated with neither FMD nor NMD. EL was significantly negatively correlated with HDL-PL/HDL-apoA-I in HV, but not in MS patients, and with serum levels of small dense HDL containing apolipoprotein A-II in MS patients, but not in HV. We conclude that MS modulates the association between HDL and endothelial function, as well as between EL and HDL. HDL cholesterol efflux capacity and arylesterase activity, as well as EL serum levels, are not associated with endothelial function in HV or MS patients.
Collapse
Affiliation(s)
- Iva Klobučar
- Department of Cardiology, Sisters of Charity University Hospital Centre, 10000 Zagreb, Croatia
| | - Julia T. Stadler
- Otto Loewi Research Center, Division of Pharmacology, Medical University of Graz, 8010 Graz, Austria
| | - Lucija Klobučar
- Department of Medicine, University Hospital Centre Osijek, 31000 Osijek, Croatia
| | - Margarete Lechleitner
- Gottfried Schatz Research Center, Molecular Biology and Biochemistry, Medical University of Graz, 8010 Graz, Austria
| | - Matias Trbušić
- Department of Cardiology, Sisters of Charity University Hospital Centre, 10000 Zagreb, Croatia
- School of Medicine, University of Zagreb, 10000 Zagreb, Croatia
| | - Gudrun Pregartner
- Institute for Medical Informatics, Statistics und Documentation, Medical University of Graz, 8036 Graz, Austria
| | - Andrea Berghold
- Institute for Medical Informatics, Statistics und Documentation, Medical University of Graz, 8036 Graz, Austria
| | - Hansjörg Habisch
- Gottfried Schatz Research Center, Molecular Biology and Biochemistry, Medical University of Graz, 8010 Graz, Austria
| | - Tobias Madl
- Gottfried Schatz Research Center, Molecular Biology and Biochemistry, Medical University of Graz, 8010 Graz, Austria
- BioTechMed-Graz, 8010 Graz, Austria
| | - Gunther Marsche
- Otto Loewi Research Center, Division of Pharmacology, Medical University of Graz, 8010 Graz, Austria
- BioTechMed-Graz, 8010 Graz, Austria
| | - Saša Frank
- Gottfried Schatz Research Center, Molecular Biology and Biochemistry, Medical University of Graz, 8010 Graz, Austria
- BioTechMed-Graz, 8010 Graz, Austria
| | - Vesna Degoricija
- School of Medicine, University of Zagreb, 10000 Zagreb, Croatia
- Department of Medicine, Sisters of Charity University Hospital Centre, 10000 Zagreb, Croatia
| |
Collapse
|
6
|
Engineering chitosan nano-cocktail containing iron oxide and ceria: A two-in-one approach for treatment of inflammatory diseases and tracking of material delivery. MATERIALS SCIENCE & ENGINEERING. C, MATERIALS FOR BIOLOGICAL APPLICATIONS 2021; 131:112477. [PMID: 34857262 DOI: 10.1016/j.msec.2021.112477] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/13/2021] [Revised: 09/19/2021] [Accepted: 10/01/2021] [Indexed: 01/07/2023]
Abstract
In this study, modular two-in-one nano-cocktails were synthesised to provide treatment of inflammatory diseases and also enable tracking of their delivery to the disease sites. Chitosan nano-cocktails loaded with treatment module (cerium oxide nanoparticles) and imaging module (iron oxide nanoparticles) were synthesised by electrostatic self-assembly (Chit-IOCO) and ionic gelation method (Chit-TPP-IOCO), respectively. Their MRI capability, anti-inflammatory and anti-fibrosis ability were investigated. Results demonstrated that Chit-IOCO significantly reduced the expression of TNF-α and COX-2, while Chit-TPP-IOCO reduced IL-6 in the LPS-stimulated macrophages RAW264.7. Cytotoxicity studies showed that the nano-cocktails inhibited the proliferation of macrophages. Additionally, Chit-IOCO exhibited higher in vitro MRI relaxivity than Chit-TPP-IOCO, indicating that Chit-IOCO is a better MRI contrast agent in macrophages. It was possible to track the delivery of Chit-IOCO to the inflamed livers of CCl4-treated C57BL/6 mice, demonstrated by a shortened T2⁎ relaxation time of the livers after injecting Chit-IOCO into mice. In vivo anti-inflammatory and blood tests demonstrated that Chit-IOCO reduced inflammation-related proteins (TNF-a, iNOS and Cox-2) and bilirubin in CCl4 treated C57BL/6. Histology images indicated that the nano-cocktails at the treatment doses did not affect the organs of the mice. Importantly, the nano-cocktail reduced fibrosis of CCl4-treated mouse liver. This is the first reported data on the anti-inflammation and anti-fibrosis efficacy of Chit-IOCO in C57BL/6 mouse liver inflammation model. Overall, Chit-IOCO nanoparticles have shown great potential in MR imaging/detecting and treating/therapeutic capabilities for inflammatory diseases.
Collapse
|
7
|
Sposito AC, de Lima-Junior JC, Moura FA, Barreto J, Bonilha I, Santana M, Virginio VW, Sun L, Carvalho LSF, Soares AA, Nadruz W, Feinstein SB, Nofer JR, Zanotti I, Kontush A, Remaley AT. Reciprocal Multifaceted Interaction Between HDL (High-Density Lipoprotein) and Myocardial Infarction. Arterioscler Thromb Vasc Biol 2019; 39:1550-1564. [DOI: 10.1161/atvbaha.119.312880] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Despite decades of therapeutic advances, myocardial infarction remains a leading cause of death worldwide. Recent studies have identified HDLs (high-density lipoproteins) as a potential candidate for mitigating coronary ischemia/reperfusion injury via a broad spectrum of signaling pathways. HDL ligands, such as S1P (sphingosine-1-phosphate), Apo (apolipoprotein) A-I, clusterin, and miRNA, may influence the opening of the mitochondrial channel, insulin sensitivity, and production of vascular autacoids, such as NO, prostacyclin, and endothelin-1. In parallel, antioxidant activity and sequestration of oxidized molecules provided by HDL can attenuate the oxidative stress that triggers ischemia/reperfusion. Nevertheless, during myocardial infarction, oxidation and the capture of oxidized and proinflammatory molecules generate large phenotypic and functional changes in HDL, potentially limiting its beneficial properties. In this review, new findings from cellular and animal models, as well as from clinical studies, will be discussed to describe the cardioprotective benefits of HDL on myocardial infarction. Furthermore, mechanisms by which HDL modulates cardiac function and potential strategies to mitigate postmyocardial infarction risk damage by HDL will be detailed throughout the review.
