1
|
Miyauchi K, Iwata H, Nishizaki Y, Inoue T, Hirayama A, Kimura K, Ozaki Y, Murohara T, Ueshima K, Kuwabara Y, Tanaka-Mizuno S, Yanagisawa N, Sato T, Daida H. Randomized Trial for Evaluation in Secondary Prevention Efficacy of Combination Therapy-Statin and Eicosapentaenoic Acid (RESPECT-EPA). Circulation 2024; 150:425-434. [PMID: 38873793 DOI: 10.1161/circulationaha.123.065520] [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: 05/09/2023] [Accepted: 05/24/2024] [Indexed: 06/15/2024]
Abstract
BACKGROUND Low plasma levels of eicosapentaenoic acid (EPA) are associated with cardiovascular events. This trial aimed to assess the clinical benefits of icosapent ethyl in patients with coronary artery disease, a low EPA/arachidonic acid (AA) ratio, and statin treatment. METHODS In this prospective, multicenter, randomized, open-label, blinded end-point study, patients with stable coronary artery disease and a low EPA/AA ratio (<0.4) were randomized to EPA (1800 of icosapent ethyl administered daily) or control group. The primary end point was a composite of cardiovascular death, nonfatal myocardial infarction, nonfatal ischemic stroke, unstable angina pectoris, and coronary revascularization. The secondary composite end points of coronary events included sudden cardiac death, fatal and nonfatal myocardial infarction, unstable angina requiring emergency hospitalization and coronary revascularization, or coronary revascularization. RESULTS Overall, 3884 patients were enrolled at 95 sites in Japan. Among them, 2506 patients had a low EPA/AA ratio, and 1249 and 1257 patients were randomized to the EPA and control group, respectively. The median EPA/AA ratio was 0.243 (interquartile range, 0.180-0.314) and 0.235 (interquartile range, 0.163-0.310) in the EPA and control group, respectively. Over a median period of 5 years, the primary end point occurred in 112 of 1225 patients (9.1%) and 155 of 1235 patients (12.6%) in the EPA and control group, respectively (hazard ratio, 0.79 [95% CI, 0.62-1.00]; P=0.055). Meanwhile, the secondary composite end point of coronary events in the EPA group was significantly lower (81/1225 [6.6%] versus 120/1235 [9.7%] patients; hazard ratio, 0.73 [95% CI, 0.55-0.97]). Adverse events did not differ between the groups, but the rate of new-onset atrial fibrillation was significantly higher in the EPA group (3.1% versus 1.6%; P=0.017). CONCLUSIONS Icosapent ethyl treatment resulted in a numerically lower risk of cardiovascular events that did not reach statistical significance in patients with chronic coronary artery disease, a low EPA/AA ratio, and statin treatment. REGISTRATION URL: https://www.umin.ac.jp/ctr/; Unique identifier: UMIN000012069.
Collapse
Affiliation(s)
- Katsumi Miyauchi
- Department of Cardiovascular Biology and Medicine, Juntendo University Graduate School of Medicine, Bunkyo-ku, Tokyo, Japan (K.M., H.I., Y.N., H.D.)
| | - Hiroshi Iwata
- Department of Cardiovascular Biology and Medicine, Juntendo University Graduate School of Medicine, Bunkyo-ku, Tokyo, Japan (K.M., H.I., Y.N., H.D.)
| | - Yuji Nishizaki
- Department of Cardiovascular Biology and Medicine, Juntendo University Graduate School of Medicine, Bunkyo-ku, Tokyo, Japan (K.M., H.I., Y.N., H.D.)
| | - Teruo Inoue
- Japan Red Cross Society, Nasu Red Cross Hospital, Otawara, Tochigi, Japan (T.I.)
- Dokkyo Medical University, Mibu, Tochigi, Japan (T.I.)
| | - Atsushi Hirayama
- Osaka Anti-Tuberculosis Association, Osaka Fukujyuji Hospital, Neyagawa, Osaka, Japan (A.H.)
| | - Kazuo Kimura
- Department of Cardiology, Yokohama City University Medical Center, Kanagawa, Japan (K.K.)
| | - Yukio Ozaki
- Department of Cardiology, Fujita Health University, Toyoake, Aichi, Japan (Y.O.)
| | - Toyoaki Murohara
- Department of Cardiology, Nagoya University Graduate School of Medicine, Nagoya, Aichi, Japan (T.M.)
| | - Kenji Ueshima
- Medical Examination Center, Uji-Takeda Hospital, Uji, Kyoto, Japan (K.U.)
| | - Yoshihiro Kuwabara
- Cancer Control Center, Osaka International Cancer Institute, Osaka, Osaka, Japan (Y.K.)
| | - Sachiko Tanaka-Mizuno
- Department of Digital Health and Epidemiology, Graduate School of Medicine, Kyoto University, Kyoto, Kyoto, Japan (S.T.-M.)
| | - Naotake Yanagisawa
- Medical Technology Innovation Center (N.Y.), Juntendo University, Bunkyo-ku, Tokyo, Japan
| | - Tosiya Sato
- Department of Biostatistics, Kyoto University School of Public Health, Kyoto, Kyoto, Japan (T.S.)
| | - Hiroyuki Daida
- Department of Cardiovascular Biology and Medicine, Juntendo University Graduate School of Medicine, Bunkyo-ku, Tokyo, Japan (K.M., H.I., Y.N., H.D.)
- Faculty of Health Science (H.D.), Juntendo University, Bunkyo-ku, Tokyo, Japan
| |
Collapse
|
2
|
Okamura T, Tsukamoto K, Arai H, Fujioka Y, Ishigaki Y, Koba S, Ohmura H, Shoji T, Yokote K, Yoshida H, Yoshida M, Deguchi J, Dobashi K, Fujiyoshi A, Hamaguchi H, Hara M, Harada-Shiba M, Hirata T, Iida M, Ikeda Y, Ishibashi S, Kanda H, Kihara S, Kitagawa K, Kodama S, Koseki M, Maezawa Y, Masuda D, Miida T, Miyamoto Y, Nishimura R, Node K, Noguchi M, Ohishi M, Saito I, Sawada S, Sone H, Takemoto M, Wakatsuki A, Yanai H. Japan Atherosclerosis Society (JAS) Guidelines for Prevention of Atherosclerotic Cardiovascular Diseases 2022. J Atheroscler Thromb 2024; 31:641-853. [PMID: 38123343 DOI: 10.5551/jat.gl2022] [Citation(s) in RCA: 50] [Impact Index Per Article: 50.0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2023] Open
Affiliation(s)
- Tomonori Okamura
- Preventive Medicine and Public Health, Keio University School of Medicine
| | | | | | - Yoshio Fujioka
- Faculty of Nutrition, Division of Clinical Nutrition, Kobe Gakuin University
| | - Yasushi Ishigaki
- Division of Diabetes, Metabolism and Endocrinology, Department of Internal Medicine, Iwate Medical University
| | - Shinji Koba
- Division of Cardiology, Department of Medicine, Showa University School of Medicine
| | - Hirotoshi Ohmura
- Department of Cardiovascular Biology and Medicine, Juntendo University Graduate School of Medicine
| | - Tetsuo Shoji
- Department of Vascular Medicine, Osaka Metropolitan University Graduate school of Medicine
| | - Koutaro Yokote
- Department of Endocrinology, Hematology and Gerontology, Chiba University Graduate School of Medicine
| | - Hiroshi Yoshida
- Department of Laboratory Medicine, The Jikei University Kashiwa Hospital
| | | | - Juno Deguchi
- Department of Vascular Surgery, Saitama Medical Center, Saitama Medical University
| | - Kazushige Dobashi
- Department of Pediatrics, School of Medicine, University of Yamanashi
| | | | | | - Masumi Hara
- Department of Internal Medicine, Mizonokuchi Hospital, Teikyo University School of Medicine
| | - Mariko Harada-Shiba
- Cardiovascular Center, Osaka Medical and Pharmaceutical University
- Department of Molecular Pathogenesis, National Cerebral and Cardiovascular Center Research Institute
| | - Takumi Hirata
- Institute for Clinical and Translational Science, Nara Medical University
| | - Mami Iida
- Department of Internal Medicine and Cardiology, Gifu Prefectural General Medical Center
| | - Yoshiyuki Ikeda
- Department of Cardiovascular Medicine and Hypertension, Graduate School of Medical and Dental Sciences, Kagoshima University
| | - Shun Ishibashi
- Division of Endocrinology and Metabolism, Department of Internal Medicine, Jichi Medical University, School of Medicine
- Current affiliation: Ishibashi Diabetes and Endocrine Clinic
| | - Hideyuki Kanda
- Department of Public Health, Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama University
| | - Shinji Kihara
- Medical Laboratory Science and Technology, Division of Health Sciences, Osaka University graduate School of medicine
| | - Kazuo Kitagawa
- Department of Neurology, Tokyo Women's Medical University Hospital
| | - Satoru Kodama
- Department of Prevention of Noncommunicable Diseases and Promotion of Health Checkup, Department of Hematology, Endocrinology and Metabolism, Niigata University Faculty of Medicine
| | - Masahiro Koseki
- Department of Cardiovascular Medicine, Osaka University Graduate School of Medicine
| | - Yoshiro Maezawa
- Department of Endocrinology, Hematology and Gerontology, Chiba University Graduate School of Medicine
| | - Daisaku Masuda
- Department of Cardiology, Center for Innovative Medicine and Therapeutics, Dementia Care Center, Doctor's Support Center, Health Care Center, Rinku General Medical Center
| | - Takashi Miida
- Department of Clinical Laboratory Medicine, Juntendo University Graduate School of Medicine
| | | | - Rimei Nishimura
- Department of Diabetes, Metabolism and Endocrinology, The Jikei University School of Medicine
| | - Koichi Node
- Department of Cardiovascular Medicine, Saga University
| | - Midori Noguchi
- Division of Public Health, Department of Social Medicine, Graduate School of Medicine, Osaka University
| | - Mitsuru Ohishi
- Department of Cardiovascular Medicine and Hypertension, Graduate School of Medical and Dental Sciences, Kagoshima University
| | - Isao Saito
- Department of Public Health and Epidemiology, Faculty of Medicine, Oita University
| | - Shojiro Sawada
- Division of Metabolism and Diabetes, Faculty of Medicine, Tohoku Medical and Pharmaceutical University
| | - Hirohito Sone
- Department of Hematology, Endocrinology and Metabolism, Niigata University Faculty of Medicine
| | - Minoru Takemoto
- Department of Diabetes, Metabolism and Endocrinology, International University of Health and Welfare
| | | | - Hidekatsu Yanai
- Department of Diabetes, Endocrinology and Metabolism, National Center for Global Health and Medicine Kohnodai Hospital
| |
Collapse
|
3
|
Luo S, Hou H, Wang Y, Li Y, Zhang L, Zhang H, Jin Q, Wu G, Wang X. Effects of omega-3, omega-6, and total dietary polyunsaturated fatty acid supplementation in patients with atherosclerotic cardiovascular disease: a systematic review and meta-analysis. Food Funct 2024; 15:1208-1222. [PMID: 38224465 DOI: 10.1039/d3fo02522e] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2024]
Abstract
Background: Uncertainty exists about the link between omega-3 fatty acid, omega-6 fatty acid, and total polyunsaturated fatty acid (PUFA) intake and mortality in atherosclerotic cardiovascular disease (ASCVD) patients, and no meta-analyses summarize the relationship between these various types of PUFAs and ASCVD. Methods: Web of Science, PubMed, EBSCO and Cochrane Library up to November 30, 2022 were searched for prospective randomized controlled studies investigating the relationships among omega-3, omega-6, and PUFA intake and mortality and cardiovascular events in ASCVD patients. This study has been registered at PROSPERO (No. CRD42023407566). Results: This meta-analysis included 21 publications from 17 studies involving 40 861 participants published between 1965 and 2022. In ASCVD patients, omega-3 may lower all-cause mortality (RR: 0.90, 95% CI [0.83, 0.98], I2 = 8%), CVD mortality (RR: 0.82, 95% CI [0.73, 0.91], I2 = 34%) and CVD events (RR: 0.90, 95% CI [0.86, 0.93], I2 = 79%). Subgroup analyses showed that EPA or EPA ethyl ester supplementation reduced CVD events, while the mixture of EPA and DHA had no significant impact. Long-chain omega-3 consumption of 1.0-4.0 g per d reduced death risk by 3.5% for each 1 g per d increase. Omega-6 and PUFA had no significant effect on mortality or CVD events, with low-quality evidence and significant heterogeneity. Conclusions: omega-3 intake is associated with a reduced risk of all-cause mortality, CVD mortality, and CVD events in ASCVD patients, while omega-6 or total PUFA intake showed no significant association. Increasing the omega-3 intake by 1 g per d resulted in a 3.5% decrease in the risk of death. These findings support the recommendation of supplements with omega-3 fatty acids for the secondary prevention of ASCVD.
Collapse
Affiliation(s)
- Siqi Luo
- State Key Laboratory of Food Science and Resources, School of Food Science and Technology, National Engineering Research Center for Functional Food, International Joint Research Laboratory for Lipid Nutrition and Safety, Collaborative Innovation Center of Food Safety and Quality Control in Jiangsu Province, Jiangnan University, Wuxi, China.
| | - Hongmei Hou
- Department of Neonatology, The Affiliated Wuxi Children's Hospital of Jiangnan University, Wuxi, China
| | - Yongjin Wang
- State Key Laboratory of Food Science and Resources, School of Food Science and Technology, National Engineering Research Center for Functional Food, International Joint Research Laboratory for Lipid Nutrition and Safety, Collaborative Innovation Center of Food Safety and Quality Control in Jiangsu Province, Jiangnan University, Wuxi, China.
| | - Yun Li
- Department of Neonatology, The Affiliated Wuxi Children's Hospital of Jiangnan University, Wuxi, China
| | - Le Zhang
- Department of Neonatology, The Affiliated Wuxi Children's Hospital of Jiangnan University, Wuxi, China
| | - Hui Zhang
- State Key Laboratory of Food Science and Resources, School of Food Science and Technology, National Engineering Research Center for Functional Food, International Joint Research Laboratory for Lipid Nutrition and Safety, Collaborative Innovation Center of Food Safety and Quality Control in Jiangsu Province, Jiangnan University, Wuxi, China.
| | - Qingzhe Jin
- State Key Laboratory of Food Science and Resources, School of Food Science and Technology, National Engineering Research Center for Functional Food, International Joint Research Laboratory for Lipid Nutrition and Safety, Collaborative Innovation Center of Food Safety and Quality Control in Jiangsu Province, Jiangnan University, Wuxi, China.
| | - Gangcheng Wu
- State Key Laboratory of Food Science and Resources, School of Food Science and Technology, National Engineering Research Center for Functional Food, International Joint Research Laboratory for Lipid Nutrition and Safety, Collaborative Innovation Center of Food Safety and Quality Control in Jiangsu Province, Jiangnan University, Wuxi, China.
| | - Xingguo Wang
- State Key Laboratory of Food Science and Resources, School of Food Science and Technology, National Engineering Research Center for Functional Food, International Joint Research Laboratory for Lipid Nutrition and Safety, Collaborative Innovation Center of Food Safety and Quality Control in Jiangsu Province, Jiangnan University, Wuxi, China.
| |
Collapse
|
4
|
Saito K, Koizumi S. A promising drug for neuropathic pain: identification of vesicular nucleotide transporter as a novel target of eicosapentaenoic acid. Purinergic Signal 2023; 19:587-589. [PMID: 36627401 PMCID: PMC10754788 DOI: 10.1007/s11302-022-09918-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2022] [Accepted: 12/26/2022] [Indexed: 01/12/2023] Open
Affiliation(s)
- Kozo Saito
- Department of Neuropharmacology, Interdisciplinary Graduate School of Medicine, University of Yamanashi, 1110 Shimokato, Chuo, Yamanashi, 409-3898, Japan
- Yamanashi GLIA Center, University of Yamanashi, Chuo, Yamanashi, Japan
| | - Schuichi Koizumi
- Department of Neuropharmacology, Interdisciplinary Graduate School of Medicine, University of Yamanashi, 1110 Shimokato, Chuo, Yamanashi, 409-3898, Japan.
- Yamanashi GLIA Center, University of Yamanashi, Chuo, Yamanashi, Japan.
| |
Collapse
|
5
|
Kim MS, Kim JM, Lee SG, Jung EJ, Lee SH, Huang WY, Han BK, Jung DE, Yang SB, Ji I, Kim YJ, Hong JY. Assessing Health and Economic Benefits of Omega-3 Fatty Acid Supplementation on Cardiovascular Disease in the Republic of Korea. Healthcare (Basel) 2023; 11:2365. [PMID: 37628562 PMCID: PMC10454021 DOI: 10.3390/healthcare11162365] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2023] [Revised: 08/06/2023] [Accepted: 08/09/2023] [Indexed: 08/27/2023] Open
Abstract
BACKGROUND Cardiovascular disease (CVD) is the primary cause of mortality worldwide and imposes a significant social burden on many countries. METHODS This study assessed the health and economic benefits of omega-3 associated with CVD. The meta-analysis estimated the risk ratio (RR) and absolute risk reduction (ARR), and the economic impact was calculated using direct and indirect costs related to CVD treatments in Korean adults. RESULTS A total of 33 studies were included in the meta-analysis on CVD outcomes, with 80,426 participants in the intervention group and 80,251 participants in the control group. The meta-analysis determined a significant reduction in omega-3 in CVD (RR = 0.92, 95% CI: 0.86~0.97) and ARR (1.48%). Additionally, the subgroup analysis indicated that higher doses and the long-term consumption of omega-3 could further enhance these effects. After applying ARR from meta-analysis to the target population of about 1,167,370 in 2021, the Republic of Korea, it was estimated that omega-3 consumption could result in an economic benefit of KRW 300 billion by subtracting the purchase expenses of omega-3 supplements from the total social cost savings. CONCLUSION Omega-3 supplements can help to reduce the risk of CVD and subsequent economic benefits in the Republic of Korea.
Collapse
Affiliation(s)
- Moon Seong Kim
- Department of Food and Regulatory Science, Korea University, Sejong 30019, Republic of Korea; (M.S.K.); (J.M.K.); (S.G.L.); (E.J.J.); (B.K.H.)
| | - Jin Man Kim
- Department of Food and Regulatory Science, Korea University, Sejong 30019, Republic of Korea; (M.S.K.); (J.M.K.); (S.G.L.); (E.J.J.); (B.K.H.)
| | - Sang Gyeong Lee
- Department of Food and Regulatory Science, Korea University, Sejong 30019, Republic of Korea; (M.S.K.); (J.M.K.); (S.G.L.); (E.J.J.); (B.K.H.)
| | - Eun Jin Jung
- Department of Food and Regulatory Science, Korea University, Sejong 30019, Republic of Korea; (M.S.K.); (J.M.K.); (S.G.L.); (E.J.J.); (B.K.H.)
| | - Sang Hoon Lee
- Department of Food and Biotechnology, Korea University, Sejong 30019, Republic of Korea; (S.H.L.); (W.Y.H.)
| | - Wen Yan Huang
- Department of Food and Biotechnology, Korea University, Sejong 30019, Republic of Korea; (S.H.L.); (W.Y.H.)
- BK21 FOUR Research Education Team for Omics-Based Bio-Health in Food Industry, Korea University, Sejong 30019, Republic of Korea
| | - Bok Kyung Han
- Department of Food and Regulatory Science, Korea University, Sejong 30019, Republic of Korea; (M.S.K.); (J.M.K.); (S.G.L.); (E.J.J.); (B.K.H.)
- Department of Food and Biotechnology, Korea University, Sejong 30019, Republic of Korea; (S.H.L.); (W.Y.H.)
| | - Da Eun Jung
- Department of Environmental and Resource Economics, Dankook University, Chungnam 16890, Republic of Korea; (D.E.J.); (S.B.Y.)
| | - Sung Bum Yang
- Department of Environmental and Resource Economics, Dankook University, Chungnam 16890, Republic of Korea; (D.E.J.); (S.B.Y.)
| | - Inbae Ji
- Department of Food Industrial Management, Dongguk University, Seoul 04620, Republic of Korea;
| | - Young Jun Kim
- Department of Food and Regulatory Science, Korea University, Sejong 30019, Republic of Korea; (M.S.K.); (J.M.K.); (S.G.L.); (E.J.J.); (B.K.H.)
- Department of Food and Biotechnology, Korea University, Sejong 30019, Republic of Korea; (S.H.L.); (W.Y.H.)
