51
|
Watts GF, Gidding SS, Hegele RA, Raal FJ, Sturm AC, Jones LK, Sarkies MN, Al-Rasadi K, Blom DJ, Daccord M, de Ferranti SD, Folco E, Libby P, Mata P, Nawawi HM, Ramaswami U, Ray KK, Stefanutti C, Yamashita S, Pang J, Thompson GR, Santos RD. International Atherosclerosis Society guidance for implementing best practice in the care of familial hypercholesterolaemia. Nat Rev Cardiol 2023; 20:845-869. [PMID: 37322181 DOI: 10.1038/s41569-023-00892-0] [Citation(s) in RCA: 99] [Impact Index Per Article: 49.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 05/12/2023] [Indexed: 06/17/2023]
Abstract
This contemporary, international, evidence-informed guidance aims to achieve the greatest good for the greatest number of people with familial hypercholesterolaemia (FH) across different countries. FH, a family of monogenic defects in the hepatic LDL clearance pathway, is a preventable cause of premature coronary artery disease and death. Worldwide, 35 million people have FH, but most remain undiagnosed or undertreated. Current FH care is guided by a useful and diverse group of evidence-based guidelines, with some primarily directed at cholesterol management and some that are country-specific. However, none of these guidelines provides a comprehensive overview of FH care that includes both the lifelong components of clinical practice and strategies for implementation. Therefore, a group of international experts systematically developed this guidance to compile clinical strategies from existing evidence-based guidelines for the detection (screening, diagnosis, genetic testing and counselling) and management (risk stratification, treatment of adults or children with heterozygous or homozygous FH, therapy during pregnancy and use of apheresis) of patients with FH, update evidence-informed clinical recommendations, and develop and integrate consensus-based implementation strategies at the patient, provider and health-care system levels, with the aim of maximizing the potential benefit for at-risk patients and their families worldwide.
Collapse
Affiliation(s)
- Gerald F Watts
- School of Medicine, University of Western Australia, Perth, WA, Australia.
- Departments of Cardiology and Internal Medicine, Royal Perth Hospital, Perth, WA, Australia.
| | | | - Robert A Hegele
- Department of Medicine and Robarts Research Institute, Schulich School of Medicine, Western University, London, ON, Canada
| | - Frederick J Raal
- Department of Medicine, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa
| | - Amy C Sturm
- Department of Genomic Health, Geisinger, Danville, PA, USA
- 23andMe, Sunnyvale, CA, USA
| | - Laney K Jones
- Department of Genomic Health, Geisinger, Danville, PA, USA
| | - Mitchell N Sarkies
- School of Health Sciences, Faculty of Medicine and Health, University of Sydney, Sydney, NSW, Australia
| | - Khalid Al-Rasadi
- Medical Research Centre, College of Medicine and Health Sciences, Sultan Qaboos University, Muscat, Oman
| | - Dirk J Blom
- Division of Lipidology and Cape Heart Institute, Department of Medicine, University of Cape Town, Cape Town, South Africa
| | | | | | | | - Peter Libby
- Division of Cardiovascular Medicine, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Pedro Mata
- Fundación Hipercolesterolemia Familiar, Madrid, Spain
| | - Hapizah M Nawawi
- Institute of Pathology, Laboratory and Forensic Medicine (I-PPerForM) and Faculty of Medicine, Universiti Teknologi MARA, Sungai Buloh, Selangor, Malaysia
- Specialist Lipid and Coronary Risk Prevention Clinics, Hospital Al-Sultan Abdullah (HASA) and Clinical Training Centre, Puncak Alam and Sungai Buloh Campuses, Universiti Teknologi MARA, Sungai Buloh, Selangor, Malaysia
| | - Uma Ramaswami
- Royal Free London NHS Foundation Trust, University College London, London, UK
| | - Kausik K Ray
- Imperial Centre for Cardiovascular Disease Prevention, Imperial College London, London, UK
| | - Claudia Stefanutti
- Department of Molecular Medicine, Extracorporeal Therapeutic Techniques Unit, Lipid Clinic and Atherosclerosis Prevention Centre, Regional Centre for Rare Diseases, Immunohematology and Transfusion Medicine, Umberto I Hospital, 'Sapienza' University of Rome, Rome, Italy
| | - Shizuya Yamashita
- Department of Cardiology, Rinku General Medical Center, Osaka, Japan
| | - Jing Pang
- School of Medicine, University of Western Australia, Perth, WA, Australia
| | | | - Raul D Santos
- Lipid Clinic, Heart Institute (InCor), University of São Paulo, São Paulo, Brazil
- Hospital Israelita Albert Einstein, São Paulo, Brazil
| |
Collapse
|
52
|
Qin J, Kurt E, LBassi T, Sa L, Xie D. Biotechnological production of omega-3 fatty acids: current status and future perspectives. Front Microbiol 2023; 14:1280296. [PMID: 38029217 PMCID: PMC10662050 DOI: 10.3389/fmicb.2023.1280296] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2023] [Accepted: 10/25/2023] [Indexed: 12/01/2023] Open
Abstract
Omega-3 fatty acids, including alpha-linolenic acids (ALA), eicosapentaenoic acid (EPA), and docosahexaenoic acid (DHA), have shown major health benefits, but the human body's inability to synthesize them has led to the necessity of dietary intake of the products. The omega-3 fatty acid market has grown significantly, with a global market from an estimated USD 2.10 billion in 2020 to a predicted nearly USD 3.61 billion in 2028. However, obtaining a sufficient supply of high-quality and stable omega-3 fatty acids can be challenging. Currently, fish oil serves as the primary source of omega-3 fatty acids in the market, but it has several drawbacks, including high cost, inconsistent product quality, and major uncertainties in its sustainability and ecological impact. Other significant sources of omega-3 fatty acids include plants and microalgae fermentation, but they face similar challenges in reducing manufacturing costs and improving product quality and sustainability. With the advances in synthetic biology, biotechnological production of omega-3 fatty acids via engineered microbial cell factories still offers the best solution to provide a more stable, sustainable, and affordable source of omega-3 fatty acids by overcoming the major issues associated with conventional sources. This review summarizes the current status, key challenges, and future perspectives for the biotechnological production of major omega-3 fatty acids.