Collapse
Affiliation(s)
- Andrei C. Sposito
- From the Atherosclerosis and Vascular Biology Laboratory, Cardiology Department, State University of Campinas, Brazil (A.C.S., J.C.d.L.-J., F.A.M., J.B., I.B., M.S., V.W.V., L.S.F.C., A.A.S.S., W.N.)
| | - José Carlos de Lima-Junior
- From the Atherosclerosis and Vascular Biology Laboratory, Cardiology Department, State University of Campinas, Brazil (A.C.S., J.C.d.L.-J., F.A.M., J.B., I.B., M.S., V.W.V., L.S.F.C., A.A.S.S., W.N.)
| | - Filipe A. Moura
- From the Atherosclerosis and Vascular Biology Laboratory, Cardiology Department, State University of Campinas, Brazil (A.C.S., J.C.d.L.-J., F.A.M., J.B., I.B., M.S., V.W.V., L.S.F.C., A.A.S.S., W.N.)
- Department of Medicine, Weill-Cornell Medical College, New York, NY (F.A.M.)
| | - Joaquim Barreto
- From the Atherosclerosis and Vascular Biology Laboratory, Cardiology Department, State University of Campinas, Brazil (A.C.S., J.C.d.L.-J., F.A.M., J.B., I.B., M.S., V.W.V., L.S.F.C., A.A.S.S., W.N.)
| | - Isabella Bonilha
- From the Atherosclerosis and Vascular Biology Laboratory, Cardiology Department, State University of Campinas, Brazil (A.C.S., J.C.d.L.-J., F.A.M., J.B., I.B., M.S., V.W.V., L.S.F.C., A.A.S.S., W.N.)
| | - Michele Santana
- From the Atherosclerosis and Vascular Biology Laboratory, Cardiology Department, State University of Campinas, Brazil (A.C.S., J.C.d.L.-J., F.A.M., J.B., I.B., M.S., V.W.V., L.S.F.C., A.A.S.S., W.N.)
| | - Vitor W. Virginio
- From the Atherosclerosis and Vascular Biology Laboratory, Cardiology Department, State University of Campinas, Brazil (A.C.S., J.C.d.L.-J., F.A.M., J.B., I.B., M.S., V.W.V., L.S.F.C., A.A.S.S., W.N.)
| | - Lufan Sun
- Lipoprotein Metabolism Section, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD (L.S., A.T.R.)
- Department of Cardiology, The First Hospital of China Medical University, Shenyang, Liaoning Province, China (L.S.)
| | - Luiz Sergio F. Carvalho
- From the Atherosclerosis and Vascular Biology Laboratory, Cardiology Department, State University of Campinas, Brazil (A.C.S., J.C.d.L.-J., F.A.M., J.B., I.B., M.S., V.W.V., L.S.F.C., A.A.S.S., W.N.)
| | - Alexandre A.S. Soares
- From the Atherosclerosis and Vascular Biology Laboratory, Cardiology Department, State University of Campinas, Brazil (A.C.S., J.C.d.L.-J., F.A.M., J.B., I.B., M.S., V.W.V., L.S.F.C., A.A.S.S., W.N.)
| | - Wilson Nadruz
- From the Atherosclerosis and Vascular Biology Laboratory, Cardiology Department, State University of Campinas, Brazil (A.C.S., J.C.d.L.-J., F.A.M., J.B., I.B., M.S., V.W.V., L.S.F.C., A.A.S.S., W.N.)
| | - Steve B. Feinstein
- Division of Cardiology, Rush University Medical Center, Chicago, IL (S.B.F.)
| | - Jerzy-Roch Nofer
- Institute of Clinical Chemistry and Laboratory Medicine, University Medical Center Hamburg-Eppendorf, Germany (J.-R.N.)
| | - Ilaria Zanotti
- Department of Food and Drugs, University of Parma, Italy (I.Z.)
| | - Anatol Kontush
- UMR-ICAN 1166, National Institute for Health and Medical Research (INSERM), Sorbonne University, Paris, France (A.K.)
| | - Alan T. Remaley
- Lipoprotein Metabolism Section, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD (L.S., A.T.R.)
| |
Collapse
|
8
|
Association of extremely high levels of high-density lipoprotein cholesterol with endothelial dysfunction in men. J Clin Lipidol 2019; 13:664-672.e1. [DOI: 10.1016/j.jacl.2019.06.004] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2019] [Revised: 05/23/2019] [Accepted: 06/06/2019] [Indexed: 11/21/2022]
|
9
|
Cabou C, Honorato P, Briceño L, Ghezali L, Duparc T, León M, Combes G, Frayssinhes L, Fournel A, Abot A, Masri B, Parada N, Aguilera V, Aguayo C, Knauf C, González M, Radojkovic C, Martinez LO. Pharmacological inhibition of the F 1 -ATPase/P2Y 1 pathway suppresses the effect of apolipoprotein A1 on endothelial nitric oxide synthesis and vasorelaxation. Acta Physiol (Oxf) 2019; 226:e13268. [PMID: 30821416 DOI: 10.1111/apha.13268] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2018] [Revised: 02/14/2019] [Accepted: 02/22/2019] [Indexed: 12/12/2022]
Abstract
AIM The contribution of apolipoprotein A1 (APOA1), the major apolipoprotein of high-density lipoprotein (HDL), to endothelium-dependent vasodilatation is unclear, and there is little information regarding endothelial receptors involved in this effect. Ecto-F1 -ATPase is a receptor for APOA1, and its activity in endothelial cells is coupled to adenosine diphosphate (ADP)-sensitive P2Y receptors (P2Y ADP receptors). Ecto-F1 -ATPase is involved in APOA1-mediated cell proliferation and HDL transcytosis. Here, we investigated the effect of lipid-free APOA1 and the involvement of ecto-F1 -ATPase and P2Y ADP receptors on nitric oxide (NO) synthesis and the regulation of vascular tone. METHOD Nitric oxide synthesis was assessed in human endothelial cells from umbilical veins (HUVECs) and isolated mouse aortas. Changes in vascular tone were evaluated by isometric force measurements in isolated human umbilical and placental veins and by assessing femoral artery blood flow in conscious mice. RESULTS Physiological concentrations of lipid-free APOA1 enhanced endothelial NO synthesis, which was abolished by inhibitors of endothelial nitric oxide synthase (eNOS) and of the ecto-F1 -ATPase/P2Y1 axis. Accordingly, APOA1 inhibited vasoconstriction induced by thromboxane A2 receptor agonist and increased femoral artery blood flow in mice. These effects were blunted by inhibitors of eNOS, ecto-F1 -ATPase and P2Y1 receptor. CONCLUSIONS Using a pharmacological approach, we thus found that APOA1 promotes endothelial NO production and thereby controls vascular tone in a process that requires activation of the ecto-F1 -ATPase/P2Y1 pathway by APOA1. Pharmacological targeting of this pathway with respect to vascular diseases should be explored.