- BK21 FOUR Research Education Team for Omics-Based Bio-Health in Food Industry, Korea University, Sejong 30019, Republic of Korea
| | - Ji Youn Hong
- Department of Food and Regulatory Science, Korea University, Sejong 30019, Republic of Korea; (M.S.K.); (J.M.K.); (S.G.L.); (E.J.J.); (B.K.H.)
- Department of Food and Biotechnology, Korea University, Sejong 30019, Republic of Korea; (S.H.L.); (W.Y.H.)
- BK21 FOUR Research Education Team for Omics-Based Bio-Health in Food Industry, Korea University, Sejong 30019, Republic of Korea
| |
Collapse
|
6
|
Brosolo G, Da Porto A, Marcante S, Picci A, Capilupi F, Capilupi P, Bertin N, Vivarelli C, Bulfone L, Vacca A, Catena C, Sechi LA. Omega-3 Fatty Acids in Arterial Hypertension: Is There Any Good News? Int J Mol Sci 2023; 24:9520. [PMID: 37298468 PMCID: PMC10253816 DOI: 10.3390/ijms24119520] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2023] [Revised: 05/28/2023] [Accepted: 05/29/2023] [Indexed: 06/12/2023] Open
Abstract
Omega-3 polyunsaturated fatty acids (ω-3 PUFAs), including alpha-linolenic acid (ALA) and its derivatives eicosapentaenoic acid (EPA) and docosahexaenoic acid (DHA), are "essential" fatty acids mainly obtained from diet sources comprising plant oils, marine blue fish, and commercially available fish oil supplements. Many epidemiological and retrospective studies suggested that ω-3 PUFA consumption decreases the risk of cardiovascular disease, but results of early intervention trials have not consistently confirmed this effect. In recent years, some large-scale randomized controlled trials have shed new light on the potential role of ω-3 PUFAs, particularly high-dose EPA-only formulations, in cardiovascular prevention, making them an attractive tool for the treatment of "residual" cardiovascular risk. ω-3 PUFAs' beneficial effects on cardiovascular outcomes go far beyond the reduction in triglyceride levels and are thought to be mediated by their broadly documented "pleiotropic" actions, most of which are directed to vascular protection. A considerable number of clinical studies and meta-analyses suggest the beneficial effects of ω-3 PUFAs in the regulation of blood pressure in hypertensive and normotensive subjects. These effects occur mostly through regulation of the vascular tone that could be mediated by both endothelium-dependent and independent mechanisms. In this narrative review, we summarize the results of both experimental and clinical studies that evaluated the effect of ω-3 PUFAs on blood pressure, highlighting the mechanisms of their action on the vascular system and their possible impact on hypertension, hypertension-related vascular damage, and, ultimately, cardiovascular outcomes.
Collapse
Affiliation(s)
- Gabriele Brosolo
- Department of Medicine, University of Udine, 33100 Udine, Italy; (A.D.P.); (S.M.); (A.P.); (F.C.); (P.C.); (N.B.); (C.V.); (L.B.); (A.V.); (C.C.)
- European Hypertension Excellence Center, Clinica Medica, University of Udine, 33100 Udine, Italy
| | - Andrea Da Porto
- Department of Medicine, University of Udine, 33100 Udine, Italy; (A.D.P.); (S.M.); (A.P.); (F.C.); (P.C.); (N.B.); (C.V.); (L.B.); (A.V.); (C.C.)
- Diabetes and Metabolism Unit, Clinica Medica, University of Udine, 33100 Udine, Italy
| | - Stefano Marcante
- Department of Medicine, University of Udine, 33100 Udine, Italy; (A.D.P.); (S.M.); (A.P.); (F.C.); (P.C.); (N.B.); (C.V.); (L.B.); (A.V.); (C.C.)
| | - Alessandro Picci
- Department of Medicine, University of Udine, 33100 Udine, Italy; (A.D.P.); (S.M.); (A.P.); (F.C.); (P.C.); (N.B.); (C.V.); (L.B.); (A.V.); (C.C.)
| | - Filippo Capilupi
- Department of Medicine, University of Udine, 33100 Udine, Italy; (A.D.P.); (S.M.); (A.P.); (F.C.); (P.C.); (N.B.); (C.V.); (L.B.); (A.V.); (C.C.)
| | - Patrizio Capilupi
- Department of Medicine, University of Udine, 33100 Udine, Italy; (A.D.P.); (S.M.); (A.P.); (F.C.); (P.C.); (N.B.); (C.V.); (L.B.); (A.V.); (C.C.)
| | - Nicole Bertin
- Department of Medicine, University of Udine, 33100 Udine, Italy; (A.D.P.); (S.M.); (A.P.); (F.C.); (P.C.); (N.B.); (C.V.); (L.B.); (A.V.); (C.C.)
- Thrombosis and Hemostasis Unit, Clinica Medica, University of Udine, 33100 Udine, Italy
| | - Cinzia Vivarelli
- Department of Medicine, University of Udine, 33100 Udine, Italy; (A.D.P.); (S.M.); (A.P.); (F.C.); (P.C.); (N.B.); (C.V.); (L.B.); (A.V.); (C.C.)
| | - Luca Bulfone
- Department of Medicine, University of Udine, 33100 Udine, Italy; (A.D.P.); (S.M.); (A.P.); (F.C.); (P.C.); (N.B.); (C.V.); (L.B.); (A.V.); (C.C.)
- European Hypertension Excellence Center, Clinica Medica, University of Udine, 33100 Udine, Italy
| | - Antonio Vacca
- Department of Medicine, University of Udine, 33100 Udine, Italy; (A.D.P.); (S.M.); (A.P.); (F.C.); (P.C.); (N.B.); (C.V.); (L.B.); (A.V.); (C.C.)
- European Hypertension Excellence Center, Clinica Medica, University of Udine, 33100 Udine, Italy
| | - Cristiana Catena
- Department of Medicine, University of Udine, 33100 Udine, Italy; (A.D.P.); (S.M.); (A.P.); (F.C.); (P.C.); (N.B.); (C.V.); (L.B.); (A.V.); (C.C.)
- European Hypertension Excellence Center, Clinica Medica, University of Udine, 33100 Udine, Italy
| | - Leonardo A. Sechi
- Department of Medicine, University of Udine, 33100 Udine, Italy; (A.D.P.); (S.M.); (A.P.); (F.C.); (P.C.); (N.B.); (C.V.); (L.B.); (A.V.); (C.C.)
- European Hypertension Excellence Center, Clinica Medica, University of Udine, 33100 Udine, Italy
- Diabetes and Metabolism Unit, Clinica Medica, University of Udine, 33100 Udine, Italy
- Thrombosis and Hemostasis Unit, Clinica Medica, University of Udine, 33100 Udine, Italy
| |
Collapse
|
7
|
Nishizaki Y, Miyauchi K, Iwata H, Inoue T, Hirayama A, Kimura K, Ozaki Y, Murohara T, Ueshima K, Kuwabara Y, Tanaka-Mizuno S, Yanagisawa N, Sato T, Daida H. Study protocol and baseline characteristics of Randomized trial for Evaluation in Secondary Prevention Efficacy of Combination Therapy-Statin and Eicosapentaenoic Acid: RESPECT-EPA, the combination of a randomized control trial and an observational biomarker study. Am Heart J 2023; 257:1-8. [PMID: 36372250 DOI: 10.1016/j.ahj.2022.11.008] [Citation(s) in RCA: 25] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/09/2022] [Revised: 10/11/2022] [Accepted: 11/03/2022] [Indexed: 05/11/2023]
Abstract
BACKGROUND Omega-3 polyunsaturated fatty acids (PUFAs) have been a hot topic since the Japan EPA Lipid Intervention Study (JELIS), the first landmark study using a highly purified eicosapentaenoic acid (EPA), indicated that EPA could decrease the incidence of cardiovascular events. Over 20 years have passed since the JELIS was conducted, and the standard treatment for dyslipidemia has altered significantly since then. The JELIS subjects did not undertake the current risk management especially current standard statins and did not exclusively target secondary prevention patients. In addition, the subjects included are relatively high EPA population. Furthermore, the clinical implication of the plasma EPA/arachidonic acid (AA) ratio as a biomarker has not yet been validated. Therefore, the Randomized trial for Evaluation in Secondary Prevention Efficacy of Combination Therapy - Statin and EPA (RESPECT-EPA) was planned and is currently underway in Japan. METHODS The RESPECT-EPA comprises two parts: the open-label randomized controlled trial (RCT) and biomarker study (prospective cohort study design). The RCT included patients with a low EPA/AA ratio. These patients were then randomized to highly purified EPA (1800 mg/day) or control groups. The primary endpoint was cardiovascular death, non-fatal myocardial infarction, non-fatal ischemic stroke, unstable angina pectoris, and clinically indicated coronary revascularization. The biomarker study assesses the EPA/AA ratio's usefulness as a biomarker for cardiovascular events prediction. RESULTS In the RCT, a total of 2,460 patients were enrolled in 95 sites in Japan. Patients' baseline characteristics were similar between intervention and control groups in the RCT. The baseline median EPA/AA ratio was 0.243 and 0.235, respectively. A total of 1,314 patients were participated in the observational part, and the baseline median EPA/AA ratio was 0.577. CONCLUSIONS After this study is completed, we will have further evidence on whether a highly purified EPA is effective in reducing cardiovascular events for secondary prevention or not, as well as whether if EPA/AA ratio is a predictor for future cardiovascular events. This study was registered in the University Hospital Medical Information Network Clinical Trials Registry (UMIN000012069).
Collapse
Affiliation(s)
- Yuji Nishizaki
- Department of Cardiovascular Biology and Medicine, Juntendo University Graduate School of Medicine, Bunkyo, Tokyo, Japan
| | - Katsumi Miyauchi
- Department of Cardiovascular Biology and Medicine, Juntendo University Graduate School of Medicine, Bunkyo, Tokyo, Japan
| | - Hiroshi Iwata
- Department of Cardiovascular Biology and Medicine, Juntendo University Graduate School of Medicine, Bunkyo, Tokyo, Japan
| | - Teruo Inoue
- Japan Red Cross Society, Nasu Red Cross Hospital, Otawara, Tochigi, Japan; Dokkyo Medical University, Mibu, Tochigi, Japan
| | - Atsushi Hirayama
- Department of Cardiology, Osaka Police Hospital, Sumiyoshi, Osaka, Japan
| | - Kazuo Kimura
- Department of Cardiology, Yokohama City University Medical Center, Yokohama, Kanagawa, Japan
| | - Yukio Ozaki
- Department of Cardiology, Fujita Health University, Toyoake, Aichi, Japan
| | - Toyoaki Murohara
- Department of Cardiology, Nagoya University Graduate School of Medicine, Nagoya, Aichi, Japan
| | - Kenji Ueshima
- Medical Examination Center, Uji-Takeda Hospital,Uji, Kyoto, Japan
| | - Yoshihiro Kuwabara
- Cancer Control Center, Osaka International Cancer Institute,Otemae, Osaka, Japan
| | - Sachiko Tanaka-Mizuno
- Department of Digital Health and Epidemiology, Graduate School of Medicine, Kyoto University, Kyoto, Kyoto, Japan
| | - Naotake Yanagisawa
- Medical Technology Innovation Center, Juntendo University, Bunkyo, Tokyo, Japan
| | - Tosiya Sato
- Department of Biostatistics, Kyoto University School of Public Health, Kyoto, Kyoto, Japan
| | - Hiroyuki Daida
- Department of Cardiovascular Biology and Medicine, Juntendo University Graduate School of Medicine, Bunkyo, Tokyo, Japan; Faculty of Health Science, Juntendo University, Bunkyo, Tokyo, Japan.
| |
Collapse
|
8
|
Asakura K, Minami Y, Nagata T, Katamine M, Katsura A, Hashimoto T, Kinoshita D, Ako J. Impact of the eicosapentaenoic acid to arachidonic acid ratio on plaque characteristics in statin-treated patients with coronary artery disease. J Clin Lipidol 2023; 17:189-196. [PMID: 36517412 DOI: 10.1016/j.jacl.2022.11.011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2022] [Revised: 11/15/2022] [Accepted: 11/24/2022] [Indexed: 12/12/2022]
Abstract
BACKGROUND A low eicosapentaenoic acid (EPA)/arachidonic acid (AA) ratio is associated with an increased risk of cardiovascular events in patients with coronary artery disease (CAD). OBJECTIVE To clarify the impact of the EPA/AA ratio on the characteristics of non-culprit coronary plaques in statin-treated patients with CAD. METHODS A total of 370 consecutive stable coronary disease patients treated with statins, who underwent percutaneous coronary intervention for the culprit lesion and optical coherence tomography (OCT) imaging of the non-culprit plaque in a culprit vessel were included. The characteristics of non-culprit plaques assessed using OCT were compared between the lower EPA/AA group (EPA/AA <0.4, n = 255) and the higher EPA/AA group (EPA/AA ≥0.4, n = 115). RESULTS The prevalence of lipid-rich plaque (58.8 vs. 41.7%, p = 0.003) and plaque with macrophages (56.5 vs. 31.3%, p <0.001) was significantly higher in the lower EPA/AA group than in the higher EPA/AA group. This association was observed even if the LDL-C level was <100 mg/dL. The prevalence of thin-cap fibroatheroma was significantly higher in patients with lower EPA/AA and higher LDL-C (≥100 mg/dL) than in those with higher EPA/AA and lower LDL-C (<100 mg/dL) (odds ratio: 2.750, 95% confidence interval: 1.182-6.988, p = 0.024). An EPA/AA <0.4 was independently associated with a higher prevalence of lipid-rich plaque, plaque with macrophages, and cholesterol crystals. CONCLUSION Lower EPA/AA ratio was associated with higher prevalence of vulnerable characteristics in non-culprit plaques. The present results suggest the importance of EPA/AA ratio on the secondary prevention of CAD.
Collapse
Affiliation(s)
- Kiyoshi Asakura
- Department of Cardiovascular Medicine, Kitasato University School of Medicine, Sagamihara, Japan
| | - Yoshiyasu Minami
- Department of Cardiovascular Medicine, Kitasato University School of Medicine, Sagamihara, Japan.
| | - Takako Nagata
- Department of Cardiovascular Medicine, Kitasato University School of Medicine, Sagamihara, Japan
| | - Masahiro Katamine
- Department of Cardiovascular Medicine, Kitasato University School of Medicine, Sagamihara, Japan
| | - Aritomo Katsura
- Department of Cardiovascular Medicine, Kitasato University School of Medicine, Sagamihara, Japan
| | - Takuya Hashimoto
- Department of Cardiovascular Medicine, Kitasato University School of Medicine, Sagamihara, Japan
| | - Daisuke Kinoshita
- Department of Cardiovascular Medicine, Kitasato University School of Medicine, Sagamihara, Japan
| | - Junya Ako
- Department of Cardiovascular Medicine, Kitasato University School of Medicine, Sagamihara, Japan
| |
Collapse
|
9
|
Alvarez Campano CG, Macleod MJ, Aucott L, Thies F. Marine-derived n-3 fatty acids therapy for stroke. Cochrane Database Syst Rev 2022; 6:CD012815. [PMID: 35766825 PMCID: PMC9241930 DOI: 10.1002/14651858.cd012815.pub3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
Abstract
BACKGROUND Currently, with stroke burden increasing, there is a need to explore therapeutic options that ameliorate the acute insult. There is substantial evidence of a neuroprotective effect of marine-derived n-3 polyunsaturated fatty acids (PUFAs) in animal models of stroke, leading to a better functional outcome. OBJECTIVES To assess the effects of administration of marine-derived n-3 PUFAs on functional outcomes and dependence in people with stroke. SEARCH METHODS We searched the Cochrane Stroke Trials Register (last searched 31 May 2021), the Cochrane Central Register of Controlled Trials (CENTRAL; 2021, Issue 5), MEDLINE Ovid (from 1948 to 31 May 2021), Embase Ovid (from 1980 to 31 May 2021), CINAHL EBSCO (Cumulative Index to Nursing and Allied Health Literature; from 1982 to 31 May 2021), Science Citation Index Expanded ‒ Web of Science (SCI-EXPANDED), Conference Proceedings Citation Index-Science - Web of Science (CPCI-S), and BIOSIS Citation Index. We also searched ongoing trial registers, reference lists, relevant systematic reviews, and used the Science Citation Index Reference Search. SELECTION CRITERIA We included randomised controlled trials (RCTs) comparing marine-derived n-3 PUFAs to placebo or open control (no placebo) in people with a history of stroke or transient ischaemic attack (TIA), or both. DATA COLLECTION AND ANALYSIS At least two review authors independently selected trials for inclusion, extracted data, assessed risk of bias, and used the GRADE approach to assess the certainty of the body of evidence. We contacted study authors for clarification and additional information on stroke/TIA participants. We conducted random-effects meta-analysis or narrative synthesis, as appropriate. The primary outcome was efficacy (functional outcome) assessed using a validated scale, for example, the Glasgow Outcome Scale Extended (GOSE) dichotomised into poor or good clinical outcome, the Barthel Index (higher score is better; scale from 0 to 100), or the Rivermead Mobility Index (higher score is better; scale from 0 to 15). Our secondary outcomes were vascular-related death, recurrent events, incidence of other type of stroke, adverse events, quality of life, and mood. MAIN RESULTS We included 30 RCTs; nine of them provided outcome data (3339 participants). Only one study included participants in the acute phase of stroke (haemorrhagic). Doses of marine-derived n-3 PUFAs ranged from 400 mg/day to 3300 mg/day. Risk of bias was generally low or unclear in most trials, with a higher risk of bias in smaller studies. We assessed results separately for short (up to three months) and longer (more than three months) follow-up studies. Short follow-up (up to three months) Functional outcome was reported in only one pilot study as poor clinical outcome assessed with the GOSE (risk ratio (RR) 0.78, 95% confidence interval (CI) 0.36 to 1.68, P = 0.52; 40 participants; very low-certainty evidence). Mood (assessed with the GHQ-30, lower score better) was reported by only one study and favoured control (mean difference (MD) 1.41, 95% CI 0.07 to 2.75, P = 0.04; 102 participants; low-certainty evidence). We found no evidence of an effect of the intervention for the remainder of the secondary outcomes: vascular-related death (two studies, not pooled due to differences in population, RR 0.33, 95% CI 0.01 to 8.00, P = 0.50, and RR 0.33, 95% CI 0.01 to 7.72, P = 0.49; 142 participants; low-certainty evidence); recurrent events (RR 0.41, 95% CI 0.02 to 8.84, P = 0.57; 18 participants; very low-certainty evidence); incidence of other type of stroke (two studies, not pooled due to different type of index stroke, RR 6.11, 95% CI 0.33 to 111.71, P = 0.22, and RR 0.63, 95% CI 0.25 to 1.58, P = 0.32; 58 participants; very low-certainty evidence); and quality of life (physical component, MD -2.31, 95% CI -4.81 to 0.19, P = 0.07, and mental component, MD -2.16, 95% CI -5.91 to 1.59, P = 0.26; 1 study; 102 participants; low-certainty evidence). Adverse events were reported by two studies (57 participants; very low-certainty evidence), one trial reporting extracranial haemorrhage (RR 0.25, 95% CI 0.04 to 1.73, P = 0.16) and the other one reporting bleeding complications (RR 0.32, 95% CI 0.01 to 7.35, P = 0.47). Longer follow-up (more than three months) One small trial assessed functional outcome with both the Barthel Index for activities of daily living (MD 7.09, 95% CI -5.16 to 19.34, P = 0.26), and the Rivermead Mobility Index for mobility (MD 1.30, 95% CI -1.31 to 3.91, P = 0.33) (52 participants; very low-certainty evidence). We carried out meta-analysis for vascular-related death (RR 1.02, 95% CI 0.78 to 1.35, P = 0.86; 5 studies; 2237 participants; low-certainty evidence) and fatal recurrent events (RR 0.69, 95% CI 0.31 to 1.55, P = 0.37; 3 studies; 1819 participants; low-certainty evidence). We found no evidence of an effect of the intervention for mood (MD 1.00, 95% CI -2.07 to 4.07, P = 0.61; 1 study; 14 participants; low-certainty evidence). Incidence of other type of stroke and quality of life were not reported. Adverse events (all combined) were reported by only one study (RR 0.94, 95% CI 0.56 to 1.58, P = 0.82; 1455 participants; low-certainty evidence). AUTHORS' CONCLUSIONS We are very uncertain of the effect of marine-derived n-3 PUFAs therapy on functional outcomes and dependence after stroke as there is insufficient high-certainty evidence. More well-designed RCTs are needed, specifically in acute stroke, to determine the efficacy and safety of the intervention. Studies assessing functional outcome might consider starting the intervention as early as possible after the event, as well as using standardised, clinically relevant measures for functional outcomes, such as the modified Rankin Scale. Optimal doses remain to be determined; delivery forms (type of lipid carriers) and mode of administration (ingestion or injection) also need further consideration.