Collapse
Affiliation(s)
| | | | | | | | - Dongming Xie
- Department of Chemical Engineering, University of Massachusetts Lowell, Lowell, MA, United States
| |
Collapse
|
53
|
Pourrajab B, Sharifi-Zahabi E, Soltani S, Shahinfar H, Shidfar F. Comparison of canola oil and olive oil consumption on the serum lipid profile in adults: a systematic review and meta-analysis of randomized controlled trials. Crit Rev Food Sci Nutr 2023; 63:12270-12284. [PMID: 35866510 DOI: 10.1080/10408398.2022.2100314] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/03/2022]
Abstract
BACKGROUND AND AIMS Several randomized clinical trials have investigated the effects of canola oil (CO) compared to olive oil (OO) on the serum lipid profiles in adults. However, the results of these studies are inconsistent. Thus, this study aimed to assess the comparison of CO and OO consumption on the serum lipid components in adults. METHODS AND RESULTS The following online databases were searched until February 4th, 2022: PubMed/Medline, Scopus, Clarivate Analytics Web of Science, Cochrane Central Register of Controlled Trials, and Google Scholar. The effect sizes were stated as the weighted mean difference (WMD) with 95% confidence intervals (CI). A total of 13 eligible trials were included in this meta-analysis. The results showed that the CO consumption, significantly reduced serum LDL-c (WMD: -6.13 mg/dl, 95%CI: -9.79, -2.46, p = 0.001), TC (WMD: -8.92 mg/dl, 95% CI: -13.52, -4.33, P < 0.001) and LDL-c/HDL-c ratio (WMD: -0.30; 95% CI, -0.53, -0.06, p = 0.01) levels compared to OO. There were no significant changes in the other components of the blood lipids. CONCLUSION The results of this review suggest that CO consumptionhas beneficial effects on LDL-c, TC, and LDL-c/HDL-c ratio compared to OO. Therefore, its replacement with OO can have cardioprotective impacts.
Collapse
Affiliation(s)
- Behnaz Pourrajab
- Department of Nutrition, School of Public Health, Iran University of Medical Sciences, Tehran, Iran
| | - Elham Sharifi-Zahabi
- Department of Nutrition, School of Public Health, Iran University of Medical Sciences, Tehran, Iran
| | - Sepideh Soltani
- Yazd Cardiovascular Research Center, Non-communicable Diseases Research Institute, Shahid Sadoughi University of Medical Sciences, Yazd, Iran
| | - Hossein Shahinfar
- Department of Nutrition, School of Public Health, Iran University of Medical Sciences, Tehran, Iran
| | - Farzad Shidfar
- Department of Nutrition, School of Public Health, Iran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
54
|
Yin S, Xu H, Xia J, Lu Y, Xu D, Sun J, Wang Y, Liao W, Sun G. Effect of Alpha-Linolenic Acid Supplementation on Cardiovascular Disease Risk Profile in Individuals with Obesity or Overweight: A Systematic Review and Meta-Analysis of Randomized Controlled Trials. Adv Nutr 2023; 14:1644-1655. [PMID: 37778442 PMCID: PMC10721518 DOI: 10.1016/j.advnut.2023.09.010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2023] [Revised: 08/22/2023] [Accepted: 09/19/2023] [Indexed: 10/03/2023] Open
Abstract
Overweight and obesity are highly prevalent worldwide and are associated with cardiovascular disease (CVD) risk factors, including systematic inflammation, dyslipidemia, and hypertension. Alpha-linolenic acid (ALA) is a plant-based essential polyunsaturated fatty acid associated with reduced CVD risks. This systematic review and meta-analysis aimed to investigate the effects of supplementation with ALA compared with the placebo on CVD risk factors in people with obesity or overweight (International Prospective Register of Systematic Reviews Registration No. CRD42023429563). This review included studies with adults using oral supplementation or food or combined interventions containing vegetable sources of ALA. All studies were randomly assigned trials with parallel or crossover designs. The Cochrane Collaboration tool was used for assessing the risk of bias (Version 1). PubMed, Web of Science, Embase, and Cochrane library databases were searched from inception to April 2023. Nineteen eligible randomized controlled trials, including 1183 participants, were included in the meta-analysis. Compared with placebo, dietary ALA supplementation significantly reduced C-reactive protein concentration (standardized mean difference [SMD] = -0.38 mg/L; 95% confidence interval [CI]: -0.72, -0.04), tumor necrosis factor-α concentration (SMD = -0.45 pg/mL; 95% CI: -0.73, -0.17), triglyceride in serum (SMD = -4.41 mg/dL; 95% CI: -5.99, -2.82), and systolic blood pressure (SMD = -0.37 mm Hg; 95% CI: -0.66, -0.08); but led to a significant increase in low-density lipoprotein cholesterol concentrations (SMD = 1.32 mg/dL; 95% CI: 0.05, 2.59). ALA supplementation had no significant effect on interleukin-6, diastolic blood pressure, total cholesterol, or high-density lipoprotein cholesterol (all P ≥ 0.05). Subgroup analysis revealed that ALA supplementation at a dose of ≥3 g/d from flaxseed and flaxseed oil had a more prominent effect on improving CVD risk profiles, particularly where the intervention duration was ≥12 wk and where the baseline CVD profile was poor.
