1
|
Shi F, Chowdhury R, Sofianopoulou E, Koulman A, Sun L, Steur M, Aleksandrova K, Dahm CC, Schulze MB, van der Schouw YT, Agnoli C, Amiano P, Boer JMA, Bork CS, Cabrera-Castro N, Eichelmann F, Elbaz A, Farràs M, Heath AK, Kaaks R, Katzke V, Keski-Rahkonen P, Masala G, Moreno-Iribas C, Panico S, Papier K, Petrova D, Quirós JR, Ricceri F, Severi G, Tjønneland A, Tong TYN, Tumino R, Wareham NJ, Weiderpass E, Di Angelantonio E, Forouhi NG, Danesh J, Butterworth AS, Kaptoge S. Association of circulating fatty acids with cardiovascular disease risk: analysis of individual-level data in three large prospective cohorts and updated meta-analysis. Eur J Prev Cardiol 2025; 32:233-246. [PMID: 39365172 PMCID: PMC11832215 DOI: 10.1093/eurjpc/zwae315] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/24/2024] [Revised: 05/21/2024] [Accepted: 08/20/2024] [Indexed: 10/05/2024]
Abstract
AIMS Associations of saturated and unsaturated fatty acids (FAs) with cardiovascular disease (CVD) remain controversial. We therefore aimed to investigate the prospective associations of objectively measured FAs with CVD, including incident coronary heart disease (CHD) and stroke, as well as CVD mortality. METHODS AND RESULTS Circulating FA concentrations expressed as the percentage of total FAs were assayed in 172 891 participants without prior vascular disease at baseline from the European Prospective Investigation into Cancer and Nutrition-CVD (EPIC-CVD) (7343 CHD; 6499 stroke), UK Biobank (1825; 1474), and INTERVAL (285; 209) cohort studies. Hazard ratio (HR) per 1-standard deviation (SD) higher FA concentrations was estimated using Cox regression models and pooled by random-effects meta-analysis. Systematic reviews with meta-analysis published by 6 May 2023 on associations between FAs and CVDs were systematically searched and updated meta-analyses using random-effects model were conducted. Evidence from randomized controlled trials (RCTs) was also summarized. Higher concentrations of total saturated FAs (SFAs) were associated with higher cardiovascular risks in the combined analysis, with differential findings noted for SFA sub-types in further analysis restricted to EPIC-CVD: positive associations for even-chain SFA [HR for CHD 1.24 (95% CI: 1.18-1.32); stroke 1.23 (1.10-1.38)] and negative associations for odd-chain [0.82 (0.76-0.87); 0.73 (0.67-0.78)] and longer-chain [0.95 (0.80-1.12); 0.84 (0.72-0.99)] SFA. In the combined analysis, total n-3 polyunsaturated FA (PUFA) [0.91 (0.85-0.97)], including docosahexaenoic acid (DHA) [0.91 (0.84-0.98)], was negatively associated with incident CHD risk. Similarly, total n-6 PUFA [0.94 (0.91-0.98)], including linoleic acid (LA) [0.89 (0.83-0.95)], was negatively associated with incident stroke risk. In contrast, more detailed analyses in EPIC-CVD revealed that several downstream n-6 PUFAs of LA were positively associated with CHD risk. Updated meta-analyses of 37 FAs including 49 non-overlapping studies, involving between 7787 and 22 802 CHD cases and between 6499 and 14 221 stroke cases, showed broadly similar results as our combined empirical analysis and further suggested significant inverse associations of individual long-chain n-3 PUFAs and LA on both CHD and stroke. The findings of long-chain n-3 PUFAs were consistent with those from published RCTs on CHD despite insufficient evidence in monotherapy, while RCT evidence remained unclear for the rest of the explored FAs. CONCLUSION Our study provides an overview of the most recent evidence on the associations between objectively measured FAs and CVD outcomes. Collectively, the data reveal notable differences in associations by SFA sub-types and call for further studies, especially RCTs, to explore these links.
Collapse
Affiliation(s)
- Fanchao Shi
- BHF Cardiovascular Epidemiology Unit, Department of Public Health and Primary Care, University of Cambridge, Cambridge CB2 0BB, UK
- Victor Phillip Dahdaleh Heart and Lung Research Institute, University of Cambridge, Papworth Road, Cambridge Biomedical Campus, Cambridge CB2 0BB, UK
| | - Rajiv Chowdhury
- Stempel College of Public Health and Social Work, Florida International University, Miami, FL, USA
| | - Eleni Sofianopoulou
- BHF Cardiovascular Epidemiology Unit, Department of Public Health and Primary Care, University of Cambridge, Cambridge CB2 0BB, UK
- Victor Phillip Dahdaleh Heart and Lung Research Institute, University of Cambridge, Papworth Road, Cambridge Biomedical Campus, Cambridge CB2 0BB, UK
| | - Albert Koulman
- Medical Research Council Epidemiology Unit, Institute of Metabolic Science, University of Cambridge, Cambridge, UK
| | - Luanluan Sun
- BHF Cardiovascular Epidemiology Unit, Department of Public Health and Primary Care, University of Cambridge, Cambridge CB2 0BB, UK
- Victor Phillip Dahdaleh Heart and Lung Research Institute, University of Cambridge, Papworth Road, Cambridge Biomedical Campus, Cambridge CB2 0BB, UK
| | - Marinka Steur
- Division of Human Nutrition and Health, Wageningen University, Wageningen, The Netherlands
| | - Krasimira Aleksandrova
- Biomarkers and Metabolism Research Group, Department of Epidemiological Methods and Etiological Research, Leibniz Institute for Prevention Research and Epidemiology, Bremen, Germany
- Faculty of Human and Health Sciences, University of Bremen, Bremen, Germany
| | | | - Matthias B Schulze
- Department of Molecular Epidemiology, German Institute of Human Nutrition Potsdam-Rehbruecke, Nuthetal, Germany
- Institute of Nutritional Science, University of Potsdam, Nuthetal, Germany
| | - Yvonne T van der Schouw
- Julius Center for Health Sciences and Primary Care, University Medical Center Utrecht, Utrecht University, Utrecht, The Netherlands
| | - Claudia Agnoli
- Epidemiology and Prevention Unit, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | - Pilar Amiano
- Centro de Investigación Biomédica en Red de Epidemiología y Salud Pública (CIBERESP), Madrid, Spain
- Ministry of Health of the Basque Government Sub Directorate for Public Health and Addictions of Gipuzkoa, San Sebastian, Spain
- Biodonostia Health Research Institute, Epidemiology of Chronic and Communicable Diseases Group, San Sebastian, Spain
| | - Jolanda M A Boer
- National Institute of Public Health and the Environment (RIVM), Bilthoven, The Netherlands
| | - Christian S Bork
- Department of Cardiology, Aalborg University Hospital, Aalborg, Denmark
| | - Natalia Cabrera-Castro
- Centro de Investigación Biomédica en Red de Epidemiología y Salud Pública (CIBERESP), Madrid, Spain
- Department of Epidemiology, Regional Health Council, IMIB-Arrixaca, Murcia, Spain
| | - Fabian Eichelmann
- Department of Molecular Epidemiology, German Institute of Human Nutrition Potsdam-Rehbruecke, Nuthetal, Germany
- German Center for Diabetes Research (DZD), Neuherberg, Germany
| | - Alexis Elbaz
- Paris-Saclay University, UVSQ, Inserm, Gustave Roussy, CESP, Villejuif, France
| | - Marta Farràs
- Unit of Nutrition and Cancer, Epidemiology Research Program, Catalan Institute of Oncology (ICO), Bellvitge Biomedical Research Institute (IDIBELL), 08908 L’Hospitalet de Llobregat, Spain
| | - Alicia K Heath
- Department of Epidemiology and Biostatistics, School of Public Health, Imperial College London, London, UK
| | - Rudolf Kaaks
- Department of Cancer Epidemiology, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Verena Katzke
- Department of Cancer Epidemiology, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Pekka Keski-Rahkonen
- Nutrition and Metabolism Branch, International Agency for Research on Cancer (IARC/WHO), Lyon, France
| | - Giovanna Masala
- Clinical Epidemiology Unit, Institute for cancer research, prevention and clinical network (ISPRO), Florence, Italy
| | - Conchi Moreno-Iribas
- Instituto de Salud Pública y Laboral de Navarra, 31003 Pamplona, Spain
- Centro de Investigación Biomédica en Red de Epidemiología y Salud Pública (CIBERESP), 28029 Madrid, Spain
- Navarra Institute for Health Research (IdiSNA), 31008 Pamplona, Spain
| | | | - Keren Papier
- Cancer Epidemiology Unit, Nuffield Department of Population Health, University of Oxford, Oxford, UK
| | - Dafina Petrova
- Centro de Investigación Biomédica en Red de Epidemiología y Salud Pública (CIBERESP), 28029 Madrid, Spain
- Escuela Andaluza de Salud Pública (EASP), 18011 Granada, Spain
- Instituto de Investigación Biosanitaria ibsGRANADA, 18012 Granada, Spain
| | | | - Fulvio Ricceri
- Centre for Biostatistics, Epidemiology, and Public Health (C-BEPH), Department of Clinical and Biological Sciences, University of Turin, Turin, Italy
| | - Gianluca Severi
- Paris-Saclay University, UVSQ, Inserm, Gustave Roussy, CESP, Villejuif, France
- Department of Statistics, Computer Science, Applications ‘G Parenti’, University of Florence, Florence, Italy
| | - Anne Tjønneland
- The Danish Cancer Institute, Strandboulevarden 49, 2100 Copenhagen O, Denmark
- Department of Public Health, University of Copenghagen, Copenhagen, Denmark
| | - Tammy Y N Tong
- Cancer Epidemiology Unit, Nuffield Department of Population Health, University of Oxford, Oxford, UK
| | - Rosario Tumino
- Hyblean Association for Epidemiological Research (AIRE-ONLUS), Ragusa, Italy
| | - Nicholas J Wareham
- Medical Research Council Epidemiology Unit, Institute of Metabolic Science, University of Cambridge, Cambridge, UK
| | | | - Emanuele Di Angelantonio
- BHF Cardiovascular Epidemiology Unit, Department of Public Health and Primary Care, University of Cambridge, Cambridge CB2 0BB, UK
- Victor Phillip Dahdaleh Heart and Lung Research Institute, University of Cambridge, Papworth Road, Cambridge Biomedical Campus, Cambridge CB2 0BB, UK
- BHF Centre of Research Excellence, School of Clinical Medicine, Addenbrooke's Hospital, University of Cambridge, Cambridge, UK
- Health Data Research UK Cambridge, Wellcome Genome Campus and University of Cambridge, Hinxton, UK
- NIHR Blood and Transplant Research Unit in Donor Health and Behaviour, University of Cambridge, Cambridge CB2 0BB, UK
- Health Data Science Centre, Human Technopole, Milan, Italy
| | - Nita G Forouhi
- Medical Research Council Epidemiology Unit, Institute of Metabolic Science, University of Cambridge, Cambridge, UK
| | - John Danesh
- BHF Cardiovascular Epidemiology Unit, Department of Public Health and Primary Care, University of Cambridge, Cambridge CB2 0BB, UK
- Victor Phillip Dahdaleh Heart and Lung Research Institute, University of Cambridge, Papworth Road, Cambridge Biomedical Campus, Cambridge CB2 0BB, UK
- BHF Centre of Research Excellence, School of Clinical Medicine, Addenbrooke's Hospital, University of Cambridge, Cambridge, UK
- Health Data Research UK Cambridge, Wellcome Genome Campus and University of Cambridge, Hinxton, UK
- NIHR Blood and Transplant Research Unit in Donor Health and Behaviour, University of Cambridge, Cambridge CB2 0BB, UK
- Department of Human Genetics, Wellcome Sanger Institute, Hinxton, UK
| | - Adam S Butterworth
- BHF Cardiovascular Epidemiology Unit, Department of Public Health and Primary Care, University of Cambridge, Cambridge CB2 0BB, UK
- Victor Phillip Dahdaleh Heart and Lung Research Institute, University of Cambridge, Papworth Road, Cambridge Biomedical Campus, Cambridge CB2 0BB, UK
- BHF Centre of Research Excellence, School of Clinical Medicine, Addenbrooke's Hospital, University of Cambridge, Cambridge, UK
- Health Data Research UK Cambridge, Wellcome Genome Campus and University of Cambridge, Hinxton, UK
- NIHR Blood and Transplant Research Unit in Donor Health and Behaviour, University of Cambridge, Cambridge CB2 0BB, UK
| | - Stephen Kaptoge
- BHF Cardiovascular Epidemiology Unit, Department of Public Health and Primary Care, University of Cambridge, Cambridge CB2 0BB, UK
- Victor Phillip Dahdaleh Heart and Lung Research Institute, University of Cambridge, Papworth Road, Cambridge Biomedical Campus, Cambridge CB2 0BB, UK
- NIHR Blood and Transplant Research Unit in Donor Health and Behaviour, University of Cambridge, Cambridge CB2 0BB, UK
| |
Collapse
|
2
|
Fock E, Parnova R. Omega-3 polyunsaturated fatty acids in the brain and visual system: Focus on invertebrates. Comp Biochem Physiol B Biochem Mol Biol 2025; 275:111023. [PMID: 39154851 DOI: 10.1016/j.cbpb.2024.111023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2024] [Revised: 08/14/2024] [Accepted: 08/14/2024] [Indexed: 08/20/2024]
Abstract
A critical role of omega-3 polyunsaturated fatty acids (PUFA), mainly docosahexaenoic acid 22:6ω3 (DHA), in the development and function of the brain and visual system is well established. DHA, the most abundant omega-3 PUFA in the vertebrate brain, contributes to neuro- and synaptogenesis, neuronal differentiation, synaptic transmission and plasticity, neuronal network formation, memory and behaviour formation. Based on these data, the unique importance of DHA and its irreplaceability in neural and retinal tissues has been postulated. In this review, we consider omega-3 PUFA composition in the brain and retina of various invertebrates, and show that DHA has only been found in marine mollusks and crustaceans. A gradual decrease in the DHA content until its disappearance can be observed in the brain lipids of the series marine-freshwater-terrestrial crustaceans and marine-terrestrial mollusks, suggesting that the transition to the land lifestyle in the evolution of invertebrates, but not vertebrates, was accompanied by a loss of DHA. As with terrestrial crustaceans and mollusks, DHA was not found in insects, either terrestrial or aquatic, or in nematodes. We show that the nervous and visual systems of various DHA-free invertebrates can be highly enriched in alpha-linolenic acid 18:3ω3 or eicosapentaenoic acid 20:5ω3, which affect neurological and visual function, stimulating synaptogenesis, synaptic transmission, visual processing, learning and even cognition. The review data show that, in animals at different levels of organization, omega-3 PUFA are required for the functioning of the nervous and visual systems and that their specific needs can be met by various omega-3 PUFA.