Collapse
Affiliation(s)
| | | | - Lorna Aucott
- Health Services Research Unit, University of Aberdeen, Aberdeen, UK
| | - Frank Thies
- The Rowett Institute, University of Aberdeen, Aberdeen, UK
| |
Collapse
|
10
|
Matsumoto I, Moriya S, Kurozumi M, Namba T, Takagi Y. Simultaneous evaluation of fatty acid and triglycerides after percutaneous coronary intervention. J Cardiol 2022; 80:149-154. [DOI: 10.1016/j.jjcc.2022.02.011] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/02/2021] [Revised: 01/13/2022] [Accepted: 02/16/2022] [Indexed: 12/13/2022]
|
11
|
Nakamura M, Yaku H, Ako J, Arai H, Asai T, Chikamori T, Daida H, Doi K, Fukui T, Ito T, Kadota K, Kobayashi J, Komiya T, Kozuma K, Nakagawa Y, Nakao K, Niinami H, Ohno T, Ozaki Y, Sata M, Takanashi S, Takemura H, Ueno T, Yasuda S, Yokoyama H, Fujita T, Kasai T, Kohsaka S, Kubo T, Manabe S, Matsumoto N, Miyagawa S, Mizuno T, Motomura N, Numata S, Nakajima H, Oda H, Otake H, Otsuka F, Sasaki KI, Shimada K, Shimokawa T, Shinke T, Suzuki T, Takahashi M, Tanaka N, Tsuneyoshi H, Tojo T, Une D, Wakasa S, Yamaguchi K, Akasaka T, Hirayama A, Kimura K, Kimura T, Matsui Y, Miyazaki S, Okamura Y, Ono M, Shiomi H, Tanemoto K. JCS 2018 Guideline on Revascularization of Stable Coronary Artery Disease. Circ J 2022; 86:477-588. [DOI: 10.1253/circj.cj-20-1282] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Affiliation(s)
- Masato Nakamura
- Division of Cardiovascular Medicine, Toho University Ohashi Medical Center
| | - Hitoshi Yaku
- Department of Cardiovascular Surgery, Kyoto Prefectural University of Medicine
| | - Junya Ako
- Department of Cardiovascular Medicine, Kitasato University Graduate School of Medical Sciences
| | - Hirokuni Arai
- Department of Cardiovascular Surgery, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University
| | - Tohru Asai
- Department of Cardiovascular Surgery, Juntendo University Graduate School of Medicine
| | | | - Hiroyuki Daida
- Department of Cardiovascular Medicine, Juntendo University Graduate School of Medicine
| | - Kiyoshi Doi
- General and Cardiothoracic Surgery, Gifu University Graduate School of Medicine
| | - Toshihiro Fukui
- Department of Cardiovascular Surgery, Graduate School of Medical Sciences, Kumamoto University
| | - Toshiaki Ito
- Department of Cardiovascular Surgery, Japanese Red Cross Nagoya Daiichi Hospital
| | | | - Junjiro Kobayashi
- Department of Cardiovascular Surgery, National Cerebral and Cardiovascular Center
| | - Tatsuhiko Komiya
- Department of Cardiovascular Surgery, Kurashiki Central Hospital
| | - Ken Kozuma
- Department of Internal Medicine, Teikyo University Faculty of Medicine
| | - Yoshihisa Nakagawa
- Department of Cardiovascular Medicine, Shiga University of Medical Science
| | - Koichi Nakao
- Division of Cardiology, Saiseikai Kumamoto Hospital Cardiovascular Center
| | - Hiroshi Niinami
- Department of Cardiovascular Surgery, Tokyo Women’s Medical University
| | - Takayuki Ohno
- Department of Cardiovascular Surgery, Mitsui Memorial Hospital
| | - Yukio Ozaki
- Department of Cardiology, Fujita Health University Hospital
| | - Masataka Sata
- Department of Cardiovascular Medicine, Tokushima University Graduate School of Biomedical Sciences
| | | | - Hirofumi Takemura
- Department of Cardiovascular Surgery, Graduate School of Medical Sciences, Kanazawa University
| | | | - Satoshi Yasuda
- Department of Cardiovascular Medicine, National Cerebral and Cardiovascular Center
| | - Hitoshi Yokoyama
- Department of Cardiovascular Surgery, Fukushima Medical University
| | - Tomoyuki Fujita
- Department of Cardiovascular Surgery, National Cerebral and Cardiovascular Center
| | - Tokuo Kasai
- Department of Cardiology, Uonuma Institute of Community Medicine, Niigata University Uonuma Kikan Hospital
| | - Shun Kohsaka
- Department of Cardiology, Keio University School of Medicine
| | - Takashi Kubo
- Department of Cardiovascular Medicine, Wakayama Medical University
| | - Susumu Manabe
- Department of Cardiovascular Surgery, Tsuchiura Kyodo General Hospital
| | | | - Shigeru Miyagawa
- Frontier of Regenerative Medicine, Graduate School of Medicine, Osaka University
| | - Tomohiro Mizuno
- Department of Cardiovascular Surgery, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University
| | - Noboru Motomura
- Department of Cardiovascular Surgery, Graduate School of Medicine, Toho University
| | - Satoshi Numata
- Department of Cardiovascular Surgery, Kyoto Prefectural University of Medicine
| | - Hiroyuki Nakajima
- Department of Cardiovascular Surgery, Saitama Medical University International Medical Center
| | - Hirotaka Oda
- Department of Cardiology, Niigata City General Hospital
| | - Hiromasa Otake
- Department of Cardiovascular Medicine, Kobe University Graduate School of Medicine
| | - Fumiyuki Otsuka
- Department of Cardiovascular Medicine, National Cerebral and Cardiovascular Center
| | - Ken-ichiro Sasaki
- Division of Cardiovascular Medicine, Kurume University School of Medicine
| | - Kazunori Shimada
- Department of Cardiovascular Medicine, Juntendo University Graduate School of Medicine
| | - Tomoki Shimokawa
- Department of Cardiovascular Surgery, Sakakibara Heart Institute
| | - Toshiro Shinke
- Division of Cardiology, Department of Medicine, Showa University School of Medicine
| | - Tomoaki Suzuki
- Department of Cardiovascular Surgery, Shiga University of Medical Science
| | - Masao Takahashi
- Department of Cardiovascular Surgery, Hiratsuka Kyosai Hospital
| | - Nobuhiro Tanaka
- Department of Cardiology, Tokyo Medical University Hachioji Medical Center
| | | | - Taiki Tojo
- Department of Cardiovascular Medicine, Kitasato University Graduate School of Medical Sciences
| | - Dai Une
- Department of Cardiovascular Surgery, Okayama Medical Center
| | - Satoru Wakasa
- Department of Cardiovascular and Thoracic Surgery, Hokkaido University Graduate School of Medicine
| | - Koji Yamaguchi
- Department of Cardiovascular Medicine, Tokushima University Graduate School of Biomedical Sciences
| | - Takashi Akasaka
- Department of Cardiovascular Medicine, Wakayama Medical University
| | | | - Kazuo Kimura
- Cardiovascular Center, Yokohama City University Medical Center
| | - Takeshi Kimura
- Department of Cardiovascular Medicine, Graduate School of Medicine, Kyoto University
| | - Yoshiro Matsui
- Department of Cardiovascular and Thoracic Surgery, Graduate School of Medicine, Hokkaido University
| | - Shunichi Miyazaki
- Division of Cardiology, Department of Internal Medicine, Faculty of Medicine, Kindai University
| | | | - Minoru Ono
- Department of Cardiac Surgery, Graduate School of Medicine, The University of Tokyo
| | - Hiroki Shiomi
- Department of Cardiovascular Medicine, Graduate School of Medicine, Kyoto University
| | - Kazuo Tanemoto
- Department of Cardiovascular Surgery, Kawasaki Medical School
| | | |
Collapse
|
12
|
Yamamoto T, Sano M. Deranged Myocardial Fatty Acid Metabolism in Heart Failure. Int J Mol Sci 2022; 23:996. [PMID: 35055179 PMCID: PMC8779056 DOI: 10.3390/ijms23020996] [Citation(s) in RCA: 34] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2021] [Revised: 01/12/2022] [Accepted: 01/14/2022] [Indexed: 01/27/2023] Open
Abstract
The heart requires fatty acids to maintain its activity. Various mechanisms regulate myocardial fatty acid metabolism, such as energy production using fatty acids as fuel, for which it is known that coordinated control of fatty acid uptake, β-oxidation, and mitochondrial oxidative phosphorylation steps are important for efficient adenosine triphosphate (ATP) production without unwanted side effects. The fatty acids taken up by cardiomyocytes are not only used as substrates for energy production but also for the synthesis of triglycerides and the replacement reaction of fatty acid chains in cell membrane phospholipids. Alterations in fatty acid metabolism affect the structure and function of the heart. Recently, breakthrough studies have focused on the key transcription factors that regulate fatty acid metabolism in cardiomyocytes and the signaling systems that modify their functions. In this article, we reviewed the latest research on the role of fatty acid metabolism in the pathogenesis of heart failure and provide an outlook on future challenges.
Collapse
Affiliation(s)
| | - Motoaki Sano
- Department of Cardiology, Keio University School of Medicine, Tokyo 160-8582, Japan;
| |
Collapse
|
13
|
Wang C, Enssle J, Pietzner A, Schmöcker C, Weiland L, Ritter O, Jaensch M, Elbelt U, Pagonas N, Weylandt KH. Essential Polyunsaturated Fatty Acids in Blood from Patients with and without Catheter-Proven Coronary Artery Disease. Int J Mol Sci 2022; 23:ijms23020766. [PMID: 35054948 PMCID: PMC8775772 DOI: 10.3390/ijms23020766] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2021] [Revised: 12/30/2021] [Accepted: 01/07/2022] [Indexed: 01/21/2023] Open
Abstract
Coronary artery disease (CAD) is the leading cause of death worldwide. Statins reduce morbidity and mortality of CAD. Intake of n-3 polyunsaturated fatty acid (n-3 PUFAs), particularly eicosapentaenoic acid (EPA), is associated with reduced morbidity and mortality in patients with CAD. Previous data indicate that a higher conversion of precursor fatty acids (FAs) to arachidonic acid (AA) is associated with increased CAD prevalence. Our study explored the FA composition in blood to assess n-3 PUFA levels from patients with and without CAD. We analyzed blood samples from 273 patients undergoing cardiac catheterization. Patients were stratified according to clinically relevant CAD (n = 192) and those without (n = 81). FA analysis in full blood was performed by gas chromatography. Indicating increased formation of AA from precursors, the ratio of dihomo-gamma-linolenic acid (DGLA) to AA, the delta-5 desaturase index (D5D index) was higher in CAD patients. CAD patients had significantly lower levels of omega-6 polyunsaturated FAs (n-6 PUFA) and n-3 PUFA, particularly EPA, in the blood. Thus, our study supports a role of increased EPA levels for cardioprotection.
Collapse
Affiliation(s)
- Chaoxuan Wang
- Division of Medicine, Department of Gastroenterology, Metabolism and Oncology, University Hospital Ruppin-Brandenburg, Brandenburg Medical School, 16816 Neuruppin, Germany; (C.W.); (J.E.); (A.P.); (C.S.); (U.E.)
- Medical Department, Division of Psychosomatic Medicine, Campus Benjamin Franklin, Charité—Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, 12203 Berlin, Germany
| | - Jörg Enssle
- Division of Medicine, Department of Gastroenterology, Metabolism and Oncology, University Hospital Ruppin-Brandenburg, Brandenburg Medical School, 16816 Neuruppin, Germany; (C.W.); (J.E.); (A.P.); (C.S.); (U.E.)
- Faculty of Health Sciences, Joint Faculty of the Brandenburg University of Technology, Brandenburg Medical School and University of Potsdam, 14469 Potsdam, Germany
| | - Anne Pietzner
- Division of Medicine, Department of Gastroenterology, Metabolism and Oncology, University Hospital Ruppin-Brandenburg, Brandenburg Medical School, 16816 Neuruppin, Germany; (C.W.); (J.E.); (A.P.); (C.S.); (U.E.)
| | - Christoph Schmöcker
- Division of Medicine, Department of Gastroenterology, Metabolism and Oncology, University Hospital Ruppin-Brandenburg, Brandenburg Medical School, 16816 Neuruppin, Germany; (C.W.); (J.E.); (A.P.); (C.S.); (U.E.)
| | - Linda Weiland
- Division of Medicine, Department of Cardiology, University Hospital Brandenburg an der Havel, Brandenburg Medical School, 14770 Brandenburg an der Havel, Germany; (L.W.); (O.R.); (M.J.); (N.P.)
| | - Oliver Ritter
- Division of Medicine, Department of Cardiology, University Hospital Brandenburg an der Havel, Brandenburg Medical School, 14770 Brandenburg an der Havel, Germany; (L.W.); (O.R.); (M.J.); (N.P.)
| | - Monique Jaensch
- Division of Medicine, Department of Cardiology, University Hospital Brandenburg an der Havel, Brandenburg Medical School, 14770 Brandenburg an der Havel, Germany; (L.W.); (O.R.); (M.J.); (N.P.)
| | - Ulf Elbelt
- Division of Medicine, Department of Gastroenterology, Metabolism and Oncology, University Hospital Ruppin-Brandenburg, Brandenburg Medical School, 16816 Neuruppin, Germany; (C.W.); (J.E.); (A.P.); (C.S.); (U.E.)
- Medical Department, Division of Psychosomatic Medicine, Campus Benjamin Franklin, Charité—Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, 12203 Berlin, Germany
| | - Nikolaos Pagonas
- Division of Medicine, Department of Cardiology, University Hospital Brandenburg an der Havel, Brandenburg Medical School, 14770 Brandenburg an der Havel, Germany; (L.W.); (O.R.); (M.J.); (N.P.)
| | - Karsten H. Weylandt
- Division of Medicine, Department of Gastroenterology, Metabolism and Oncology, University Hospital Ruppin-Brandenburg, Brandenburg Medical School, 16816 Neuruppin, Germany; (C.W.); (J.E.); (A.P.); (C.S.); (U.E.)
- Faculty of Health Sciences, Joint Faculty of the Brandenburg University of Technology, Brandenburg Medical School and University of Potsdam, 14469 Potsdam, Germany
- Correspondence: ; Tel.: +49-(0)3391-39-3210
| |
Collapse
|
14
|
Yousaf M, Razmovski-Naumovski V, Zubair M, Chang D, Zhou X. Synergistic Effects of Natural Product Combinations in Protecting the Endothelium Against Cardiovascular Risk Factors. J Evid Based Integr Med 2022; 27:2515690X221113327. [PMID: 35849068 PMCID: PMC9297466 DOI: 10.1177/2515690x221113327] [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] [Indexed: 11/22/2022] Open
Abstract
Endothelial dysfunction is an early hallmark of cardiovascular diseases (CVDs). Monotherapies are limited due to the complex, multifactorial pathways. The multi-component and multi-targeted approach of natural products have the potential to manage CVDs. This review aims to provide a comprehensive insight into the synergistic mechanism of natural product combinations in protecting the endothelium against various cardiovascular risk factors. Databases (PubMed, MEDLINE and EMBASE) and Google Scholar were searched, and studies in English published between January 2000 and February 2022 were collated. Clinical and pre-clinical studies of natural product combinations with or without pharmaceutical medicines, compared with monotherapy and/or proposing the underlying mechanism in protecting endothelial function, were included. Four clinical studies demonstrated that natural product combinations or natural product-pharmaceutical combinations improved endothelial function. This was associated with multi-targeted effects or improved absorption of the active substances in the body. Seventeen preclinical studies showed that natural product combinations produced synergistic (demonstrated by combination index or Bliss independence model) or enhanced effects in protecting the endothelium against hyperlipidemia, hypertension, diabetes mellitus, platelet activation, oxidative stress and hyperhomocysteinemia. The molecular targets included reactive oxygen species, Nrf2-HO-1, p38MAPK, P13K/Akt and NF-κB. Thus, the current available evidence of natural product combinations in targeting endothelial dysfunction is predominantly from preclinical studies. These have demonstrated synergistic/enhanced pharmacological activities and proposed associated mechanisms. However, evidence from larger, well-designed clinical trials remains weak. More cohesion is required between preclinical and clinical data to support natural product combinations in preventing or slowing the progression of CVDs.
Collapse
Affiliation(s)
- Muhammad Yousaf
- Department of Bioinformatics and Biotechnology, 72594Government College University, Faisalabad, Pakistan
| | - Valentina Razmovski-Naumovski
- NICM Health Research Institute, 6489Western Sydney University, Westmead, Australia.,South Western Sydney Clinical School, School of Medicine & Health, 7800University of New South Wales (UNSW), Sydney, Australia
| | - Muhammad Zubair
- Department of Bioinformatics and Biotechnology, 72594Government College University, Faisalabad, Pakistan
| | - Dennis Chang
- NICM Health Research Institute, 6489Western Sydney University, Westmead, Australia
| | - Xian Zhou
- NICM Health Research Institute, 6489Western Sydney University, Westmead, Australia
| |
Collapse
|
15
|
Tomlinson B, Patil NG, Fok M, Lam CWK. Managing dyslipidemia in patients with Type 2 diabetes. Expert Opin Pharmacother 2021; 22:2221-2234. [PMID: 33823719 DOI: 10.1080/14656566.2021.1912734] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
INTRODUCTION Type 2 diabetes mellitus (T2DM) is associated with increased risk for atherosclerotic cardiovascular disease (ASCVD) which is partly related to atherogenic dyslipidemia with raised triglycerides, reduced high-density lipoprotein cholesterol levels, and accompanying lipid changes. Treatment of this dyslipidemia is regarded as a priority to reduce the ASCVD risk in T2DM. AREAS COVERED This article reviews the relevant studies and guidelines from the publications related to this area. EXPERT OPINION Lifestyle modification should always be encouraged, and statin treatment is indicated in most patients with T2DM based on the outcome of randomized controlled trials. If LDL-C goals are not achieved, first, ezetimibe and subsequently proprotein convertase subtilisin-kexin type 9 (PCSK9) inhibitors should be added. Patients with T2DM derive greater benefits from ezetimibe and PCSK9 inhibitors due to their higher absolute ASCVD risk compared to patients without T2DM. If triglyceride levels remain elevated, a high dose of eicosapentaenoic acid ethyl ester should be added. Fibrates should be used for severe hypertriglyceridemia to prevent acute pancreatitis. Novel treatments including pemafibrate and inclisiran are undergoing cardiovascular outcome trials, and RNA-based therapies may help to target residual hypertriglyceridemia and high lipoprotein(a) with the long acting treatments offering potential improved adherence to therapy.
Collapse
Affiliation(s)
- Brian Tomlinson
- Faculty of Medicine, Macau University of Science and Technology, Macau China
| | | | - Manson Fok
- Faculty of Medicine, Macau University of Science and Technology, Macau China
| | | |
Collapse
|
16
|
Yasuzawa T, Nakamura T, Ueshima S, Mima A. Protective Effects of Eicosapentaenoic Acid on the Glomerular Endothelium via Inhibition of EndMT in Diabetes. J Diabetes Res 2021; 2021:2182225. [PMID: 34977254 PMCID: PMC8720008 DOI: 10.1155/2021/2182225] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/31/2021] [Revised: 11/29/2021] [Accepted: 11/30/2021] [Indexed: 12/11/2022] Open
Abstract
Diabetes-induced endothelial pathologies are hypothesized to lead to the progression of diabetic kidney disease (DKD). The endothelial to mesenchymal transition (EndMT) possibly induces fibrosis, leading to glomerulosclerosis in the kidney. Furthermore, this could lead to albuminuria in diabetic nephropathy due to glomerular endothelial dysfunction. Eicosapentaenoic acid (EPA), purified from fish oil, decreases inflammatory cytokine levels in glomerulonephritis. Here, we aimed at finding whether ethyl eicosapentaenoate (EPA-E) exerts renal protective effects via EndMT inhibition. To find out whether EPA inhibits EndMT in vitro, the changes in CD31 expression were studied in cultured mouse endothelial cells. The addition of the conditioned medium from the adipocyte culture significantly decreased the protein levels of CD31, while the addition of EPA-E partially reversed this inhibition. Further, EndMT inhibition by EPA-E treatment might occur via the inhibition of the protein kinase Cβ (PKCβ)/transforming growth factor-β (TGF-β)/plasminogen activator inhibitor-1 (PAI-1) signaling and not via microRNAs. Streptozotocin-induced diabetic mice fed a high-fat diet (60% from fat) exhibited mesangial expansion and albuminuria. Induction of EPA-E ameliorated the mesangial expansion and decreased albuminuria without affecting blood pressure, triglyceride and free fatty acid levels, and intraperitoneal glucose. These findings suggest that EPA-E exerts renal protective effects on endothelial cells, by normalizing EndMT followed by the PKCβ/TGF-β/PAI-1 signaling. Thus, EPA-E has the potential for imparting renal protection by regulating EndMT in DKD.