Collapse
Affiliation(s)
- Cendrine Cabou
- INSERM, UMR1048, Institute of Metabolic and Cardiovascular Diseases University of Toulouse, Paul Sabatier University Toulouse France
- Department of Human Physiology, Faculty of Pharmacy University Paul Sabatier Toulouse France
| | - Paula Honorato
- Department of Clinical Biochemistry and Immunology, Faculty of Pharmacy Universidad de Concepción Concepción Chile
| | - Luis Briceño
- Department of Clinical Biochemistry and Immunology, Faculty of Pharmacy Universidad de Concepción Concepción Chile
| | - Lamia Ghezali
- INSERM, UMR1048, Institute of Metabolic and Cardiovascular Diseases University of Toulouse, Paul Sabatier University Toulouse France
| | - Thibaut Duparc
- INSERM, UMR1048, Institute of Metabolic and Cardiovascular Diseases University of Toulouse, Paul Sabatier University Toulouse France
| | - Marcelo León
- Department of Clinical Biochemistry and Immunology, Faculty of Pharmacy Universidad de Concepción Concepción Chile
| | - Guillaume Combes
- INSERM, UMR1048, Institute of Metabolic and Cardiovascular Diseases University of Toulouse, Paul Sabatier University Toulouse France
| | - Laure Frayssinhes
- INSERM, UMR1048, Institute of Metabolic and Cardiovascular Diseases University of Toulouse, Paul Sabatier University Toulouse France
| | - Audren Fournel
- UMR 1220, IRSD, INSERM, INRA, ENVT, European Associated Laboratory NeuroMicrobiota (INSERM/UCL) University of Toulouse Toulouse France
| | - Anne Abot
- UMR 1220, IRSD, INSERM, INRA, ENVT, European Associated Laboratory NeuroMicrobiota (INSERM/UCL) University of Toulouse Toulouse France
| | - Bernard Masri
- INSERM, UMR1048, Institute of Metabolic and Cardiovascular Diseases University of Toulouse, Paul Sabatier University Toulouse France
| | - Nicol Parada
- Department of Clinical Biochemistry and Immunology, Faculty of Pharmacy Universidad de Concepción Concepción Chile
| | - Valeria Aguilera
- Department of Clinical Biochemistry and Immunology, Faculty of Pharmacy Universidad de Concepción Concepción Chile
| | - Claudio Aguayo
- Department of Clinical Biochemistry and Immunology, Faculty of Pharmacy Universidad de Concepción Concepción Chile
- Group of Research and Innovation in Vascular Health (GRIVAS Health) Chillan Chile
| | - Claude Knauf
- UMR 1220, IRSD, INSERM, INRA, ENVT, European Associated Laboratory NeuroMicrobiota (INSERM/UCL) University of Toulouse Toulouse France
| | - Marcelo González
- Group of Research and Innovation in Vascular Health (GRIVAS Health) Chillan Chile
- Vascular Physiology Laboratory, Department of Physiology, Faculty of Biological Sciences, and Department of Obstetrics and Gynecology, Faculty of Medicine Universidad de Concepción Concepción Chile
| | - Claudia Radojkovic
- Department of Clinical Biochemistry and Immunology, Faculty of Pharmacy Universidad de Concepción Concepción Chile
| | - Laurent O. Martinez
- INSERM, UMR1048, Institute of Metabolic and Cardiovascular Diseases University of Toulouse, Paul Sabatier University Toulouse France
| |
Collapse
|
10
|
Hernáez Á, Soria-Florido MT, Schröder H, Ros E, Pintó X, Estruch R, Salas-Salvadó J, Corella D, Arós F, Serra-Majem L, Martínez-González MÁ, Fiol M, Lapetra J, Elosua R, Lamuela-Raventós RM, Fitó M. Role of HDL function and LDL atherogenicity on cardiovascular risk: A comprehensive examination. PLoS One 2019; 14:e0218533. [PMID: 31246976 PMCID: PMC6597156 DOI: 10.1371/journal.pone.0218533] [Citation(s) in RCA: 31] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2018] [Accepted: 06/04/2019] [Indexed: 11/18/2022] Open
Abstract
BACKGROUND High-density lipoprotein (HDL) functionality and low-density lipoprotein (LDL) atherogenic traits can describe the role of both particles on cardiovascular diseases more accurately than HDL- or LDL-cholesterol levels. However, it is unclear how these lipoprotein properties are particularly affected by different cardiovascular risk factors. OBJECTIVE To determine which lipoprotein properties are associated with greater cardiovascular risk scores and each cardiovascular risk factor. METHODS In two cross-sectional baseline samples of PREDIMED trial volunteers, we assessed the associations of HDL functionality (N = 296) and LDL atherogenicity traits (N = 210) with: 1) the 10-year predicted coronary risk (according to the Framingham-REGICOR score), and 2) classical cardiovascular risk factors. RESULTS Greater cardiovascular risk scores were associated with low cholesterol efflux values; oxidized, triglyceride-rich, small HDL particles; and small LDLs with low resistance against oxidation (P-trend<0.05, all). After adjusting for the rest of risk factors; 1) type-2 diabetic individuals presented smaller and more oxidized LDLs (P<0.026, all); 2) dyslipidemic participants had smaller HDLs with an impaired capacity to metabolize cholesterol (P<0.035, all); 3) high body mass index values were associated to lower HDL and LDL size and a lower HDL capacity to esterify cholesterol (P<0.037, all); 4) men presented a greater HDL oxidation and lower HDL vasodilatory capacity (P<0.046, all); and 5) greater ages were related to small, oxidized, cytotoxic LDL particles (P<0.037, all). CONCLUSIONS Dysfunctional HDL and atherogenic LDL particles are present in high cardiovascular risk patients. Dyslipidemia and male sex are predominantly linked to HDL dysfunctionality, whilst diabetes and advanced age are associated with LDL atherogenicity.