Collapse
Affiliation(s)
- Ekaterina Fock
- Sechenov Institute of Evolutionary Physiology and Biochemistry of the Russian Academy of Sciences, 194223, Torez Av., 44, Saint-Petersburg, Russia
| | - Rimma Parnova
- Sechenov Institute of Evolutionary Physiology and Biochemistry of the Russian Academy of Sciences, 194223, Torez Av., 44, Saint-Petersburg, Russia.
| |
Collapse
|
3
|
Capece U, Gugliandolo S, Morciano C, Avolio A, Splendore A, Di Giuseppe G, Ciccarelli G, Soldovieri L, Brunetti M, Mezza T, Pontecorvi A, Giaccari A, Cinti F. Erythrocyte Membrane Fluidity and Omega-3 Fatty Acid Intake: Current Outlook and Perspectives for a Novel, Nutritionally Modifiable Cardiovascular Risk Factor. Nutrients 2024; 16:4318. [PMID: 39770939 PMCID: PMC11676811 DOI: 10.3390/nu16244318] [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: 11/11/2024] [Revised: 12/07/2024] [Accepted: 12/12/2024] [Indexed: 01/11/2025] Open
Abstract
Omega-3 fatty acids reduce triglycerides and have several positive effects on different organs and systems. They are also found in the plasma membrane in variable amounts in relation to genetics and diet. However, it is still unclear whether omega-3 supplementation can reduce the occurrence of major cardiovascular events (MACEs). Two trials, REDUCE-IT (Reduction of Cardiovascular Events with Icosapent Ethyl-Intervention Trial), with highly purified EPA, and STRENGTH (Effect of High-Dose Omega-3 Fatty Acids vs. Corn Oil on Major Adverse Cardiovascular Events in Patients at High Cardiovascular Risk), with a combination of EPA and DHA, have produced different outcomes, triggering a scientific debate on possible explanations for the discrepancies. Furthermore, doubts have arisen as to the anti-inflammatory and anti-aggregating activity of these compounds. Recent studies have, however, highlighted interesting effects of EPA and DHA on erythrocyte membrane fluidity (EMF). EMF is governed by a complex and dynamic biochemical framework, with fatty acids playing a central role. Furthermore, it can be easily measured in erythrocytes from a blood sample using fluorescent probes. Recent research has also shown that EMF could act as a possible cardiovascular risk factor biomarker. This review aims to synthetize the latest evidence on erythrocyte membrane fluidity, exploring its potential role as a biomarker of residual cardiovascular risk and discussing its clinical relevance. Further, we aim to dissect the possible biological mechanisms that link omega-3 modifiable membrane fluidity to cardiovascular health.
Collapse
Affiliation(s)
- Umberto Capece
- Endocrinologia e Diabetologia, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, 00136 Rome, Italy
- Dipartimento di Medicina e Chirurgia Traslazionale, Università Cattolica del Sacro Cuore, 00168 Rome, Italy
| | - Shawn Gugliandolo
- Endocrinologia e Diabetologia, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, 00136 Rome, Italy
- Dipartimento di Medicina e Chirurgia Traslazionale, Università Cattolica del Sacro Cuore, 00168 Rome, Italy
| | - Cassandra Morciano
- Endocrinologia e Diabetologia, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, 00136 Rome, Italy
- Dipartimento di Medicina e Chirurgia Traslazionale, Università Cattolica del Sacro Cuore, 00168 Rome, Italy
| | - Adriana Avolio
- Endocrinologia e Diabetologia, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, 00136 Rome, Italy
- Dipartimento di Medicina e Chirurgia Traslazionale, Università Cattolica del Sacro Cuore, 00168 Rome, Italy
| | - Amelia Splendore
- Endocrinologia e Diabetologia, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, 00136 Rome, Italy
- Dipartimento di Medicina e Chirurgia Traslazionale, Università Cattolica del Sacro Cuore, 00168 Rome, Italy
| | - Gianfranco Di Giuseppe
- Endocrinologia e Diabetologia, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, 00136 Rome, Italy
- Dipartimento di Medicina e Chirurgia Traslazionale, Università Cattolica del Sacro Cuore, 00168 Rome, Italy
| | - Gea Ciccarelli
- Endocrinologia e Diabetologia, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, 00136 Rome, Italy
- Dipartimento di Medicina e Chirurgia Traslazionale, Università Cattolica del Sacro Cuore, 00168 Rome, Italy
| | - Laura Soldovieri
- Endocrinologia e Diabetologia, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, 00136 Rome, Italy
- Dipartimento di Medicina e Chirurgia Traslazionale, Università Cattolica del Sacro Cuore, 00168 Rome, Italy
| | - Michela Brunetti
- Endocrinologia e Diabetologia, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, 00136 Rome, Italy
- Dipartimento di Medicina e Chirurgia Traslazionale, Università Cattolica del Sacro Cuore, 00168 Rome, Italy
| | - Teresa Mezza
- Endocrinologia e Diabetologia, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, 00136 Rome, Italy
- Dipartimento di Medicina e Chirurgia Traslazionale, Università Cattolica del Sacro Cuore, 00168 Rome, Italy
| | - Alfredo Pontecorvi
- Endocrinologia e Diabetologia, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, 00136 Rome, Italy
- Dipartimento di Medicina e Chirurgia Traslazionale, Università Cattolica del Sacro Cuore, 00168 Rome, Italy
| | - Andrea Giaccari
- Endocrinologia e Diabetologia, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, 00136 Rome, Italy
- Dipartimento di Medicina e Chirurgia Traslazionale, Università Cattolica del Sacro Cuore, 00168 Rome, Italy
| | - Francesca Cinti
- Endocrinologia e Diabetologia, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, 00136 Rome, Italy
- Dipartimento di Medicina e Chirurgia Traslazionale, Università Cattolica del Sacro Cuore, 00168 Rome, Italy
| |
Collapse
|
4
|
Oye Mintsa Mi-Mba MF, Lebbadi M, Alata W, Julien C, Emond V, Tremblay C, Fortin S, Barrow CJ, Bilodeau JF, Calon F. Differential impact of eicosapentaenoic acid and docosahexaenoic acid in an animal model of Alzheimer's disease. J Lipid Res 2024; 65:100682. [PMID: 39490923 DOI: 10.1016/j.jlr.2024.100682] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2024] [Revised: 10/14/2024] [Accepted: 10/16/2024] [Indexed: 11/05/2024] Open
Abstract
Dietary supplementation with n-3 polyunsaturated fatty acids improves cognitive performance in several animal models of Alzheimer's disease (AD), an effect often associated with reduced amyloid-beta and/or tau pathologies. However, it remains unclear to what extent eicosapentaenoic (EPA) provides additional benefits compared to docosahexaenoic acid (DHA). Here, male and female 3xTg-AD mice were fed for 3 months (13-16 months of age) the following diets: (1) control (no DHA/EPA), (2) DHA (1.1g/kg) and low EPA (0.4g/kg), or (3) DHA (0.9g/kg) with high EPA (9.2g/kg). The DHA and DHA + EPA diets respectively increased DHA by 19% and 8% in the frontal cortex of 3xTg-AD mice, compared to controls. Levels of EPA, which were below the detection limit after the control diet, reached 0.14% and 0.29% of total brain fatty acids after the DHA and DHA + EPA diet, respectively. DHA and DHA + EPA diets lowered brain arachidonic acid levels and the n-6:n-3 docosapentaenoic acid ratio. Brain uptake of free 14C-DHA measured through intracarotid brain perfusion, but not of 14C-EPA, was lower in 3xTg-AD than in NonTg mice. DHA and DHA + EPA diets in 3xTg-AD mice reduced cortical soluble phosphorylated tau (pS202) (-34% high-DHA, -34% DHA + EPA, P < 0.05) while increasing p21-activated kinase (+58% and +83%, P < 0.001; respectively). High EPA intake lowered insoluble phosphorylated tau (-31% vs. DHA, P < 0.05). No diet effect on amyloid-beta levels was observed. In conclusion, dietary intake of DHA and EPA leads to differential changes in brain PUFA while altering cerebral biomarkers consistent with beneficial effects against AD-like neuropathology.
Collapse
Affiliation(s)
- Méryl-Farelle Oye Mintsa Mi-Mba
- Faculty of Pharmacy, Laval University, Quebec, QC, Canada; Centre Hospitalier de l'Université Laval (CHUL) Research Center, Quebec, QC, Canada
| | - Meryem Lebbadi
- Faculty of Pharmacy, Laval University, Quebec, QC, Canada; Centre Hospitalier de l'Université Laval (CHUL) Research Center, Quebec, QC, Canada
| | - Waël Alata
- Faculty of Pharmacy, Laval University, Quebec, QC, Canada; Centre Hospitalier de l'Université Laval (CHUL) Research Center, Quebec, QC, Canada
| | - Carl Julien
- Faculty of Pharmacy, Laval University, Quebec, QC, Canada; Centre Hospitalier de l'Université Laval (CHUL) Research Center, Quebec, QC, Canada
| | - Vincent Emond
- Centre Hospitalier de l'Université Laval (CHUL) Research Center, Quebec, QC, Canada
| | - Cyntia Tremblay
- Centre Hospitalier de l'Université Laval (CHUL) Research Center, Quebec, QC, Canada
| | - Samuel Fortin
- Centre de recherche sur les biotechnologies marines, Rimouski, QC, Canada
| | - Colin J Barrow
- Centre for Sustainable Bioproducts, Deakin University Geelong, Victoria, Australia
| | - Jean-François Bilodeau
- Centre Hospitalier de l'Université Laval (CHUL) Research Center, Quebec, QC, Canada; Department of medicine, Faculty of Medecine, Laval University, Quebec, QC, Canada
| | - Frédéric Calon
- Faculty of Pharmacy, Laval University, Quebec, QC, Canada; Centre Hospitalier de l'Université Laval (CHUL) Research Center, Quebec, QC, Canada.
| |
Collapse
|
5
|
Dinu M, Sofi F, Lotti S, Colombini B, Mattioli AV, Catapano AL, Casula M, Baragetti A, Wong ND, Steg PG, Ambrosio G. Effects of omega-3 fatty acids on coronary revascularization and cardiovascular events: a meta-analysis. Eur J Prev Cardiol 2024; 31:1863-1875. [PMID: 38869144 DOI: 10.1093/eurjpc/zwae184] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/14/2024] [Revised: 04/16/2024] [Accepted: 05/18/2024] [Indexed: 06/14/2024]
Abstract
AIMS Benefits of pharmacologic omega-3 fatty acid administration in cardiovascular prevention are controversial. Particularly, effects on coronary revascularization are unclear; also debated are specific benefits of eicosapentaenoic acid (EPA). We investigated incident coronary revascularizations, myocardial infarction (MI), stroke, heart failure (HF), unstable angina, and cardiovascular death, in subjects randomized to receive EPA or EPA + docosahexaenoic acid (EPA + DHA) vs. control. METHODS AND RESULTS Meta-analysis of randomized controlled trials (RCTs) was conducted after MEDLINE, Embase, Scopus, Web of Science, and Cochrane Library search. Preferred Reporting Items for Systematic Reviews and Meta-analysis guidelines were followed for abstracting data and assessing data quality and validity. Data were pooled using a random effects model. Eighteen RCTs with 134 144 participants (primary and secondary cardiovascular prevention) receiving DHA + EPA (n = 52 498), EPA alone (n = 14 640), or control/placebo (n = 67 006) were included. Follow-up ranged from 4.5 months to 7.4 years. Overall, compared with controls, omega-3 supplementation reduced the risk of revascularization [0.90, 95% confidence interval (CI) 0.84-0.98; P = 0.001; P-heterogeneity = 0.0002; I2 = 68%], MI (0.89, 95% CI 0.81-0.98; P = 0.02; P-heterogeneity = 0.06; I2 = 41%), and cardiovascular death (0.92, 95% CI 0.85-0.99; P = 0.02; P-heterogeneity = 0.13; I2 = 33%). Lower risk was still observed in trials where most participants (≥60%) were on statin therapy. Compared with DHA + EPA, EPA alone showed a further significant risk reduction of revascularizations (0.76, 95% CI 0.65-0.88; P = 0.0002; P-interaction = 0.005) and all outcomes except HF. CONCLUSION Omega-3 fatty acid supplementation reduced the risk of cardiovascular events and coronary revascularization, regardless of background statin use. Eicosapentaenoic acid alone produced greater benefits. The role of specific omega-3 molecules in primary vs. secondary prevention and the potential benefits of reduced revascularizations on overall health status and cost savings warrant further research.
Collapse
Affiliation(s)
- Monica Dinu
- Department of Experimental and Clinical Medicine, University of Florence, Florence, Italy
| | - Francesco Sofi
- Department of Experimental and Clinical Medicine, University of Florence, Florence, Italy
- Unit of Clinical Nutrition, Careggi University Hospital, Florence, Italy
| | - Sofia Lotti
- Department of Experimental and Clinical Medicine, University of Florence, Florence, Italy
| | - Barbara Colombini
- Department of Experimental and Clinical Medicine, University of Florence, Florence, Italy
| | - Anna Vittoria Mattioli
- Department of Medical and Surgical Sciences, University of Modena and Reggio Emilia, Modena, Italy
| | - Alberico L Catapano
- IRCCS MultiMedica, Milan, Italy
- Department of Pharmacological and Biomolecular Sciences, University of Milan, Milan, Italy
| | - Manuela Casula
- IRCCS MultiMedica, Milan, Italy
- Department of Pharmacological and Biomolecular Sciences, University of Milan, Milan, Italy
| | - Andrea Baragetti
- IRCCS MultiMedica, Milan, Italy
- Department of Pharmacological and Biomolecular Sciences, University of Milan, Milan, Italy
| | - Nathan D Wong
- Division of Cardiology, University of California, Irvine, USA
| | - Philippe Gabriel Steg
- Université Paris-Cité, INSERM U1148, FACT French Alliance for Cardiovascular Trials, AP-HP Hopital Bichat, Paris, France
| | - Giuseppe Ambrosio
- Division of Cardiology, Center for Clinical and Translational Research-CERICLET, University of Perugia School of Medicine, Ospedale S. Maria della Misericordia, Via S. Andrea delle Fratte, 06156 Perugia, Italy
| |
Collapse
|
6
|
Sherratt SCR. REDUCE-IT, biomarkers, and confirmation bias: are we missing the forest for the trees? Eur J Prev Cardiol 2024; 31:e113-e114. [PMID: 37195316 DOI: 10.1093/eurjpc/zwad169] [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: 03/23/2023] [Accepted: 05/15/2023] [Indexed: 05/18/2023]
Affiliation(s)
- Samuel C R Sherratt
- Department of Molecular, Cellular, and Biomedical Sciences, University of New Hampshire, Durham, NH, USA
| |
Collapse
|
7
|
Chao TH, Lin TH, Cheng CI, Wu YW, Ueng KC, Wu YJ, Lin WW, Leu HB, Cheng HM, Huang CC, Wu CC, Lin CF, Chang WT, Pan WH, Chen PR, Ting KH, Su CH, Chu CS, Chien KL, Yen HW, Wang YC, Su TC, Liu PY, Chang HY, Chen PW, Juang JMJ, Lu YW, Lin PL, Wang CP, Ko YS, Chiang CE, Hou CJY, Wang TD, Lin YH, Huang PH, Chen WJ. 2024 Guidelines of the Taiwan Society of Cardiology on the Primary Prevention of Atherosclerotic Cardiovascular Disease --- Part II. ACTA CARDIOLOGICA SINICA 2024; 40:669-715. [PMID: 39582845 PMCID: PMC11579689 DOI: 10.6515/acs.202411_40(6).20240724b] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Received: 02/12/2024] [Accepted: 07/24/2024] [Indexed: 11/26/2024]
Abstract
For the primary prevention of atherosclerotic cardiovascular disease (ASCVD), the recommended treatment target for each modifiable risk factor is as follows: reducing body weight by 5-10%; blood pressure < 130/80 mmHg (systolic pressure < 120 mmHg in high-risk individuals); low-density lipoprotein cholesterol (LDL-C) < 100 mg/dL in high-risk individuals, LDL-C < 115 mg/dL in moderate-risk individuals, LDL-C < 130 mg/dL in low-risk individuals, and LDL-C < 160 mg/dL in those with a minimal; complete and persistent abstinence from cigarette smoking; hemoglobin A1C < 7.0%; fulfilling recommended amounts of the six food groups according to the Taiwan food guide; and moderate-intensity physical activity 150 min/wk or vigorous physical activity 75 min/wk. For the primary prevention of ASCVD by pharmacological treatment in individuals with modifiable risk factors/clinical conditions, statins are the first-line therapy for reducing LDL-C levels; some specific anti-diabetic drugs proven to be effective in randomized controlled trials for the primary prevention of ASCVD are recommended in patients with type 2 diabetes mellitus; pharmacological treatment is recommended to assist in weight management for obese patients with a body mass index ≥ 30 kg/m2 (or 27 kg/m2 who also have at least one ASCVD risk factor or obesity-related comorbidity); an angiotensin-converting enzyme inhibitor, a glucagon-like peptide-1 receptor agonist, a sodium-dependent glucose cotransporter-2 inhibitor, and finerenone can be used in diabetic patients with chronic kidney disease for the primary prevention of ASCVD. Of note, healthcare providers are at full discretion in clinical practice, owing to the diversity of individuals and practice, and the availability of resources and facilities.