Collapse
Affiliation(s)
- Toshinori Yasuzawa
- Department of Nephrology, Osaka Medical and Pharmaceutical University, Osaka, Japan
- Department of Food Science and Nutrition, Faculty of Agriculture, Kindai University, Nara, Japan
- Department of Health and Nutrition, Faculty of Health Science, Kio University, Nara, Japan
| | - Tomomi Nakamura
- Department of Food Science and Nutrition, Faculty of Agriculture, Kindai University, Nara, Japan
| | - Shigeru Ueshima
- Department of Food Science and Nutrition, Faculty of Agriculture, Kindai University, Nara, Japan
| | - Akira Mima
- Department of Nephrology, Osaka Medical and Pharmaceutical University, Osaka, Japan
| |
Collapse
|
17
|
Yoosefee S, Rahimi M, Razavinia FS, Vahedian M, Bagheri AR. Serum Eicosapentaenoic and Arachidonic Acids Concentration and Cognitive Function in Patients With Coronary Artery Disease. Basic Clin Neurosci 2021; 12:63-68. [PMID: 33995928 PMCID: PMC8114866 DOI: 10.32598/bcn.12.1.606.3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2020] [Revised: 05/22/2020] [Accepted: 07/19/2020] [Indexed: 11/20/2022] Open
Abstract
Introduction: Recent studies have shown that Polyunsaturated Fatty Acids (PUFAs), including Eicosapentaenoic Acid (EPA), and Arachidonic Acid (AA), are associated with cognitive functions in patients with Coronary Artery Disease (CAD). Nevertheless, controversial results have been reported, too. The current study aimed to clarify the association of serum EPA and AA levels with cognitive decline in an Iranian sample with CAD. Methods: We evaluated cognitive function with the Mini-Mental State Examination (MMSE) and Montreal Cognitive Assessment (MoCA), in relation to serum levels of EPA and AA, in 179 CAD patients. The associations between the MMSE and MoCA scores and the other demographic parameters were evaluated. Results: Patients with CAD generally had mild cognitive impairment. But we could not find any significant correlation between PUFAs and cognitive function. However, BMI was associated with EPA/MoCA; age was associated with MMSE/MoCA and BMI. Finally, the correlation between sex and MMSE/MoCA was significant. Conclusion: Subjects generally had mild cognitive impairment, but we could not find any significant correlation between serum EPA and AA levels with cognitive function.
Collapse
Affiliation(s)
- Sadegh Yoosefee
- Neurosciences Research Center, Qom University of Medical Sciences, Qom, Iran.,Spiritual Health Research Center, Qom University of Medical Sciences, Qom, Iran
| | - Maryam Rahimi
- Neurosciences Research Center, Qom University of Medical Sciences, Qom, Iran
| | | | - Mostafa Vahedian
- Research Center for Environmental Pollutants, Qom University of Medical Sciences, Qom, Iran
| | - Ahmad Reza Bagheri
- Department of Cardiology, Faculty of Medicine, Qom University of Medical Sciences, Qom, Iran
| |
Collapse
|
18
|
Watanabe Y, Tatsuno I. Omega-3 polyunsaturated fatty acids focusing on eicosapentaenoic acid and docosahexaenoic acid in the prevention of cardiovascular diseases: a review of the state-of-the-art. Expert Rev Clin Pharmacol 2020; 14:79-93. [PMID: 33306922 DOI: 10.1080/17512433.2021.1863784] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
INTRODUCTION : An epidemiological study of Greenlandic Inuit suggested the importance of omega-3 polyunsaturated fatty acids (PUFAs) in preventing ischemic heart disease. After this landmark study, large-scale epidemiological studies have examined the benefits of omega-3 PUFAs in the prevention of cardiovascular diseases. AREAS COVERED : This article reviews studies on omega-3 PUFAs, and identifies issues relevant to cardiovascular risk. EXPERT OPINION : Recent studies have focused on the anti-inflammatory effects of omega-3 PUFAs and specialized pro-resolving mediators. High-purity eicosapentaenoic acid (EPA) ethyl ester and EPA/docosahexaenoic acid (DHA) preparations have been developed primarily for the treatment of hypertriglyceridemia. Various trials on the cardiovascular protective effects of omega-3 PUFAs have been reported, but the results have not been consistent. Some issues of the trials have been suggested, such as using low-dose omega-3 PUFAs and not including hypertriglyceridemia in subject selection criteria. REDUCE-IT study that used a high dose of high-purity EPA preparation showed a relative reduction in cardiovascular events, but, the STRENGTH study that used a high dose of EPA/DHA preparation did not support this benefit. This article reviews the roles of omega-3 PUFAs in cardiovascular diseases, including progress in understanding the molecular mechanisms and recent large-scale clinical trials.
Collapse
Affiliation(s)
- Yasuhiro Watanabe
- Center for Diabetes, Metabolism and Endocrinology, Toho University Sakura Medical Center , Chiba, Japan
| | - Ichiro Tatsuno
- Center for Diabetes, Metabolism and Endocrinology, Toho University Sakura Medical Center , Chiba, Japan
| |
Collapse
|
19
|
Abstract
PURPOSE OF REVIEW Increasing interest has focused on the potential cardioprotective effects of the omega-3 fatty acids, eicosapentaenoic acid (EPA) and docosahexaenoic acid (DHA) on the basis of findings from epidemiology and cohort studies. This review will summarize the findings of contemporary clinical trials of omega-3 fatty acids. RECENT FINDINGS Although a large clinical trial performed prior to the widespread use of statins demonstrated cardiovascular benefit with fish oils, subsequent studies have failed to reproduce this result. More recent studies have demonstrated a reduction in cardiovascular risk with administration of high-dose EPA, but not a carboxylic acid formulation containing both EPA and DHA or with lower doses of omega-3 fatty acids. SUMMARY Administration of omega-3 fatty acids differing in either composition or dose produce variable effects on cardiovascular outcomes. This has implications for both the public health and pharmacological approach to cardiovascular prevention.
Collapse
Affiliation(s)
- Stephen J Nicholls
- Monash Cardiovascular Research Centre, Victorian Heart Institute, Monash University, Melbourne, Victoria, Australia
| | - Adam J Nelson
- Duke Clinical Research Institute, Durham, North Carolina, USA
| |
Collapse
|
20
|
Zambon A, Pirillo A, Zambon S, Norata GD, Catapano AL. Omega n-3 Supplementation: Exploring the Cardiovascular Benefits Beyond Lipoprotein Reduction. Curr Atheroscler Rep 2020; 22:74. [PMID: 33009961 DOI: 10.1007/s11883-020-00893-1] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/16/2020] [Indexed: 12/18/2022]
Abstract
PURPOSE OF REVIEW Hypertriglyceridaemia is a highly prevalent disorder worldwide. Genetic and Mendelian randomization studies have suggested that triglyceride (TG)-rich lipoproteins are causal risk factors for coronary heart disease and contribute to the residual cardiovascular risk observed in patients optimally treated with statins. However, clinical trials failed to show cardiovascular benefits of TG-lowering; in this context, trials with omega-3 fatty acids provided contrasting results. Few trials have tested the supplementation of EPA alone rather than the combination of EPA + DHA. The JELIS study showed that EPA 1.8 g/day significantly reduced CV events in hypercholesterolaemic patients given statins, an effect that was independent on lipid reduction. RECENT FINDINGS The REDUCE-IT trial showed that high-dose (4 g/day) EPA significantly reduces the incidence of major cardiovascular events compared with placebo in patients with elevated TG levels. The clinical benefit was higher than expected by the reduction of TG-rich lipoprotein levels. Recent data support the efficacy of high-dose EPA supplementation on a background of optimal LDL-C-lowering therapy as a key approach to achieve a further and significant reduction of CV events in very-high CV risk patients with persistent hypertriglyceridaemia. The effect on lipids does not appear to fully explain the CV benefit, and additional mechanisms of action of EPA likely contribute to the cardiovascular protection, including the reduction of inflammation and platelet aggregation. Current guidelines recommend using high-dose EPA in combination with a statin in high/very-high CV risk patients with mild-to-moderate elevation of plasma TG to reduce the residual CV risk.
Collapse
Affiliation(s)
- Alberto Zambon
- Department of Medicine - DIMED, University of Padua, Via Giustiniani, 2, 35128, Padua, Italy.
- IRCCS MultiMedica, Sesto S. Giovanni, Milan, Italy.
| | - Angela Pirillo
- IRCCS MultiMedica, Sesto S. Giovanni, Milan, Italy
- Center for the Study of Atherosclerosis, E. Bassini Hospital, Cinisello Balsamo, Milan, Italy
| | - Sabina Zambon
- Department of Medicine - DIMED, University of Padua, Via Giustiniani, 2, 35128, Padua, Italy
| | - Giuseppe D Norata
- Center for the Study of Atherosclerosis, E. Bassini Hospital, Cinisello Balsamo, Milan, Italy
- Department of Pharmacological and Biomolecular Sciences, University of Milan and IRCCS Multimedica, Via Balzaretti, 9, 20133, Milan, Italy
| | - Alberico L Catapano
- IRCCS MultiMedica, Sesto S. Giovanni, Milan, Italy.
- Department of Pharmacological and Biomolecular Sciences, University of Milan and IRCCS Multimedica, Via Balzaretti, 9, 20133, Milan, Italy.
| |
Collapse
|
21
|
Watanabe Y, Tatsuno I. Prevention of Cardiovascular Events with Omega-3 Polyunsaturated Fatty Acids and the Mechanism Involved. J Atheroscler Thromb 2020; 27:183-198. [PMID: 31582621 PMCID: PMC7113138 DOI: 10.5551/jat.50658] [Citation(s) in RCA: 55] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2019] [Accepted: 08/05/2019] [Indexed: 12/16/2022] Open
Abstract
An epidemiological study of Greenlandic Inuit suggested that fish oil, or omega-3 polyunsaturated fatty acids (PUFA), was important in preventing atherosclerotic disease. After this landmark study, many large-scale epidemiological studies and meta-analyses have examined the health benefits of omega-3 PUFA as part of a fatty acid-rich diet to demonstrate its beneficial roles in the prevention of cardiovascular diseases. Recent research has also focused attention on the anti-inflammatory effects of omega-3 PUFA and on specialized pro-resolving mediators. Findings of these studies have led to the development of omega-3 PUFA preparations for the treatment of dyslipidemia, including a highly purified eicosapentaenoic acid (EPA)-ethyl ester product (Epadel®) in Japan and an EPA/docosahexaenoic acid (DHA) preparation (Lotriga®) in the United States and Europe. Although various large-scale clinical trials on the cardiovascular preventive effect of omega-3 PUFA were conducted and reported, the results were not always consistent. The issues of not targeting subjects with hypertriglyceridemia and using low dose of omega-3 PUFA have been suggested to contribute to the failure of demonstrating the preventive effect of omega-3 PUFA in these clinical trials. Taking into account the above issues, the REDUCE-IT trial evaluated a highly purified EPA preparation at a high dose of 4 g/day in patients with hypertriglyceridemia and high cardiovascular risk, and demonstrated an extraordinary outcome of 25% relative reduction in cardiovascular events. This article reviews studies on omega-3 fatty acids during the last 50 years, including the progress in elucidating molecular mechanisms and recent large-scale clinical studies.
Collapse
Affiliation(s)
- Yasuhiro Watanabe
- Center for Diabetes, Metabolism and Endocrinology, Toho University Sakura Medical Center, Chiba, Japan
| | - Ichiro Tatsuno
- Center for Diabetes, Metabolism and Endocrinology, Toho University Sakura Medical Center, Chiba, Japan
| |
Collapse
|
22
|
Abdelhamid AS, Brown TJ, Brainard JS, Biswas P, Thorpe GC, Moore HJ, Deane KHO, Summerbell CD, Worthington HV, Song F, Hooper L. Omega-3 fatty acids for the primary and secondary prevention of cardiovascular disease. Cochrane Database Syst Rev 2020; 3:CD003177. [PMID: 32114706 PMCID: PMC7049091 DOI: 10.1002/14651858.cd003177.pub5] [Citation(s) in RCA: 118] [Impact Index Per Article: 23.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
BACKGROUND Omega-3 polyunsaturated fatty acids from oily fish (long-chain omega-3 (LCn3)), including eicosapentaenoic acid (EPA) and docosahexaenoic acid (DHA)), as well as from plants (alpha-linolenic acid (ALA)) may benefit cardiovascular health. Guidelines recommend increasing omega-3-rich foods, and sometimes supplementation, but recent trials have not confirmed this. OBJECTIVES To assess the effects of increased intake of fish- and plant-based omega-3 fats for all-cause mortality, cardiovascular events, adiposity and lipids. SEARCH METHODS We searched CENTRAL, MEDLINE and Embase to February 2019, plus ClinicalTrials.gov and World Health Organization International Clinical Trials Registry to August 2019, with no language restrictions. We handsearched systematic review references and bibliographies and contacted trial authors. SELECTION CRITERIA We included randomised controlled trials (RCTs) that lasted at least 12 months and compared supplementation or advice to increase LCn3 or ALA intake, or both, versus usual or lower intake. DATA COLLECTION AND ANALYSIS Two review authors independently assessed trials for inclusion, extracted data and assessed validity. We performed separate random-effects meta-analysis for ALA and LCn3 interventions, and assessed dose-response relationships through meta-regression. MAIN RESULTS We included 86 RCTs (162,796 participants) in this review update and found that 28 were at low summary risk of bias. Trials were of 12 to 88 months' duration and included adults at varying cardiovascular risk, mainly in high-income countries. Most trials assessed LCn3 supplementation with capsules, but some used LCn3- or ALA-rich or enriched foods or dietary advice compared to placebo or usual diet. LCn3 doses ranged from 0.5 g a day to more than 5 g a day (19 RCTs gave at least 3 g LCn3 daily). Meta-analysis and sensitivity analyses suggested little or no effect of increasing LCn3 on all-cause mortality (risk ratio (RR) 0.97, 95% confidence interval (CI) 0.93 to 1.01; 143,693 participants; 11,297 deaths in 45 RCTs; high-certainty evidence), cardiovascular mortality (RR 0.92, 95% CI 0.86 to 0.99; 117,837 participants; 5658 deaths in 29 RCTs; moderate-certainty evidence), cardiovascular events (RR 0.96, 95% CI 0.92 to 1.01; 140,482 participants; 17,619 people experienced events in 43 RCTs; high-certainty evidence), stroke (RR 1.02, 95% CI 0.94 to 1.12; 138,888 participants; 2850 strokes in 31 RCTs; moderate-certainty evidence) or arrhythmia (RR 0.99, 95% CI 0.92 to 1.06; 77,990 participants; 4586 people experienced arrhythmia in 30 RCTs; low-certainty evidence). Increasing LCn3 may slightly reduce coronary heart disease mortality (number needed to treat for an additional beneficial outcome (NNTB) 334, RR 0.90, 95% CI 0.81 to 1.00; 127,378 participants; 3598 coronary heart disease deaths in 24 RCTs, low-certainty evidence) and coronary heart disease events (NNTB 167, RR 0.91, 95% CI 0.85 to 0.97; 134,116 participants; 8791 people experienced coronary heart disease events in 32 RCTs, low-certainty evidence). Overall, effects did not differ by trial duration or LCn3 dose in pre-planned subgrouping or meta-regression. There is little evidence of effects of eating fish. Increasing ALA intake probably makes little or no difference to all-cause mortality (RR 1.01, 95% CI 0.84 to 1.20; 19,327 participants; 459 deaths in 5 RCTs, moderate-certainty evidence),cardiovascular mortality (RR 0.96, 95% CI 0.74 to 1.25; 18,619 participants; 219 cardiovascular deaths in 4 RCTs; moderate-certainty evidence), coronary heart disease mortality (RR 0.95, 95% CI 0.72 to 1.26; 18,353 participants; 193 coronary heart disease deaths in 3 RCTs; moderate-certainty evidence) and coronary heart disease events (RR 1.00, 95% CI 0.82 to 1.22; 19,061 participants; 397 coronary heart disease events in 4 RCTs; low-certainty evidence). However, increased ALA may slightly reduce risk of cardiovascular disease events (NNTB 500, RR 0.95, 95% CI 0.83 to 1.07; but RR 0.91, 95% CI 0.79 to 1.04 in RCTs at low summary risk of bias; 19,327 participants; 884 cardiovascular disease events in 5 RCTs; low-certainty evidence), and probably slightly reduces risk of arrhythmia (NNTB 91, RR 0.73, 95% CI 0.55 to 0.97; 4912 participants; 173 events in 2 RCTs; moderate-certainty evidence). Effects on stroke are unclear. Increasing LCn3 and ALA had little or no effect on serious adverse events, adiposity, lipids and blood pressure, except increasing LCn3 reduced triglycerides by ˜15% in a dose-dependent way (high-certainty evidence). AUTHORS' CONCLUSIONS This is the most extensive systematic assessment of effects of omega-3 fats on cardiovascular health to date. Moderate- and low-certainty evidence suggests that increasing LCn3 slightly reduces risk of coronary heart disease mortality and events, and reduces serum triglycerides (evidence mainly from supplement trials). Increasing ALA slightly reduces risk of cardiovascular events and arrhythmia.
Collapse
Affiliation(s)
- Asmaa S Abdelhamid
- University of East AngliaNorwich Medical SchoolNorwich Research ParkNorwichNorfolkUKNR4 7TJ
| | - Tracey J Brown
- University of East AngliaNorwich Medical SchoolNorwich Research ParkNorwichNorfolkUKNR4 7TJ
| | - Julii S Brainard
- University of East AngliaNorwich Medical SchoolNorwich Research ParkNorwichNorfolkUKNR4 7TJ
| | - Priti Biswas
- University of East AngliaMED/HSCNorwich Research ParkNorwichUKNR4 7TJ
| | - Gabrielle C Thorpe
- University of East AngliaSchool of Health SciencesEarlham RoadNorwichUKNR4 7TJ
| | - Helen J Moore
- Teesside UniversitySchool of Social Sciences, Humanities and LawMiddlesboroughUKTS1 3BA
| | - Katherine HO Deane
- University of East AngliaSchool of Health SciencesEarlham RoadNorwichUKNR4 7TJ
| | - Carolyn D Summerbell
- Durham UniversityDepartment of Sport and Exercise Sciences42 Old ElvetDurhamUKDH13HN
| | - Helen V Worthington
- Division of Dentistry, School of Medical Sciences, Faculty of Biology, Medicine and Health, The University of ManchesterCochrane Oral HealthCoupland Building 3Oxford RoadManchesterUKM13 9PL
| | - Fujian Song
- University of East AngliaNorwich Medical SchoolNorwich Research ParkNorwichNorfolkUKNR4 7TJ
| | - Lee Hooper
- University of East AngliaNorwich Medical SchoolNorwich Research ParkNorwichNorfolkUKNR4 7TJ
| | | |
Collapse
|
23
|
High-Dose Omega-3 Fatty Acids in Cardiovascular Prevention: Finally Living Up to Their Potential? Am J Cardiovasc Drugs 2020; 20:11-18. [PMID: 31485851 DOI: 10.1007/s40256-019-00363-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/26/2022]
Abstract
Despite the widespread use of statins in the setting of high cardiovascular risk, many patients continue to experience clinical events. This highlights the need to identify additional therapeutic strategies for high-risk patients. Interest in the use of omega-3 polyunsaturated fatty acids to prevent cardiovascular disease has been high for several decades. Despite promising results from before the statin era, many clinical trials have produced disappointing findings regarding products containing conventional doses of omega-3 fatty acids. More recent clinical trials using high doses of omega-3 fatty acids in targeted populations have suggested potential benefit when targeting the risk driven by atherogenic dyslipidemia. We review the clinical implications of completed and ongoing trials.