Collapse
Affiliation(s)
- Álvaro Hernáez
- Cardiovascular Risk, Nutrition and Aging Research Unit, August Pi i Sunyer Biomedical Research Institute (IDIBAPS), Barcelona, Spain
- Blanquerna School of Life Sciences, Universitat Ramón Llull, Barcelona, Spain
- CIBER de Fisiopatología de la Obesidad y Nutrición (CIBEROBN), Instituto de Salud Carlos III (ISCIII), Madrid, Spain
| | - María Trinidad Soria-Florido
- Cardiovascular Risk and Nutrition Research Group, Hospital del Mar Medical Research Institute (IMIM), Barcelona, Spain
- PhD Program in Food Sciences and Nutrition, Universitat de Barcelona, Barcelona, Spain
| | - Helmut Schröder
- Cardiovascular Risk and Nutrition Research Group, Hospital del Mar Medical Research Institute (IMIM), Barcelona, Spain
- CIBER de Epidemiología y Salud Pública (CIBERESP), ISCIII, Madrid, Spain
| | - Emilio Ros
- Cardiovascular Risk, Nutrition and Aging Research Unit, August Pi i Sunyer Biomedical Research Institute (IDIBAPS), Barcelona, Spain
- CIBER de Fisiopatología de la Obesidad y Nutrición (CIBEROBN), Instituto de Salud Carlos III (ISCIII), Madrid, Spain
- Lipid Clinic, Endocrinology and Nutrition Service, Hospital Clínic, Barcelona, Spain
| | - Xavier Pintó
- CIBER de Fisiopatología de la Obesidad y Nutrición (CIBEROBN), Instituto de Salud Carlos III (ISCIII), Madrid, Spain
- Lipids and Vascular Risk Unit, Internal Medicine Service, Hospital Universitario de Bellvitge, Hospitalet de Llobregat, Spain
| | - Ramón Estruch
- Cardiovascular Risk, Nutrition and Aging Research Unit, August Pi i Sunyer Biomedical Research Institute (IDIBAPS), Barcelona, Spain
- CIBER de Fisiopatología de la Obesidad y Nutrición (CIBEROBN), Instituto de Salud Carlos III (ISCIII), Madrid, Spain
- Internal Medicine Service, Hospital Clínic, Barcelona, Spain
| | - Jordi Salas-Salvadó
- CIBER de Fisiopatología de la Obesidad y Nutrición (CIBEROBN), Instituto de Salud Carlos III (ISCIII), Madrid, Spain
- Human Nutrition Unit, Hospital Universitari Sant Joan, Institut d’Investigació Sanitaria Pere Virgili, Universitat Rovira i Virgili, Reus, Spain
| | - Dolores Corella
- CIBER de Fisiopatología de la Obesidad y Nutrición (CIBEROBN), Instituto de Salud Carlos III (ISCIII), Madrid, Spain
- Department of Preventive Medicine, Universidad de Valencia, Valencia, Spain
| | - Fernando Arós
- CIBER de Fisiopatología de la Obesidad y Nutrición (CIBEROBN), Instituto de Salud Carlos III (ISCIII), Madrid, Spain
- Department of Cardiology, Hospital Universitario de Álava, Vitoria, Spain
| | - Lluis Serra-Majem
- Department of Clinical Sciences & Research Institute of Biomedical and Health Sciences, Universidad de Las Palmas de Gran Canaria, Las Palmas, Spain
| | - Miguel Ángel Martínez-González
- CIBER de Fisiopatología de la Obesidad y Nutrición (CIBEROBN), Instituto de Salud Carlos III (ISCIII), Madrid, Spain
- Department of Preventive Medicine and Public Health, Universidad de Navarra, Pamplona, Spain
- Department of Nutrition, Harvard TH Chan School of Public Health, Boston, Massachusetts, United States of America
| | - Miquel Fiol
- CIBER de Fisiopatología de la Obesidad y Nutrición (CIBEROBN), Instituto de Salud Carlos III (ISCIII), Madrid, Spain
- Balearic Islands Health Research Institute, Hospital Son Espases, Palma de Mallorca, Spain
| | - José Lapetra
- CIBER de Fisiopatología de la Obesidad y Nutrición (CIBEROBN), Instituto de Salud Carlos III (ISCIII), Madrid, Spain
- Department of Family Medicine, Research Unit, Distrito Sanitario Atención Primaria Sevilla, Sevilla, Spain
| | - Roberto Elosua
- Cardiovascular Epidemiology and Genetics-REGICOR Research Group, Hospital del Mar Medical Research Institute (IMIM), Barcelona, Spain
- CIBER de Enfermedades Cardiovasculares (CIBERCV), ISCIII, Madrid, Spain
| | - Rosa María Lamuela-Raventós
- CIBER de Fisiopatología de la Obesidad y Nutrición (CIBEROBN), Instituto de Salud Carlos III (ISCIII), Madrid, Spain
- Department of Nutrition and Bromatology, Faculty of Pharmacy, Universitat de Barcelona, Barcelona, Spain
| | - Montserrat Fitó
- CIBER de Fisiopatología de la Obesidad y Nutrición (CIBEROBN), Instituto de Salud Carlos III (ISCIII), Madrid, Spain
- Cardiovascular Risk and Nutrition Research Group, Hospital del Mar Medical Research Institute (IMIM), Barcelona, Spain
| |
Collapse
|
11
|
|
12
|
Shahzad S, Mateen S, Naeem SS, Akhtar K, Rizvi W, Moin S. Syringic acid protects from isoproterenol induced cardiotoxicity in rats. Eur J Pharmacol 2019; 849:135-145. [PMID: 30731086 DOI: 10.1016/j.ejphar.2019.01.056] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2018] [Revised: 01/23/2019] [Accepted: 01/30/2019] [Indexed: 01/26/2023]
Abstract
Identification of pharmacologically potent antioxidant compounds for their use in preventive medicine is thrust area of current research. This study was undertaken with the aim of determining the protective role of syringic acid (SA) on isoproterenol (ISO) induced myocardial infarction (MI) in rats. SA was orally given to rats for 21 days at three different concentrations (12.5, 25 and 50 mg/kg). At 20th and 21st day, rats were subcutaneously injected with ISO and at the end of experimental period, rats were killed. ISO induced myocardial damage was averted by pre-co-treatment of SA, as decrease was found in serum level of marker enzymes (CKMB, LDH, AST, ALT), lipid peroxidation, protein carbonyl (PC) and proinflammatory cytokines (TNFα, IL 6). Furthermore, content of glutathione (GSH) and activities of antioxidant enzymes in heart tissue were significantly raised. Improvement in infarct size and erythrocyte (RBCs) morphology was also observed. The biochemical findings were supported by histopathological outcome and protective effect of SA was found to be dose dependent. The results of our study demonstrated that the cardioprotective potential of SA in rat model of ISO induced MI might be due to anti-lipid peroxidative and endogenous antioxidant system enhancement effects.