Collapse
Affiliation(s)
- Ting-Hsing Chao
- Division of Cardiology, Department of Internal Medicine, National Cheng Kung University Hospital, College of Medicine, National Cheng Kung University, Tainan
- Division of Cardiology, Department of Internal Medicine, Chung-Shan Medical University Hospital; School of Medicine, Chung Shan Medical University, Taichung
| | - Tsung-Hsien Lin
- Division of Cardiology, Department of Internal Medicine Kaohsiung Medical University Hospital
- Faculty of Medicine and Graduate Institute of Medicine, College of Medicine, Kaohsiung Medical University
| | - Cheng-I Cheng
- Division of Cardiology, Department of Internal Medicine, Kaohsiung Chang Gung Memorial Hospital, Kaohsiung; School of Medicine, College of Medicine, Chang Gung University, Taoyuan
| | - Yen-Wen Wu
- Division of Cardiology, Cardiovascular Medical Center, Far Eastern Memorial Hospital, New Taipei City
- School of Medicine, National Yang Ming Chiao Tung University, Taipei
- Graduate Institute of Medicine, Yuan Ze University, Taoyuan
| | - Kwo-Chang Ueng
- Division of Cardiology, Department of Internal Medicine, Chung-Shan Medical University Hospital; School of Medicine, Chung Shan Medical University, Taichung
| | - Yih-Jer Wu
- Department of Medicine and Institute of Biomedical Sciences, MacKay Medical College, New Taipei City
- Cardiovascular Center, Department of Internal Medicine, MacKay Memorial Hospital, Taipei
| | - Wei-Wen Lin
- Cardiovascular center, Taichung Veterans General Hospital, Taichung
| | - Hsing-Ban Leu
- School of Medicine, National Yang Ming Chiao Tung University, Taipei
- Cardiovascular Research Center, National Yang Ming Chiao Tung University
- Healthcare and Management Center
- Division of Cardiology, Department of Medicine, Taipei Veterans General Hospital
| | - Hao-Min Cheng
- Ph.D. Program of Interdisciplinary Medicine (PIM), National Yang Ming Chiao Tung University College of Medicine; Division of Faculty Development; Center for Evidence-based Medicine, Taipei Veterans General Hospital; Institute of Public Health; Institute of Health and Welfare Policy, National Yang Ming Chiao Tung University College of Medicine
| | - Chin-Chou Huang
- School of Medicine, National Yang Ming Chiao Tung University, Taipei
- Division of Cardiology, Department of Medicine, Taipei Veterans General Hospital
- Institute of Pharmacology, National Yang Ming Chiao Tung University, Taipei
| | - Chih-Cheng Wu
- Center of Quality Management, National Taiwan University Hospital Hsinchu Branch, Hsinchu; College of Medicine, National Taiwan University, Taipei; Institute of Biomedical Engineering, National Tsing-Hua University, Hsinchu; Institute of Cellular and System Medicine, National Health Research Institutes, Zhunan
| | - Chao-Feng Lin
- Department of Medicine, MacKay Medical College, New Taipei City; Department of Cardiology, MacKay Memorial Hospital, Taipei
| | - Wei-Ting Chang
- School of Medicine and Doctoral Program of Clinical and Experimental Medicine, College of Medicine and Center of Excellence for Metabolic Associated Fatty Liver Disease, National Sun Yat-sen University, Kaohsiung; Division of Cardiology, Department of Internal Medicine, Chi Mei Medical Center, Tainan
| | - Wen-Han Pan
- Institute of Biomedical Sciences, Academia Sinica, Taipei; Institute of Population Health Sciences, National Health Research Institutes, Miaoli; Institute of Biochemistry and Biotechnology, National Taiwan University
| | - Pey-Rong Chen
- Department of Dietetics, National Taiwan University Hospital, Taipei
| | - Ke-Hsin Ting
- Division of Cardiology, Department of Internal Medicine, Yunlin Christian Hospital, Yunlin
| | - Chun-Hung Su
- Division of Cardiology, Department of Internal Medicine, Chung-Shan Medical University Hospital; School of Medicine, Chung Shan Medical University, Taichung
| | - Chih-Sheng Chu
- Division of Cardiology, Department of Internal Medicine, Kaohsiung Municipal Ta-Tung Hospital, Kaohsiung Medical University, Kaohsiung
| | - Kuo-Liong Chien
- Institute of Epidemiology and Preventive Medicine, College of Public Health, National Taiwan University; Department of Internal Medicine, National Taiwan University Hospital and College of Medicine; Population Health Research Center, National Taiwan University, Taipei
| | - Hsueh-Wei Yen
- Division of Cardiology, Department of Internal Medicine Kaohsiung Medical University Hospital
| | - Yu-Chen Wang
- Division of Cardiology, Asia University Hospital; Department of Medical Laboratory Science and Biotechnology, Asia University; Division of Cardiology, China Medical University College of Medicine and Hospital, Taichung
| | - Ta-Chen Su
- Cardiovascular Center, Department of Internal Medicine, National Taiwan University Hospital
- Department of Environmental and Occupational Medicine, National Taiwan University College of Medicine
| | - Pang-Yen Liu
- Division of Cardiology, Department of Internal Medicine, Tri-Service General Hospital, National Defense Medical Center
| | - Hsien-Yuan Chang
- Division of Cardiology, Department of Internal Medicine, National Cheng Kung University Hospital, College of Medicine, National Cheng Kung University, Tainan
| | - Po-Wei Chen
- Division of Cardiology, Department of Internal Medicine, National Cheng Kung University Hospital, College of Medicine, National Cheng Kung University, Tainan
| | - Jyh-Ming Jimmy Juang
- Heart Failure Center and Division of Cardiology, Department of Internal Medicine, National Taiwan University College of Medicine, and National Taiwan University Hospital
| | - Ya-Wen Lu
- Cardiovascular center, Taichung Veterans General Hospital, Taichung
- Cardiovascular Research Center, National Yang Ming Chiao Tung University
- Institute of Clinical Medicine, National Yang Ming Chiao Tung University, Taipei
| | - Po-Lin Lin
- Division of Cardiology, Department of Internal Medicine, Hsinchu MacKay Memorial Hospital, Hsinchu
| | - Chao-Ping Wang
- Division of Cardiology, E-Da Hospital; School of Medicine for International Students, College of Medicine, I-Shou University, Kaohsiung
| | - Yu-Shien Ko
- Cardiovascular Division, Chang Gung Memorial Hospital; College of Medicine, Chang Gung University, Taoyuan
| | - Chern-En Chiang
- General Clinical Research Center and Division of Cardiology, Taipei Veterans General Hospital and National Yang Ming Chiao Tung University
| | - Charles Jia-Yin Hou
- Cardiovascular Center, Department of Internal Medicine, MacKay Memorial Hospital, Taipei
| | - Tzung-Dau Wang
- Cardiovascular Center and Divisions of Hospital Medicine and Cardiology, Department of Internal Medicine, National Taiwan University Hospital and National Taiwan University College of Medicine
| | - Yen-Hung Lin
- Division of Cardiology, Department of Internal Medicine, National Taiwan University Hospital and National Taiwan University College of Medicine, Taipei
| | - Po-Hsun Huang
- Cardiovascular Research Center, National Yang Ming Chiao Tung University
- Division of Cardiology, Department of Medicine, Taipei Veterans General Hospital
| | - Wen-Jone Chen
- Department of Internal Medicine, Min-Sheng General Hospital, Taoyuan; Department of Internal Medicine, College of Medicine, National Taiwan University, Taipei, Taiwan
| |
Collapse
|
8
|
Massaro M, Quarta S, Calabriso N, Carluccio MA, Scoditti E, Mancuso P, De Caterina R, Madonna R. Omega-3 polyunsaturated fatty acids and pulmonary arterial hypertension: Insights and perspectives. Eur J Clin Invest 2024; 54:e14277. [PMID: 38940236 PMCID: PMC11490397 DOI: 10.1111/eci.14277] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/27/2024] [Accepted: 06/17/2024] [Indexed: 06/29/2024]
Abstract
Pulmonary arterial hypertension (PAH) is a rare and progressive disorder that affects the pulmonary vasculature. Although recent developments in pharmacotherapy have extended the life expectancy of PAH patients, their 5-year survival remains unacceptably low, underscoring the need for multitarget and more comprehensive approaches to managing the disease. This should incorporate not only medical, but also lifestyle interventions, including dietary changes and the use of nutraceutical support. Among these strategies, n-3 polyunsaturated fatty acids (n-3 PUFAs) are emerging as promising agents able to counteract the inflammatory component of PAH. In this narrative review, we aim at analysing the preclinical evidence for the impact of n-3 PUFAs on the pathogenesis and the course of PAH. Although evidence for the role of n-3 PUFAs deficiencies in the development and progression of PAH in humans is limited, preclinical studies suggest that these dietary components may influence several aspects of the pathobiology of PAH. Further clinical research should test the efficacy of n-3 PUFAs on top of approved clinical management. These studies will provide evidence on whether n-3 PUFAs can genuinely serve as a valuable tool to enhance the efficacy of pharmacotherapy in the treatment of PAH.
Collapse
Affiliation(s)
- Marika Massaro
- Institute of Clinical Physiology (IFC), National Research Council (CNR), 73100 Lecce, Italy
| | - Stefano Quarta
- Institute of Clinical Physiology (IFC), National Research Council (CNR), 73100 Lecce, Italy
| | - Nadia Calabriso
- Institute of Clinical Physiology (IFC), National Research Council (CNR), 73100 Lecce, Italy
| | | | - Egeria Scoditti
- Institute of Clinical Physiology (IFC), National Research Council (CNR), 73100 Lecce, Italy
| | - Peter Mancuso
- Department of Nutritional Sciences and the Program in Immunology, School of Public Health, University of Michigan, 1415 Washington Hts., Ann Arbor, Michigan 481009
| | | | | |
Collapse
|
9
|
Shimizu K, Kuramochi Y, Hayamizu K. Effect of omega-3 fatty acids on sleep: a systematic review and meta-analysis of randomized controlled trials. J Clin Biochem Nutr 2024; 75:204-212. [PMID: 39583980 PMCID: PMC11579846 DOI: 10.3164/jcbn.24-36] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2024] [Accepted: 06/24/2024] [Indexed: 11/26/2024] Open
Abstract
Omega-3 long-chain polyunsaturated fatty acids (LC-PUFAs) have been reported to improve sleep quality in several studies, but meta-analyses have been inconclusive. We conducted this study to investigate the effects of omega-3 LC-PUFAs on sleep in clinical trials. The study was planned in accordance with the criteria of the Preferred Reporting Items for Systematic Review and Meta-Analysis Protocols (PRISMA-2020), and was performed by searching PubMed, The Cochrane Library, and Ichushi-web databases. Randomized controlled trials and clinical trials with control groups were included. Finally, eight studies were selected for inclusion in this study. Sleep efficiency was significantly higher in the omega-3 LC-PUFA group than in the control group, while sleep latency and total sleep duration did not differ significantly. Subjectively assessed sleep was significantly improved by omega-3 LC-PUFA, but heterogeneity was so high that a subgroup analysis based on dose of omega-3 supplementation was performed. It showed low heterogeneity and significant improvement in the omega-3 LC-PUFA group compared with the control group. Omega-3 LC-PUFAs have been shown to may improve sleep quality. Further studies are needed to confirm the relationship between omega-3 LC-PUFAs and sleep. The protocol for this review was registered in UMIN000052527.
Collapse
Affiliation(s)
- Kaori Shimizu
- Department of Pharmacy, Yokohama University of Pharmacy, Totsuka-ku, Yokohama, Kanagawa 245-0066, Japan
| | - Yui Kuramochi
- Department of Pharmacy, Yokohama University of Pharmacy, Totsuka-ku, Yokohama, Kanagawa 245-0066, Japan
| | - Kohsuke Hayamizu
- Department of Pharmacy, Yokohama University of Pharmacy, Totsuka-ku, Yokohama, Kanagawa 245-0066, Japan
| |
Collapse
|
10
|
Zhu J, Li S, Chen W, Xu X, Wang X, Wang X, Han J, Jouhet J, Amato A, Maréchal E, Hu H, Allen AE, Gong Y, Jiang H. Delta-5 elongase knockout reduces docosahexaenoic acid and lipid synthesis and increases heat sensitivity in a diatom. PLANT PHYSIOLOGY 2024; 196:1356-1373. [PMID: 38796833 DOI: 10.1093/plphys/kiae297] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/24/2024] [Revised: 04/19/2024] [Accepted: 04/28/2024] [Indexed: 05/29/2024]
Abstract
Recent global marine lipidomic analysis reveals a strong relationship between ocean temperature and phytoplanktonic abundance of omega-3 long-chain polyunsaturated fatty acids, especially eicosapentaenoic acid (EPA) and docosahexaenoic acid (DHA), which are essential for human nutrition and primarily sourced from phytoplankton in marine food webs. In phytoplanktonic organisms, EPA may play a major role in regulating the phase transition temperature of membranes, while the function of DHA remains unexplored. In the oleaginous diatom Phaeodactylum tricornutum, DHA is distributed mainly on extraplastidial phospholipids, which is very different from the EPA enriched in thylakoid lipids. Here, clustered regularly interspaced short palindromic repeats/CRISPR-associated protein 9-mediated knockout of delta-5 elongase (ptELO5a), which encodes a delta-5 elongase (ELO5) catalyzing the elongation of EPA to synthesize DHA, led to a substantial interruption of DHA synthesis in P. tricornutum. The ptELO5a mutants showed some alterations in transcriptome and glycerolipidomes, including membrane lipids and triacylglycerols under normal temperature (22 °C), and were more sensitive to elevated temperature (28 °C) than wild type. We conclude that PtELO5a-mediated synthesis of small amounts of DHA has indispensable functions in regulating membrane lipids, indirectly contributing to storage lipid accumulation, and maintaining thermomorphogenesis in P. tricornutum. This study also highlights the significance of DHA synthesis and lipid composition for environmental adaptation of P. tricornutum.