Collapse
|
24
|
Infante M, Ricordi C, Padilla N, Alvarez A, Linetsky E, Lanzoni G, Mattina A, Bertuzzi F, Fabbri A, Baidal D, Alejandro R. The Role of Vitamin D and Omega-3 PUFAs in Islet Transplantation. Nutrients 2019; 11:E2937. [PMID: 31816979 PMCID: PMC6950335 DOI: 10.3390/nu11122937] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2019] [Revised: 11/21/2019] [Accepted: 11/26/2019] [Indexed: 12/15/2022] Open
Abstract
Recurrence of autoimmunity and allograft rejection represent major challenges that impact the success of islet transplantation. Despite the remarkable improvements achieved in immunosuppression strategies after the publication of the Edmonton protocol, long-term data of intra-hepatic islet transplantation show a gradual decline in beta-cell function. Therefore, there is a growing interest in the investigation of novel, safe and effective anti-inflammatory and immunomodulatory strategies able to promote long-term islet graft survival and notable improvements in clinical outcomes of islet transplant recipients. Vitamin D has been shown to exert anti-inflammatory and immunomodulatory effects. Pre-clinical studies investigating the use of vitamin D and its analogs (alone or in combination with immunosuppressive agents and/or other anti-inflammatory agents, such as omega-3 polyunsaturated fatty acids) showed beneficial results in terms of islet graft survival and prevention of recurrence of autoimmunity/allograft rejection in animal models of syngeneic and allogeneic islet transplantation. Moreover, epidemiologic studies demonstrated that vitamin D deficiency is highly prevalent after solid organ transplantation (e.g., heart, liver or kidney transplantation). However, studies that critically assess the prevalence of vitamin D deficiency among islet transplant recipients have yet to be conducted. In addition, prospective studies aimed to address the safety and efficacy of vitamin D supplementation as an adjuvant immunomodulatory strategy in islet transplant recipients are lacking and are therefore awaited in the future.
Collapse
Affiliation(s)
- Marco Infante
- Diabetes Research Institute (DRI) and Clinical Cell Transplant Program, Miller School of Medicine, University of Miami, Miami, FL 33136, USA; (C.R.); (N.P.); (A.A.); (G.L.); (D.B.); (R.A.)
- Department of Systems Medicine, University of Rome “Tor Vergata”, 00133 Rome, Italy;
| | - Camillo Ricordi
- Diabetes Research Institute (DRI) and Clinical Cell Transplant Program, Miller School of Medicine, University of Miami, Miami, FL 33136, USA; (C.R.); (N.P.); (A.A.); (G.L.); (D.B.); (R.A.)
| | - Nathalia Padilla
- Diabetes Research Institute (DRI) and Clinical Cell Transplant Program, Miller School of Medicine, University of Miami, Miami, FL 33136, USA; (C.R.); (N.P.); (A.A.); (G.L.); (D.B.); (R.A.)
| | - Ana Alvarez
- Diabetes Research Institute (DRI) and Clinical Cell Transplant Program, Miller School of Medicine, University of Miami, Miami, FL 33136, USA; (C.R.); (N.P.); (A.A.); (G.L.); (D.B.); (R.A.)
| | - Elina Linetsky
- Diabetes Research Institute (DRI) and Cell Transplant Center, cGMP Cell Processing Facility, Miller School of Medicine, University of Miami, Miami, FL 33136, USA;
| | - Giacomo Lanzoni
- Diabetes Research Institute (DRI) and Clinical Cell Transplant Program, Miller School of Medicine, University of Miami, Miami, FL 33136, USA; (C.R.); (N.P.); (A.A.); (G.L.); (D.B.); (R.A.)
| | - Alessandro Mattina
- Diabetes and Islet Transplantation Unit, Department of Diagnostic and Therapeutic Services, IRCCS-ISMETT (Istituto Mediterraneo per i Trapianti e Terapie ad alta specializzazione), UPMC, 90127 Palermo, Italy;
| | | | - Andrea Fabbri
- Department of Systems Medicine, University of Rome “Tor Vergata”, 00133 Rome, Italy;
| | - David Baidal
- Diabetes Research Institute (DRI) and Clinical Cell Transplant Program, Miller School of Medicine, University of Miami, Miami, FL 33136, USA; (C.R.); (N.P.); (A.A.); (G.L.); (D.B.); (R.A.)
| | - Rodolfo Alejandro
- Diabetes Research Institute (DRI) and Clinical Cell Transplant Program, Miller School of Medicine, University of Miami, Miami, FL 33136, USA; (C.R.); (N.P.); (A.A.); (G.L.); (D.B.); (R.A.)
| |
Collapse
|
25
|
Harris WS. Understanding why REDUCE-IT was positive - Mechanistic overview of eicosapentaenoic acid. Prog Cardiovasc Dis 2019; 62:401-405. [PMID: 31666183 DOI: 10.1016/j.pcad.2019.10.008] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/23/2019] [Accepted: 10/23/2019] [Indexed: 11/16/2022]
Abstract
The REDUCE-IT study found that patients at elevated risk for cardiovascular disease (CVD) who were already taking statins obtained a marked benefit by taking 4 g/d of eicosapentaenoic acid ethyl esters (icosapent ethyl, IPE; Vascepa) over about 5 years. Although approved for triglyceride (TG) lowering, IPE had only a modest TG-lowering effect in REDUCE-IT, largely because median TG levels were relatively low already. Hence the question of what mechanisms IPE might be working through is of great interest. At present, it appears that the best mechanistic candidates would be anti-platelet effects and/or anti-inflammatory effects. Whatever the cause, the powerful effects of IPE on CVD risk have renewed interest in the clinical utility of omega-3 fatty acids.
Collapse
Affiliation(s)
- William S Harris
- Department of Internal Medicine, Sanford School of Medicine, University of South Dakota, and OmegaQuant Analytics, LLC, Sioux Falls, SD, United States of America.
| |
Collapse
|
26
|
Simonetto M, Infante M, Sacco RL, Rundek T, Della-Morte D. A Novel Anti-Inflammatory Role of Omega-3 PUFAs in Prevention and Treatment of Atherosclerosis and Vascular Cognitive Impairment and Dementia. Nutrients 2019; 11:E2279. [PMID: 31547601 PMCID: PMC6835717 DOI: 10.3390/nu11102279] [Citation(s) in RCA: 89] [Impact Index Per Article: 14.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2019] [Revised: 09/11/2019] [Accepted: 09/19/2019] [Indexed: 12/13/2022] Open
Abstract
Atherosclerosis is an inflammatory chronic disease affecting arterial vessels and leading to vascular diseases, such as stroke and myocardial infarction. The relationship between atherosclerosis and risk of neurodegeneration has been established, in particular with vascular cognitive impairment and dementia (VCID). Systemic atherosclerosis increases the risk of VCID by inducing cerebral infarction, or through systemic or local inflammatory factors that underlie both atherosclerosis and cognition. Omega-3 and omega-6 polyunsaturated fatty acids (PUFAs) are involved in inflammatory processes, but with opposite roles. Specifically, omega-3 PUFAs exert anti-inflammatory properties by competing with omega-6 PUFAs and displacing arachidonic acid in membrane phospholipids, decreasing the production of pro-inflammatory eicosanoids. Experimental studies and some clinical trials have demonstrated that omega-3 PUFA supplementation may reduce the risk of different phenotypes of atherosclerosis and cardiovascular disease. This review describes the link between atherosclerosis, VCID and inflammation, as well as how omega-3 PUFA supplementation may be useful to prevent and treat inflammatory-related diseases.
Collapse
Affiliation(s)
- Marialaura Simonetto
- Department of Neurology and The Evelyn F. McKnight Brain Institute, University of Miami Miller School of Medicine, Miami, FL 33136, USA.
| | - Marco Infante
- Diabetes Research Institute (DRI) and Clinical Cell Transplant Program, University of Miami Miller School of Medicine, Miami, FL 33136, USA.
- Department of Systems Medicine, University of Rome "Tor Vergata", 00133 Rome, Italy.
| | - Ralph L Sacco
- Department of Neurology and The Evelyn F. McKnight Brain Institute, University of Miami Miller School of Medicine, Miami, FL 33136, USA.
| | - Tatjana Rundek
- Department of Neurology and The Evelyn F. McKnight Brain Institute, University of Miami Miller School of Medicine, Miami, FL 33136, USA.
| | - David Della-Morte
- Department of Neurology and The Evelyn F. McKnight Brain Institute, University of Miami Miller School of Medicine, Miami, FL 33136, USA.
- Department of Systems Medicine, University of Rome "Tor Vergata", 00133 Rome, Italy.
- San Raffaele Roma Open University, 00166 Rome, Italy.
| |
Collapse
|
27
|
Abstract
BACKGROUND Currently, with stroke burden increasing, there is a need to explore therapeutic options that ameliorate the acute insult. There is substantial evidence of a neuroprotective effect of marine-derived n-3 polyunsaturated fatty acids (PUFAs) in experimental stroke, leading to a better functional outcome. OBJECTIVES To assess the effects of administration of marine-derived n-3 PUFAs on functional outcomes and dependence in people with stroke.Our secondary outcomes were vascular-related death, recurrent events, incidence of other type of stroke, adverse events, quality of life, and mood. SEARCH METHODS We searched the Cochrane Stroke Group trials register (6 August 2018), the Cochrane Central Register of Controlled Trials (CENTRAL; Issue 1, January 2019), MEDLINE Ovid (from 1948 to 6 August 2018), Embase Ovid (from 1980 to 6 August 2018), CINAHL EBSCO (Cumulative Index to Nursing and Allied Health Literature; from 1982 to 6 August 2018), Science Citation Index Expanded ‒ Web of Science (SCI-EXPANDED), Conference Proceedings Citation Index-Science - Web of Science (CPCI-S), and BIOSIS Citation Index. We also searched ongoing trial registers, reference lists, relevant systematic reviews, and used the Science Citation Index Reference Search. SELECTION CRITERIA We included randomised controlled trials (RCTs) comparing marine-derived n-3 PUFAs to placebo or open control (no placebo) in people with a history of stroke or transient ischaemic attack (TIA), or both. DATA COLLECTION AND ANALYSIS At least two review authors independently selected trials for inclusion, extracted data, assessed risk of bias, and used the GRADE approach to assess the quality of the body of evidence. We contacted study authors for clarification and additional information on stroke/TIA participants. We conducted random-effects meta-analysis or narrative synthesis, as appropriate. The primary outcome was efficacy (functional outcome) assessed using a validated scale e.g. Glasgow Outcome Scale Extended (GOSE) dichotomised into poor or good clinical outcome, Barthel Index (higher score is better; scale from 0 to 100) or Rivermead Mobility Index (higher score is better; scale from 0 to 15). MAIN RESULTS We included 29 RCTs; nine of them provided outcome data (3339 participants). Only one study included participants in the acute phase of stroke (haemorrhagic). Doses of marine-derived n-3 PUFAs ranged from 400 mg/day to 3300 mg/day. Risk of bias was generally low or unclear in most trials, with a higher risk of bias in smaller studies. We assessed results separately for short (up to three months) and longer (more than three months) follow-up studies.Short follow-up (up to three months)Functional outcome was reported in only one pilot study as poor clinical outcome assessed with GOSE (risk ratio (RR) 0.78, 95% confidence interval (CI) 0.36 to 1.68; 40 participants; very low quality evidence). Mood (assessed with GHQ-30, lower score better), was reported by only one study and favoured control (mean difference (MD) 1.41, 95% CI 0.07 to 2.75; 102 participants; low-quality evidence).We found no evidence of an effect of the intervention for the remainder of the secondary outcomes: vascular-related death (two studies, not pooled due to differences in population, RR 0.33, 95% CI 0.01 to 8.00, and RR 0.33, 95% CI 0.01 to 7.72; 142 participants; low-quality evidence); recurrent events (RR 0.41, 95% CI 0.02 to 8.84; 18 participants; very low quality evidence); incidence of other type of stroke (two studies, not pooled due to different type of index stroke, RR 6.11, 95% CI 0.33 to 111.71, and RR 0.63, 95% CI 0.25 to 1.58; 58 participants; very low quality evidence); and quality of life (physical component mean difference (MD) -2.31, 95% CI -4.81 to 0.19, and mental component MD -2.16, 95% CI -5.91 to 1.59; one study; 102 participants; low-quality evidence).Adverse events were reported by two studies (57 participants; very low quality evidence), one trial reporting extracranial haemorrhage (RR 0.25, 95% CI 0.04 to 1.73) and the other one reporting bleeding complications (RR 0.32, 95% CI 0.01 to 7.35).Longer follow-up (more than three months)One small trial assessed functional outcome with both Barthel Index (MD 7.09, 95% CI -5.16 to 19.34) for activities of daily living, and Rivermead Mobility Index (MD 1.30, 95% CI -1.31 to 3.91) for mobility (52 participants; very low quality evidence). We carried out meta-analysis for vascular-related death (RR 1.02, 95% CI 0.78 to 1.35; five studies; 2237 participants; low-quality evidence) and fatal recurrent events (RR 0.69, 95% CI 0.31 to 1.55; three studies; 1819 participants; low-quality evidence).We found no evidence of an effect of the intervention for mood (MD 1.00, 95% CI -2.07 to 4.07; one study; 14 participants; low-quality evidence). Incidence of other type of stroke and quality of life were not reported.Adverse events (all combined) were reported by only one study (RR 0.94, 95% CI 0.56 to 1.58; 1455 participants; low-quality evidence). AUTHORS' CONCLUSIONS We are very uncertain of the effect of marine-derived n-3 PUFAs therapy on functional outcomes and dependence after stroke as there is insufficient high-quality evidence. More well-designed RCTs are needed, specifically in acute stroke, to determine the efficacy and safety of the intervention.Studies assessing functionality might consider starting the intervention as early as possible after the event, as well as using standardised clinically-relevant measures for functional outcomes, such as the modified Rankin Scale. Optimal doses remain to be determined; delivery forms (type of lipid carriers) and mode of administration (ingestion or injection) also need further consideration.
Collapse
|
28
|
Nelson JR, Raskin S. The eicosapentaenoic acid:arachidonic acid ratio and its clinical utility in cardiovascular disease. Postgrad Med 2019; 131:268-277. [PMID: 31063407 DOI: 10.1080/00325481.2019.1607414] [Citation(s) in RCA: 104] [Impact Index Per Article: 17.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Eicosapentaenoic acid (EPA) is a key anti-inflammatory/anti-aggregatory long-chain polyunsaturated omega-3 fatty acid. Conversely, the omega-6 fatty acid, arachidonic acid (AA) is a precursor to a number of pro-inflammatory/pro-aggregatory mediators. EPA acts competitively with AA for the key cyclooxygenase and lipoxygenase enzymes to form less inflammatory products. As a result, the EPA:AA ratio may be a marker of chronic inflammation, with a lower ratio corresponding to higher levels of inflammation. It is now well established that inflammation plays an important role in cardiovascular disease. This review examines the role of the EPA:AA ratio as a marker of cardiovascular disease and the relationship between changes in the ratio (mediated by EPA intake) and changes in cardiovascular risk. Epidemiological studies have shown that a lower EPA:AA ratio is associated with an increased risk of coronary artery disease, acute coronary syndrome, myocardial infarction, stroke, chronic heart failure, peripheral artery disease, and vascular disease. Increasing the EPA:AA ratio through treatment with purified EPA has been shown in clinical studies to be effective in primary and secondary prevention of coronary artery disease and reduces the risk of cardiovascular events following percutaneous coronary intervention. The EPA:AA ratio is a valuable predictor of cardiovascular risk. Results from ongoing clinical trials will help to define thresholds for EPA treatment associated with better clinical outcomes.
Collapse
Affiliation(s)
- J R Nelson
- a California Cardiovascular Institute , Fresno , CA , USA
| | - S Raskin
- b Lipid Clinic , Sutter East Bay Medical Foundation , Oakland , CA , USA
| |
Collapse
|
29
|
Nakamura H, Tsujiguchi H, Kambayashi Y, Hara A, Miyagi S, Yamada Y, Nguyen TTT, Shimizu Y, Hori D, Nakamura H. Relationship between saturated fatty acid intake and hypertension and oxidative stress. Nutrition 2019; 61:8-15. [DOI: 10.1016/j.nut.2018.10.020] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2018] [Revised: 09/14/2018] [Accepted: 10/14/2018] [Indexed: 01/11/2023]
|
30
|
Abdelhamid AS, Brown TJ, Brainard JS, Biswas P, Thorpe GC, Moore HJ, Deane KHO, AlAbdulghafoor FK, Summerbell CD, Worthington HV, Song F, Hooper L. Omega-3 fatty acids for the primary and secondary prevention of cardiovascular disease. Cochrane Database Syst Rev 2018; 11:CD003177. [PMID: 30521670 PMCID: PMC6517311 DOI: 10.1002/14651858.cd003177.pub4] [Citation(s) in RCA: 95] [Impact Index Per Article: 13.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
BACKGROUND Researchers have suggested that omega-3 polyunsaturated fatty acids from oily fish (long-chain omega-3 (LCn3), including eicosapentaenoic acid (EPA) and docosahexaenoic acid (DHA)), as well as from plants (alpha-linolenic acid (ALA)) benefit cardiovascular health. Guidelines recommend increasing omega-3-rich foods, and sometimes supplementation, but recent trials have not confirmed this. OBJECTIVES To assess effects of increased intake of fish- and plant-based omega-3 for all-cause mortality, cardiovascular (CVD) events, adiposity and lipids. SEARCH METHODS We searched CENTRAL, MEDLINE and Embase to April 2017, plus ClinicalTrials.gov and World Health Organization International Clinical Trials Registry to September 2016, with no language restrictions. We handsearched systematic review references and bibliographies and contacted authors. SELECTION CRITERIA We included randomised controlled trials (RCTs) that lasted at least 12 months and compared supplementation and/or advice to increase LCn3 or ALA intake versus usual or lower intake. DATA COLLECTION AND ANALYSIS Two review authors independently assessed studies for inclusion, extracted data and assessed validity. We performed separate random-effects meta-analysis for ALA and LCn3 interventions, and assessed dose-response relationships through meta-regression. MAIN RESULTS We included 79 RCTs (112,059 participants) in this review update and found that 25 were at low summary risk of bias. Trials were of 12 to 72 months' duration and included adults at varying cardiovascular risk, mainly in high-income countries. Most studies assessed LCn3 supplementation with capsules, but some used LCn3- or ALA-rich or enriched foods or dietary advice compared to placebo or usual diet. LCn3 doses ranged from 0.5g/d LCn3 to > 5 g/d (16 RCTs gave at least 3g/d LCn3).Meta-analysis and sensitivity analyses suggested little or no effect of increasing LCn3 on all-cause mortality (RR 0.98, 95% CI 0.90 to 1.03, 92,653 participants; 8189 deaths in 39 trials, high-quality evidence), cardiovascular mortality (RR 0.95, 95% CI 0.87 to 1.03, 67,772 participants; 4544 CVD deaths in 25 RCTs), cardiovascular events (RR 0.99, 95% CI 0.94 to 1.04, 90,378 participants; 14,737 people experienced events in 38 trials, high-quality evidence), coronary heart disease (CHD) mortality (RR 0.93, 95% CI 0.79 to 1.09, 73,491 participants; 1596 CHD deaths in 21 RCTs), stroke (RR 1.06, 95% CI 0.96 to 1.16, 89,358 participants; 1822 strokes in 28 trials) or arrhythmia (RR 0.97, 95% CI 0.90 to 1.05, 53,796 participants; 3788 people experienced arrhythmia in 28 RCTs). There was a suggestion that LCn3 reduced CHD events (RR 0.93, 95% CI 0.88 to 0.97, 84,301 participants; 5469 people experienced CHD events in 28 RCTs); however, this was not maintained in sensitivity analyses - LCn3 probably makes little or no difference to CHD event risk. All evidence was of moderate GRADE quality, except as noted.Increasing ALA intake probably makes little or no difference to all-cause mortality (RR 1.01, 95% CI 0.84 to 1.20, 19,327 participants; 459 deaths, 5 RCTs),cardiovascular mortality (RR 0.96, 95% CI 0.74 to 1.25, 18,619 participants; 219 cardiovascular deaths, 4 RCTs), and CHD mortality (1.1% to 1.0%, RR 0.95, 95% CI 0.72 to 1.26, 18,353 participants; 193 CHD deaths, 3 RCTs) and ALA may make little or no difference to CHD events (RR 1.00, 95% CI 0.80 to 1.22, 19,061 participants, 397 CHD events, 4 RCTs, low-quality evidence). However, increased ALA may slightly reduce risk of cardiovascular events (from 4.8% to 4.7%, RR 0.95, 95% CI 0.83 to 1.07, 19,327 participants; 884 CVD events, 5 RCTs, low-quality evidence with greater effects in trials at low summary risk of bias), and probably reduces risk of arrhythmia (3.3% to 2.6%, RR 0.79, 95% CI 0.57 to 1.10, 4,837 participants; 141 events, 1 RCT). Effects on stroke are unclear.Sensitivity analysis retaining only trials at low summary risk of bias moved effect sizes towards the null (RR 1.0) for all LCn3 primary outcomes except arrhythmias, but for most ALA outcomes, effect sizes moved to suggest protection. LCn3 funnel plots suggested that adding in missing studies/results would move effect sizes towards null for most primary outcomes. There were no dose or duration effects in subgrouping or meta-regression.There was no evidence that increasing LCn3 or ALA altered serious adverse events, adiposity or lipids, except LCn3 reduced triglycerides by ˜15% in a dose-dependant way (high-quality evidence). AUTHORS' CONCLUSIONS This is the most extensive systematic assessment of effects of omega-3 fats on cardiovascular health to date. Moderate- and high-quality evidence suggests that increasing EPA and DHA has little or no effect on mortality or cardiovascular health (evidence mainly from supplement trials). Previous suggestions of benefits from EPA and DHA supplements appear to spring from trials with higher risk of bias. Low-quality evidence suggests ALA may slightly reduce CVD event and arrhythmia risk.