Collapse
Affiliation(s)
- Shiyu Yin
- Department of Nutrition and Food Hygiene, Key Laboratory of Environmental Medicine and Engineering of Ministry of Education, School of Public Health, Southeast University, Nanjing, China
| | - Hai Xu
- Department of Nutrition and Food Hygiene, Key Laboratory of Environmental Medicine and Engineering of Ministry of Education, School of Public Health, Southeast University, Nanjing, China; Department of Food Processing and Safety, College of Biology and Food Engineering, Chongqing Three Gorges University, Chongqing, China
| | - Jiayue Xia
- Department of Nutrition and Food Hygiene, Key Laboratory of Environmental Medicine and Engineering of Ministry of Education, School of Public Health, Southeast University, Nanjing, China
| | - Yifei Lu
- Department of Nutrition and Food Hygiene, Key Laboratory of Environmental Medicine and Engineering of Ministry of Education, School of Public Health, Southeast University, Nanjing, China
| | - Dengfeng Xu
- Department of Nutrition and Food Hygiene, Key Laboratory of Environmental Medicine and Engineering of Ministry of Education, School of Public Health, Southeast University, Nanjing, China
| | - Jihan Sun
- Department of Nutrition and Food Hygiene, Key Laboratory of Environmental Medicine and Engineering of Ministry of Education, School of Public Health, Southeast University, Nanjing, China
| | - Yuanyuan Wang
- Department of Nutrition and Food Hygiene, Key Laboratory of Environmental Medicine and Engineering of Ministry of Education, School of Public Health, Southeast University, Nanjing, China
| | - Wang Liao
- Department of Nutrition and Food Hygiene, Key Laboratory of Environmental Medicine and Engineering of Ministry of Education, School of Public Health, Southeast University, Nanjing, China; China-DRIs Expert Committee on Macronutrients, Chinese Nutrition Society, Beijing, China
| | - Guiju Sun
- Department of Nutrition and Food Hygiene, Key Laboratory of Environmental Medicine and Engineering of Ministry of Education, School of Public Health, Southeast University, Nanjing, China; China-DRIs Expert Committee on Macronutrients, Chinese Nutrition Society, Beijing, China.
| |
Collapse
|
55
|
Liu H, Wang F, Xia H, Pan D, Yang L, Wang S, Zhao F, Sun G. Comparison of the effects of 3 kinds of oils rich in omega-3 polyunsaturated fatty acids on glycolipid metabolism and lipoprotein subfractions. FOOD SCIENCE AND HUMAN WELLNESS 2023; 12:2221-2231. [DOI: 10.1016/j.fshw.2023.03.042] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/31/2024]
|
56
|
Malick WA, Do R, Rosenson RS. Severe hypertriglyceridemia: Existing and emerging therapies. Pharmacol Ther 2023; 251:108544. [PMID: 37848164 DOI: 10.1016/j.pharmthera.2023.108544] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2023] [Revised: 09/19/2023] [Accepted: 09/28/2023] [Indexed: 10/19/2023]
Abstract
Severe hypertriglyceridemia (sHTG), defined as a triglyceride (TG) concentration ≥ 500 mg/dL (≥ 5.7 mmol/L) is an important risk factor for acute pancreatitis. Although lifestyle, some medications, and certain conditions such as diabetes may lead to HTG, sHTG results from a combination of major and minor genetic defects in proteins that regulate TG lipolysis. Familial chylomicronemia syndrome (FCS) is a rare disorder caused by complete loss of function in lipoprotein lipase (LPL) or LPL activating proteins due to two homozygous recessive traits or compound heterozygous traits. Multifactorial chylomicronemia syndrome (MCS) and sHTG are due to the accumulation of rare heterozygous variants and polygenic defects that predispose individuals to sHTG phenotypes. Until recently, treatment of sHTG focused on lifestyle interventions, control of secondary factors, and nonselective pharmacotherapies that had modest TG-lowering efficacy and no corresponding reductions in atherosclerotic cardiovascular disease events. Genetic discoveries have allowed for the development of novel pathway-specific therapeutics targeting LPL modulating proteins. New targets directed towards inhibition of apolipoprotein C-III (apoC-III), angiopoietin-like protein 3 (ANGPTL3), angiopoietin-like protein 4 (ANGPTL4), and fibroblast growth factor-21 (FGF21) offer far more efficacy in treating the various phenotypes of sHTG and opportunities to reduce the risk of acute pancreatitis and atherosclerotic cardiovascular disease events.
Collapse
Affiliation(s)
- Waqas A Malick
- Metabolism and Lipids Program, The Zena and Michael A. Wiener Cardiovascular Institute, Mount Sinai Heart, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Ron Do
- The Charles Bronfman Institute for Personalized Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Robert S Rosenson
- Metabolism and Lipids Program, The Zena and Michael A. Wiener Cardiovascular Institute, Mount Sinai Heart, Icahn School of Medicine at Mount Sinai, New York, NY, USA.
| |
Collapse
|
57
|
Engert LC, Mullington JM, Haack M. Prolonged experimental sleep disturbance affects the inflammatory resolution pathways in healthy humans. Brain Behav Immun 2023; 113:12-20. [PMID: 37369338 PMCID: PMC10528069 DOI: 10.1016/j.bbi.2023.06.018] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/10/2023] [Revised: 05/26/2023] [Accepted: 06/22/2023] [Indexed: 06/29/2023] Open
Abstract
BACKGROUND Sleep disturbances, as manifested in insomnia symptoms of difficulties falling asleep or frequent nighttime awakenings, are a strong risk factor for a diverse range of diseases involving immunopathology. Low-grade systemic inflammation has been frequently found associated with sleep disturbances and may mechanistically contribute to increased disease risk. Effects of sleep disturbances on inflammation have been observed to be long lasting and remain after recovery sleep has been obtained, suggesting that sleep disturbances may not only affect inflammatory mediators, but also the so-called specialized pro-resolving mediators (SPMs) that actively resolve inflammation. The goal of this investigation was to test for the first time whether the omega-3 fatty acid-derived D- (RvD) and E-series (RvE) resolvins are impacted by prolonged experimental sleep disturbance (ESD). METHODS Twenty-four healthy participants (12 F, age 20-42 years) underwent two 19-day in-hospital protocols (ESD/control), separated by > 2 months. The ESD protocol consisted of repeated nights of short and disrupted sleep with intermittent nights of undisturbed sleep, followed by three nights of recovery sleep at the end of the protocol. Under the control sleep condition, participants had an undisturbed sleep opportunity of 8 h/night throughout the protocol. The D- and E-series resolvins were measured in plasma using liquid chromatography-tandem mass spectrometry (LC-MS/MS). RESULTS The precursor of the D-series resolvins, 17-HDHA, was downregulated in the ESD compared to the control sleep condition (p <.001 for condition), and this effect remained after the third night of recovery sleep has been obtained. This effect was also observed for the resolvins RvD3, RvD4, and RvD5 (p <.001 for condition), while RvD1 was higher in the ESD compared to the control sleep condition (p <.01 for condition) and RvD2 showed a mixed effect of a decrease during disturbed sleep followed by an increase during recovery sleep in the ESD condition (p <.001 for condition*day interaction). The precursor of E-series resolvins, 18-HEPE, was downregulated in the ESD compared to the control sleep condition (p <.01 for condition) and remained low after recovery sleep has been obtained. This effect of downregulation was also observed for RvE2 (p <.01 for condition), while there was no effect for RvE1 (p >.05 for condition or condition*day interaction). Sex-differential effects were found for two of the D-series resolvins, i.e., RvD2 and RvD4. CONCLUSION This first investigation on the effects of experimental sleep disturbance on inflammatory resolution processes shows that SPMs, particularly resolvins of the D-series, are profoundly downregulated by sleep disturbances and remain downregulated after recovery sleep has been obtained, suggesting a longer lasting impact of sleep disturbances on these mediators. These findings also suggest that sleep disturbances contribute to the development and progression of a wide range of diseases characterized by immunopathology by interfering with processes that actively resolve inflammation. Pharmacological interventions aimed at promoting inflammatory resolution physiology may help to prevent future disease risk as a common consequence of sleep disturbances. TRIAL REGISTRATION ClinicalTrials.gov NCT02484742.