Collapse
Affiliation(s)
- Junkai Zhu
- School of Marine Sciences, Ningbo University, Ningbo 315211, China
- Key Laboratory of Biology and Genetic Improvement of Oil Crops, Oil Crops Research Institute, Chinese Academy of Agricultural Sciences, Wuhan 430062, China
| | - Shuangqing Li
- School of Marine Sciences, Ningbo University, Ningbo 315211, China
- Life and Ecology Center, Southern Marine Science and Engineering Guangdong Laboratory (Zhuhai), Zhuhai 519080, China
| | - Weizhong Chen
- School of Marine Sciences, Ningbo University, Ningbo 315211, China
| | - Xinde Xu
- Department of Human Nutrition, Zhejiang Medicine Co. Ltd., Xinchang 312500, China
- Department of Human Nutrition, Zhejiang Keming Biopharmaceuticals Co. Ltd., Xinchang 312500, China
| | - Xiaoping Wang
- Department of Human Nutrition, Zhejiang Medicine Co. Ltd., Xinchang 312500, China
- Department of Human Nutrition, Zhejiang Keming Biopharmaceuticals Co. Ltd., Xinchang 312500, China
| | - Xinwei Wang
- School of Marine Sciences, Ningbo University, Ningbo 315211, China
| | - Jichang Han
- College of Food Science and Engineering, Ningbo University, Ningbo 315211, China
| | - Juliette Jouhet
- Laboratoire de Physiologie Cellulaire et Végétale, Centre National de la Recherche Scientifique, Commissariat à l'Energie Atomique et aux Energies Alternatives, INRAE, Université Grenoble Alpes, Unité mixte de recherche 5168, IRIG, CEA Grenoble, F-38041 Grenoble, France
| | - Alberto Amato
- Laboratoire de Physiologie Cellulaire et Végétale, Centre National de la Recherche Scientifique, Commissariat à l'Energie Atomique et aux Energies Alternatives, INRAE, Université Grenoble Alpes, Unité mixte de recherche 5168, IRIG, CEA Grenoble, F-38041 Grenoble, France
| | - Eric Maréchal
- Laboratoire de Physiologie Cellulaire et Végétale, Centre National de la Recherche Scientifique, Commissariat à l'Energie Atomique et aux Energies Alternatives, INRAE, Université Grenoble Alpes, Unité mixte de recherche 5168, IRIG, CEA Grenoble, F-38041 Grenoble, France
| | - Hanhua Hu
- Key Laboratory of Algal Biology, Institute of Hydrobiology, Chinese Academy of Sciences, Wuhan 430072, China
| | - Andrew E Allen
- Scripps Institution of Oceanography, University of California, San Diego, La Jolla, CA 92093, USA
- Department of Environment and Sustainability, J. Craig Venter Institute, La Jolla, CA 92037, USA
| | - Yangmin Gong
- Key Laboratory of Biology and Genetic Improvement of Oil Crops, Oil Crops Research Institute, Chinese Academy of Agricultural Sciences, Wuhan 430062, China
| | - Haibo Jiang
- School of Marine Sciences, Ningbo University, Ningbo 315211, China
- Life and Ecology Center, Southern Marine Science and Engineering Guangdong Laboratory (Zhuhai), Zhuhai 519080, China
| |
Collapse
|
11
|
Parolini A, Da Dalt L, Norata GD, Baragetti A. Dietary fats as regulators of neutrophil plasticity: an update on molecular mechanisms. Curr Opin Clin Nutr Metab Care 2024; 27:434-442. [PMID: 39083430 PMCID: PMC11309349 DOI: 10.1097/mco.0000000000001055] [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] [Indexed: 08/02/2024]
Abstract
PURPOSE OF REVIEW Contemporary guidelines for the prevention of cardio-metabolic diseases focus on the control of dietary fat intake, because of their adverse metabolic effects. Moreover, fats alter innate immune defenses, by eliciting pro-inflammatory epigenetic mechanisms on the long-living hematopoietic cell progenitors which, in the bone marrow, mainly give rise to short-living neutrophils. Nevertheless, the heterogenicity of fats and the complexity of the biology of neutrophils pose challenges in the understanding on how this class of nutrients could contribute to the development of cardio-metabolic diseases via specific molecular mechanisms activating the inflammatory response. RECENT FINDINGS The knowledge on the biology of neutrophils is expanding and there are now different cellular networks orchestrating site-specific reprogramming of these cells to optimize the responses against pathogens. The innate immune competence of neutrophil is altered in response to high fat diet and contributes to the development of metabolic alterations, although the precise mechanisms are still poorly understood. SUMMARY Defining the different molecular mechanisms involved in the fat-neutrophil crosstalk will help to reconcile the sparse data about the interaction of dietary fats with neutrophils and to tailor strategies to target neutrophils in the context of cardio-metabolic diseases.
Collapse
Affiliation(s)
- Anna Parolini
- Department of Pharmacological and Biomolecular Sciences "Rodolfo Paoletti", University of Milan, Milan, Italy
| | | | | | | |
Collapse
|
12
|
Marcus MD, Link MS. Omega-3 Fatty Acids and Arrhythmias. Circulation 2024; 150:488-503. [PMID: 39102482 DOI: 10.1161/circulationaha.123.065769] [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] [Indexed: 08/07/2024]
Abstract
The pro- and antiarrhythmic effects of omega-3 polyunsaturated fatty acids (n-3 PUFAs) have been extensively studied in preclinical and human trials. Despite early evidence of an antiarrhythmic role of n-3 PUFA in the prevention of sudden cardiac death and postoperative and persistent atrial fibrillation (AF), subsequent well-designed randomized trials have largely not shown an antiarrhythmic benefit. Two trials that tested moderate and high-dose n-3 PUFA demonstrated a reduction in sudden cardiac death, but these findings have not been widely replicated, and the potential of eicosapentaenoic acid (EPA) and docosahexaenoic acid (DHA) to reduce arrhythmic death in combination, or as monotherapy, remains uncertain. The accumulated clinical evidence does not support supplementation of n-3 PUFA for postoperative AF or secondary prevention of AF. Several large, contemporary, randomized controlled trials of high-dose n-3 PUFA for primary or secondary cardiovascular prevention have demonstrated a small, significant, dose-dependent increased risk of incident AF compared with mineral oil or corn oil comparator. These findings were reproduced with both icosapent ethyl monotherapy and a mixed EPA+DHA formulation. The proarrhythmic mechanism of increased AF in contemporary cohorts exposed to high-dose n-3 PUFA is unknown. EPA and DHA and their metabolites have pleiotropic cardiometabolic and pro- and antiarrhythmic effects, including modification of the lipid raft microenvironment; alteration of cell membrane structure and fluidity; modulation of sodium, potassium, and calcium currents; and regulation of gene transcription, cell proliferation, and inflammation. Further characterization of the complex association between EPA, EPA+DHA, and DHA and AF is needed. Which formulations, dose ranges, and patient subgroups are at highest risk, remain unclear.
Collapse
Affiliation(s)
- Mason D Marcus
- Department of Medicine, Division of Cardiology, UT Southwestern Medical Center, Dallas, TX
| | - Mark S Link
- Department of Medicine, Division of Cardiology, UT Southwestern Medical Center, Dallas, TX
| |
Collapse
|
13
|
Kaur G, Mason RP, Steg PG, Bhatt DL. Omega-3 fatty acids for cardiovascular event lowering. Eur J Prev Cardiol 2024; 31:1005-1014. [PMID: 38169319 DOI: 10.1093/eurjpc/zwae003] [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: 11/28/2023] [Revised: 12/28/2023] [Accepted: 12/31/2023] [Indexed: 01/05/2024]
Abstract
Low-density lipoprotein cholesterol (LDL-C) is the main target for therapeutics aimed at reducing the risk of atherosclerotic cardiovascular disease (ASCVD) and downstream cardiovascular (CV) events. However, multiple studies have demonstrated that high-risk patient populations harbour residual risk despite effective LDL-C lowering. While data support the causal relationship between triglycerides and ASCVD risk, triglyceride-lowering therapies such as omega-3 fatty acids have shown mixed results in CV outcomes trials. Notably, icosapent ethyl, a purified formulation of eicosapentaenoic acid (EPA), has garnered compelling evidence in lowering residual CV risk in patients with hypertriglyceridaemia and treated with statins. In this review, we summarize studies that have investigated omega-3-fatty acids for CV event lowering and discuss the clinical implementation of these agents based on trial data and guidelines.
Collapse
Affiliation(s)
- Gurleen Kaur
- Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - R Preston Mason
- Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
- Elucida Research LLC, Beverly, MA, USA
| | - Ph Gabriel Steg
- Paris Cité University, Public Hospitals of Paris (AP-HP), Bichat Hospital, Paris, France
| | - Deepak L Bhatt
- Mount Sinai Fuster Heart Hospital, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| |
Collapse
|
14
|
Zirpoli H, Bernis ME, Sabir H, Manual Kollareth DJ, Hamilton JA, Huang N, Ng J, Sosunov SA, Gaebler B, Ten VS, Deckelbaum RJ. Omega-3 fatty acid diglyceride emulsions as a novel injectable acute therapeutic in neonatal hypoxic-ischemic brain injury. Biomed Pharmacother 2024; 175:116749. [PMID: 38761420 PMCID: PMC11156760 DOI: 10.1016/j.biopha.2024.116749] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2024] [Revised: 05/10/2024] [Accepted: 05/10/2024] [Indexed: 05/20/2024] Open
Abstract
Hypoxic-ischemic encephalopathy (HIE), resulting from a lack of blood flow and oxygen before or during newborn delivery, is a leading cause of cerebral palsy and neurological disability in children. Therapeutic hypothermia (TH), the current standard of care in HIE, is only beneficial in 1 of 7-8 cases. Therefore, there is a critical need for more efficient treatments. We have previously reported that omega-3 (n-3) fatty acids (FA) carried by triglyceride (TG) lipid emulsions provide neuroprotection after experimental hypoxic-ischemic (HI) injury in neonatal mice. Herein, we propose a novel acute therapeutic approach using an n-3 diglyceride (DG) lipid emulsions. Importantly, n-3 DG preparations had much smaller particle size compared to commercially available or lab-made n-3 TG emulsions. We showed that n-3 DG molecules have the advantage of incorporating at substantially higher levels than n-3 TG into an in vitro model of phospholipid membranes. We also observed that n-3 DG after parenteral administration in neonatal mice reaches the bloodstream more rapidly than n-3 TG. Using neonatal HI brain injury models in mice and rats, we found that n-3 DG emulsions provide superior neuroprotection than n-3 TG emulsions or TH in decreasing brain infarct size. Additionally, we found that n-3 DGs attenuate microgliosis and astrogliosis. Thus, n-3 DG emulsions are a superior, promising, and novel therapy for treating HIE.
Collapse
Affiliation(s)
- Hylde Zirpoli
- Institute of Human Nutrition, Columbia University Irving Medical Center, New York, NY 10032, USA; Department of Pediatrics, Vagelos College of Physicians and Surgeons, Columbia University Irving Medical Center, New York, NY 10032, USA.
| | - Maria Eugenia Bernis
- Department of Neonatology and Pediatric Intensive Care, Children's Hospital, University of Bonn, Germany; Deutsches Zentrum für Neurodegenerative Erkrankungen (DZNE), Bonn 53127, Germany
| | - Hemmen Sabir
- Department of Neonatology and Pediatric Intensive Care, Children's Hospital, University of Bonn, Germany; Deutsches Zentrum für Neurodegenerative Erkrankungen (DZNE), Bonn 53127, Germany
| | - Denny Joseph Manual Kollareth
- Department of Pediatrics, Vagelos College of Physicians and Surgeons, Columbia University Irving Medical Center, New York, NY 10032, USA
| | - James A Hamilton
- Department of Physiology & Biophysics, Department of Biomedical Engineering, Boston University School of Medicine, Boston, MA 02215, USA
| | - Nasi Huang
- Department of Physiology & Biophysics, Department of Biomedical Engineering, Boston University School of Medicine, Boston, MA 02215, USA
| | - Jesse Ng
- Department of Physiology & Biophysics, Department of Biomedical Engineering, Boston University School of Medicine, Boston, MA 02215, USA
| | - Sergey A Sosunov
- Department of Pediatrics, Vagelos College of Physicians and Surgeons, Columbia University Irving Medical Center, New York, NY 10032, USA; Division of Neonatology, Department of Pediatrics, Robert Wood Johnson Medical School, Rutgers University, New Brunswick, NJ 08901, USA
| | | | - Vadim S Ten
- Department of Pediatrics, Vagelos College of Physicians and Surgeons, Columbia University Irving Medical Center, New York, NY 10032, USA; Division of Neonatology, Department of Pediatrics, Robert Wood Johnson Medical School, Rutgers University, New Brunswick, NJ 08901, USA
| | - Richard J Deckelbaum
- Institute of Human Nutrition, Columbia University Irving Medical Center, New York, NY 10032, USA; Department of Pediatrics, Vagelos College of Physicians and Surgeons, Columbia University Irving Medical Center, New York, NY 10032, USA
| |
Collapse
|
15
|
Chai Z, Zhang H, Ji X, Hu X, He Y, Zhao F, Song C, Zhou Y, Li T, He C, Zhou D, Zhang X. The disparate effects of omega-3 PUFAs on intestinal microbial homeostasis in experimental rodents under physiological condition. Prostaglandins Leukot Essent Fatty Acids 2024; 203:102643. [PMID: 39317024 DOI: 10.1016/j.plefa.2024.102643] [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: 07/03/2024] [Revised: 08/25/2024] [Accepted: 08/27/2024] [Indexed: 09/26/2024]
Abstract
The health benefits of omega-3 polyunsaturated fatty acids (omega-3 PUFAs), primarily eicosapentaenoic acid (EPA) and docosahexaenoic acid (DHA), are linked to their regulatory effects on the composition of the gut microbiota. However, there is a lack of direct evidence on whether omega-3 PUFAs regulate the gut microbial homeostasis under physiological conditions. This study investigated the impact of equivalent doses of EPA, DHA, and fish oil (FO) with a DHA to EPA ratio of approximately 1:1 on the bacterial and fungal composition of normal young mice. This study also analyzed changes in key components of the gut microenvironment, including the colonic mucus barrier and short-chain fatty acids, to address the prebiotic potential of omega-3 PUFAs. The results showed that all three omega-3 PUFAs interventions induced significant fluctuations in the gut bacteria and fungi, leading to an increase in the abundance of some probiotics. Notably, DHA, EPA, and FO interventions significantly increased the abundance of the probiotic Lactobacillus, Bifidobacterium, and Akkermansia, respectively. Both DHA and fish oil interventions also significantly reduced the abundance of potentially pathogenic fungi, such as Aspergillus and Penicillium. Association analysis of the top 19 differential fungal and bacterial genera in abundance revealed a much more bacteria-bacteria and bacteria-fungi connections, but fewer fungi-fungi connections. This highlights the importance of bacteria in the gut microecological network. Furthermore, the levels of butyric acid and valeric acid in the colonic contents of DHA intervention group were significantly increased, and the colonic mucus layer thickness was increased in three treatment groups. In summary, DHA, EPA and FO interventions showed targeted enhancement of different probiotics and enhanced colon defense barrier (mucus barrier), showing potential prebiotic effects.