Collapse
Affiliation(s)
- Asmaa S Abdelhamid
- University of East AngliaNorwich Medical SchoolNorwich Research ParkNorwichNorfolkUKNR4 7TJ
| | - Tracey J Brown
- University of East AngliaNorwich Medical SchoolNorwich Research ParkNorwichNorfolkUKNR4 7TJ
| | - Julii S Brainard
- University of East AngliaNorwich Medical SchoolNorwich Research ParkNorwichNorfolkUKNR4 7TJ
| | - Priti Biswas
- University of East AngliaMED/HSCNorwich Research ParkNorwichUKNR4 7TJ
| | - Gabrielle C Thorpe
- University of East AngliaSchool of Health SciencesEarlham RoadNorwichUKNR4 7TJ
| | - Helen J Moore
- Durham UniversityWolfson Research InstituteDurhamUKDH1 3LE
| | - Katherine HO Deane
- University of East AngliaSchool of Health SciencesEarlham RoadNorwichUKNR4 7TJ
| | - Fai K AlAbdulghafoor
- University of East AngliaNorwich Medical SchoolNorwich Research ParkNorwichNorfolkUKNR4 7TJ
| | - Carolyn D Summerbell
- Durham UniversityDepartment of Sport and Exercise Science42 Old ElvetDurhamUKDH13HN
| | - Helen V Worthington
- Division of Dentistry, School of Medical Sciences, Faculty of Biology, Medicine and Health, The University of ManchesterCochrane Oral HealthJR Moore BuildingOxford RoadManchesterUKM13 9PL
| | - Fujian Song
- University of East AngliaNorwich Medical SchoolNorwich Research ParkNorwichNorfolkUKNR4 7TJ
| | - Lee Hooper
- University of East AngliaNorwich Medical SchoolNorwich Research ParkNorwichNorfolkUKNR4 7TJ
| | | |
Collapse
|
31
|
Abdelhamid AS, Martin N, Bridges C, Brainard JS, Wang X, Brown TJ, Hanson S, Jimoh OF, Ajabnoor SM, Deane KHO, Song F, Hooper L. Polyunsaturated fatty acids for the primary and secondary prevention of cardiovascular disease. Cochrane Database Syst Rev 2018; 11:CD012345. [PMID: 30484282 PMCID: PMC6517012 DOI: 10.1002/14651858.cd012345.pub3] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
BACKGROUND Evidence on the health effects of total polyunsaturated fatty acids (PUFA) is equivocal. Fish oils are rich in omega-3 PUFA and plant oils in omega-6 PUFA. Evidence suggests that increasing PUFA-rich foods, supplements or supplemented foods can reduce serum cholesterol, but may increase body weight, so overall cardiovascular effects are unclear. OBJECTIVES To assess effects of increasing total PUFA intake on cardiovascular disease and all-cause mortality, lipids and adiposity in adults. SEARCH METHODS We searched CENTRAL, MEDLINE and Embase to April 2017 and clinicaltrials.gov and the World Health Organization International Clinical Trials Registry Platform to September 2016, without language restrictions. We checked trials included in relevant systematic reviews. SELECTION CRITERIA We included randomised controlled trials (RCTs) comparing higher with lower PUFA intakes in adults with or without cardiovascular disease that assessed effects over 12 months or longer. We included full texts, abstracts, trials registry entries and unpublished data. Outcomes were all-cause mortality, cardiovascular disease mortality and events, risk factors (blood lipids, adiposity, blood pressure), and adverse events. We excluded trials where we could not separate effects of PUFA intake from other dietary, lifestyle or medication interventions. DATA COLLECTION AND ANALYSIS Two review authors independently screened titles and abstracts, assessed trials for inclusion, extracted data, and assessed risk of bias. We wrote to authors of included trials for further data. Meta-analyses used random-effects analysis, sensitivity analyses included fixed-effects and limiting to low summary risk of bias. We assessed GRADE quality of evidence. MAIN RESULTS We included 49 RCTs randomising 24,272 participants, with duration of one to eight years. Eleven included trials were at low summary risk of bias, 33 recruited participants without cardiovascular disease. Baseline PUFA intake was unclear in most trials, but 3.9% to 8% of total energy intake where reported. Most trials gave supplemental capsules, but eight gave dietary advice, eight gave supplemental foods such as nuts or margarine, and three used a combination of methods to increase PUFA.Increasing PUFA intake probably has little or no effect on all-cause mortality (risk 7.8% vs 7.6%, risk ratio (RR) 0.98, 95% confidence interval (CI) 0.89 to 1.07, 19,290 participants in 24 trials), but probably slightly reduces risk of coronary heart disease events from 14.2% to 12.3% (RR 0.87, 95% CI 0.72 to 1.06, 15 trials, 10,076 participants) and cardiovascular disease events from 14.6% to 13.0% (RR 0.89, 95% CI 0.79 to 1.01, 17,799 participants in 21 trials), all moderate-quality evidence. Increasing PUFA may slightly reduce risk of coronary heart disease death (6.6% to 6.1%, RR 0.91, 95% CI 0.78 to 1.06, 9 trials, 8810 participants) andstroke (1.2% to 1.1%, RR 0.91, 95% CI 0.58 to 1.44, 11 trials, 14,742 participants, though confidence intervals include important harms), but has little or no effect on cardiovascular mortality (RR 1.02, 95% CI 0.82 to 1.26, 16 trials, 15,107 participants) all low-quality evidence. Effects of increasing PUFA on major adverse cardiac and cerebrovascular events and atrial fibrillation are unclear as evidence is of very low quality.Increasing PUFA intake probably slightly decreases triglycerides (by 15%, MD -0.12 mmol/L, 95% CI -0.20 to -0.04, 20 trials, 3905 participants), but has little or no effect on total cholesterol (mean difference (MD) -0.12 mmol/L, 95% CI -0.23 to -0.02, 26 trials, 8072 participants), high-density lipoprotein (HDL) (MD -0.01 mmol/L, 95% CI -0.02 to 0.01, 18 trials, 4674 participants) or low-density lipoprotein (LDL) (MD -0.01 mmol/L, 95% CI -0.09 to 0.06, 15 trials, 3362 participants). Increasing PUFA probably has little or no effect on adiposity (body weight MD 0.76 kg, 95% CI 0.34 to 1.19, 12 trials, 7100 participants).Effects of increasing PUFA on serious adverse events such as pulmonary embolism and bleeding are unclear as the evidence is of very low quality. AUTHORS' CONCLUSIONS This is the most extensive systematic review of RCTs conducted to date to assess effects of increasing PUFA on cardiovascular disease, mortality, lipids or adiposity. Increasing PUFA intake probably slightly reduces risk of coronary heart disease and cardiovascular disease events, may slightly reduce risk of coronary heart disease mortality and stroke (though not ruling out harms), but has little or no effect on all-cause or cardiovascular disease mortality. The mechanism may be via TG reduction.
Collapse
Affiliation(s)
- Asmaa S Abdelhamid
- University of East AngliaNorwich Medical SchoolNorwich Research ParkNorwichNorfolkUKNR4 7TJ
| | - Nicole Martin
- University College LondonInstitute of Health Informatics Research222 Euston RoadLondonUKNW1 2DA
| | - Charlene Bridges
- University College LondonInstitute of Health Informatics Research222 Euston RoadLondonUKNW1 2DA
| | - Julii S Brainard
- University of East AngliaNorwich Medical SchoolNorwich Research ParkNorwichNorfolkUKNR4 7TJ
| | - Xia Wang
- University of East AngliaNorwich Medical SchoolNorwich Research ParkNorwichNorfolkUKNR4 7TJ
| | - Tracey J Brown
- University of East AngliaNorwich Medical SchoolNorwich Research ParkNorwichNorfolkUKNR4 7TJ
| | - Sarah Hanson
- University of East AngliaSchool of Health SciencesEdith Cavell BuildingNorwichUKNR4 7TJ
| | - Oluseyi F Jimoh
- University of East AngliaNorwich Medical SchoolNorwich Research ParkNorwichNorfolkUKNR4 7TJ
| | - Sarah M Ajabnoor
- University of East AngliaNorwich Medical SchoolNorwich Research ParkNorwichNorfolkUKNR4 7TJ
| | - Katherine HO Deane
- University of East AngliaSchool of Health SciencesEdith Cavell BuildingNorwichUKNR4 7TJ
| | - Fujian Song
- University of East AngliaNorwich Medical SchoolNorwich Research ParkNorwichNorfolkUKNR4 7TJ
| | - Lee Hooper
- University of East AngliaNorwich Medical SchoolNorwich Research ParkNorwichNorfolkUKNR4 7TJ
| | | |
Collapse
|
32
|
Nakamura H, Hara A, Tsujiguchi H, Thi Thu Nguyen T, Kambayashi Y, Miyagi S, Yamada Y, Suzuki K, Shimizu Y, Nakamura H. Relationship between Dietary n-6 Fatty Acid Intake and Hypertension: Effect of Glycated Hemoglobin Levels. Nutrients 2018; 10:E1825. [PMID: 30477201 PMCID: PMC6316654 DOI: 10.3390/nu10121825] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2018] [Revised: 11/19/2018] [Accepted: 11/20/2018] [Indexed: 12/16/2022] Open
Abstract
The relationship between dietary n-6 fatty acids and hypertension is not clear. The metabolic products of n-6 fatty acids include those that control blood pressure, such as prostaglandin and thromboxane, and that differ depending on the extent of glucose tolerance. This cross-sectional study investigated the association of dietary n-6 fatty acid intake on hypertension, and the effects of glycated hemoglobin (HbA1c) value in 633 Japanese subjects aged 40 years and older. Dietary intake was measured using a validated brief self-administered diet history questionnaire. We defined hypertension as the use of antihypertensive medication or a blood pressure of 140/90 mmHg. The prevalence of hypertension was 55.3%. A high n-6 fatty acids intake inversely correlated with hypertension in subjects with HbA1c values less than 6.5% (odds ratio, 0.857; 95% confidence interval, 0.744 to 0.987). On the contrary, in subjects with an HbA1c value of 6.5% or higher, the n-6 fatty acids intake was significantly associated with hypertension (odds ratio, 3.618; 95% confidence interval, 1.019 to 12.84). Regular dietary n-6 fatty acid intake may contribute to the prevention and treatment of hypertension in a healthy general population. By contrast, in subjects with diabetes, regular n-6 fatty acids intake may increase the risk of hypertension.
Collapse
Affiliation(s)
- Haruki Nakamura
- Department of Environmental and Preventive Medicine, Graduate School of Medical Science, Kanazawa University, 13-1 Takara-machi, Kanazawa, Ishikawa 920-8640, Japan.
| | - Akinori Hara
- Department of Environmental and Preventive Medicine, Graduate School of Medical Science, Kanazawa University, 13-1 Takara-machi, Kanazawa, Ishikawa 920-8640, Japan.
| | - Hiromasa Tsujiguchi
- Department of Environmental and Preventive Medicine, Graduate School of Medical Science, Kanazawa University, 13-1 Takara-machi, Kanazawa, Ishikawa 920-8640, Japan.
| | - Thao Thi Thu Nguyen
- Department of Environmental and Preventive Medicine, Graduate School of Medical Science, Kanazawa University, 13-1 Takara-machi, Kanazawa, Ishikawa 920-8640, Japan.
| | - Yasuhiro Kambayashi
- Department of Environmental and Preventive Medicine, Graduate School of Medical Science, Kanazawa University, 13-1 Takara-machi, Kanazawa, Ishikawa 920-8640, Japan.
| | - Sakae Miyagi
- Department of Environmental and Preventive Medicine, Graduate School of Medical Science, Kanazawa University, 13-1 Takara-machi, Kanazawa, Ishikawa 920-8640, Japan.
| | - Yohei Yamada
- Department of Environmental and Preventive Medicine, Graduate School of Medical Science, Kanazawa University, 13-1 Takara-machi, Kanazawa, Ishikawa 920-8640, Japan.
| | - Keita Suzuki
- Department of Environmental and Preventive Medicine, Graduate School of Medical Science, Kanazawa University, 13-1 Takara-machi, Kanazawa, Ishikawa 920-8640, Japan.
| | - Yukari Shimizu
- Department of Environmental and Preventive Medicine, Graduate School of Medical Science, Kanazawa University, 13-1 Takara-machi, Kanazawa, Ishikawa 920-8640, Japan.
| | - Hiroyuki Nakamura
- Department of Environmental and Preventive Medicine, Graduate School of Medical Science, Kanazawa University, 13-1 Takara-machi, Kanazawa, Ishikawa 920-8640, Japan.
| |
Collapse
|
33
|
Ishii H, Murohara T. Trade-Off Between Lipid-Lowering Therapy and Costs in Patients With Cardiovascular Disease. Circ J 2018; 82:2481-2482. [PMID: 30089745 DOI: 10.1253/circj.cj-18-0811] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/05/2024]
Affiliation(s)
- Hideki Ishii
- Department of Cardiology, Nagoya University Graduate School of Medicine
| | - Toyoaki Murohara
- Department of Cardiology, Nagoya University Graduate School of Medicine
| |
Collapse
|
34
|
Abe S, Sugimura H, Watanabe S, Murakami Y, Ebisawa K, Ioka T, Takahashi T, Ando T, Kono K, Inoue T. Eicosapantaenoic acid treatment based on the EPA/AA ratio in patients with coronary artery disease: follow-up data from the Tochigi Ryomo EPA/AA Trial in Coronary Artery Disease (TREAT-CAD) study. Hypertens Res 2018; 41:939-946. [DOI: 10.1038/s41440-018-0102-9] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2017] [Revised: 04/02/2018] [Accepted: 05/05/2018] [Indexed: 01/06/2023]
|
35
|
Yagi S, Kondo D, Ise T, Fukuda D, Yamaguchi K, Wakatsuki T, Kawabata Y, Ito H, Saijo Y, Seno H, Sutou K, Ueno R, Todoroki T, Kusunose K, Matsuura T, Tobiume T, Yamada H, Soeki T, Shimabukuro M, Aihara KI, Akaike M, Sata M. Association of Decreased Docosahexaenoic Acid Level After Statin Therapy and Low Eicosapentaenoic Acid Level with In-Stent Restenosis in Patients with Acute Coronary Syndrome. J Atheroscler Thromb 2018; 26:272-281. [PMID: 30135329 PMCID: PMC6402885 DOI: 10.5551/jat.44735] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023] Open
Abstract
Aim: It is speculated that statin therapy modulates the synthesis of polyunsaturated fatty acids (PUFA), including eicosapentaenoic acid (EPA) and docosahexaenoic acid (DHA). However, the data available on the effects of statin therapy on the serum levels of PUFA and the subsequent impact on in-stent restenosis (ISR) in patients with acute coronary syndrome (ACS) are limited. Methods: A total of 120 ACS patients who received emergent coronary stent implantation, follow-up coronary angiography to evaluate ISR, and new statin therapy were enrolled. We measured the serum levels of the PUFA and lipids at the onset of ACS and at the follow-up coronary angiography. Results: The follow-up coronary angiography revealed 38 ISR cases. New statin therapy significantly reduced the serum levels of DHA and low-density lipoprotein cholesterol (LDL-C), while it did not affect EPA level. Single regression analysis revealed that a decreased serum level of LDL-C was associated with decreased DHA level. The multiple logistic regression analysis revealed that the decreased DHA level after statin therapy and low serum level of EPA on admission were determinants of prevalence of ISR. Conclusion: Statin therapy decreased the serum level of DHA with a parallel reduction in LDL-C level in patients with ACS. Decreased DHA level after statin therapy and low EPA level on admission are risk factors for ISR, indicating that in patients with ACS, decreased serum levels of DHA may be a residual target for the prevention of ISR.