Collapse
Affiliation(s)
- Sumayya Shahzad
- Department of Biochemistry, Jawaharlal Nehru Medical College, Faculty of Medicine, Aligarh Muslim University, Aligarh 202002, Uttar Pradesh, India
| | - Somaiya Mateen
- Department of Biochemistry, Jawaharlal Nehru Medical College, Faculty of Medicine, Aligarh Muslim University, Aligarh 202002, Uttar Pradesh, India
| | - Syed Shariq Naeem
- Department of Pharmacology, Jawaharlal Nehru Medical College, Faculty of Medicine, Aligarh Muslim University, Aligarh 202002, Uttar Pradesh, India
| | - Kafil Akhtar
- Department of Pathology, Jawaharlal Nehru Medical College, Faculty of Medicine, Aligarh Muslim University, Aligarh 202002, Uttar Pradesh, India
| | - Waseem Rizvi
- Department of Pharmacology, Jawaharlal Nehru Medical College, Faculty of Medicine, Aligarh Muslim University, Aligarh 202002, Uttar Pradesh, India
| | - Shagufta Moin
- Department of Biochemistry, Jawaharlal Nehru Medical College, Faculty of Medicine, Aligarh Muslim University, Aligarh 202002, Uttar Pradesh, India.
| |
Collapse
|
13
|
Soares AA, Carvalho LSF, Bonilha I, Virginio VW, Nadruz Junior W, Coelho-Filho OR, Quinaglia e Silva JC, Petrucci Junior O, Sposito AC. Adverse interaction between HDL and the mass of myocardial infarction. Atherosclerosis 2019; 281:9-16. [DOI: 10.1016/j.atherosclerosis.2018.12.002] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/26/2018] [Revised: 11/21/2018] [Accepted: 12/04/2018] [Indexed: 01/20/2023]
|
14
|
Shahzad S, Mateen S, Mubeena Mariyath PM, Naeem SS, Akhtar K, Rizvi W, Moin S. Protective effect of syringaldehyde on biomolecular oxidation, inflammation and histopathological alterations in isoproterenol induced cardiotoxicity in rats. Biomed Pharmacother 2018; 108:625-633. [PMID: 30245462 DOI: 10.1016/j.biopha.2018.09.055] [Citation(s) in RCA: 31] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2018] [Revised: 09/07/2018] [Accepted: 09/08/2018] [Indexed: 11/15/2022] Open
Abstract
BACKGROUND Ischemic injury during myocardial infarction (MI) is responsible for increased deaths among patients with cardiovascular disorders. Recently, research has been directed for finding treatment using natural compounds. This study was performed to investigate the effects of syrigaldehyde (SYD), a phytochemical against isoproterenol (ISO) induced cardiotoxicity model. METHODS For induction of MI, rats were intoxicated with two doses of ISO and were treated with SYD at three different concentrations (12.5, 25 & 50 mg/kg) both prior and simultaneous to ISO administration. RESULTS ISO group revealed amplified activity of marker enzymes (CKMB, LDH, AST, ALT), increased oxidation of proteins and lipid molecules. Moreover, augmentation in pro-inflammatory markers was also found. The same group also displayed marked changes in histopathology and erythrocyte (RBCs) morphology. SYD treated groups showed diminished levels of serum markers enzymes, lipid peroxidation and protein carbonyl (PC) with increment in antioxidant defense in cardiac tissues of ISO administered rats. Our findings also revealed the modulatory effect of SYD on membrane bound ATPases, showing that SYD significantly improved the ISO induced changes in membrane fluidity. Furthermore, decline in infarct size, alleviation of structural RBC damage and improved myocardial histopathological outcome were observed in treated groups. In addition, mitigation of biochemical and histopathological changes by SYD was found to be dependent on its concentration. CONCLUSION SYD had cardioprotective efficacy owing to its antioxidative and anti-inflammatory properties. Our results support incorporation of SYD in regular diet for prevention of MI.
Collapse
Affiliation(s)
- Sumayya Shahzad
- Department of Biochemistry, Jawaharlal Nehru Medical College, Faculty of Medicine, Aligarh Muslim University, Aligarh, 202002, Uttar Pradesh, India
| | - Somaiya Mateen
- Department of Biochemistry, Jawaharlal Nehru Medical College, Faculty of Medicine, Aligarh Muslim University, Aligarh, 202002, Uttar Pradesh, India
| | - P M Mubeena Mariyath
- Interdisciplinary Brain Research Unit, Jawaharlal Nehru Medical College, Faculty of Medicine, Aligarh Muslim University, Aligarh, 202002, Uttar Pradesh, India
| | - Syed Shariq Naeem
- Department of Pharmacology, Jawaharlal Nehru Medical College, Faculty of Medicine, Aligarh Muslim University, Aligarh, 202002, Uttar Pradesh, India
| | - Kafil Akhtar
- Department of Pathology, Jawaharlal Nehru Medical College, Faculty of Medicine, Aligarh Muslim University, Aligarh, 202002, Uttar Pradesh, India
| | - Waseem Rizvi
- Department of Pharmacology, Jawaharlal Nehru Medical College, Faculty of Medicine, Aligarh Muslim University, Aligarh, 202002, Uttar Pradesh, India
| | - Shagufta Moin
- Department of Biochemistry, Jawaharlal Nehru Medical College, Faculty of Medicine, Aligarh Muslim University, Aligarh, 202002, Uttar Pradesh, India.
| |
Collapse
|
15
|
Haghikia A, Landmesser U. Lipoproteins and Cardiovascular Redox Signaling: Role in Atherosclerosis and Coronary Disease. Antioxid Redox Signal 2018; 29:337-352. [PMID: 28817963 DOI: 10.1089/ars.2017.7052] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/21/2023]
Abstract
SIGNIFICANCE Lipoproteins, such as low-density lipoprotein, play a causal role in the development of atherosclerosis and coronary disease. Recent Advances: Lipoproteins can stimulate vascular production of reactive oxygen species, which act as important signaling molecules in the cardiovascular system contributing to the pathophysiology of endothelial dysfunction, hypertension, and atherosclerosis. CRITICAL ISSUES Modified lipoproteins have emerged as important regulators of redox signaling, such as oxidized or carbamylated low-density lipoprotein or modified high-density lipoproteins, that contain oxidized lipids, an altered protein cargo, and associated small molecules, such as symmetric dimethylarginine. FUTURE DIRECTIONS In this review, we provide an overview on signaling pathways stimulated by modified lipoproteins in the cardiovascular system and their potential role in cardiovascular disease development. Moreover, we highlight novel aspects of how gut microbiome-related mechanisms-a growing research field-may contribute to lipoprotein modification with subsequent impact on cardiovascular redox signaling. Antioxid. Redox Signal. 29, 337-352.