Collapse
Affiliation(s)
- Zhenglong Chai
- School of Public Health, Health Science Center, Ningbo University, Zhejiang Key Laboratory of Pathophysiology, Ningbo, Zhejiang, 315211, China
| | - Hui Zhang
- School of Public Health, Health Science Center, Ningbo University, Zhejiang Key Laboratory of Pathophysiology, Ningbo, Zhejiang, 315211, China; Academy of Integrative Medicine Institute, The First Donguan Affiliated Hospital, Guangdong Medical University, Donguan, Guangdong, 523000, China
| | - Xinyue Ji
- School of Public Health, Health Science Center, Ningbo University, Zhejiang Key Laboratory of Pathophysiology, Ningbo, Zhejiang, 315211, China
| | - Xinyi Hu
- School of Public Health, Health Science Center, Ningbo University, Zhejiang Key Laboratory of Pathophysiology, Ningbo, Zhejiang, 315211, China
| | - Yannan He
- OmegaBandz. Inc Shanghai, 1180 Xingxian Road, Shanghai, 201815, China; Institute of Nutrition and Health of Qingdao University, Qingdao, Shandong, 266021, China
| | - Feng Zhao
- Xi'an University, Xi'an, Shanxi, 710065, China
| | - Chunyan Song
- School of Public Health, Health Science Center, Ningbo University, Zhejiang Key Laboratory of Pathophysiology, Ningbo, Zhejiang, 315211, China
| | - Yiqiu Zhou
- The First Affiliated Hospital of Ningbo University, Ningbo, Zhejiang, 315020, China
| | - Tao Li
- HEALTH BioMed Research & Development Center, Health BioMed Co. Ltd., Ningbo, Zhejiang 315801, China
| | - Canxia He
- School of Public Health, Health Science Center, Ningbo University, Zhejiang Key Laboratory of Pathophysiology, Ningbo, Zhejiang, 315211, China
| | - Dezheng Zhou
- School of Public Health, Health Science Center, Ningbo University, Zhejiang Key Laboratory of Pathophysiology, Ningbo, Zhejiang, 315211, China
| | - Xiaohong Zhang
- School of Public Health, Health Science Center, Ningbo University, Zhejiang Key Laboratory of Pathophysiology, Ningbo, Zhejiang, 315211, China.
| |
Collapse
|
16
|
Djuricic I, Calder PC. Omega-3 ( n-3) Fatty Acid-Statin Interaction: Evidence for a Novel Therapeutic Strategy for Atherosclerotic Cardiovascular Disease. Nutrients 2024; 16:962. [PMID: 38612996 PMCID: PMC11013773 DOI: 10.3390/nu16070962] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2024] [Revised: 03/24/2024] [Accepted: 03/25/2024] [Indexed: 04/14/2024] Open
Abstract
Managing atherosclerotic cardiovascular disease (ASCVD) often involves a combination of lifestyle modifications and medications aiming to decrease the risk of cardiovascular outcomes, such as myocardial infarction and stroke. The aim of this article is to discuss possible omega-3 (n-3) fatty acid-statin interactions in the prevention and treatment of ASCVD and to provide evidence to consider for clinical practice, highlighting novel insights in this field. Statins and n-3 fatty acids (eicosapentaenoic acid (EPA) and docosahexaenoic acid (DHA)) are commonly used to control cardiovascular risk factors in order to treat ASCVD. Statins are an important lipid-lowering therapy, primarily targeting low-density lipoprotein cholesterol (LDL-C) levels, while n-3 fatty acids address triglyceride (TG) concentrations. Both statins and n-3 fatty acids have pleiotropic actions which overlap, including improving endothelial function, modulation of inflammation, and stabilizing atherosclerotic plaques. Thus, both statins and n-3 fatty acids potentially mitigate the residual cardiovascular risk that remains beyond lipid lowering, such as persistent inflammation. EPA and DHA are both substrates for the synthesis of so-called specialized pro-resolving mediators (SPMs), a relatively recently recognized feature of their ability to combat inflammation. Interestingly, statins seem to have the ability to promote the production of some SPMs, suggesting a largely unrecognized interaction between statins and n-3 fatty acids with relevance to the control of inflammation. Although n-3 fatty acids are the major substrates for the production of SPMs, these signaling molecules may have additional therapeutic benefits beyond those provided by the precursor n-3 fatty acids themselves. In this article, we discuss the accumulating evidence that supports SPMs as a novel therapeutic tool and the possible statin-n-3 fatty acid interactions relevant to the prevention and treatment of ASCVD.
Collapse
Affiliation(s)
- Ivana Djuricic
- Department of Bromatology, Faculty of Pharmacy, University of Belgrade, 11221 Belgrade, Serbia;
| | - Philip C. Calder
- School of Human Development and Health, Faculty of Medicine, University of Southampton, Southampton SO16 6YD, UK
- NIHR Southampton Biomedical Research Centre, University Hospital Southampton NHS Foundation Trust and University of Southampton, Southampton SO16 6YD, UK
| |
Collapse
|
17
|
Bianchetti G, Cefalo CMA, Ferreri C, Sansone A, Vitale M, Serantoni C, Abeltino A, Mezza T, Ferraro PM, De Spirito M, Riccardi G, Giaccari A, Maulucci G. Erythrocyte membrane fluidity: A novel biomarker of residual cardiovascular risk in type 2 diabetes. Eur J Clin Invest 2024; 54:e14121. [PMID: 37929812 DOI: 10.1111/eci.14121] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/21/2023] [Revised: 10/18/2023] [Accepted: 10/21/2023] [Indexed: 11/07/2023]
Abstract
AIMS Improving the composition of circulating fatty acids (FA) leads to a reduction in cardiovascular diseases (CVD) in high-risk individuals. The membrane fluidity of red blood cells (RBC), which reflects circulating FA status, may be a valid biomarker of cardiovascular (CV) risk in type 2 diabetes (T2D). METHODS Red blood cell membrane fluidity, quantified as general polarization (GP), was assessed in 234 subjects with T2D, 86 with prior major CVD. Based on GP distribution, a cut-off of .445 was used to divide the study cohort into two groups: the first with higher GP, called GEL, and the second, defined as lower GP (LGP). Lipidomic analysis was performed to evaluate FA composition of RBC membranes. RESULTS Although with comparable CV risk factors, the LGP group had a greater percentage of patients with major CVD than the GEL group (40% vs 24%, respectively, p < .05). Moreover, in a logistic regression analysis, a lower GP value was independently associated with the presence of macrovascular complications. Lipidomic analysis showed a clear shift of LGP membranes towards a pro-inflammatory condition due to higher content of arachidonic acid and increased omega 6/omega 3 index. CONCLUSIONS Increased membrane fluidity is associated with a higher CV risk in subjects with T2D. If confirmed in prospective studies, membrane fluidity could be a new biomarker for residual CV risk assessment in T2D.
Collapse
Affiliation(s)
- Giada Bianchetti
- Department of Neurosciences, Biophysics Section, Catholic University of the Sacred Heart, Rome, Italy
- Fondazione Policlinico Universitario A. Gemelli IRCCS, Rome, Italy
| | - Chiara Maria Assunta Cefalo
- Department of Translational Medicine and Surgery, Catholic University of the Sacred Heart, Rome, Italy
- Department of Medical and Surgical Sciences, Center for Endocrine and Metabolic Diseases, Fondazione Policlinico Universitario A. Gemelli IRCCS, Rome, Italy
| | - Carla Ferreri
- CNR ISOF, National Research Council, ISOF, Bologna, Italy
| | - Anna Sansone
- CNR ISOF, National Research Council, ISOF, Bologna, Italy
| | - Marilena Vitale
- Department of Clinical Medicine and Surgery, University of Naples Federico II, Naples, Italy
| | - Cassandra Serantoni
- Department of Neurosciences, Biophysics Section, Catholic University of the Sacred Heart, Rome, Italy
- Fondazione Policlinico Universitario A. Gemelli IRCCS, Rome, Italy
| | - Alessio Abeltino
- Department of Neurosciences, Biophysics Section, Catholic University of the Sacred Heart, Rome, Italy
- Fondazione Policlinico Universitario A. Gemelli IRCCS, Rome, Italy
| | - Teresa Mezza
- Department of Translational Medicine and Surgery, Catholic University of the Sacred Heart, Rome, Italy
- Department of Medical and Surgical Sciences, Center for Endocrine and Metabolic Diseases, Fondazione Policlinico Universitario A. Gemelli IRCCS, Rome, Italy
| | - Pietro Manuel Ferraro
- Unit of Nephrology, Fondazione Policlinico Universitario A. Gemelli IRCCS, Rome, Italy
| | - Marco De Spirito
- Department of Neurosciences, Biophysics Section, Catholic University of the Sacred Heart, Rome, Italy
- Fondazione Policlinico Universitario A. Gemelli IRCCS, Rome, Italy
| | - Gabriele Riccardi
- Department of Clinical Medicine and Surgery, University of Naples Federico II, Naples, Italy
| | - Andrea Giaccari
- Department of Translational Medicine and Surgery, Catholic University of the Sacred Heart, Rome, Italy
- Department of Medical and Surgical Sciences, Center for Endocrine and Metabolic Diseases, Fondazione Policlinico Universitario A. Gemelli IRCCS, Rome, Italy
| | - Giuseppe Maulucci
- Department of Neurosciences, Biophysics Section, Catholic University of the Sacred Heart, Rome, Italy
- Fondazione Policlinico Universitario A. Gemelli IRCCS, Rome, Italy
| |
Collapse
|
18
|
Sherratt SCR, Mason RP, Libby P, Steg PG, Bhatt DL. Do patients benefit from omega-3 fatty acids? Cardiovasc Res 2024; 119:2884-2901. [PMID: 38252923 PMCID: PMC10874279 DOI: 10.1093/cvr/cvad188] [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: 05/01/2023] [Revised: 08/11/2023] [Accepted: 09/26/2023] [Indexed: 01/24/2024] Open
Abstract
Omega-3 fatty acids (O3FAs) possess beneficial properties for cardiovascular (CV) health and elevated O3FA levels are associated with lower incident risk for CV disease (CVD.) Yet, treatment of at-risk patients with various O3FA formulations has produced disparate results in large, well-controlled and well-conducted clinical trials. Prescription formulations and fish oil supplements containing low-dose mixtures of eicosapentaenoic acid (EPA) and docosahexaenoic acid (DHA) have routinely failed to prevent CV events in primary and secondary prevention settings when added to contemporary care, as shown most recently in the STRENGTH and OMEMI trials. However, as observed in JELIS, REDUCE-IT, and RESPECT-EPA, EPA-only formulations significantly reduce CVD events in high-risk patients. The CV mechanism of action of EPA, while certainly multifaceted, does not depend solely on reductions of circulating lipids, including triglycerides (TG) and LDL, and event reduction appears related to achieved EPA levels suggesting that the particular chemical and biological properties of EPA, as compared to DHA and other O3FAs, may contribute to its distinct clinical efficacy. In vitro and in vivo studies have shown different effects of EPA compared with DHA alone or EPA/DHA combination treatments, on atherosclerotic plaque morphology, LDL and membrane oxidation, cholesterol distribution, membrane lipid dynamics, glucose homeostasis, endothelial function, and downstream lipid metabolite function. These findings indicate that prescription-grade, EPA-only formulations provide greater benefit than other O3FAs formulations tested. This review summarizes the clinical findings associated with various O3FA formulations, their efficacy in treating CV disease, and their underlying mechanisms of action.
Collapse
Affiliation(s)
- Samuel C R Sherratt
- Department of Molecular, Cellular, and Biomedical Sciences, University of New Hampshire, Durham, NH, USA
- Elucida Research LLC, Beverly, MA, USA
| | - R Preston Mason
- Elucida Research LLC, Beverly, MA, USA
- Department of Medicine, Cardiovascular Division, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA, USA
| | - Peter Libby
- Department of Medicine, Cardiovascular Division, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA, USA
| | - Ph Gabriel Steg
- Université Paris-Cité, INSERM_UMR1148/LVTS, FACT (French Alliance for Cardiovascular Trials), Assistance Publique–Hôpitaux de Paris, Hôpital Bichat, Paris, France
| | - Deepak L Bhatt
- Mount Sinai Fuster Heart Hospital, Icahn School of Medicine at Mount Sinai, 1 Gustave L. Levy Place, NewYork 10029-5674, NY, USA
| |
Collapse
|
19
|
Sublette ME, Daray FM, Ganança L, Shaikh SR. The role of polyunsaturated fatty acids in the neurobiology of major depressive disorder and suicide risk. Mol Psychiatry 2024; 29:269-286. [PMID: 37993501 DOI: 10.1038/s41380-023-02322-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/01/2023] [Revised: 10/19/2023] [Accepted: 11/07/2023] [Indexed: 11/24/2023]
Abstract
Long-chain polyunsaturated fatty acids (LC-PUFAs) are obtained from diet or derived from essential shorter-chain fatty acids, and are crucial for brain development and functioning. Fundamentally, LC-PUFAs' neurobiological effects derive from their physicochemical characteristics, including length and double bond configuration, which differentiate LC-PUFA species and give rise to functional differences between n(omega)-3 and n-6 LC-PUFAs. LC-PUFA imbalances are implicated in psychiatric disorders, including major depression and suicide risk. Dietary intake and genetic variants in enzymes involved in biosynthesis of LC-PUFAs from shorter chain fatty acids influence LC-PUFA status. Domains impacted by LC-PUFAs include 1) cell signaling, 2) inflammation, and 3) bioenergetics. 1) As major constituents of lipid bilayers, LC-PUFAs are determinants of cell membrane properties of viscosity and order, affecting lipid rafts, which play a role in regulation of membrane-bound proteins involved in cell-cell signaling, including monoaminergic receptors and transporters. 2) The n-3:n-6 LC-PUFA balance profoundly influences inflammation. Generally, metabolic products of n-6 LC-PUFAs (eicosanoids) are pro-inflammatory, while those of n-3 LC-PUFAs (docosanoids) participate in the resolution of inflammation. Additionally, n-3 LC-PUFAs suppress microglial activation and the ensuing proinflammatory cascade. 3) N-3 LC-PUFAs in the inner mitochondrial membrane affect oxidative stress, suppressing production of and scavenging reactive oxygen species (ROS), with neuroprotective benefits. Until now, this wealth of knowledge about LC-PUFA biomechanisms has not been adequately tapped to develop translational studies of LC-PUFA clinical effects in humans. Future studies integrating neurobiological mechanisms with clinical outcomes may suggest ways to identify depressed individuals most likely to respond to n-3 LC-PUFA supplementation, and mechanistic research may generate new treatment strategies.
Collapse
Affiliation(s)
- M Elizabeth Sublette
- Department of Psychiatry, Columbia University, New York, NY, USA.
- Molecular Imaging & Neuropathology Area, New York State Psychiatric Institute, New York, NY, USA.
| | - Federico Manuel Daray
- University of Buenos Aires, School of Medicine, Institute of Pharmacology, Buenos Aires, Argentina
- National Scientific and Technical Research Council, Buenos Aires, Argentina
| | - Licínia Ganança
- Clínica Universitária de Psiquiatria e Psicologia Médica, Faculdade de Medicina, Universidade de Lisboa, Lisbon, Portugal
- Departamento de Psiquiatria e Saúde Mental, Centro Hospitalar Universitário Lisboa Norte, Lisbon, Portugal
| | - Saame Raza Shaikh
- Nutritional Obesity Research Center, Department of Nutrition, Gillings School of Global Public Health and School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| |
Collapse
|
20
|
Chen Z, Kong Y, Huang Z, Zheng X, Zheng Z, Yao D, Yang S, Zhang Y, Aweya JJ. Exogenous alpha-linolenic acid and Vibrio parahaemolyticus induce EPA and DHA levels mediated by delta-6 desaturase to enhance shrimp immunity. Int J Biol Macromol 2024; 257:128583. [PMID: 38056755 DOI: 10.1016/j.ijbiomac.2023.128583] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2023] [Revised: 11/18/2023] [Accepted: 12/01/2023] [Indexed: 12/08/2023]
Abstract
Globally, penaeid shrimp are the most farmed and traded aquatic organisms, although they are easily susceptible to microbial pathogens. Moreover, there is a desire to increase the nutritional value of shrimp, especially the levels of n-3 polyunsaturated fatty acids (PUFAs), such as eicosapentaenoic acid (EPA) and docosahexaenoic acid (DHA), which also possess immunomodulatory and anti-inflammatory properties. Some aquatic animals can synthesize EPA and DHA from dietary plant-sourced alpha-linolenic acid (ALA), but penaeid shrimps' ability to synthesize these n-3 PUFAs is unknown. Here, molecular biology techniques, including gas chromatography-mass spectrometry, qPCR, ELISA, etc., were used to demonstrate that exogenous ALA or Vibrio parahaemolyticus could modulate EPA and DHA levels and immune genes in Penaeus vannamei by inducing key enzymes involved in n-3 PUFAs biosynthesis, such as delta desaturases and elongation of very long-chain fatty acid (ELOVLs). Most importantly, knockdown or inhibition of ∆6 desaturase significantly decreased EPA and DHA levels and immune gene expression even with exogenous ALA treatment, consequently affecting shrimp antibacterial immunity and survival. This study provides new insight into the potential of P. vannamei to synthesize n-3 PUFAs from exogenous ALA or upon bacteria challenge, which could be leveraged to increase their nutritional content and antimicrobial immunity.