Collapse
Affiliation(s)
- Shusuke Yagi
- Department of Cardiovascular Medicine, Tokushima University Graduate School of Biomedical Sciences.,Department of Community Medicine and Human Resource Development, Tokushima University Graduate School of Biomedical Sciences
| | - Daisuke Kondo
- Student Laboratory, Faculty of Medicine, Tokushima University
| | - Takayuki Ise
- Department of Cardiovascular Medicine, Tokushima University Graduate School of Biomedical Sciences
| | - Daiju Fukuda
- Department of Cardio-Diabetes Medicine, Tokushima University Graduate School of Biomedical Sciences
| | - Koji Yamaguchi
- Department of Cardiovascular Medicine, Tokushima University Graduate School of Biomedical Sciences
| | - Tetsuzo Wakatsuki
- Department of Cardiovascular Medicine, Tokushima University Graduate School of Biomedical Sciences
| | - Yutaka Kawabata
- Department of Cardiovascular Medicine, Tokushima University Graduate School of Biomedical Sciences
| | - Hiroyuki Ito
- Department of Cardiovascular Medicine, Tokushima University Graduate School of Biomedical Sciences
| | - Yoshihito Saijo
- Department of Cardiovascular Medicine, Tokushima University Graduate School of Biomedical Sciences
| | - Hiromitsu Seno
- Department of Cardiovascular Medicine, Tokushima University Graduate School of Biomedical Sciences
| | - Kumiko Sutou
- Department of Cardiovascular Medicine, Tokushima University Graduate School of Biomedical Sciences
| | - Rie Ueno
- Department of Cardiovascular Medicine, Tokushima University Graduate School of Biomedical Sciences
| | - Takafumi Todoroki
- Department of Cardiovascular Medicine, Tokushima University Graduate School of Biomedical Sciences
| | - Kenya Kusunose
- Department of Cardiovascular Medicine, Tokushima University Graduate School of Biomedical Sciences
| | - Tomomi Matsuura
- Department of Cardiovascular Medicine, Tokushima University Graduate School of Biomedical Sciences
| | - Takeshi Tobiume
- Department of Cardiovascular Medicine, Tokushima University Graduate School of Biomedical Sciences
| | - Hirotsugu Yamada
- Department of Cardiovascular Medicine, Tokushima University Graduate School of Biomedical Sciences
| | - Takeshi Soeki
- Department of Cardiovascular Medicine, Tokushima University Graduate School of Biomedical Sciences
| | - Michio Shimabukuro
- Department of Cardio-Diabetes Medicine, Tokushima University Graduate School of Biomedical Sciences.,Department of Diabetes, Endocrinology and Metabolism, School of Medicine, Fukushima Medical University
| | - Ken-Ichi Aihara
- Department of Community Medicine for Diabetes and Metabolic Disorders, Tokushima University Graduate School of Biomedical Sciences
| | - Masashi Akaike
- Department of Medical Education, Tokushima University Graduate School of Biomedical Sciences
| | - Masataka Sata
- Department of Cardiovascular Medicine, Tokushima University Graduate School of Biomedical Sciences
| |
Collapse
|
36
|
Kimura Y, Yamada M, Hanada K, Endo T, Narita N, Narita M, Senoo M, Yonekura M, Ichikawa H, Kurauchi S, Nakaji S, Okumura K, Tomita H. Relationship Between Serum Eicosapentaenoic Acid Levels and J-Waves in a General Population in Japan. Int Heart J 2018; 59:736-740. [DOI: 10.1536/ihj.17-434] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
Affiliation(s)
- Yoshihiro Kimura
- Department of Cardiology, Hirosaki University Graduate School of Medicine
| | - Masahiro Yamada
- Department of Cardiology, Hirosaki University Graduate School of Medicine
| | - Kenji Hanada
- Department of Cardiology, Hirosaki University Graduate School of Medicine
| | - Tomohide Endo
- Department of Cardiology, Hirosaki University Graduate School of Medicine
| | - Noritomo Narita
- Department of Cardiology, Hirosaki University Graduate School of Medicine
| | - Masato Narita
- Department of Cardiology, Hirosaki University Graduate School of Medicine
| | - Maiko Senoo
- Department of Cardiology, Hirosaki University Graduate School of Medicine
| | - Manabu Yonekura
- Department of Cardiology, Hirosaki University Graduate School of Medicine
| | - Hiroaki Ichikawa
- Department of Cardiology, Hirosaki University Graduate School of Medicine
| | - Shizuka Kurauchi
- Department of Social Medicine, Hirosaki University Graduate School of Medicine
| | - Shigeyuki Nakaji
- Department of Social Medicine, Hirosaki University Graduate School of Medicine
| | - Ken Okumura
- Division of Cardiology, Saiseikai Kumamoto Hospital
| | - Hirofumi Tomita
- Department of Cardiology, Hirosaki University Graduate School of Medicine
| |
Collapse
|
37
|
Abdelhamid AS, Martin N, Bridges C, Brainard JS, Wang X, Brown TJ, Hanson S, Jimoh OF, Ajabnoor SM, Deane KHO, Song F, Hooper L. Polyunsaturated fatty acids for the primary and secondary prevention of cardiovascular disease. Cochrane Database Syst Rev 2018; 7:CD012345. [PMID: 30019767 PMCID: PMC6513571 DOI: 10.1002/14651858.cd012345.pub2] [Citation(s) in RCA: 49] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
BACKGROUND Evidence on the health effects of total polyunsaturated fatty acids (PUFA) is equivocal. Fish oils are rich in omega-3 PUFA and plant oils in omega-6 PUFA. Evidence suggests that increasing PUFA-rich foods, supplements or supplemented foods can reduce serum cholesterol, but may increase body weight, so overall cardiovascular effects are unclear. OBJECTIVES To assess effects of increasing total PUFA intake on cardiovascular disease and all-cause mortality, lipids and adiposity in adults. SEARCH METHODS We searched CENTRAL, MEDLINE and Embase to April 2017 and clinicaltrials.gov and the World Health Organization International Clinical Trials Registry Platform to September 2016, without language restrictions. We checked trials included in relevant systematic reviews. SELECTION CRITERIA We included randomised controlled trials (RCTs) comparing higher with lower PUFA intakes in adults with or without cardiovascular disease that assessed effects over 12 months or longer. We included full texts, abstracts, trials registry entries and unpublished data. Outcomes were all-cause mortality, cardiovascular disease mortality and events, risk factors (blood lipids, adiposity, blood pressure), and adverse events. We excluded trials where we could not separate effects of PUFA intake from other dietary, lifestyle or medication interventions. DATA COLLECTION AND ANALYSIS Two review authors independently screened titles and abstracts, assessed trials for inclusion, extracted data, and assessed risk of bias. We wrote to authors of included trials for further data. Meta-analyses used random-effects analysis, sensitivity analyses included fixed-effects and limiting to low summary risk of bias. We assessed GRADE quality of evidence. MAIN RESULTS We included 49 RCTs randomising 24,272 participants, with duration of one to eight years. Eleven included trials were at low summary risk of bias, 33 recruited participants without cardiovascular disease. Baseline PUFA intake was unclear in most trials, but 3.9% to 8% of total energy intake where reported. Most trials gave supplemental capsules, but eight gave dietary advice, eight gave supplemental foods such as nuts or margarine, and three used a combination of methods to increase PUFA.Increasing PUFA intake probably has little or no effect on all-cause mortality (risk 7.8% vs 7.6%, risk ratio (RR) 0.98, 95% confidence interval (CI) 0.89 to 1.07, 19,290 participants in 24 trials), but probably slightly reduces risk of coronary heart disease events from 14.2% to 12.3% (RR 0.87, 95% CI 0.72 to 1.06, 15 trials, 10,076 participants) and cardiovascular disease events from 14.6% to 13.0% (RR 0.89, 95% CI 0.79 to 1.01, 17,799 participants in 21 trials), all moderate-quality evidence. Increasing PUFA may slightly reduce risk of coronary heart disease death (6.6% to 6.1%, RR 0.91, 95% CI 0.78 to 1.06, 9 trials, 8810 participants) andstroke (1.2% to 1.1%, RR 0.91, 95% CI 0.58 to 1.44, 11 trials, 14,742 participants, though confidence intervals include important harms), but has little or no effect on cardiovascular mortality (RR 1.02, 95% CI 0.82 to 1.26, 16 trials, 15,107 participants) all low-quality evidence. Effects of increasing PUFA on major adverse cardiac and cerebrovascular events and atrial fibrillation are unclear as evidence is of very low quality.Increasing PUFA intake slightly reduces total cholesterol (mean difference (MD) -0.12 mmol/L, 95% CI -0.23 to -0.02, 26 trials, 8072 participants) and probably slightly decreases triglycerides (MD -0.12 mmol/L, 95% CI -0.20 to -0.04, 20 trials, 3905 participants), but has little or no effect on high-density lipoprotein (HDL) (MD -0.01 mmol/L, 95% CI -0.02 to 0.01, 18 trials, 4674 participants) or low-density lipoprotein (LDL) (MD -0.01 mmol/L, 95% CI -0.09 to 0.06, 15 trials, 3362 participants). Increasing PUFA probably causes slight weight gain (MD 0.76 kg, 95% CI 0.34 to 1.19, 12 trials, 7100 participants).Effects of increasing PUFA on serious adverse events such as pulmonary embolism and bleeding are unclear as the evidence is of very low quality. AUTHORS' CONCLUSIONS This is the most extensive systematic review of RCTs conducted to date to assess effects of increasing PUFA on cardiovascular disease, mortality, lipids or adiposity. Increasing PUFA intake probably slightly reduces risk of coronary heart disease and cardiovascular disease events, may slightly reduce risk of coronary heart disease mortality and stroke (though not ruling out harms), but has little or no effect on all-cause or cardiovascular disease mortality. The mechanism may be via lipid reduction, but increasing PUFA probably slightly increases weight.
Collapse
Affiliation(s)
- Asmaa S Abdelhamid
- University of East AngliaNorwich Medical SchoolNorwich Research ParkNorwichUKNR4 7TJ
| | - Nicole Martin
- University College LondonFarr Institute of Health Informatics Research222 Euston RoadLondonUKNW1 2DA
| | - Charlene Bridges
- University College LondonFarr Institute of Health Informatics Research222 Euston RoadLondonUKNW1 2DA
| | - Julii S Brainard
- University of East AngliaNorwich Medical SchoolNorwich Research ParkNorwichUKNR4 7TJ
| | - Xia Wang
- University of East AngliaNorwich Medical SchoolNorwich Research ParkNorwichUKNR4 7TJ
| | - Tracey J Brown
- University of East AngliaNorwich Medical SchoolNorwich Research ParkNorwichUKNR4 7TJ
| | - Sarah Hanson
- University of East AngliaSchool of Health SciencesEdith Cavell BuildingNorwichUKNR4 7TJ
| | - Oluseyi F Jimoh
- University of East AngliaNorwich Medical SchoolNorwich Research ParkNorwichUKNR4 7TJ
| | - Sarah M Ajabnoor
- University of East AngliaNorwich Medical SchoolNorwich Research ParkNorwichUKNR4 7TJ
| | - Katherine HO Deane
- University of East AngliaSchool of Health SciencesEdith Cavell BuildingNorwichUKNR4 7TJ
| | - Fujian Song
- University of East AngliaNorwich Medical SchoolNorwich Research ParkNorwichUKNR4 7TJ
| | - Lee Hooper
- University of East AngliaNorwich Medical SchoolNorwich Research ParkNorwichUKNR4 7TJ
| |
Collapse
|
38
|
Abdelhamid AS, Brown TJ, Brainard JS, Biswas P, Thorpe GC, Moore HJ, Deane KHO, AlAbdulghafoor FK, Summerbell CD, Worthington HV, Song F, Hooper L. Omega-3 fatty acids for the primary and secondary prevention of cardiovascular disease. Cochrane Database Syst Rev 2018; 7:CD003177. [PMID: 30019766 PMCID: PMC6513557 DOI: 10.1002/14651858.cd003177.pub3] [Citation(s) in RCA: 122] [Impact Index Per Article: 17.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
BACKGROUND Researchers have suggested that omega-3 polyunsaturated fatty acids from oily fish (long-chain omega-3 (LCn3), including eicosapentaenoic acid (EPA) and docosahexaenoic acid (DHA)), as well as from plants (alpha-linolenic acid (ALA)) benefit cardiovascular health. Guidelines recommend increasing omega-3-rich foods, and sometimes supplementation, but recent trials have not confirmed this. OBJECTIVES To assess effects of increased intake of fish- and plant-based omega-3 for all-cause mortality, cardiovascular (CVD) events, adiposity and lipids. SEARCH METHODS We searched CENTRAL, MEDLINE and Embase to April 2017, plus ClinicalTrials.gov and World Health Organization International Clinical Trials Registry to September 2016, with no language restrictions. We handsearched systematic review references and bibliographies and contacted authors. SELECTION CRITERIA We included randomised controlled trials (RCTs) that lasted at least 12 months and compared supplementation and/or advice to increase LCn3 or ALA intake versus usual or lower intake. DATA COLLECTION AND ANALYSIS Two review authors independently assessed studies for inclusion, extracted data and assessed validity. We performed separate random-effects meta-analysis for ALA and LCn3 interventions, and assessed dose-response relationships through meta-regression. MAIN RESULTS We included 79 RCTs (112,059 participants) in this review update and found that 25 were at low summary risk of bias. Trials were of 12 to 72 months' duration and included adults at varying cardiovascular risk, mainly in high-income countries. Most studies assessed LCn3 supplementation with capsules, but some used LCn3- or ALA-rich or enriched foods or dietary advice compared to placebo or usual diet.Meta-analysis and sensitivity analyses suggested little or no effect of increasing LCn3 on all-cause mortality (RR 0.98, 95% CI 0.90 to 1.03, 92,653 participants; 8189 deaths in 39 trials, high-quality evidence), cardiovascular mortality (RR 0.95, 95% CI 0.87 to 1.03, 67,772 participants; 4544 CVD deaths in 25 RCTs), cardiovascular events (RR 0.99, 95% CI 0.94 to 1.04, 90,378 participants; 14,737 people experienced events in 38 trials, high-quality evidence), coronary heart disease (CHD) mortality (RR 0.93, 95% CI 0.79 to 1.09, 73,491 participants; 1596 CHD deaths in 21 RCTs), stroke (RR 1.06, 95% CI 0.96 to 1.16, 89,358 participants; 1822 strokes in 28 trials) or arrhythmia (RR 0.97, 95% CI 0.90 to 1.05, 53,796 participants; 3788 people experienced arrhythmia in 28 RCTs). There was a suggestion that LCn3 reduced CHD events (RR 0.93, 95% CI 0.88 to 0.97, 84,301 participants; 5469 people experienced CHD events in 28 RCTs); however, this was not maintained in sensitivity analyses - LCn3 probably makes little or no difference to CHD event risk. All evidence was of moderate GRADE quality, except as noted.Increasing ALA intake probably makes little or no difference to all-cause mortality (RR 1.01, 95% CI 0.84 to 1.20, 19,327 participants; 459 deaths, 5 RCTs),cardiovascular mortality (RR 0.96, 95% CI 0.74 to 1.25, 18,619 participants; 219 cardiovascular deaths, 4 RCTs), and it may make little or no difference to CHD events (RR 1.00, 95% CI 0.80 to 1.22, 19,061 participants, 397 CHD events, 4 RCTs, low-quality evidence). However, increased ALA may slightly reduce risk of cardiovascular events (from 4.8% to 4.7%, RR 0.95, 95% CI 0.83 to 1.07, 19,327 participants; 884 CVD events, 5 RCTs, low-quality evidence), and probably reduces risk of CHD mortality (1.1% to 1.0%, RR 0.95, 95% CI 0.72 to 1.26, 18,353 participants; 193 CHD deaths, 3 RCTs), and arrhythmia (3.3% to 2.6%, RR 0.79, 95% CI 0.57 to 1.10, 4,837 participants; 141 events, 1 RCT). Effects on stroke are unclear.Sensitivity analysis retaining only trials at low summary risk of bias moved effect sizes towards the null (RR 1.0) for all LCn3 primary outcomes except arrhythmias, but for most ALA outcomes, effect sizes moved to suggest protection. LCn3 funnel plots suggested that adding in missing studies/results would move effect sizes towards null for most primary outcomes. There were no dose or duration effects in subgrouping or meta-regression.There was no evidence that increasing LCn3 or ALA altered serious adverse events, adiposity or lipids, although LCn3 slightly reduced triglycerides and increased HDL. ALA probably reduces HDL (high- or moderate-quality evidence). AUTHORS' CONCLUSIONS This is the most extensive systematic assessment of effects of omega-3 fats on cardiovascular health to date. Moderate- and high-quality evidence suggests that increasing EPA and DHA has little or no effect on mortality or cardiovascular health (evidence mainly from supplement trials). Previous suggestions of benefits from EPA and DHA supplements appear to spring from trials with higher risk of bias. Low-quality evidence suggests ALA may slightly reduce CVD event risk, CHD mortality and arrhythmia.
Collapse
Affiliation(s)
- Asmaa S Abdelhamid
- University of East AngliaNorwich Medical SchoolNorwich Research ParkNorwichUKNR4 7TJ
| | - Tracey J Brown
- University of East AngliaNorwich Medical SchoolNorwich Research ParkNorwichUKNR4 7TJ
| | - Julii S Brainard
- University of East AngliaNorwich Medical SchoolNorwich Research ParkNorwichUKNR4 7TJ
| | - Priti Biswas
- University of East AngliaMED/HSCNorwich Research ParkNorwichUKNR4 7TJ
| | - Gabrielle C Thorpe
- University of East AngliaSchool of Health SciencesEarlham RoadNorwichUKNR4 7TJ
| | - Helen J Moore
- Durham UniversityWolfson Research InstituteDurhamUKDH1 3LE
| | - Katherine HO Deane
- University of East AngliaSchool of Health SciencesEarlham RoadNorwichUKNR4 7TJ
| | - Fai K AlAbdulghafoor
- University of East AngliaNorwich Medical SchoolNorwich Research ParkNorwichUKNR4 7TJ
| | - Carolyn D Summerbell
- Durham UniversityDepartment of Sport and Exercise Science42 Old ElvetDurhamUKDH13HN
| | - Helen V Worthington
- Division of Dentistry, School of Medical Sciences, Faculty of Biology, Medicine and Health, The University of ManchesterCochrane Oral HealthJR Moore BuildingOxford RoadManchesterUKM13 9PL
| | - Fujian Song
- University of East AngliaNorwich Medical SchoolNorwich Research ParkNorwichUKNR4 7TJ
| | - Lee Hooper
- University of East AngliaNorwich Medical SchoolNorwich Research ParkNorwichUKNR4 7TJ
| |
Collapse
|
39
|
The Role of n-3 Long Chain Polyunsaturated Fatty Acids in Cardiovascular Disease Prevention, and Interactions with Statins. Nutrients 2018; 10:nu10060775. [PMID: 29914111 PMCID: PMC6024670 DOI: 10.3390/nu10060775] [Citation(s) in RCA: 47] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2018] [Revised: 06/09/2018] [Accepted: 06/13/2018] [Indexed: 01/08/2023] Open
Abstract
Decreases in global cardiovascular disease (CVD) mortality and morbidity in recent decades can be partly attributed to cholesterol reduction through statin use. n-3 long chain polyunsaturated fatty acids are recommended by some authorities for primary and secondary CVD prevention, and for triglyceride reduction. The residual risk of CVD that remains after statin therapy may potentially be reduced by n-3 long chain polyunsaturated fatty acids. However, the effects of concomitant use of statins and n-3 long chain polyunsaturated fatty acids are not well understood. Pleiotropic effects of statins and n-3 long chain polyunsaturated fatty acids overlap. For example, cytochrome P450 enzymes that metabolize statins may affect n-3 long chain polyunsaturated fatty acid metabolism and vice versa. Clinical and mechanistic study results show both synergistic and antagonistic effects of statins and n-3 long chain polyunsaturated fatty acids when used in combination.
Collapse
|
40
|
Mason RP, Dawoud H, Jacob RF, Sherratt SCR, Malinski T. Eicosapentaenoic acid improves endothelial function and nitric oxide bioavailability in a manner that is enhanced in combination with a statin. Biomed Pharmacother 2018; 103:1231-1237. [PMID: 29864903 DOI: 10.1016/j.biopha.2018.04.118] [Citation(s) in RCA: 62] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2018] [Revised: 04/04/2018] [Accepted: 04/16/2018] [Indexed: 11/27/2022] Open
Abstract
The endothelium exerts many vasoprotective effects that are largely mediated by release of nitric oxide (NO). Endothelial dysfunction represents an early but reversible step in atherosclerosis and is characterized by a reduction in the bioavailability of NO. Previous studies have shown that eicosapentaenoic acid (EPA), an omega-3 fatty acid (O3FA), and statins individually improve endothelial cell function, but their effects in combination have not been tested. Through a series of in vitro experiments, this study evaluated the effects of a combined treatment of EPA and the active metabolite of atorvastatin (ATM) on endothelial cell function under conditions of oxidative stress. Specifically, the comparative and time-dependent effects of these agents on endothelial dysfunction were examined by measuring the levels of NO and peroxynitrite (ONOO-) released from human umbilical vein endothelial cells (HUVECs). The data suggest that combined treatment with EPA and ATM is beneficial to endothelial function and was unique to EPA and ATM since similar improvements could not be recapitulated by substituting another O3FA docosahexaenoic acid (DHA) or other TG-lowering agents such as fenofibrate, niacin, or gemfibrozil. Comparable beneficial effects were observed when HUVECs were pretreated with EPA and ATM before exposure to oxidative stress. Interestingly, the kinetics of EPA-based protection of endothelial function in response to oxidation were found to be significantly different than those of DHA. Lastly, the beneficial effects on endothelial function generated by combined treatment of EPA and ATM were reproduced when this study was expanded to an ex vivo model utilizing rat glomerular endothelial cells. Taken together, these findings suggest that a combined treatment of EPA and ATM can inhibit endothelial dysfunction that occurs in response to conditions such as hyperglycemia, oxidative stress, and dyslipidemia.
Collapse
Affiliation(s)
- R Preston Mason
- Elucida Research LLC, Beverly, MA, 01915, United States; Cardiovascular Division, Department of Medicine, Brigham & Women's Hospital, Harvard Medical School, Boston, MA 02115, United States.
| | - Hazem Dawoud
- Nanomedical Research Laboratory, Ohio University, Athens, OH, 45701, United States
| | | | | | - Tadeusz Malinski
- Nanomedical Research Laboratory, Ohio University, Athens, OH, 45701, United States
| |
Collapse
|
41
|
Chen LS, Horton A, Bierut L. Pathways to precision medicine in smoking cessation treatments. Neurosci Lett 2018; 669:83-92. [PMID: 27208830 PMCID: PMC5115988 DOI: 10.1016/j.neulet.2016.05.033] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2016] [Revised: 05/12/2016] [Accepted: 05/17/2016] [Indexed: 02/06/2023]
Abstract
Cigarette smoking is highly addictive and modern genetic research has identified robust genetic influences on nicotine dependence. An important step in translating these genetic findings to clinical practice is identifying the genetic factors affecting smoking cessation in order to enhance current smoking cessation treatments. We reviewed the significant genetic variants that predict nicotine dependence, smoking cessation, and response to cessation pharmacotherapy. These data suggest that genetic risks can predict smoking cessation outcomes and moderate the effect of pharmacological treatments. Some pharmacogenetic findings have been replicated in meta-analyses or in multiple smoking cessation trials. The variation in efficacy between smokers with different genetic markers supports the notion that personalized smoking cessation intervention based upon genotype could maximize the efficiency of such treatment while minimizing side effects, thus influencing the number needed to treat (NNT) and the number needed to harm. In summary, as precision medicine is revolutionizing healthcare, smoking cessation may be one of the first areas where genetic variants may identify individuals at increased risk. Current evidence strongly suggests that genetic variants predict cessation failure and that cessation pharmacotherapy effectiveness is modulated by biomarkers such as nicotinic cholinergic receptor α5 subunit (CHRNA5) genotypes or nicotine metabolism ratio (NMR). These findings strengthen the case for the development and rigorous testing of treatments that target patients with different biological risk profiles.