Collapse
Affiliation(s)
- Arash Haghikia
- 1 Department of Cardiology, Charité Universitätsmedizin Berlin , Berlin, Germany
- 2 German Center for Cardiovascular Research (DZHK) , partner site Berlin, Berlin, Germany
| | - Ulf Landmesser
- 1 Department of Cardiology, Charité Universitätsmedizin Berlin , Berlin, Germany
- 2 German Center for Cardiovascular Research (DZHK) , partner site Berlin, Berlin, Germany
- 3 Berlin Institute of Health (BIH) , Berlin, Germany
| |
Collapse
|
16
|
Shao Y, Zhao H, Wang Y, Liu J, Li J, Chai H, Xing M. Arsenic and/or copper caused inflammatory response via activation of inducible nitric oxide synthase pathway and triggered heat shock protein responses in testis tissues of chicken. ENVIRONMENTAL SCIENCE AND POLLUTION RESEARCH INTERNATIONAL 2018; 25:7719-7729. [PMID: 29288301 DOI: 10.1007/s11356-017-1042-7] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/01/2017] [Accepted: 12/12/2017] [Indexed: 06/07/2023]
Abstract
The aim of this study is to investigate the effects of arsenic (As) and copper (Cu) on the inflammatory response, and the protective roles of heat shock proteins (Hsps) in chicken testes. Seventy-two 1-day-old male Hy-line chickens were treated with 30 mg/kg feed of arsenic trioxide (As2O3) and/or 300 mg/kg feed of copper sulfate (CuSO4) for 4, 8, and 12 weeks. The histological changes, inducible nitric oxide synthase (iNOS) activity, and the expressions of Hsps and inflammatory cytokines were detected. The results showed that slight histology changes were obvious in the testis tissue exposure to treatment groups. The activity and the protein level of iNOS were increased compared to the control group. The mRNA levels of proinflammatory cytokines and inflammatory factors were increased as a whole. However, anti-inflammatory cytokines were inhibited. The mRNA and protein levels of Hsp60, Hsp70, and Hsp90 were upregulated. These results suggested that sub-chronic exposure to As and/or Cu induced testicular poisoning in chickens. Increased Hsps tried to protect chicken testis tissues from tissues damage caused by inflammation. In conclusion, testicular poisoning induced by As and/or Cu caused inflammatory response and heat shock protein response in chicken testis tissues.
Collapse
Affiliation(s)
- Yizhi Shao
- College of Wildlife Resource, Northeast Forestry University, Harbin, 150040, Heilongjiang Province, People's Republic of China
| | - Hongjing Zhao
- College of Wildlife Resource, Northeast Forestry University, Harbin, 150040, Heilongjiang Province, People's Republic of China
| | - Yu Wang
- College of Wildlife Resource, Northeast Forestry University, Harbin, 150040, Heilongjiang Province, People's Republic of China
| | - Juanjuan Liu
- College of Wildlife Resource, Northeast Forestry University, Harbin, 150040, Heilongjiang Province, People's Republic of China
| | - Jinglun Li
- College of Wildlife Resource, Northeast Forestry University, Harbin, 150040, Heilongjiang Province, People's Republic of China
| | - Hongliang Chai
- College of Wildlife Resource, Northeast Forestry University, Harbin, 150040, Heilongjiang Province, People's Republic of China.
| | - Mingwei Xing
- College of Wildlife Resource, Northeast Forestry University, Harbin, 150040, Heilongjiang Province, People's Republic of China.
| |
Collapse
|
17
|
HDL acceptor capacities for cholesterol efflux from macrophages and lipid transfer are both acutely reduced after myocardial infarction. Clin Chim Acta 2018; 478:51-56. [DOI: 10.1016/j.cca.2017.12.031] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2017] [Revised: 12/15/2017] [Accepted: 12/20/2017] [Indexed: 01/16/2023]
|
18
|
Momordica charantia polysaccharides ameliorate oxidative stress, hyperlipidemia, inflammation, and apoptosis during myocardial infarction by inhibiting the NF-κB signaling pathway. Int J Biol Macromol 2017; 97:544-551. [PMID: 28109806 DOI: 10.1016/j.ijbiomac.2017.01.074] [Citation(s) in RCA: 90] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2016] [Revised: 01/02/2017] [Accepted: 01/15/2017] [Indexed: 01/07/2023]
Abstract
The polysaccharide extract of Momordica charantia has various biological activities; however, its effect on endothelial dysfunction in myocardial infarction remains unclear. To elucidate this, myocardial infarction was induced in rats using isoproterenol (ISP). Pretreatment with M. charantia polysaccharides (MCP; 150 or 300mg/kg) for 25days significantly inhibited increases in heart weight, the heart-weight-to-body-weight ratio, and infarction size, and ameliorated the increased serum levels of aspartate transaminase, creatine kinase, lactate dehydrogenase, total cholesterol, triglycerides, very-low-density lipoprotein cholesterol, low-density lipoprotein cholesterol, and high-density lipoprotein cholesterol. In addition, MCP enhanced the activity of superoxide dismutase, catalase, and non-protein sulfhydryls, and decreased the level of lipid peroxidation. Moreover, MCP pretreatment downregulated the expression of proinflammatory cytokines (tumor necrosis factor alpha, interleukin (IL)-6, and IL-10), inflammatory markers (nitric oxide, myeloperoxidase, and inducible nitric oxide synthase), and apoptotic markers (caspase-3 and BAX), and upregulated Bcl-2 expression. Pretreatment with MCP reduced myonecrosis, edema, and inflammatory cell infiltration, and restored cardiomyocytes architecture. This myocardial protective effect could be related to the enhancement of the antioxidant defense system through the nuclear factor kappa B (NF-kB) pathways, and to anti-apoptosis through regulation of Bax, caspase-3, and Bcl-2.