Collapse
Affiliation(s)
- Zeyan Chen
- Guangdong Provincial Key Laboratory of Marine Biotechnology, Institute of Marine Sciences, Shantou University, Shantou 515063, China
| | - Yonghui Kong
- Guangdong Provincial Key Laboratory of Marine Biotechnology, Institute of Marine Sciences, Shantou University, Shantou 515063, China
| | - Zishu Huang
- Guangdong Provincial Key Laboratory of Marine Biotechnology, Institute of Marine Sciences, Shantou University, Shantou 515063, China
| | - Xiaoyu Zheng
- Guangdong Provincial Key Laboratory of Marine Biotechnology, Institute of Marine Sciences, Shantou University, Shantou 515063, China
| | - Zhihong Zheng
- Guangdong Provincial Key Laboratory of Marine Biotechnology, Institute of Marine Sciences, Shantou University, Shantou 515063, China
| | - Defu Yao
- Guangdong Provincial Key Laboratory of Marine Biotechnology, Institute of Marine Sciences, Shantou University, Shantou 515063, China
| | - Shen Yang
- Fujian Provincial Key Laboratory of Food Microbiology and Enzyme Engineering, College of Ocean Food and Biological Engineering, Jimei University, Xiamen 361021, China
| | - Yueling Zhang
- Guangdong Provincial Key Laboratory of Marine Biotechnology, Institute of Marine Sciences, Shantou University, Shantou 515063, China.
| | - Jude Juventus Aweya
- Fujian Provincial Key Laboratory of Food Microbiology and Enzyme Engineering, College of Ocean Food and Biological Engineering, Jimei University, Xiamen 361021, China; Guangdong Provincial Key Laboratory of Marine Biotechnology, Institute of Marine Sciences, Shantou University, Shantou 515063, China.
| |
Collapse
|
21
|
Chen H, Leng X, Liu S, Zeng Z, Huang F, Huang R, Zou Y, Xu Y. Association between dietary intake of omega-3 polyunsaturated fatty acids and all-cause and cardiovascular mortality among hypertensive adults: Results from NHANES 1999-2018. Clin Nutr 2023; 42:2434-2442. [PMID: 37871484 DOI: 10.1016/j.clnu.2023.09.011] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2023] [Revised: 08/30/2023] [Accepted: 09/10/2023] [Indexed: 10/25/2023]
Abstract
BACKGROUND Hypertensive adults are at a higher risk of cardiovascular morbidity and mortality. Dietary omega-3 polyunsaturated fatty acids (N3-PUFA) intake has been associated with cardiovascular benefits. However, few studies have specifically investigated whether dietary intake of N3-PUFA is associated with lower risk of all-cause and cardiovascular mortality among hypertensive adults in the U.S. METHODS This prospective cohort study included 26,914 hypertensive individuals 18 years or older who participated in 10 NHANES cycles from 1999 to 2018. Dietary levels of N3-PUFA were obtained from the 24-hour dietary recalls. The dietary data were linked to mortality records from the National Death Index through December 31, 2019. The associations between dietary N3-PUFA levels and mortality were evaluated by constructing the Multivariable Cox Proportional Hazards models. RESULTS We observed an increasing trend of dietary N3-PUFA intake levels over the years, mainly driven by alpha-linolenic acid (ALA). Lower all-cause mortality risk was observed among hypertensive adults with higher consumption of total N3-PUFA [adjusted hazards ratio, 95% confidence interval: 0.91 (0.86, 0.97)], plant-based ALA [0.88 (0.83, 0.93)], fish oil-based eicosapentaenoic acid (EPA) + docosahexaenoic acid (DHA) [0.91 (0.83, 0.99)], EPA [0.93 (0.88, 0.98)], docosapentaenoic acid (DPA) [0.73 (0.58, 0.91)], or DHA [0.95 (0.90, 0.99)]. Hypertensive adults were at lower risk of cardiovascular mortality if their diet contained higher levels of total N3-PUFA [0.68 (0.53, 0.88)], ALA [0.89 (0.80, 0.99)], EPA [0.87 (0.79, 0.97)] or DPA [0.86 (0.78, 0.95)]. Weighted quantile sum analysis showed that ALA, EPA, and DPA were the main contributors of the N3-PUFA benefits against mortality among hypertensive adults. CONCLUSIONS Dietary intake of N3-PUFA, particularly ALA, EPA, and DPA, was associated with lower risk of all-cause and cardiovascular mortality among U.S. hypertensive adults. These findings suggest that increasing dietary intake of N3-PUFA may serve as a potential strategy to lower hypertension-associated mortality risk.
Collapse
Affiliation(s)
- Hao Chen
- Department of Occupational and Environmental Health, School of Public Health, Guangxi Medical University, Nanning, 530021, China
| | - Xuebing Leng
- Department of Microbiology and Immunology, Miller School of Medicine, University of Miami, Miami, FL, 33146, USA
| | - Shaohui Liu
- Department of Toxicology, School of Public Health, Guangxi Medical University, Nanning, 530021, China
| | - Ziqi Zeng
- Department of Toxicology, School of Public Health, Guangxi Medical University, Nanning, 530021, China
| | - Feng Huang
- Department of Cardiology, The First Affiliated Hospital of Guangxi Medical University, Nanning, 530021, China; Department of Medical Research, The First Affiliated Hospital of Guangxi Medical University, Nanning, 530021, China
| | - Rongjie Huang
- Department of Cardiology, The First Affiliated Hospital of Guangxi Medical University, Nanning, 530021, China
| | - Yunfeng Zou
- Department of Toxicology, School of Public Health, Guangxi Medical University, Nanning, 530021, China.
| | - Yunan Xu
- Department of Medical Research, The First Affiliated Hospital of Guangxi Medical University, Nanning, 530021, China.
| |
Collapse
|
22
|
Qin J, Kurt E, LBassi T, Sa L, Xie D. Biotechnological production of omega-3 fatty acids: current status and future perspectives. Front Microbiol 2023; 14:1280296. [PMID: 38029217 PMCID: PMC10662050 DOI: 10.3389/fmicb.2023.1280296] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2023] [Accepted: 10/25/2023] [Indexed: 12/01/2023] Open
Abstract
Omega-3 fatty acids, including alpha-linolenic acids (ALA), eicosapentaenoic acid (EPA), and docosahexaenoic acid (DHA), have shown major health benefits, but the human body's inability to synthesize them has led to the necessity of dietary intake of the products. The omega-3 fatty acid market has grown significantly, with a global market from an estimated USD 2.10 billion in 2020 to a predicted nearly USD 3.61 billion in 2028. However, obtaining a sufficient supply of high-quality and stable omega-3 fatty acids can be challenging. Currently, fish oil serves as the primary source of omega-3 fatty acids in the market, but it has several drawbacks, including high cost, inconsistent product quality, and major uncertainties in its sustainability and ecological impact. Other significant sources of omega-3 fatty acids include plants and microalgae fermentation, but they face similar challenges in reducing manufacturing costs and improving product quality and sustainability. With the advances in synthetic biology, biotechnological production of omega-3 fatty acids via engineered microbial cell factories still offers the best solution to provide a more stable, sustainable, and affordable source of omega-3 fatty acids by overcoming the major issues associated with conventional sources. This review summarizes the current status, key challenges, and future perspectives for the biotechnological production of major omega-3 fatty acids.
Collapse
Affiliation(s)
| | | | | | | | - Dongming Xie
- Department of Chemical Engineering, University of Massachusetts Lowell, Lowell, MA, United States
| |
Collapse
|
23
|
Ly R, MacIntyre BC, Philips SM, McGlory C, Mutch DM, Britz-McKibbin P. Lipidomic studies reveal two specific circulating phosphatidylcholines as surrogate biomarkers of the omega-3 index. J Lipid Res 2023; 64:100445. [PMID: 37730162 PMCID: PMC10622695 DOI: 10.1016/j.jlr.2023.100445] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2023] [Revised: 09/13/2023] [Accepted: 09/14/2023] [Indexed: 09/22/2023] Open
Abstract
Optimal dietary intake of omega-3 long-chain polyunsaturated fatty acids (n3-LCPUFAs) is critical to human health across the lifespan. However, omega-3 index (O3I) determination is not routinely assessed due to complicated procedures for n3-LCPUFA analysis from the phospholipid (PL) fraction of erythrocytes. Herein, a high-throughput method for lipidomics based on multisegment injection-nonaqueous capillary electrophoresis-mass spectrometry was applied to identify circulating PLs as surrogate biomarkers of O3I in two randomized placebo-controlled trials. An untargeted lipidomic data workflow using a subgroup analysis of serum extracts from sunflower oil versus high-dose fish oil (FO)-supplemented participants revealed that ingested n3-LCPUFAs were primarily distributed as their phosphatidylcholines (PCs) relative to other PL classes. In both high-dose FO (5.0 g/day) and EPA-only trials (3.0 g/day), PC (16:0_20:5) was the most responsive PL, whereas PC (16:0_22:6) was selective to DHA-only supplementation. We also demonstrated that the sum concentration of both these PCs in fasting serum or plasma samples was positively correlated to the O3I following FO (r = 0.708, P = 1.02 × 10-11, n = 69) and EPA- or DHA-only supplementation (r = 0.768, P = 1.01 × 10-33, n = 167). Overall, DHA was more effective in improving the O3I (ΔO3I = 4.90 ± 1.33%) compared to EPA (ΔO3I = 2.99 ± 1.19%) in young Canadian adults who had a poor nutritional status with an O3I (3.50 ± 0.68%) at baseline. Our method enables the rapid assessment of the O3I by directly measuring two circulating PC species in small volumes of blood, which may facilitate screening applications for population and precision health.
Collapse
Affiliation(s)
- Ritchie Ly
- Department of Chemistry and Chemical Biology, McMaster University, Hamilton, ON, Canada
| | - Brittany C MacIntyre
- Department of Human Health and Nutritional Sciences, University of Guelph, Guelph, ON, Canada
| | - Stuart M Philips
- Department of Kinesiology, McMaster University, Hamilton, ON, Canada
| | - Chris McGlory
- Department of Kinesiology, McMaster University, Hamilton, ON, Canada; School of Kinesiology and Health Studies, Queen's University, Kingston, ON, Canada
| | - David M Mutch
- Department of Human Health and Nutritional Sciences, University of Guelph, Guelph, ON, Canada
| | - Philip Britz-McKibbin
- Department of Chemistry and Chemical Biology, McMaster University, Hamilton, ON, Canada.
| |
Collapse
|
24
|
Ali O, Szabó A. Review of Eukaryote Cellular Membrane Lipid Composition, with Special Attention to the Fatty Acids. Int J Mol Sci 2023; 24:15693. [PMID: 37958678 PMCID: PMC10649022 DOI: 10.3390/ijms242115693] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2023] [Revised: 10/24/2023] [Accepted: 10/25/2023] [Indexed: 11/15/2023] Open
Abstract
Biological membranes, primarily composed of lipids, envelop each living cell. The intricate composition and organization of membrane lipids, including the variety of fatty acids they encompass, serve a dynamic role in sustaining cellular structural integrity and functionality. Typically, modifications in lipid composition coincide with consequential alterations in universally significant signaling pathways. Exploring the various fatty acids, which serve as the foundational building blocks of membrane lipids, provides crucial insights into the underlying mechanisms governing a myriad of cellular processes, such as membrane fluidity, protein trafficking, signal transduction, intercellular communication, and the etiology of certain metabolic disorders. Furthermore, comprehending how alterations in the lipid composition, especially concerning the fatty acid profile, either contribute to or prevent the onset of pathological conditions stands as a compelling area of research. Hence, this review aims to meticulously introduce the intricacies of membrane lipids and their constituent fatty acids in a healthy organism, thereby illuminating their remarkable diversity and profound influence on cellular function. Furthermore, this review aspires to highlight some potential therapeutic targets for various pathological conditions that may be ameliorated through dietary fatty acid supplements. The initial section of this review expounds on the eukaryotic biomembranes and their complex lipids. Subsequent sections provide insights into the synthesis, membrane incorporation, and distribution of fatty acids across various fractions of membrane lipids. The last section highlights the functional significance of membrane-associated fatty acids and their innate capacity to shape the various cellular physiological responses.
Collapse
Affiliation(s)
- Omeralfaroug Ali
- Agrobiotechnology and Precision Breeding for Food Security National Laboratory, Institute of Physiology and Animal Nutrition, Department of Animal Physiology and Health, Hungarian University of Agriculture and Life Sciences, Guba Sándor Str. 40, 7400 Kaposvár, Hungary;
| | - András Szabó
- Agrobiotechnology and Precision Breeding for Food Security National Laboratory, Institute of Physiology and Animal Nutrition, Department of Animal Physiology and Health, Hungarian University of Agriculture and Life Sciences, Guba Sándor Str. 40, 7400 Kaposvár, Hungary;
- HUN-REN-MATE Mycotoxins in the Food Chain Research Group, Hungarian University of Agriculture and Life Sciences, Guba Sándor Str. 40, 7400 Kaposvár, Hungary
| |
Collapse
|
25
|
Sherratt SCR, Libby P, Dawoud H, Bhatt DL, Malinski T, Mason RP. Eicosapentaenoic acid (EPA) reduces pulmonary endothelial dysfunction and inflammation due to changes in protein expression during exposure to particulate matter air pollution. Biomed Pharmacother 2023; 162:114629. [PMID: 37027984 DOI: 10.1016/j.biopha.2023.114629] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2023] [Revised: 03/27/2023] [Accepted: 03/29/2023] [Indexed: 04/09/2023] Open
Abstract
AIMS Inhalation of air pollution small particle matter (PM) is a leading cause of cardiovascular (CV) disease. Exposure to PMs causes endothelial cell (EC) dysfunction as evidenced by nitric oxide (NO) synthase uncoupling, vasoconstriction and inflammation. Eicosapentaenoic acid (EPA) has been shown to mitigate PM-induced adverse cardiac changes in patients receiving omega-3 fatty acid supplementation. We set out to determine the pro-inflammatory effects of multiple PMs (urban and fine) on pulmonary EC NO bioavailability and protein expression, and whether EPA restores EC function under these conditions. METHODS AND RESULTS We pretreated pulmonary ECs with EPA and then exposed them to urban or fine air pollution PMs. LC/MS-based proteomic analysis to assess relative expression levels. Expression of adhesion molecules was measured by immunochemistry. The ratio of NO to peroxynitrite (ONOO-) release, an indication of eNOS coupling, was measured using porphyrinic nanosensors following calcium stimulation. Urban/fine PMs also modulated 9/12 and 13/36 proteins, respectively, linked to platelet and neutrophil degranulation pathways and caused > 50% (p < 0.001) decrease in the stimulated NO/ONOO- release ratio. EPA treatment altered expression of proteins involved in these inflammatory pathways, including a decrease in peroxiredoxin-5 and an increase in superoxide dismutase-1. EPA also increased expression of heme oxygenase-1 (HMOX1), a cytoprotective protein, by 2.1-fold (p = 0.024). EPA reduced elevations in sICAM-1 levels by 22% (p < 0.01) and improved the NO/ONOO- release ratio by > 35% (p < 0.05). CONCLUSION These cellular changes may contribute to anti-inflammatory, cytoprotective and lipid changes associated with EPA treatment during air pollution exposure.