Collapse
Affiliation(s)
- Li-Shiun Chen
- Department of Psychiatry, Washington University School of Medicine, St. Louis, MO 63110, United States.
| | - Amy Horton
- Department of Psychiatry, Washington University School of Medicine, St. Louis, MO 63110, United States
| | - Laura Bierut
- Department of Psychiatry, Washington University School of Medicine, St. Louis, MO 63110, United States
| |
Collapse
|
42
|
Kim M, Kim M, Yoo HJ, Lee A, Jeong S, Lee JH. Associations among FADS1 rs174547, eicosapentaenoic acid/arachidonic acid ratio, and arterial stiffness in overweight subjects. Prostaglandins Leukot Essent Fatty Acids 2018; 130:11-18. [PMID: 29549917 DOI: 10.1016/j.plefa.2018.02.004] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/15/2017] [Revised: 12/29/2017] [Accepted: 02/14/2018] [Indexed: 01/09/2023]
Abstract
We aimed to evaluate the longitudinal interaction effects between the minor allele of FADS1 rs174547 and overweight on n-3 and n-6 long-chain polyunsaturated fatty acid (PUFA) levels and pulse wave velocity (PWV). Plasma PUFA levels were measured via GC-MS, and arterial stiffness was determined as brachial-ankle PWV (ba-PWV) at baseline and after a mean follow-up of 3 years. The FADS1 rs174547 T > C genotype was analyzed. At 3-years of follow-up, after adjustment for age, sex, smoking and drinking, there were interaction effects between the FADS1 rs174547 T > C genotype and baseline BMI on the changes (from baseline) in plasma arachidonic acid (AA) levels, in the eicosapentaenoic acid (EPA)/AA ratio, and in ba-PWV (p for interaction = 0.036, 0.022, and 0.001, respectively). There were smaller increases in AA levels from baseline among normal-weight C allele carriers (n = 112) and overweight TT subjects (n = 47) than among normal-weight TT subjects (n = 91). Overweight C allele carriers (n = 37) showed greater reductions in the plasma EPA/AA ratio and greater increases in ba-PWV than the 3 other populations studied. The minor allele of the FADS1 rs174547 polymorphism is associated with age-related decreases in the EPA/AA ratio and increases in ba-PWV among overweight subjects.
Collapse
Affiliation(s)
- M Kim
- Research Center for Silver Science, Institute of Symbiotic Life-TECH, Yonsei University, Seoul, Republic of Korea
| | - M Kim
- Research Center for Silver Science, Institute of Symbiotic Life-TECH, Yonsei University, Seoul, Republic of Korea
| | - H J Yoo
- Department of Food and Nutrition, Brain Korea 21 PLUS Project, College of Human Ecology, Yonsei University, Seoul, Republic of Korea
| | - A Lee
- National Leading Research Laboratory of Clinical Nutrigenetics/Nutrigenomics, Department of Food and Nutrition, College of Human Ecology, Yonsei University, Seoul, Republic of Korea; Department of Food and Nutrition, Brain Korea 21 PLUS Project, College of Human Ecology, Yonsei University, Seoul, Republic of Korea
| | - S Jeong
- National Leading Research Laboratory of Clinical Nutrigenetics/Nutrigenomics, Department of Food and Nutrition, College of Human Ecology, Yonsei University, Seoul, Republic of Korea; Department of Food and Nutrition, Brain Korea 21 PLUS Project, College of Human Ecology, Yonsei University, Seoul, Republic of Korea
| | - J H Lee
- Research Center for Silver Science, Institute of Symbiotic Life-TECH, Yonsei University, Seoul, Republic of Korea; National Leading Research Laboratory of Clinical Nutrigenetics/Nutrigenomics, Department of Food and Nutrition, College of Human Ecology, Yonsei University, Seoul, Republic of Korea; Department of Food and Nutrition, Brain Korea 21 PLUS Project, College of Human Ecology, Yonsei University, Seoul, Republic of Korea.
| |
Collapse
|
43
|
A lower eicosapentaenoic acid/arachidonic acid ratio is associated with in-hospital fatal arrhythmic events in patients with acute myocardial infarction: a J-MINUET substudy. Heart Vessels 2017; 33:481-488. [DOI: 10.1007/s00380-017-1084-2] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/10/2017] [Accepted: 11/10/2017] [Indexed: 10/18/2022]
|
44
|
The Ratio of Eicosapentaenoic Acid (EPA) to Arachidonic Acid may be a Residual Risk Marker in Stable Coronary Artery Disease Patients Receiving Treatment with Statin Following EPA Therapy. Am J Cardiovasc Drugs 2017. [PMID: 28634822 DOI: 10.1007/s40256-017-0238-z] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
BACKGROUND We investigated the relationship between the eicosapentaenoic acid (EPA)/arachidonic acid (AA) ratio and non-high-density lipoprotein cholesterol (non-HDL-C) level, a major residual risk of coronary artery disease (CAD), in statin-treated CAD patients following EPA therapy. METHODS We conducted a 6-month, prospective, randomized clinical trial to investigate the effect of the additional administration of EPA on the EPA/AA ratio and the serum non-HDL-C level in stable CAD patients receiving statin treatment. We assigned CAD patients already receiving statin therapy to an EPA group (1800 mg/day; n = 50) or a control group (n = 50). RESULTS A significant reduction in the serum non-HDL-C level was observed in the EPA group, compared with the control group (-9.7 vs. -1.2%, p = 0.01). A multiple-regression analysis with adjustments for coronary risk factors revealed that achieved EPA/AA ratio was more reliable as an independent and significant predictor of a reduction in the non-HDL-C level at a 6-month follow-up examination (β = -0.324, p = 0.033) than the absolute change in the EPA/AA ratio. Interestingly, significant negative correlations were found between the baseline levels and the absolute change values of both non-HDL-C and triglyceride-rich lipoproteins, both markers of residual risk of CAD, indicating that patients with a higher baseline residual risk achieved a greater reduction. CONCLUSION The present results suggest that the achieved EPA/AA ratio, but not the absolute change in EPA/AA ratio, following EPA therapy might be a useful marker for the risk stratification of CAD among statin-treated patients with a high non-HDL-C level. CLINICAL TRIAL REGISTRATION UMIN ( http://www.umin.ac.jp/ ) Study ID: UMIN000010452.
Collapse
|
45
|
Clinical implications of eicosapentaenoic acid/arachidonic acid ratio (EPA/AA) in adult patients with congenital heart disease. Heart Vessels 2017; 32:1513-1522. [PMID: 28681101 DOI: 10.1007/s00380-017-1015-2] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/24/2017] [Accepted: 06/30/2017] [Indexed: 01/01/2023]
Abstract
Recent studies showed that a low ratio between the levels of eicosapentaenoic acid and those of arachidonic acid (EPA/AA) is associated with higher incidence of coronary artery disease and poor prognosis of heart failure, arrhythmia, and cardiac sudden death. However, the clinical implications of EPA/AA in adult patients with congenital heart disease remain unclear. We aimed to assess the prognostic value of EPA/AA regarding cardiac events in adult patients with congenital heart disease. We measured the serum levels of eicosapentaenoic acid and arachidonic acid in 130 adult patients (median age, 31 years) stratified into two groups according to their EPA/AA (low, ≤0.22; high, >0.22). We prospectively analyzed the association between EPA/AA and incidence of cardiac events during a mean observation period of 15 months, expressed in terms of hazard ratio (HR) with 95% confidence interval (95% CI). In the subgroup of patients with biventricular circulation (2VC) (n = 76), we analyzed the same clinical endpoints. In our study population, EPA/AA was not associated with the incidence of arrhythmic events (HR, 1.52; 95% CI, 0.82-2.85; p = 0.19), but low EPA/AA was a predictor of heart failure hospitalization (HR, 2.83; 95% CI, 1.35-6.30; p < 0.01). Among patients with 2VC, an EPA/AA of ≤0.25 was associated with a significantly higher risk of arrhythmic events (HR, 2.55; 95% CI, 1.11-6.41; p = 0.03) and heart failure hospitalization (HR, 5.20; 95% CI, 1.78-18.1; p < 0.01). EPA/AA represents a useful predictor of cardiac events in adult patients with congenital heart disease.
Collapse
|
46
|
Noda Y, Nilsson C, Shimada H, Kim H, Lundström T, Yajima T. Safety, Tolerability, and Pharmacokinetics of Single and Multiple Oral Doses of an Omega-3-Carboxylic Acid Formulation in Healthy Male Japanese Subjects: A Phase 1 Single-Blind, Randomized, Placebo-Controlled Trial. Clin Pharmacol Drug Dev 2017; 7:177-187. [PMID: 28597973 DOI: 10.1002/cpdd.360] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2016] [Accepted: 04/05/2017] [Indexed: 11/07/2022]
Abstract
OM3-CA (omega-3-carboxylic acids) is a complex mixture of omega-3 carboxylic acids, particularly eicosapentaenoic acid (EPA) and docosahexaenoic acid (DHA), which is approved in the United States for the treatment of hypertriglyceridemia. As part of its clinical development in Japan, we performed a phase 1 study to investigate the safety, tolerability, and pharmacokinetics after single and multiple doses of OM3-CA in healthy male Japanese subjects. Eighteen Japanese subjects were allocated to receive 2 or 4 g/day OM3-CA, or placebo (n = 6 per group). In addition, 6 white subjects received 4 g/day OM3-CA. The primary objective was to determine the safety and tolerability of OM3-CA. Plasma concentrations of EPA and DHA were adjusted for baseline values for pharmacokinetic analysis. Overall, OM3-CA was well tolerated in healthy Japanese subjects. Two Japanese subjects in each group and 5 white subjects experienced adverse events (AEs). Alanine aminotransferase increase was the most common AE in Japanese subjects, also seen with placebo, and diarrhea was the most common AE in white subjects. The maximum plasma concentrations of EPA and DHA were observed 5-6 hours postdose. The pharmacokinetic profiles of EPA and DHA after administration of OM3-CA were comparable between Japanese and white subjects.
Collapse
|
47
|
Shimada H, Nilsson C, Noda Y, Kim H, Lundström T, Yajima T. Effects of Food on the Pharmacokinetics of Omega-3-Carboxylic Acids in Healthy Japanese Male Subjects: A Phase I, Randomized, Open-label, Three-period, Crossover Trial. J Atheroscler Thromb 2017; 24:980-987. [PMID: 28344197 PMCID: PMC5587524 DOI: 10.5551/jat.38737] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022] Open
Abstract
Aims: Omega-3-carboxylic acids (OM3-CA) contain omega-3 free fatty acids, such as eicosapentaenoic acid (EPA) and docosahexaenoic acid (DHA), as carboxylic acids. Food intake is known to affect the bioavailability of ethyl ester fatty acid formulations. We conducted a phase I study to investigate the effects of the timing of OM3-CA administration relative to food intake on the pharmacokinetics of EPA and DHA. Methods: In this randomized, open-label, three-period crossover study, Japanese healthy male subjects were administered 4 × 1 g OM3-CA capsules with continued fasting, before a meal, or after a meal. All subjects fasted for ≥ 10 h prior to drug/meal administration. The primary objective was to examine the effect of meal timing on the pharmacokinetics of EPA and DHA after OM3-CA administration. The secondary objectives were to examine the safety and tolerability of OM3-CA. Results: A total of 42 Japanese subjects was enrolled in the study. The baseline-adjusted maximum concentration and area under the concentration–time curve from 0 to 72 h for EPA, DHA, and EPA + DHA were lower in the fasting and before meal conditions than in the after meal condition. The maximum total EPA, total DHA, and total EPA + DHA concentrations were reached later when administered in fasting conditions than in fed conditions, indicating slower absorption in fasting conditions. Diarrhea was reported by five, six, and no subjects in the fasting, before meal, and after meal conditions, respectively. Conclusions: The timing of OM3-CA administration relative to food intake influences the systemic bioavailability of EPA and DHA in healthy Japanese male subjects. Trial registration: NCT02372344
Collapse
|
48
|
Brinton EA, Mason RP. Prescription omega-3 fatty acid products containing highly purified eicosapentaenoic acid (EPA). Lipids Health Dis 2017; 16:23. [PMID: 28137294 PMCID: PMC5282870 DOI: 10.1186/s12944-017-0415-8] [Citation(s) in RCA: 46] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2016] [Accepted: 01/16/2017] [Indexed: 11/10/2022] Open
Abstract
The omega-3 fatty acid eicosapentaenoic acid (EPA) has multiple actions potentially conferring cardiovascular benefit, including lowering serum triglyceride (TG) and non-high-density lipoprotein cholesterol (non-HDL-C) levels and potentially reducing key steps in atherogenesis. Dietary supplements are a common source of omega-3 fatty acids in the US, but virtually all contain docosahexaenoic acid (DHA) in addition to EPA, and lipid effects differ between DHA and EPA. Contrary to popular belief, no over-the-counter omega-3 products are available in the US, only prescription products and dietary supplements. Among the US prescription omega-3 products, only one contains EPA exclusively (Vascepa); another closely related prescription omega-3 product also contains highly purified EPA, but is approved only in Japan and is provided in different capsule sizes. These high-purity EPA products do not raise low-density lipoprotein cholesterol (LDL-C) levels, even in patients with TG levels >500 mg/dL, in contrast to the increase in LDL-C levels with prescription omega-3 products that also contain DHA. The Japanese prescription EPA product was shown to significantly reduce major coronary events in hypercholesterolemic patients when added to statin therapy in the Japan EPA Lipid Intervention Study (JELIS). The effects of Vascepa on cardiovascular outcomes are being investigated in statin-treated patients with high TG levels in the Reduction of Cardiovascular Events With EPA-Intervention Trial (REDUCE-IT).
Collapse
Affiliation(s)
- Eliot A. Brinton
- Utah Foundation for Biomedical Research and the Utah Lipid Center, 419 Wakara Way, Suite 211, Salt Lake City, UT 84108 USA
| | - R. Preston Mason
- Department of Medicine, Cardiovascular Division, Brigham & Women’s Hospital, Harvard Medical School, Boston, MA and Elucida Research LLC, PO Box 7100, Beverly, MA 01915-6127 USA
| |
Collapse
|
49
|
Takahashi M, Ando J, Shimada K, Nishizaki Y, Tani S, Ogawa T, Yamamoto M, Nagao K, Hirayama A, Yoshimura M, Daida H, Nagai R, Komuro I. The ratio of serum n-3 to n-6 polyunsaturated fatty acids is associated with diabetes mellitus in patients with prior myocardial infarction: a multicenter cross-sectional study. BMC Cardiovasc Disord 2017; 17:41. [PMID: 28125968 PMCID: PMC5270364 DOI: 10.1186/s12872-017-0479-4] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2016] [Accepted: 01/20/2017] [Indexed: 12/16/2022] Open
Abstract
Background In prior myocardial infarction (PMI) patients, diabetes mellitus (DM), dyslipidemia, and hypertension increase the risk of secondary cardiovascular events. Although a decreased ratio of serum eicosapentaenoic acid (EPA) to arachidonic acid (AA; EPA/AA) has been shown to significantly correlate with the onset of acute coronary syndrome, the associations between polyunsaturated fatty acid (PUFA) levels and coronary risk factors in PMI patients have not been evaluated thoroughly. This study aimed to assess the associations between PUFAs levels and the risk factors in PMI patients. Methods We enrolled 1733 patients with known PUFA levels who were treated in five divisions of cardiology in a metropolitan area of Japan, including 303 patients with PMI. EPA/AA and docosahexaenoic acid (DHA) to AA level ratio (DHA/AA) in patients with and without PMI were analyzed according to presence of coronary risk factors. Results Diabetes patients with PMI had significantly lower EPA/AA and DHA/AA than diabetes patients without PMI (EPA/AA: P <0.01; DHA/AA: P =0.003), with no such differences in dyslipidemia and hypertension patients. In DM patients with high high-sensitivity C-reactive protein (hs-CRP) levels (>0.1 mg/dL), EPA/AA was low in individuals who also had PMI, whereas DHA/AA was not (EPA/AA, with PMI: 0.43 ± 0.24; without PMI: 0.53 ± 0.30, P < 0.05). Moreover, patients on statins had significantly lower DHA/AA ratios, whereas the EPA/AA ratio did not depend on statin use. Multiple regression analysis revealed that statin use in DM patients was associated with low DHA/AA but not EPA/AA. Conclusion PMI patients with DM have low EPA/AA and DHA/AA. EPA/AA and DHA/AA are differently related to hs-CRP level in DM patients with PMI. Statin use can potentially affect DHA/AA but not EPA/AA, and therefore EPA/AA ratio is a better marker of assessment for cardiovascular events. Electronic supplementary material The online version of this article (doi:10.1186/s12872-017-0479-4) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Masao Takahashi
- Department of Cardiovascular Medicine, Graduate School of Medicine, The University of Tokyo, Hongo 7-3-1, Bunkyo-ku, Tokyo, 113-8655, Japan.
| | - Jiro Ando
- Department of Cardiovascular Medicine, Graduate School of Medicine, The University of Tokyo, Hongo 7-3-1, Bunkyo-ku, Tokyo, 113-8655, Japan
| | - Kazunori Shimada
- Department of Cardiology, Juntendo University Graduate School of Medicine, Hongo 3-1-3, Bunkyo-ku, Tokyo, 113-8431, Japan
| | - Yuji Nishizaki
- Department of Cardiology, Juntendo University Graduate School of Medicine, Hongo 3-1-3, Bunkyo-ku, Tokyo, 113-8431, Japan
| | - Shigemasa Tani
- Department of Cardiology, Nihon University Hospital, 1-6 Kanda Surugadai Chiyoda-ku, Tokyo, 101-8309, Japan
| | - Takayuki Ogawa
- Divison of Cardiology, Department of Internal Medicine, The Jikei University School of Medicine, Nishishinbashi 3-19-18, Minato-ku, Tokyo, 105-8471, Japan
| | - Masato Yamamoto
- Department of Cardiology, Sempo Takanawa Hospital, Takanawa 3-10-11, Minato-ku, Tokyo, 108-8606, Japan
| | - Ken Nagao
- Department of Cardiology, Nihon University Hospital, 1-6 Kanda Surugadai Chiyoda-ku, Tokyo, 101-8309, Japan
| | - Atsushi Hirayama
- Division of Cardiology, Department of Medicine, Nihon University School of Medicine, 30-1Ohyaguchi Kamichou Itabashi-ku, Tokyo, 173-8610, Japan
| | - Michihiro Yoshimura
- Divison of Cardiology, Department of Internal Medicine, The Jikei University School of Medicine, Nishishinbashi 3-19-18, Minato-ku, Tokyo, 105-8471, Japan
| | - Hiroyuki Daida
- Department of Cardiology, Juntendo University Graduate School of Medicine, Hongo 3-1-3, Bunkyo-ku, Tokyo, 113-8431, Japan
| | - Ryozo Nagai
- Jichi Medical University, Yakushiji 3311-159, Shimotsuke city, Tochigi, 329-0498, Japan
| | - Issei Komuro
- Department of Cardiovascular Medicine, Graduate School of Medicine, The University of Tokyo, Hongo 7-3-1, Bunkyo-ku, Tokyo, 113-8655, Japan
| |
Collapse
|
50
|
Watanabe S, Yoshihisa A, Kanno Y, Takiguchi M, Yokokawa T, Sato A, Miura S, Shimizu T, Abe S, Sato T, Suzuki S, Oikawa M, Sakamoto N, Yamaki T, Sugimoto K, Kunii H, Nakazato K, Suzuki H, Saitoh SI, Takeishi Y. Associations With Eicosapentaenoic Acid to Arachidonic Acid Ratio and Mortality in Hospitalized Heart Failure Patients. J Card Fail 2016; 22:962-969. [DOI: 10.1016/j.cardfail.2016.04.017] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2015] [Revised: 04/19/2016] [Accepted: 04/25/2016] [Indexed: 10/21/2022]
|