Collapse
|
19
|
Sposito AC. HDL metrics, let’s call the number thing off? Atherosclerosis 2016; 251:525-527. [DOI: 10.1016/j.atherosclerosis.2016.06.044] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/19/2016] [Revised: 06/24/2016] [Accepted: 06/28/2016] [Indexed: 10/21/2022]
|
20
|
Papageorgiou N, Zacharia E, Androulakis E, Briasoulis A, Charakida M, Tousoulis D. HDL as a prognostic biomarker for coronary atherosclerosis: the role of inflammation. Expert Opin Ther Targets 2016; 20:907-921. [PMID: 26854521 DOI: 10.1517/14728222.2016.1152264] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2015] [Accepted: 02/05/2016] [Indexed: 01/18/2023]
Abstract
INTRODUCTION Emerging evidence suggests that the role of high density lipoprotein (HDL) in the atherosclerotic process is not as clear as previously thought, since atheroprotective HDL becomes atherogenic in states of increased inflammatory processes. AREAS COVERED In this review we aim to elucidate the role of HDL as a prognostic biomarker and we discuss therapeutic approaches that aim to increase HDL and their possible clinical benefit. EXPERT OPINION Given the structural variability and biological complexity of the HDL particle, its role in the atherosclerotic process is far from clear. According to current evidence, the atheroprotective role of HDL turns atherogenic in states of increased inflammatory processes, while even minor alterations in systemic inflammation are likely to hinder the endothelial protective effects of HDL. In accordance, significant data have revealed that HDL-related drugs may be effective in reducing cardiovascular mortality; however they are not as encouraging or unanimous as expected. Possible future goals could be to quantify either HDL subclasses or functions in an attempt to reach safer conclusions as to the prognostic importance of HDL in coronary atherosclerosis. Having achieved that, a more targeted therapy that would aim to raise either HDL functionality or to remodel HDL structure would be more easily designed.
Collapse
Affiliation(s)
| | - Effimia Zacharia
- b 1st Department of Cardiology , Hippokration Hospital, University of Athens , Athens , Greece
| | | | - Alexandros Briasoulis
- d Division of Cardiology , Wayne State University/Detroit Medical Center , Detroit , MI , USA
| | - Marietta Charakida
- e Vascular Physiology Unit, Institute of Cardiovascular Science , University College London , London , UK
| | - Dimitris Tousoulis
- b 1st Department of Cardiology , Hippokration Hospital, University of Athens , Athens , Greece
| |
Collapse
|
21
|
Distelmaier K, Schrutka L, Wurm R, Seidl V, Arfsten H, Cho A, Manjunatha S, Perkmann T, Strunk G, Lang IM, Adlbrecht C. Gender-related impact on outcomes of high density lipoprotein in acute ST-elevation myocardial infarction. Atherosclerosis 2016; 251:460-466. [PMID: 27381657 DOI: 10.1016/j.atherosclerosis.2016.06.037] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/12/2016] [Revised: 06/08/2016] [Accepted: 06/22/2016] [Indexed: 01/22/2023]
Abstract
BACKGROUND AND AIMS There is rising evidence that cardioprotective functions of high-density lipoprotein (HDL) have significant impact on clinical outcomes. ST-elevation myocardial infarction (STEMI) represents a high-risk vascular condition. Whether higher HDL-cholesterol concentrations in women correspond to protective anti-oxidant properties in the setting of STEMI is unknown. METHODS We prospectively assessed gender related differences in the anti-oxidant function of HDL, and the impact of HDL properties on mortality in 242 women and men with STEMI. Blood samples to determine HDL function and sex hormone levels were collected during primary percutaneous coronary intervention. RESULTS Patients were stratified according to preserved anti-oxidant HDL function (HDL oxidant index (HOI) < 1) and pro-oxidant HDL (HOI≥1). Despite higher serum levels of HDL-cholesterol in postmenopausal women (48 mg/dl, IQR 42-54, versus 39 mg/dl, IQR33-47, p < 0.001 in men), the proportion of patients with pro-oxidant HDL was not different between women (35%) and men (46%, p = 0.132). Kaplan-Meier analysis revealed higher cardiovascular mortality in both women (p = 0.021) and men (p = 0.045) with pro-oxidant HDL. We identified pro-oxidant HDL as strong and independent predictor of cardiovascular mortality with an adjusted HR of 8.33 (95% CI, 1.55-44.63; p = 0.013) in women and with an adjusted HR of 5.14 (95% CI, 1.61-16.42; p = 0.006) in men. Higher levels of free sex hormones (estradiol and testosterone) were associated with pro-oxidant HDL. HDL-cholesterol levels showed no association with mortality (HR in women 1.03, 95% CI 0.96-1.11, p = 0.45 and HR in men 0.99, 95% CI 0.94-1.05, p = 0.72). CONCLUSIONS Total HDL-cholesterol serum levels were not associated with mortality in STEMI patients. Pro-oxidant HDL was a strong and independent predictor of mortality in women and men with STEMI. The present study provides a link between sex hormones, HDL function and clinical events in STEMI patients. In clinical practice and future clinical trials, anti-oxidant properties of HDL rather than total HDL serum levels should be used for risk stratification.
Collapse
Affiliation(s)
- K Distelmaier
- Department of Internal Medicine II, Division of Cardiology, Medical University of Vienna, Austria
| | - L Schrutka
- Department of Internal Medicine II, Division of Cardiology, Medical University of Vienna, Austria
| | - R Wurm
- Department of Internal Medicine II, Division of Cardiology, Medical University of Vienna, Austria
| | - V Seidl
- Department of Internal Medicine II, Division of Cardiology, Medical University of Vienna, Austria
| | - H Arfsten
- Department of Internal Medicine II, Division of Cardiology, Medical University of Vienna, Austria
| | - A Cho
- Department of Internal Medicine II, Division of Cardiology, Medical University of Vienna, Austria
| | - S Manjunatha
- Division of Endocrinology and Metabolism, Mayo Clinic College of Medicine, Rochester, MN, USA
| | - T Perkmann
- Department of Medical and Chemical Laboratory Diagnostics, Medical University of Vienna, Vienna, Austria
| | - G Strunk
- Complexity-Research, Research Institute for Complex Systems, Vienna, Austria
| | - I M Lang
- Department of Internal Medicine II, Division of Cardiology, Medical University of Vienna, Austria.