Collapse
Affiliation(s)
- Larissa C Engert
- Department of Neurology, Beth Israel Deaconess Medical Center, Boston, MA, USA; Harvard Medical School, Boston, MA, USA
| | - Janet M Mullington
- Department of Neurology, Beth Israel Deaconess Medical Center, Boston, MA, USA; Harvard Medical School, Boston, MA, USA
| | - Monika Haack
- Department of Neurology, Beth Israel Deaconess Medical Center, Boston, MA, USA; Harvard Medical School, Boston, MA, USA.
| |
Collapse
|
58
|
Tran M, Stanger L, Narendra S, Holinstat M, Holman TR. Investigating the catalytic efficiency of C22-Fatty acids with LOX human isozymes and the platelet response of the C22-oxylipin products. Arch Biochem Biophys 2023; 747:109742. [PMID: 37696384 PMCID: PMC10821779 DOI: 10.1016/j.abb.2023.109742] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2023] [Revised: 08/28/2023] [Accepted: 09/05/2023] [Indexed: 09/13/2023]
Abstract
Polyunsaturated fatty acids (PUFAs) have been extensively studied for their health benefits because they can be oxidized by lipoxygenases to form bioactive oxylipins. In this study, we investigated the impact of double bond placement on the kinetic properties and product profiles of human platelet 12-lipoxygenase (h12-LOX), human reticulocyte 15-lipoxygenase-1 (h15-LOX-1), and human endothelial 15-lipoxygenase-2 (h15-LOX-2) by using 22-carbon (C22) fatty acid substrates with differing double bond content. With respect to kcat/KM values, the loss of Δ4 and Δ19 led to an 18-fold loss of kinetic activity for h12-LOX, no change in kinetic capability for h15-LOX-1, but a 24-fold loss for h15-LOX-2 for both C22-FAs. With respect to the product profiles, h12-LOX produced mainly 14-oxylipins. For h15-LOX-1, the 14-oxylipin production increased with the loss of either Δ4 and Δ19, however, the 17-oxylipin became the major species upon loss of both Δ4 and Δ19. h15-LOX-2 produced mostly the 17-oxylipin products throughout the fatty acid series. This study also investigated the effects of various 17-oxylipins on platelet activation. The results revealed that both 17(S)-hydroxy-4Z,7Z,10Z,13Z,15E,19Z-DHA (17-HDHA) and 17-hydroxy-4Z,7Z,10Z,13Z,15E-DPAn6 (17-HDPAn6) demonstrated anti-aggregation properties with thrombin or collagen stimulation. 17-hydroxy-7Z,10Z,13Z,15E,19Z-DPAn3 (17-HDPAn3) exhibited agonistic properties, and 17-hydroxy-7Z,10Z,13Z,15E-DTA (17-HDTA) showed biphasic effects, inhibiting collagen-induced aggregation at lower concentrationsbut promoting aggregation at higher concentrations. Both 17-hydroxy-13Z,15E,19Z-DTrA (17-HDTrA), and 17-hydroxy-13Z,15E-DDiA (17-HDDiA) induced platelet aggregation. In summary, the number and placement of the double bonds affect platelet activation, with the general trend being that more double bonds generally inhibit aggregation, while less double bonds promote aggregation. These findings provide insights into the potential role of specific fatty acids and their metabolizing LOX isozymes with respect to cardiovascular health.
Collapse
Affiliation(s)
- Michelle Tran
- Department of Chemistry and Biochemistry, University of California Santa Cruz, Santa Cruz, CA 95064, USA
| | - Livia Stanger
- Department of Pharmacology, University of Michigan Medical School, Ann Arbor, MI, 48109, USA
| | - Srihari Narendra
- Department of Chemistry and Biochemistry, University of California Santa Cruz, Santa Cruz, CA 95064, USA
| | - Michael Holinstat
- Department of Pharmacology, University of Michigan Medical School, Ann Arbor, MI, 48109, USA
| | - Theodore R Holman
- Department of Chemistry and Biochemistry, University of California Santa Cruz, Santa Cruz, CA 95064, USA.
| |
Collapse
|
59
|
Assadourian JN, Peterson ED, McDonald SA, Gupta A, Navar AM. Health Claims and Doses of Fish Oil Supplements in the US. JAMA Cardiol 2023; 8:984-988. [PMID: 37610733 PMCID: PMC10448371 DOI: 10.1001/jamacardio.2023.2424] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/22/2023] [Accepted: 06/09/2023] [Indexed: 08/24/2023]
Abstract
Importance One in 5 US adults older than 60 years takes fish oil supplements often for heart health despite multiple randomized clinical trials showing no data for cardiovascular benefit for supplement-range doses. Statements on the supplement labels may influence consumer beliefs about health benefits. Objectives To evaluate health claims made on the labels of fish oil supplements in the US, and to examine doses of eicosapentaenoic acid (EPA) and docosahexaenoic acid (DHA) in commonly available formulations. Design, Setting, and Participants This cross-sectional study used data from labels of on-market fish oil (and nonfish ω-3 fatty acid) supplements obtained from the National Institutes of Health Dietary Supplement Label Database. The study was conducted and data analyzed from February to June 2022. Main Outcome and Measures The frequency and types of health claims made on fish oil labels (US Food and Drug Administration [FDA]-reviewed qualified health claim vs a structure/function claim) and the organ system referenced were evaluated. The total daily doses of combined EPA and DHA (EPA+DHA) were assessed for supplements from 16 leading manufacturers and retailers. Results Across 2819 unique fish oil supplements, 2082 (73.9%) made at least 1 health claim. Of these, only 399 (19.2%) used an FDA-approved qualified health claim; the rest (1683 [80.8%]) made only structure/function claims (eg, "promotes heart health"). Cardiovascular health claims were the most common (1747 [62.0%]). Across 16 leading brands/manufacturers, 255 fish oil supplements were identified. Among these, substantial variability was found in the daily dose of EPA (median [IQR], 340 [135-647] mg/d), DHA (median [IQR], 270 [140-500] mg/d), and total EPA+DHA (median [IQR], 600 [300-1100] mg/d). Only 24 of 255 supplements (9.4%) evaluated contained a daily dose of 2 g or more EPA+DHA. Conclusions Results of this cross-sectional study suggest that the majority of fish oil supplement labels make health claims, usually in the form of structure/function claims, that imply a health benefit across a variety of organ systems despite a lack of trial data showing efficacy. Significant heterogeneity exists in the daily dose of EPA+DHA in available supplements, leading to potential variability in safety and efficacy between supplements. Increasing regulation of dietary supplement labeling may be needed to prevent consumer misinformation.