Collapse
Affiliation(s)
- Samuel C R Sherratt
- Department of Molecular, Cellular, and Biomedical Sciences, University of New Hampshire, Durham, NH, USA; Elucida Research LLC, Beverly, MA, USA
| | - Peter Libby
- Department of Medicine, Cardiovascular Division, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Hazem Dawoud
- Nanomedical Research Laboratory, Ohio University, Athens, OH, USA
| | - Deepak L Bhatt
- Mount Sinai Heart, Icahn School of Medicine at Mount Sinai Health System, New York, NY, USA
| | - Tadeusz Malinski
- Nanomedical Research Laboratory, Ohio University, Athens, OH, USA.
| | - R Preston Mason
- Elucida Research LLC, Beverly, MA, USA; Department of Medicine, Cardiovascular Division, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| |
Collapse
|
26
|
Joardar A, Chakraborty H. Differential Behavior of Eicosapentaenoic and Docosahexaenoic Acids on the Organization, Dynamics, and Fusion of Homogeneous and Heterogeneous Membranes. LANGMUIR : THE ACS JOURNAL OF SURFACES AND COLLOIDS 2023; 39:4439-4449. [PMID: 36931902 DOI: 10.1021/acs.langmuir.3c00119] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/18/2023]
Abstract
Membrane fusion is a common course in innumerable biological processes that helps in the survival of eukaryotes. Enveloped viruses utilize this process to enter the host cells. Generally, the membrane lipid compositions play an important role in membrane fusion by modulating the membrane's physical properties and the behavior of membrane proteins in the cellular milieu. In this work, we have demonstrated the role of polyunsaturated fatty acids (PUFAs), such as eicosapentaenoic (EPA) and docosahexaenoic (DHA) acids, on the organization, dynamics, and fusion of homogeneous and heterogeneous membranes. We have exploited arrays of steady-state and time-resolved fluorescence spectroscopic methods and polyethylene glycol-induced membrane fusion assay to elucidate the behavior of EPA and DHA on dioleoyl phosphatidylcholine (DOPC)/cholesterol (CH) homogeneous and DOPC/sphingomyelin/CH heterogeneous membranes. Our results suggest that EPA and DHA display differential effects on two different membranes. The effects of PUFAs in homogeneous membranes are majorly attributed to their flexible chain dynamics, whereas the ability of PUFA-induced cholesterol transfer from the lo to the ld phase rules their behavior in heterogeneous membranes. Overall, our results provide detailed information on the effect of PUFAs on homogeneous and heterogeneous membranes.
Collapse
Affiliation(s)
- Ankita Joardar
- School of Chemistry, Sambalpur University, Jyoti Vihar, Burla, Odisha 768 019, India
| | - Hirak Chakraborty
- School of Chemistry, Sambalpur University, Jyoti Vihar, Burla, Odisha 768 019, India
| |
Collapse
|
27
|
Untargeted Metabolomics Based Prediction of Therapeutic Potential for Apigenin and Chrysin. Int J Mol Sci 2023; 24:ijms24044066. [PMID: 36835484 PMCID: PMC9967419 DOI: 10.3390/ijms24044066] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2022] [Revised: 02/05/2023] [Accepted: 02/10/2023] [Indexed: 02/22/2023] Open
Abstract
The prominent flavonoids apigenin and chrysin have been demonstrated to have systemic benefits. Our previous work was first to establish the impact of apigenin and chrysin on cellular transcriptome. In the current study, we have revealed the ability of apigenin and chrysin to alter the cellular metabolome based on our untargeted metabolomics. Based on our metabolomics data, both these structurally related flavonoids demonstrate diverging and converging properties. Apigenin demonstrated the potential to possess anti-inflammatory and vasorelaxant properties through the upregulation of intermediate metabolites of alpha-linolenic acid and linoleic acid pathways. Chrysin, on the other hand, exhibited abilities to inhibit protein and pyrimidine synthesis along with downregulation of gluconeogenesis pathways based on the altered metabolites detected. Chrysin-mediated metabolite changes are mostly due to its ability to modulate L-alanine metabolism and the urea cycle. On the other hand, both the flavonoids also demonstrated converging properties. Apigenin and chrysin were able to downregulate metabolites involved in cholesterol biosynthesis and uric acid synthesis, namely 7-dehydrocholesterol and xanthosine, respectively. This work will provide understanding regarding the diverse therapeutic potential of these naturally occurring flavonoids and help us in curbing an array of metabolic complications.
Collapse
|
28
|
Role of Omega-3 Fatty Acids in Cardiovascular Disease: the Debate Continues. Curr Atheroscler Rep 2023; 25:1-17. [PMID: 36580204 PMCID: PMC9834373 DOI: 10.1007/s11883-022-01075-x] [Citation(s) in RCA: 48] [Impact Index Per Article: 24.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/15/2022] [Indexed: 12/30/2022]
Abstract
PURPOSE OF REVIEW The omega-3 fatty acids (n3-FAs), eicosapentaenoic acid (EPA) and docosahexaenoic acid (DHA), have recently undergone testing for their ability to reduce residual cardiovascular (CV) risk among statin-treated subjects. The outcome trials have yielded highly inconsistent results, perhaps attributable to variations in dosage, formulation, and composition. In particular, CV trials using icosapent ethyl (IPE), a highly purified ethyl ester of EPA, reproducibly reduced CV events and progression of atherosclerosis compared with mixed EPA/DHA treatments. This review summarizes the mechanistic evidence for differences among n3-FAs on the development and manifestations of atherothrombotic disease. RECENT FINDINGS Large randomized clinical trials with n3-FAs have produced discordant outcomes despite similar patient profiles, doses, and triglyceride (TG)-lowering effects. A large, randomized trial with IPE, a prescription EPA only formulation, showed robust reduction in CV events in statin treated patients in a manner proportional to achieved blood EPA concentrations. Multiple trials using mixed EPA/DHA formulations have not shown such benefits, despite similar TG lowering. These inconsistencies have inspired investigations into mechanistic differences among n3-FAs, as EPA and DHA have distinct membrane interactions, metabolic products, effects on cholesterol efflux, antioxidant properties, and tissue distribution. EPA maintains normal membrane cholesterol distribution, enhances endothelial function, and in combination with statins improves features implicated in plaque stability and reduces lipid content of plaques. Insights into reductions in residual CV risk have emerged from clinical trials using different formulations of n3-FAs. Among high-risk patients on contemporary care, mixed n3-FA formulations showed no reduction in CV events. The distinct benefits of IPE in multiple trials may arise from pleiotropic actions that correlate with on-treatment EPA levels beyond TG-lowering. These effects include altered platelet function, inflammation, cholesterol distribution, and endothelial dysfunction. Elucidating such mechanisms of vascular protection for EPA may lead to new interventions for atherosclerosis, a disease that continues to expand worldwide.
Collapse
|
29
|
Venegas-Calerón M, Napier JA. New alternative sources of omega-3 fish oil. ADVANCES IN FOOD AND NUTRITION RESEARCH 2023. [PMID: 37516467 DOI: 10.1016/bs.afnr.2023.01.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/08/2023]
Abstract
Long-chain omega-3 polyunsaturated fatty acids such as eicosapentaenoic and docosahexaenoic acids play an important role in brain growth and development, as well as in the health of the body. These fatty acids are traditionally found in seafood, such as fish, fish oils, and algae. They can also be added to food or consumed through dietary supplements. Due to a lack of supply to meet current demand and the potential for adverse effects from excessive consumption of fish and seafood, new alternatives are being sought to achieve the recommended levels in a safe and sustainable manner. New sources have been studied and new production mechanisms have been developed. These new proposals, as well as the importance of these fatty acids, are discussed in this paper.
Collapse
|
30
|
Shaikh SR, Virk R, Van Dyke TE. Potential Mechanisms by Which Hydroxyeicosapentaenoic Acids Regulate Glucose Homeostasis in Obesity. Adv Nutr 2022; 13:2316-2328. [PMID: 35709423 PMCID: PMC9776734 DOI: 10.1093/advances/nmac073] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2022] [Revised: 02/16/2022] [Accepted: 06/13/2022] [Indexed: 01/29/2023] Open
Abstract
Dysregulation of glucose metabolism in response to diet-induced obesity contributes toward numerous complications, such as insulin resistance and hepatic steatosis. Therefore, there is a need to develop effective strategies to improve glucose homeostasis. In this review, we first discuss emerging evidence from epidemiological studies and rodent experiments that increased consumption of EPA (either as oily fish, or dietary/pharmacological supplements) may have a role in preventing impairments in insulin and glucose homeostasis. We then review the current evidence on how EPA-derived metabolites known as hydroxyeicosapentaenoic acids (HEPEs) may be a major mode of action by which EPA exerts its beneficial effects on glucose and lipid metabolism. Notably, cell culture and rodent studies show that HEPEs prevent fat accumulation in metabolic tissues through peroxisome proliferator activated receptor (PPAR)-mediated mechanisms. In addition, activation of the resolvin E1 pathway, either by administration of EPA in the diet or via intraperitoneal administration of resolvin E1, improves hyperglycemia, hyperinsulinemia, and liver steatosis through multiple mechanisms. These mechanisms include shifting immune cell phenotypes toward resolution of inflammation and preventing dysbiosis of the gut microbiome. Finally, we present the next steps for this line of research that will drive future precision randomized clinical trials with EPA and its downstream metabolites. These include dissecting the variables that drive heterogeneity in the response to EPA, such as the baseline microbiome profile and fatty acid status, circadian rhythm, genetic variation, sex, and age. In addition, there is a critical need to further investigate mechanisms of action for HEPEs and to establish the concentration of HEPEs in differing tissues, particularly in response to consumption of oily fish and EPA-enriched supplements.
Collapse
Affiliation(s)
- Saame Raza Shaikh
- Department of Nutrition, Gillings School of Global Public Health and School
of Medicine, The University of North Carolina at Chapel Hill, Chapel
Hill, NC, USA
| | - Rafia Virk
- Department of Nutrition, Gillings School of Global Public Health and School
of Medicine, The University of North Carolina at Chapel Hill, Chapel
Hill, NC, USA
| | - Thomas E Van Dyke
- Center for Clinical and Translational Research, The Forsyth
Institute, Cambridge, MA, USA
- Department of Oral Medicine, Infection, and Immunity, Harvard School of
Dental Medicine, Harvard Medical School, Boston, MA, USA
| |
Collapse
|
31
|
Cholesterol crystals and atherosclerotic plaque instability: Therapeutic potential of Eicosapentaenoic acid. Pharmacol Ther 2022; 240:108237. [PMID: 35772589 DOI: 10.1016/j.pharmthera.2022.108237] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2021] [Revised: 06/22/2022] [Accepted: 06/23/2022] [Indexed: 12/15/2022]
Abstract
Atherosclerotic plaques associated with acute coronary syndromes (ACS), i.e. culprit lesions, frequently feature a ruptured fibrous cap with thrombotic complications. On imaging, these plaques exhibit a low attenuation, lipid-rich, necrotic core containing cholesterol crystals and are inherently unstable. Indeed, cholesterol crystals are causally associated with plaque vulnerability in vivo; their formation results from spontaneous self-assembly of cholesterol molecules. Cholesterol homeostasis is a central determinant of the physicochemical conditions leading to crystal formation, which are favored by elevated membrane free cholesterol content in plaque endothelial cells, smooth muscle cells, monocyte-derived macrophages, and foam cells, and equally by lipid oxidation. Emerging evidence from imaging trials in patients with coronary heart disease has highlighted the impact of intervention involving the omega-3 fatty acid, eicosapentaenoic acid (EPA), on vulnerable, low attenuation atherosclerotic plaques. Thus, EPA decreased features associated with unstable plaque by increasing fibrous cap thickness in statin-treated patients, by reducing lipid volume and equally attenuating intraplaque inflammation. Importantly, atherosclerotic plaques rapidly incorporate EPA; indeed, a high content of EPA in plaque tissue is associated with decreased plaque inflammation and increased stability. These findings are entirely consistent with the major reduction seen in cardiovascular events in the REDUCE-IT trial, in which high dose EPA was administered as its esterified precursor, icosapent ethyl (IPE); moreover, clinical benefit was proportional to circulating EPA levels. Eicosapentaenoic acid is efficiently incorporated into phospholipids, where it modulates cholesterol-enriched domains in cell membranes through physicochemical lipid interactions and changes in rates of lipid oxidation. Indeed, biophysical analyses indicate that EPA exists in an extended conformation in membranes, thereby enhancing normal cholesterol distribution while reducing propagation of free radicals. Such effects mitigate cholesterol aggregation and crystal formation. In addition to its favorable effect on cholesterol domain structure, EPA/IPE exerts pleiotropic actions, including antithrombotic, antiplatelet, anti-inflammatory, and proresolving effects, whose plaque-stabilizing potential cannot be excluded. Docosahexaenoic acid is distinguished from EPA by a higher degree of unsaturation and longer carbon chain length; DHA is thus predisposed to changes in its conformation with ensuing increase in membrane lipid fluidity and promotion of cholesterol aggregation into discrete domains. Such distinct molecular effects between EPA and DHA are pronounced under conditions of high cellular cholesterol content and oxidative stress. This review will focus on the formation and role of cholesterol monohydrate crystals in destabilizing atherosclerotic plaques, and on the potential of EPA as a therapeutic agent to attenuate the formation of deleterious cholesterol membrane domains and of cholesterol crystals. Such a therapeutic approach may translate to enhanced plaque stability and ultimately to reduction in cardiovascular risk.
Collapse
|
32
|
Effect of Docosahexaenoic Acid and Eicosapentaenoic Acid Supplementation on Sleep Quality in Healthy Subjects: A Randomized, Double-Blinded, Placebo-Controlled Trial. Nutrients 2022; 14:nu14194136. [PMID: 36235788 PMCID: PMC9573173 DOI: 10.3390/nu14194136] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2022] [Revised: 09/30/2022] [Accepted: 10/01/2022] [Indexed: 11/17/2022] Open
Abstract
Docosahexaenoic acid (DHA) and eicosapentaenoic acid (EPA)-omega-3 fatty acids with various functions-influence sleep in children and young adults. However, only limited studies on their effects on sleep in middle- and old-aged adults have been reported. Therefore, we investigated the effects of DHA and EPA on sleep quality in subjects aged ≥ 45 years. We performed a randomized, placebo-controlled, double-blinded, parallel-grouped study, in which we randomly assigned 66 healthy Japanese males and females. Each individual received six 480 mg capsules containing 576 mg DHA and 284 mg EPA per day (DHA/EPA group, n = 33), or corn oil (placebo group, n = 33), for 12 weeks. Before and after the intervention, the Oguri-Shirakawa-Azumi sleep inventory MA version (OSA-MA) and the sleep state test were conducted. In the DHA/EPA group, factor III (frequent dreaming) scores among the OSA-MA scores were significantly improved compared to the placebo group. Additionally, sleep state tests revealed that sleep efficiency improved in the DHA/EPA group. To our knowledge, this study is the first to report that DHA/EPA improves sleep quality in middle- and old-aged individuals, even at doses lower than those administered in previous studies.
Collapse
|
33
|
Ruscica M, Sirtori CR, Carugo S, Calder PC, Corsini A. OMEGA-3 AND CARDIOVASCULAR PREVENTION – IS THIS STILL A CHOICE? Pharmacol Res 2022; 182:106342. [DOI: 10.1016/j.phrs.2022.106342] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/24/2022] [Revised: 07/01/2022] [Accepted: 07/01/2022] [Indexed: 02/07/2023]
|
34
|
Sherratt SCR, Libby P, Bhatt DL, Mason RP. A biological rationale for the disparate effects of omega-3 fatty acids on cardiovascular disease outcomes. Prostaglandins Leukot Essent Fatty Acids 2022; 182:102450. [PMID: 35690002 DOI: 10.1016/j.plefa.2022.102450] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/01/2022] [Revised: 05/09/2022] [Accepted: 05/19/2022] [Indexed: 12/29/2022]
Abstract
The omega-3 fatty acids (n3-FAs) eicosapentaenoic acid (EPA) and docosahexaenoic acid (DHA) rapidly incorporate into cell membranes where they modulate signal transduction pathways, lipid raft formation, and cholesterol distribution. Membrane n3-FAs also form specialized pro-resolving mediators and other intracellular oxylipins that modulate inflammatory pathways, including T-cell differentiation and gene expression. Cardiovascular (CV) trials have shown that EPA, administered as icosapent ethyl (IPE), reduces composite CV events, along with plaque volume, in statin-treated, high-risk patients. Mixed EPA/DHA regimens have not shown these benefits, perhaps as the result of differences in formulation, dosage, or potential counter-regulatory actions of DHA. Indeed, EPA and DHA have distinct, tissue-specific effects on membrane structural organization and cell function. This review summarizes: (1) results of clinical outcome and imaging trials using n3-FA formulations; (2) membrane interactions of n3-FAs; (3) effects of n3-FAs on membrane oxidative stress and cholesterol crystalline domain formation during hyperglycemia; (4) n3-FA effects on endothelial function; (5) role of n3-FA-generated metabolites in inflammation; and (6) ongoing and future clinical investigations exploring treatment targets for n3-FAs, including COVID-19.