| | - C Adlbrecht
- Department of Internal Medicine II, Division of Cardiology, Medical University of Vienna, Austria; 4th Medical Department, Hietzing Hospital, Vienna, Austria
| |
Collapse
|
22
|
Wang YF, Han Y, Zhang R, Qin L, Wang MX, Ma L. CETP/LPL/LIPC gene polymorphisms and susceptibility to age-related macular degeneration. Sci Rep 2015; 5:15711. [PMID: 26503844 PMCID: PMC4621603 DOI: 10.1038/srep15711] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2015] [Accepted: 08/18/2015] [Indexed: 12/17/2022] Open
Abstract
Three high-density lipoprotein (HDL)-related loci have been reported to be associated with age-related macular degeneration (AMD), but the results were inconsistent. In this study, the cholesteryl ester transfer protein (CETP) rs3764261 variant was significantly associated with an increased risk of AMD (odds ratio [OR] = 1.13, 95% confidence interval [CI]: 1.05–1.21, P < 0.001), and the hepatic lipase (LIPC) rs10468017 variant was associated with a significantly decreased risk of AMD (OR = 0.81, CI: 0.76–0.86, P < 0.001). Individuals carrying the lipoprotein lipase (LPL) rs12678919 polymorphism (A → G) had no significant change in the risk of developing AMD (OR = 1.01, CI: 0.92–1.10, P = 0.17). After adjusting for the complement factor H (CFH) gene, both CETP and LPL conferred a significantly increased AMD risk (ORCETP = 1.17, CI: 1.08–1.26, P < 0.001; ORLPL = 1.11, CI: 1.01–1.22, P = 0.02). Subgroup analysis based on ethnicity revealed a significant association between the CETP variant and AMD in both Americans (OR = 1.12, CI: 1.02–1.23, P = 0.01) and Europeans (OR = 1.10, CI: 1.01–1.19, P = 0.011). This meta-analysis revealed that both CETP rs3764261 and LIPC rs10468017 polymorphisms were significantly associated with AMD risk. After adjustment for the CFH gene, CETP/LPL conferred a significantly increased susceptibility to the disease, indicating potential interactions among genes in the complement system and the lipid metabolism pathway.
Collapse
Affiliation(s)
- Ya-Feng Wang
- School of Public Health, Xi'an Jiao tong University Health Science Center, Xi'an, China
| | - Yue Han
- School of Public Health, Xi'an Jiao tong University Health Science Center, Xi'an, China
| | - Rui Zhang
- School of Public Health, Xi'an Jiao tong University Health Science Center, Xi'an, China
| | - Li Qin
- The First Affiliated Hospital, Xi'an Jiao tong University College of Medicine, Xi'an, China
| | - Ming-Xu Wang
- School of Public Health, Xi'an Jiao tong University Health Science Center, Xi'an, China
| | - Le Ma
- School of Public Health, Xi'an Jiao tong University Health Science Center, Xi'an, China
| |
Collapse
|
23
|
Abstract
High-density lipoproteins (HDLs) protect against atherosclerosis by removing excess cholesterol from macrophages through the ATP-binding cassette transporter A1 (ABCA1) and ATP-binding cassette transporter G1 (ABCG1) pathways involved in reverse cholesterol transport. Factors that impair the availability of functional apolipoproteins or the activities of ABCA1 and ABCG1 could, therefore, strongly influence atherogenesis. HDL also inhibits lipid oxidation, restores endothelial function, exerts anti-inflammatory and antiapoptotic actions, and exerts anti-inflammatory actions in animal models. Such properties could contribute considerably to the capacity of HDL to inhibit atherosclerosis. Systemic and vascular inflammation has been proposed to convert HDL to a dysfunctional form that has impaired antiatherogenic effects. A loss of anti-inflammatory and antioxidative proteins, perhaps in combination with a gain of proinflammatory proteins, might be another important component in rendering HDL dysfunctional. The proinflammatory enzyme myeloperoxidase induces both oxidative modification and nitrosylation of specific residues on plasma and arterial apolipoprotein A-I to render HDL dysfunctional, which results in impaired ABCA1 macrophage transport, the activation of inflammatory pathways, and an increased risk of coronary artery disease. Understanding the features of dysfunctional HDL or apolipoprotein A-I in clinical practice might lead to new diagnostic and therapeutic approaches to atherosclerosis.
Collapse
|
24
|
Thymoquinone Protects against Myocardial Ischemic Injury by Mitigating Oxidative Stress and Inflammation. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE 2015; 2015:143629. [PMID: 26101531 PMCID: PMC4458551 DOI: 10.1155/2015/143629] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/05/2014] [Accepted: 02/21/2015] [Indexed: 02/01/2023]
Abstract
The present study was aimed at investigating the cardioprotective activity of thymoquinone (TMQ), an active principle of the herb, Nigella sativa, which is used for the management of various diseases. The present study examined the cardioprotective effect of TMQ in isoproterenol- (ISP-) induced myocardial infarction in rats. Myocardial infarction was induced by two subcutaneous injections of ISP (85 mg/kg) at an interval of 24 hr. TMQ (20 mg/kg) was administered orally for 21 days. ISP-treated rats showed depletion of antioxidants and marker enzymes from myocardium along with lipid peroxidation and enhanced levels of proinflammatory cytokines. ISP also induced histopathological alterations in myocardium. Treatment with TMQ prevented the depletion of endogenous antioxidants and myocyte injury marker enzymes and inhibited lipid peroxidation as well as reducing the levels of proinflammatory cytokines. TMQ pretreatment also reduced myonecrosis, edema, and infiltration of inflammatory cells and showed preservation of cardiomyocytes histoarchitecture. The present study results demonstrate that TMQ exerts cardioprotective effect by mitigating oxidative stress, augmenting endogenous antioxidants, and maintaining structural integrity. The results of the present study indicate that TMQ may serve as an excellent agent alone or as adjuvant to prevent the onset and progression of myocardial injury.
Collapse
|
25
|
Martin SS, Joshi PH, Blaha MJ. High-density lipoprotein and endothelial function in patients with myocardial infarction: Pieces in a puzzle. Atherosclerosis 2014; 237:838-9. [PMID: 25463130 PMCID: PMC4470401 DOI: 10.1016/j.atherosclerosis.2014.10.026] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/08/2014] [Accepted: 10/19/2014] [Indexed: 11/21/2022]
Affiliation(s)
- Seth S Martin
- Ciccarone Center for the Prevention of Heart Disease, Johns Hopkins University School of Medicine, Baltimore, MD, USA.
| | - Parag H Joshi
- Ciccarone Center for the Prevention of Heart Disease, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Michael J Blaha
- Ciccarone Center for the Prevention of Heart Disease, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| |
Collapse
|