Collapse
Affiliation(s)
| | | | | | - Anand Gupta
- University of Texas Southwestern Medical Center, Dallas
| | | |
Collapse
|
60
|
Jia M, Xu T, Xu YJ, Liu Y. Dietary fatty acids activate or deactivate brown and beige fat. Life Sci 2023; 330:121978. [PMID: 37516433 DOI: 10.1016/j.lfs.2023.121978] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2022] [Revised: 06/10/2023] [Accepted: 07/23/2023] [Indexed: 07/31/2023]
Abstract
Brown adipose tissue (BAT) and beige fat have been documented to rapidly consume fatty acids (FAs) rather than deposit of lipid, and they have high capacity to dissipate energy via nonshivering thermogenesis, making BAT and beige fat potential organs to fight obesity and related chronic diseases. As the main substrate for thermogenesis and the basic constituent unit of triacylglycerol, FAs could modify BAT and remodel white adipose tissue (WAT) to beige fat. However, there are few comprehensive review covering the link between dietary FAs and thermogenic adipocyte..In this review, we described the metabolism of thermogenic adipose upon activation and comprehensively summarized publications on the dietary FAs that activate or deactivate BAT and beige fat. Specifically, eicosapentaenoic acid/docosahexaenoic acid (EPA/DHA), α-linolenic acid (α-ALA), conjugated linoleic acid (CLA), oleic acid (OA), long-chain saturated fatty acid (LC-SFA) and medium-chain fatty acid (MCFA). in addition, the influences on BAT function, WAT remodeling, and lipid metabolism, as well as delineated the possible mechanisms are also reviewed. Characterizing thermogenic or obesogenic dietary FAs may offer novel insight into dietary oil and nutritional treatment.
Collapse
Affiliation(s)
- Min Jia
- School of Food Science and Technology, State Key Laboratory of Food Science and Technology, National Engineering Laboratory for Cereal Fermentation Technology, National Engineering Research Center for Functional Food, Jiangnan University, 1800 Lihu Road, Wuxi 214122, Jiangsu, PR China; Institute of Food & Nutrition Science and Technology, Shandong Engineering Research Center of Food for Special Medical Purpose, Key Laboratory of Agro-Products Processing Technology of Shandong Province, Key Laboratory of Novel Food Resources Processing, Ministry of Agriculture and Rural Affairs, Shandong Academy of Agricultural Sciences, 23788 Gongyebei Road, Jinan 250100, Shandong, PR China
| | - Tongcheng Xu
- Institute of Food & Nutrition Science and Technology, Shandong Engineering Research Center of Food for Special Medical Purpose, Key Laboratory of Agro-Products Processing Technology of Shandong Province, Key Laboratory of Novel Food Resources Processing, Ministry of Agriculture and Rural Affairs, Shandong Academy of Agricultural Sciences, 23788 Gongyebei Road, Jinan 250100, Shandong, PR China
| | - Yong-Jiang Xu
- School of Food Science and Technology, State Key Laboratory of Food Science and Technology, National Engineering Laboratory for Cereal Fermentation Technology, National Engineering Research Center for Functional Food, Jiangnan University, 1800 Lihu Road, Wuxi 214122, Jiangsu, PR China.
| | - Yuanfa Liu
- School of Food Science and Technology, State Key Laboratory of Food Science and Technology, National Engineering Laboratory for Cereal Fermentation Technology, National Engineering Research Center for Functional Food, Jiangnan University, 1800 Lihu Road, Wuxi 214122, Jiangsu, PR China.
| |
Collapse
|
61
|
LI JJ, ZHAO SP, ZHAO D, LU GP, PENG DQ, LIU J, CHEN ZY, GUO YL, WU NQ, YAN SK, WANG ZW, GAO RL. 2023 China Guidelines for Lipid Management. J Geriatr Cardiol 2023; 20:621-663. [PMID: 37840633 PMCID: PMC10568545 DOI: 10.26599/1671-5411.2023.09.008] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/17/2023] Open
Abstract
Atherosclerotic cardiovascular disease (ASCVD) is the leading cause of death among urban and rural residents in China, and elevated low-density lipoprotein cholesterol (LDL-C) is a risk factor for ASCVD. Considering the increasing burden of ASCVD, lipid management is of the utmost importance. In recent years, research on blood lipids has made breakthroughs around the world, hence a revision of China guidelines for lipid management is imperative, especially since the target lipid levels in the general population vary in respect to the risk of ASCVD. The level of LDL-C, which can be regarded as appropriate in a population without frisk factors, can be considered abnormal in people at high risk of developing ASCVD. As a result, the "Guidelines for the prevention and treatment of dyslipidemia" were adapted into the "China Guidelines for Lipid Management" (henceforth referred to as the new guidelines) by an Experts' committee after careful deliberation. The new guidelines still recommend LDL-C as the primary target for lipid control, with CVD risk stratification to determine its target value. These guidelines recommend that moderate intensity statin therapy in adjunct with a heart-healthy lifestyle, be used as an initial line of treatment, followed by cholesterol absorption inhibitors or/and proprotein convertase subtilisin/kexin type 9 (PCSK9) inhibitors, as necessary. The new guidelines provide guidance for lipid management across various age groups, from children to the elderly. The aim of these guidelines is to comprehensively improve the management of lipids and promote the prevention and treatment of ASCVD by guiding clinical practice.