Collapse
Affiliation(s)
- Samuel C R Sherratt
- Department of Molecular, Cellular, and Biomedical Sciences, University of New Hampshire, Durham, NH 03823, USA; Elucida Research LLC, Beverly, MA 01915-0091, USA
| | - Peter Libby
- Department of Medicine, Cardiovascular Division, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115-6110, USA
| | - Deepak L Bhatt
- Department of Medicine, Cardiovascular Division, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115-6110, USA
| | - R Preston Mason
- Department of Medicine, Cardiovascular Division, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115-6110, USA; Elucida Research LLC, Beverly, MA 01915-0091, USA.
| |
Collapse
|
35
|
Mason RP, Sherratt SCR, Eckel RH. Omega-3-fatty acids: Do they prevent cardiovascular disease? Best Pract Res Clin Endocrinol Metab 2022; 37:101681. [PMID: 35739003 DOI: 10.1016/j.beem.2022.101681] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
Abstract
Despite cardiovascular disease (CVD) reductions with high-intensity statins, there remains residual risk among patients with metabolic disorders. Alongside low-density lipoproteins (LDL-C), elevated triglycerides (TG) are associated with incident CVD events. Omega-3 fatty acids (n3-FAs), specifically eicosapentaenoic acid (EPA) and docosahexaenoic acid (DHA), lower TG levels, but their ability to reduce CV risk has been highly inconsistent. Trials using icosapent ethyl (IPE), a purified EPA ethyl ester, produced reductions in CVD events and atherosclerotic plaque regression compared with mixed EPA/DHA formulations despite similar TG-reductions. The separate effects of EPA and DHA on tissue distribution, oxidative stress, inflammation, membrane structure and endothelial function may contribute to these discordant outcomes. Additional mechanistic trials will provide further insights into the role of n3-FAs in reducing CVD risk beyond TG lowering.
Collapse
Affiliation(s)
- R Preston Mason
- Cardiovascular Division, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA 02115, USA.
| | - Samuel C R Sherratt
- Department of Molecular, Cellular and Biomedical Sciences, University of New Hampshire, Durham, NH 03823, USA
| | - Robert H Eckel
- Division of Endocrinology, Metabolism & Diabetes, Division of Cardiology, University of Colorado Anschutz Medical Campus, 1635 Aurora Court, Aurora, CO 80045, USA
| |
Collapse
|
36
|
Benefits of Icosapent Ethyl for Enhancing Residual Cardiovascular Risk Reduction A Review of Key Findings from REDUCE-IT. J Clin Lipidol 2022; 16:389-402. [DOI: 10.1016/j.jacl.2022.05.067] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2022] [Revised: 05/13/2022] [Accepted: 05/17/2022] [Indexed: 11/20/2022]
|
37
|
Gaba P, Bhatt DL, Steg PG, Miller M, Brinton EA, Jacobson TA, Ketchum SB, Juliano RA, Jiao L, Doyle RT, Granowitz C, Tardif JC, Giugliano RP, Martens FMAC, Gibson CM, Ballantyne CM. Prevention of Cardiovascular Events and Mortality With Icosapent Ethyl in Patients With Prior Myocardial Infarction. J Am Coll Cardiol 2022; 79:1660-1671. [PMID: 35483753 DOI: 10.1016/j.jacc.2022.02.035] [Citation(s) in RCA: 35] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/01/2021] [Revised: 01/19/2022] [Accepted: 02/11/2022] [Indexed: 12/21/2022]
Abstract
BACKGROUND REDUCE-IT was a double-blind trial that randomized 8,179 statin-treated patients with controlled low-density lipoprotein cholesterol and moderately elevated triglycerides to icosapent ethyl (IPE) or placebo. There was a significant reduction in the primary endpoint, including death from cardiovascular (CV) causes. The specific impact of IPE among patients with prior myocardial infarction (MI) was unknown. OBJECTIVES Our goal was to examine the benefit of IPE on ischemic events among patients with prior MI in REDUCE-IT. METHODS We performed post hoc analyses of patients with prior MI. The primary endpoint was CV death, MI, stroke, coronary revascularization, or hospitalization for unstable angina. The key secondary endpoint was CV death, MI, or stroke. RESULTS A total of 3,693 patients had a history of prior MI. The primary endpoint was reduced from 26.1% to 20.2% with IPE vs placebo; HR: 0.74 (95% CI: 0.65-0.85; P = 0.00001). The key secondary endpoint was reduced from 18.0% to 13.3%; HR: 0.71 (95% CI: 0.61-0.84; P = 0.00006). There was also a significant 35% relative risk reduction in total ischemic events (P = 0.0000001), a 34% reduction in MI (P = 0.00009), a 30% reduction in CV death (P = 0.01), and a 20% lower rate of all-cause mortality (P = 0.054), although there was a slight increase in atrial fibrillation. Sudden cardiac death and cardiac arrest were also significantly reduced by 40% and 56%, respectively. CONCLUSIONS Patients with a history of prior MI in REDUCE-IT treated with IPE demonstrated large and significant relative and absolute risk reductions in ischemic events, including CV death. (A Study of AMR101 to Evaluate Its Ability to Reduce Cardiovascular Events in High Risk Patients With Hypertriglyceridemia and on Statin. The Primary Objective is to Evaluate the Effect of 4 g/Day AMR101 for Preventing the Occurrence of a First Major Cardiovascular Event. [REDUCE-IT]; NCT01492361).
Collapse
Affiliation(s)
- Prakriti Gaba
- Division of Cardiovascular Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Deepak L Bhatt
- Division of Cardiovascular Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts, USA.
| | - Ph Gabriel Steg
- Université de Paris, FACT (French Alliance for Cardiovascular Trials), Assistance Publique-Hôpitaux de Paris, Hôpital Bichat, INSERM Unité 1148, Paris, France
| | - Michael Miller
- Department of Medicine, University of Maryland School of Medicine, Baltimore, Maryland, USA
| | | | - Terry A Jacobson
- Office of Health Promotion and Disease Prevention, Department of Medicine, Emory University School of Medicine, Atlanta, Georgia, USA
| | | | | | - Lixia Jiao
- Amarin Pharma, Inc (Amarin), Bridgewater, New Jersey, USA
| | - Ralph T Doyle
- Amarin Pharma, Inc (Amarin), Bridgewater, New Jersey, USA
| | | | - Jean-Claude Tardif
- Montreal Heart Institute, Université de Montréal, Montreal, Quebec, Canada
| | - Robert P Giugliano
- Division of Cardiovascular Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Fabrice M A C Martens
- Werkgroep Cardiologische centra Nederland (WCN: Dutch Network for Cardiovascular Research) and the Department of Cardiology, Deventer Hospital, Deventer, the Netherlands
| | - C Michael Gibson
- Department of Cardiovascular Diseases, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts, USA
| | - Christie M Ballantyne
- Department of Medicine, Baylor College of Medicine, Houston, Texas, USA; Center for Cardiovascular Disease Prevention, Methodist DeBakey Heart and Vascular Center, Houston, Texas, USA
| | | |
Collapse
|
38
|
Toth PP, Chapman MJ, Parhofer KG, Nelson JR. Differentiating EPA from EPA/DHA in cardiovascular risk reduction. AMERICAN HEART JOURNAL PLUS : CARDIOLOGY RESEARCH AND PRACTICE 2022; 17:100148. [PMID: 38559888 PMCID: PMC10978325 DOI: 10.1016/j.ahjo.2022.100148] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/15/2022] [Revised: 05/19/2022] [Accepted: 05/19/2022] [Indexed: 04/04/2024]
Abstract
None of the clinical trials of omega-3 fatty acids using combinations of eicosapentaenoic acid (EPA) and docosahexaenoic acid (DHA) were able to show any effect on cardiovascular outcomes, despite reductions in triglyceride levels. In contrast, the Reduction of Cardiovascular Events With Icosapent Ethyl-Intervention Trial (REDUCE-IT), which employed high-dose (4 g) purified EPA, demonstrated a 25% reduction in atherosclerotic cardiovascular disease-related events compared with placebo (hazard ratio 0.75; 95% confidence interval 0.68-0.83; P < 0.001). Moreover, REDUCE-IT is the first clinical trial using a lipid-lowering agent as adjuvant therapy to a statin to show a significant reduction in cardiovascular mortality. Significant reductions in stroke, need for revascularization, and myocardial infarction were also observed. The pharmacology of EPA is distinct from that of DHA, with a differential effect on membrane structure, lipoprotein oxidation, and the production of downstream metabolites that promote the resolution of inflammation. Attained plasma levels of EPA may be an important determinant of efficacy, with a substudy of REDUCE-IT suggesting that the threshold for clinical benefit of EPA is approximately 100 μg/mL, a level achieved in only a minority of patients in other studies. No similar clinical trials of DHA monotherapy have been conducted, so no such threshold has been established. The results of the REDUCE-IT and the Japan EPA Lipid Intervention Study (JELIS) together affirm the efficacy of EPA therapy for cardiovascular disease risk reduction in certain patient populations.
Collapse
Affiliation(s)
- Peter P. Toth
- CGH Medical Center, Sterling, IL, USA
- Cicarrone Center for the Prevention of Cardiovascular Disease, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | | | - Klaus G. Parhofer
- Medical Clinic IV – Grosshadern Hospital of the University of Munich, Munich, Germany
| | | |
Collapse
|
39
|
Peterson BE, Bhatt DL, Steg PG, Miller M, Brinton EA, Jacobson TA, Ketchum SB, Juliano RA, Jiao L, Doyle RT, Granowitz C, Gibson CM, Pinto D, Giugliano RP, Budoff MJ, Tardif JC, Verma S, Ballantyne CM. Treatment With Icosapent Ethyl to Reduce Ischemic Events in Patients With Prior Percutaneous Coronary Intervention: Insights From REDUCE-IT PCI. J Am Heart Assoc 2022; 11:e022937. [PMID: 35261279 PMCID: PMC9075300 DOI: 10.1161/jaha.121.022937] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Background Patients who undergo percutaneous coronary intervention (PCI) are at increased risk for recurrent cardiovascular events despite aggressive medical therapy. Methods and Results This post hoc analysis focused on the subset of patients with prior PCI enrolled in REDUCE‐IT (Reduction of Cardiovascular Events With Icosapent Ethyl–Intervention Trial), a multicenter, randomized, double‐blind, placebo‐controlled trial of icosapent ethyl versus placebo. Icosapent ethyl was added to statins in patients with low‐density lipoprotein cholesterol <100 mg/dL and fasting triglycerides 135–499 mg/dL. The primary end point was a composite of cardiovascular death, nonfatal myocardial infarction, nonfatal stroke, coronary revascularization, or unstable angina requiring hospitalization. There were 8179 patients randomized in REDUCE‐IT followed for a median of 4.9 years, and 3408 (41.7%) of them had a prior PCI with a median follow‐up of 4.8 years. These patients were randomized a median of 2.9 years (11 days to 30.7 years) after PCI. Among patients treated with icosapent ethyl versus placebo, there was a 34% reduction in the primary composite end point (hazard ratio [HR], 0.66; 95% CI, 0.58–0.76; P<0.001; number needed to treat4.8 years=12) and a 34% reduction in the key secondary composite end point of cardiovascular death, nonfatal myocardial infarction, or nonfatal stroke (HR, 0.66; 95% CI, 0.56–0.79; P<0.001; NNT4.8 years=19) versus placebo. Similarly, large reductions occurred in total coronary revascularizations and revascularization subtypes. There was also a 39% reduction in total events (rate ratio, 0.61; 95% CI, 0.52–0.72; P<0.001). Conclusions Among patients treated with statins with elevated triglycerides and a history of prior PCI, icosapent ethyl substantially reduced the risk of recurrent events during an average of ~5 years of follow‐up with a number needed to treat of only 12. Registration URL: https://www.clinicaltrials.gov; Unique identifier: NCT01492361.
Collapse
Affiliation(s)
- Benjamin E Peterson
- Brigham and Women's Hospital Heart and Vascular Center Harvard Medical School Boston MA
| | - Deepak L Bhatt
- Brigham and Women's Hospital Heart and Vascular Center Harvard Medical School Boston MA
| | - Ph Gabriel Steg
- Université de ParisAP-HP (Assistance Publique-Hôpitaux de Paris)Hôpital BichatFACT (French Alliance for Cardiovascular Trials)INSERM U-1148 Paris France
| | - Michael Miller
- Department of Medicine University of Maryland School of Medicine Baltimore MD
| | | | - Terry A Jacobson
- Department of Medicine Office of Health Promotion and Disease Prevention Emory University School of Medicine Atlanta GA
| | | | | | - Lixia Jiao
- Amarin Pharma, Inc. (Amarin) Bridgewater NJ
| | | | | | | | | | - Robert P Giugliano
- Brigham and Women's Hospital Heart and Vascular Center Harvard Medical School Boston MA
| | | | | | - Subodh Verma
- Division of Cardiac Surgery St Michael's HospitalUniversity of Toronto Ontario Canada
| | - Christie M Ballantyne
- Department of Medicine Baylor College of MedicineCenter for Cardiovascular Disease PreventionMethodist DeBakey Heart and Vascular Center Houston TX
| | | |
Collapse
|
40
|
Bhatt RD, Libby P, Verma S, Mason RP, Bhatt DL. The role of eicosapentaenoic acid in reducing important cardiovascular events, including coronary revascularization. Prog Cardiovasc Dis 2021; 69:3-10. [PMID: 34419485 DOI: 10.1016/j.pcad.2021.08.003] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/17/2021] [Accepted: 08/17/2021] [Indexed: 12/23/2022]
Abstract
The omega-3 fatty acid eicosapentaenoic acid has an important role in human health. The Reduction of Cardiovascular Events with Icosapent Ethyl-Intervention Trial (REDUCE-IT) examined the prescription omega-3 fatty acid icosapent ethyl (IPE) in patients with established cardiovascular disease (CVD) or with diabetes plus additional CVD risk factors. The trial found a large reduction in CVD events, including significant reductions in CVD death, myocardial infarction, stroke, coronary revascularization, and hospitalization for unstable angina. These results led to the regulatory approval of IPE in a population similar to REDUCE-IT participants in the United States, Canada, United Kingdom, and the European Union. Moreover, multiple international guidelines have endorsed the use of IPE in such individuals. A secondary analysis of REDUCE-IT examined the endpoint of coronary artery revascularization. This analysis showed a significant reduction not only in coronary revascularization overall but also in elective, urgent, and emergent coronary revascularization. Additionally, IPE significantly reduced the need for both percutaneous coronary intervention and for coronary artery bypass graft surgery. Coronary imaging studies have demonstrated significant decreases in rates of plaque progression with IPE, with significant effects within 6-9 months. In parallel, experimental findings corroborate several effects of IPE that provide mechanisms that could contribute to the profound reductions in multiple types of ischemic events, including percutaneous and surgical coronary revascularization. Future trials should explore potential benefits of initiation of IPE at the time of revascularization in broader populations, potentially in conjunction with loading doses.
Collapse
Affiliation(s)
| | - Peter Libby
- Brigham and Women's Hospital Heart and Vascular Center, Harvard Medical School, Boston, MA, USA
| | - Subodh Verma
- Division of Cardiac Surgery, St Michael's Hospital, University of Toronto, ON, Canada
| | - R Preston Mason
- Brigham and Women's Hospital Heart and Vascular Center, Harvard Medical School, Boston, MA, USA; Elucida Research, Beverly, MA, USA
| | - Deepak L Bhatt
- Brigham and Women's Hospital Heart and Vascular Center, Harvard Medical School, Boston, MA, USA.
| |
Collapse
|