Collapse
Affiliation(s)
- Jian-Jun LI
- National Center for Cardiovascular Diseases, Fuwai Hospital, Chinese Academy of Medical Sciences, Beijing, China
| | - Shui-Ping ZHAO
- The Second Xiangya Hospital of Central South University, Changsha, Hunan, China
| | - Dong ZHAO
- Beijing Anzhen Hospital, Capital Medical University, Beijing, China
| | - Guo-Ping LU
- Ruijin Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Dao-Quan PENG
- The Second Xiangya Hospital of Central South University, Changsha, Hunan, China
| | - Jing LIU
- Beijing Anzhen Hospital, Capital Medical University, Beijing, China
| | - Zhen-Yue CHEN
- Ruijin Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Yuan-Lin GUO
- National Center for Cardiovascular Diseases, Fuwai Hospital, Chinese Academy of Medical Sciences, Beijing, China
| | - Na-Qiong WU
- National Center for Cardiovascular Diseases, Fuwai Hospital, Chinese Academy of Medical Sciences, Beijing, China
| | - Sheng-Kai YAN
- Affiliated Hospital of Zunyi Medical University, School of Laboratory Medicine of Zunyi Medical University, Zunyi, Guizhou, China
| | - Zeng-Wu WANG
- National Center for Cardiovascular Diseases, Fuwai Hospital, Chinese Academy of Medical Sciences, Beijing, China
| | - Run-Lin GAO
- National Center for Cardiovascular Diseases, Fuwai Hospital, Chinese Academy of Medical Sciences, Beijing, China
| |
Collapse
|
62
|
Sun Y, McDonald T, Baur A, Xu H, Bateman NB, Shen Y, Li C, Ye K. Fish oil supplementation modifies the genetic potential for blood lipids. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2023:2023.09.22.23295987. [PMID: 37808791 PMCID: PMC10557817 DOI: 10.1101/2023.09.22.23295987] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/10/2023]
Abstract
Background Dyslipidemia is a well-known risk factor for cardiovascular disease, which has been the leading cause of mortality worldwide. Although habitual intake of fish oil has been implicated in offering cardioprotective effects through triglyceride reduction, the interactions of fish oil with the genetic predisposition to dysregulated lipids remain elusive. Objectives We examined whether fish oil supplementation can modify the genetic potential for the circulating levels of four lipids, including total cholesterol, low-density lipoprotein cholesterol (LDL-C), high-density lipoprotein cholesterol (HDL-C), and triglycerides. Methods A total of 441,985 participants with complete genetic and phenotypic data from the UK Biobank were included in our study. Polygenic scores (PGS) were calculated in participants of diverse ancestries. Multivariable linear regression models were used to assess associations with adjustment for relevant risk factors. Results Fish oil supplementation mitigated genetic susceptibility to elevated levels of total cholesterol, LDL-C, and triglycerides, while amplifying genetic potential for increased HDL-C among 424,090 participants of European ancestry P interaction < 0.05 . Consistent significant findings were obtained using PGS calculated based on multiple genome-wide association studies or alternative PGS methods. We also showed that fish oil significantly attenuated genetic predisposition to high triglycerides in African-ancestry participants. Conclusions Fish oil supplementation attenuated the genetic susceptibility to elevated blood levels of total cholesterol, LDL-C, and triglycerides, while accentuating genetic potential for higher HDL-C. These results suggest that fish oil may have a beneficial impact on modifying genome-wide genetic effects on elevated lipid levels in the general population.
Collapse
Affiliation(s)
- Yitang Sun
- Department of Genetics, Franklin College of Arts and Sciences, University of Georgia, Athens, GA, USA
| | - Tryggvi McDonald
- Department of Genetics, Franklin College of Arts and Sciences, University of Georgia, Athens, GA, USA
| | - Abigail Baur
- Department of Genetics, Franklin College of Arts and Sciences, University of Georgia, Athens, GA, USA
| | - Huifang Xu
- Department of Genetics, Franklin College of Arts and Sciences, University of Georgia, Athens, GA, USA
| | - Naveen Brahman Bateman
- Department of Genetics, Franklin College of Arts and Sciences, University of Georgia, Athens, GA, USA
| | - Ye Shen
- Department of Epidemiology and Biostatistics, College of Public Health, University of Georgia, Athens, GA, USA
| | - Changwei Li
- Department of Epidemiology, School of Public Health and Tropical Medicine, Tulane University, New Orleans, LA, USA
| | - Kaixiong Ye
- Department of Genetics, Franklin College of Arts and Sciences, University of Georgia, Athens, GA, USA
- Institute of Bioinformatics, University of Georgia, Athens, GA, USA
| |
Collapse
|
63
|
Matveyenko A, Seid H, Kim K, Ramakrishnan R, Thomas T, Matienzo N, Reyes-Soffer G. Association of free-living diet composition with plasma lipoprotein(a) levels in healthy adults. Lipids Health Dis 2023; 22:144. [PMID: 37670291 PMCID: PMC10478368 DOI: 10.1186/s12944-023-01884-2] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2023] [Accepted: 07/27/2023] [Indexed: 09/07/2023] Open
Abstract
BACKGROUND Lipoprotein (a) [Lp(a)] is an apoB100-containing lipoprotein with high levels being positively associated with atherosclerotic cardiovascular disease. Lp(a) levels are genetically determined. However, previous studies report a negative association between Lp(a) and saturated fatty acid intake. Currently, apoB100 lowering therapies are used to lower Lp(a) levels, and apheresis therapy is FDA approved for patients with extreme elevations of Lp(a). The current study analyzed the association of free-living diet components with plasma Lp(a) levels. METHODS Dietary composition data was collected during screening visits for enrollment in previously completed lipid and lipoprotein metabolism studies at Columbia University Irving Medical Center via a standardized protocol by registered dietitians using 24 hour recalls. Data were analyzed with the Nutrition Data System for Research (Version 2018). Diet quality was calculated using the Healthy Eating Index (HEI) score. Fasting plasma Lp(a) levels were measured via an isoform-independent ELISA and apo(a) isoforms were measured using gel electrophoresis. RESULTS We enrolled 28 subjects [Black (n = 18); Hispanic (n = 7); White (n = 3)]. The mean age was 48.3 ± 12.5 years with 17 males. Median level of Lp(a) was 79.9 nmol/L (34.4-146.0) and it was negatively associated with absolute (grams/day) and relative (percent of total calories) intake of dietary saturated fatty acids (SFA) (R = -0.43, P = 0.02, SFA …(% CAL): R = -0.38, P = 0.04), palmitic acid intake (R = -0.38, P = 0.05), and stearic acid intake (R = -0.40, P = 0.03). Analyses of associations with HEI score when stratified based on Lp(a) levels > or ≤ 100 nmol/L revealed no significant associations with any of the constituent factors. CONCLUSIONS Using 24 hour recall, we confirm previous findings that Lp(a) levels are negatively associated with dietary saturated fatty acid intake. Additionally, Lp(a) levels are not related to diet quality, as assessed by the HEI score. The mechanisms underlying the relationship of SFA with Lp(a) require further investigation.
Collapse
Affiliation(s)
- Anastasiya Matveyenko
- Department of Medicine, Columbia University Vagelos College of Physicians and Surgeons, New York, N.Y, USA
| | - Heather Seid
- Irving Institute for Clinical and Translational Research, Columbia University, New York, N.Y, USA
| | - Kyungyeon Kim
- Institute of Human Nutrition, Columbia University, New York, N.Y, USA
| | - Rajasekhar Ramakrishnan
- Center for Biomathematics, Department of Pediatrics, Columbia University Vagelos College of Physicians and Surgeons, New York, N.Y, USA
| | - Tiffany Thomas
- Department of Pathology and Cell Biology, Columbia University Vagelos College of Physicians and Surgeons, New York, N.Y, USA
| | - Nelsa Matienzo
- Department of Medicine, Columbia University Vagelos College of Physicians and Surgeons, New York, N.Y, USA
| | - Gissette Reyes-Soffer
- Department of Medicine, Columbia University Vagelos College of Physicians and Surgeons, New York, N.Y, USA.
| |
Collapse
|
64
|
Li JJ, Zhao SP, Zhao D, Lu GP, Peng DQ, Liu J, Chen ZY, Guo YL, Wu NQ, Yan SK, Wang ZW, Gao RL. 2023 Chinese guideline for lipid management. Front Pharmacol 2023; 14:1190934. [PMID: 37711173 PMCID: PMC10498001 DOI: 10.3389/fphar.2023.1190934] [Citation(s) in RCA: 44] [Impact Index Per Article: 22.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2023] [Accepted: 06/26/2023] [Indexed: 09/16/2023] Open
Abstract
Atherosclerotic cardiovascular disease (ASCVD) is the leading cause of death among urban and rural residents in China, and elevated low-density lipoprotein cholesterol (LDL-C) is a risk factor for ASCVD. Considering the increasing burden of ASCVD, lipid management is of the utmost importance. In recent years, research on blood lipids has made breakthroughs around the world, hence a revision of Chinese guideline for lipid management is imperative, especially since the target lipid levels in the general population vary in respect to the risk of ASCVD. The level of LDL-C, which can be regarded as appropriate in a population without frisk factors, can be considered abnormal in people at high risk of developing ASCVD. As a result, the "Guidelines for the prevention and treatment of dyslipidemia" were adapted into the "Chinese guideline for Lipid Management" (henceforth referred to as the new guidelines) by an Experts' committee after careful deliberation. The new guidelines still recommend LDL-C as the primary target for lipid control, with cardiovascular disease (CVD) risk stratification to determine its target value. These guidelines recommend that moderate intensity statin therapy in adjunct with a heart-healthy lifestyle, be used as an initial line of treatment, followed by cholesterol absorption inhibitors or/and proprotein convertase subtilisin/kexin type 9 (PCSK9) inhibitors, as necessary. The new guidelines provide guidance for lipid management across various age groups, from children to the elderly. The aim of these guidelines is to comprehensively improve the management of lipids and promote the prevention and treatment of ASCVD by guiding clinical practice.
Collapse
Affiliation(s)
- Jian-Jun Li
- National Center for Cardiovascular Diseases, FuWai Hospital, Chinese Academy of Medical Sciences, Beijing, China
| | - Shui-Ping Zhao
- The Second Xiangya Hospital of Central South University, Changsha, Hunan, China
| | - Dong Zhao
- Beijing Anzhen Hospital, Capital Medical University, Beijing, China
| | - Guo-Ping Lu
- Ruijin Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Dao-Quan Peng
- The Second Xiangya Hospital of Central South University, Changsha, Hunan, China
| | - Jing Liu
- Beijing Anzhen Hospital, Capital Medical University, Beijing, China
| | - Zhen-Yue Chen
- Ruijin Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Yuan-Lin Guo
- National Center for Cardiovascular Diseases, FuWai Hospital, Chinese Academy of Medical Sciences, Beijing, China
| | - Na-Qiong Wu
- National Center for Cardiovascular Diseases, FuWai Hospital, Chinese Academy of Medical Sciences, Beijing, China
| | - Sheng-Kai Yan
- Affiliated Hospital of Zunyi Medical University, School of Laboratory Medicine of Zunyi Medical University, Zunyi, Guizhou, China
| | - Zeng-Wu Wang
- National Center for Cardiovascular Diseases, FuWai Hospital, Chinese Academy of Medical Sciences, Beijing, China
| | - Run-Lin Gao
- National Center for Cardiovascular Diseases, FuWai Hospital, Chinese Academy of Medical Sciences, Beijing, China
| |
Collapse
|
65
|
Le VT, Knight S, Watrous JD, Najhawan M, Dao K, McCubrey RO, Bair TL, Horne BD, May HT, Muhlestein JB, Nelson JR, Carlquist JF, Knowlton KU, Jain M, Anderson JL. Higher docosahexaenoic acid levels lower the protective impact of eicosapentaenoic acid on long-term major cardiovascular events. Front Cardiovasc Med 2023; 10:1229130. [PMID: 37680562 PMCID: PMC10482040 DOI: 10.3389/fcvm.2023.1229130] [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: 05/25/2023] [Accepted: 07/26/2023] [Indexed: 09/09/2023] Open
Abstract
Introduction Long-chain omega-3 polyunsaturated fatty acids (OM3 PUFA) are commonly used for cardiovascular disease prevention. High-dose eicosapentaenoic acid (EPA) is reported to reduce major adverse cardiovascular events (MACE); however, a combined EPA and docosahexaenoic acid (DHA) supplementation has not been proven to do so. This study aimed to evaluate the potential interaction between EPA and DHA levels on long-term MACE. Methods We studied a cohort of 987 randomly selected subjects enrolled in the INSPIRE biobank registry who underwent coronary angiography. We used rapid throughput liquid chromatography-mass spectrometry to quantify the EPA and DHA plasma levels and examined their impact unadjusted, adjusted for one another, and fully adjusted for comorbidities, EPA + DHA, and the EPA/DHA ratio on long-term (10-year) MACE (all-cause death, myocardial infarction, stroke, heart failure hospitalization). Results The average subject age was 61.5 ± 12.2 years, 57% were male, 41% were obese, 42% had severe coronary artery disease (CAD), and 311 (31.5%) had a MACE. The 10-year MACE unadjusted hazard ratio (HR) for the highest (fourth) vs. lowest (first) quartile (Q) of EPA was HR = 0.48 (95% CI: 0.35, 0.67). The adjustment for DHA changed the HR to 0.30 (CI: 0.19, 0.49), and an additional adjustment for baseline differences changed the HR to 0.36 (CI: 0.22, 0.58). Conversely, unadjusted DHA did not significantly predict MACE, but adjustment for EPA resulted in a 1.81-fold higher risk of MACE (CI: 1.14, 2.90) for Q4 vs. Q1. However, after the adjustment for baseline differences, the risk of MACE was not significant for DHA (HR = 1.37; CI: 0.85, 2.20). An EPA/DHA ratio ≥1 resulted in a lower rate of 10-year MACE outcomes (27% vs. 37%, adjusted p-value = 0.013). Conclusions Higher levels of EPA, but not DHA, are associated with a lower risk of MACE. When combined with EPA, higher DHA blunts the benefit of EPA and is associated with a higher risk of MACE in the presence of low EPA. These findings can help explain the discrepant results of EPA-only and EPA/DHA mixed clinical supplementation trials.
Collapse
Affiliation(s)
- Viet T. Le
- Intermountain Medical Center, Intermountain Heart Institute, Salt Lake City, UT, United States
- Department of Physician Assistant Studies, Rocky Mountain University of Health Professions, Provo, UT, United States
| | - Stacey Knight
- Intermountain Medical Center, Intermountain Heart Institute, Salt Lake City, UT, United States
- The University of Utah, School of Medicine, Salt Lake City, UT, United States
| | - Jeramie D. Watrous
- Department of Medicine, University of California San Diego, San Diego, CA, United States
| | - Mahan Najhawan
- Department of Medicine, University of California San Diego, San Diego, CA, United States
| | - Khoi Dao
- Department of Medicine, University of California San Diego, San Diego, CA, United States
| | - Raymond O. McCubrey
- Intermountain Medical Center, Intermountain Heart Institute, Salt Lake City, UT, United States
| | - Tami L. Bair
- Intermountain Medical Center, Intermountain Heart Institute, Salt Lake City, UT, United States
| | - Benjamin D. Horne
- Intermountain Medical Center, Intermountain Heart Institute, Salt Lake City, UT, United States
- Division of Cardiovascular Medicine, Department of Medicine, Stanford University, Stanford, CA, United States
| | - Heidi T. May
- Intermountain Medical Center, Intermountain Heart Institute, Salt Lake City, UT, United States
| | - Joseph B. Muhlestein
- Intermountain Medical Center, Intermountain Heart Institute, Salt Lake City, UT, United States
- The University of Utah, School of Medicine, Salt Lake City, UT, United States
| | - John R. Nelson
- California Cardiovascular Institute, Fresno, CA, United States
| | - John F. Carlquist
- Intermountain Medical Center, Intermountain Heart Institute, Salt Lake City, UT, United States
- The University of Utah, School of Medicine, Salt Lake City, UT, United States
| | - Kirk U. Knowlton
- Intermountain Medical Center, Intermountain Heart Institute, Salt Lake City, UT, United States
- The University of Utah, School of Medicine, Salt Lake City, UT, United States
- Department of Medicine, University of California San Diego, San Diego, CA, United States
| | - Mohit Jain
- Department of Medicine, University of California San Diego, San Diego, CA, United States
| | - Jeffrey L. Anderson
- Intermountain Medical Center, Intermountain Heart Institute, Salt Lake City, UT, United States
- The University of Utah, School of Medicine, Salt Lake City, UT, United States
| |
Collapse
|
66
|
Abstract
PURPOSE In this review, the regulation, proposed hypolipidemic mechanism, and efficacy of common dietary supplements (DSs) marketed for cardiovascular health are discussed. RECENT FINDINGS Data demonstrate modest but inconsistent lipid-lowering effects with common DSs such as probiotics, soluble fibers, plant sterols, green tea, berberine, guggul, niacin, and garlic. Furthermore, data is limited regarding turmeric, hawthorn, and cinnamon. Red yeast rice has shown to be a beneficial DS, but its safety and efficacy are dependent upon its production quality and monacolin K content, respectively. Finally, soy proteins and omega-3 fatty acid-rich foods can have significant health benefits if used to displace other animal products as part of a healthier diet. Despite the rising use of DSs, data demonstrate unpredictable results. Patients should be educated on the difference between these DSs and evidence-based lipid-lowering medications proven to improve cardiovascular outcomes.
Collapse
Affiliation(s)
- Saeid Mirzai
- Department of Internal Medicine, Cleveland Clinic Foundation, Cleveland, OH, USA
| | - Luke J Laffin
- Section of Preventive Cardiology and Rehabilitation, Department of Cardiovascular Medicine, Cleveland Clinic Foundation, 9500 Euclid Avenue, Mail Code JB1, Cleveland, OH, 44195, USA.
| |
Collapse
|
67
|
Chong SY, Wang X, van Bloois L, Huang C, Syeda NS, Zhang S, Ting HJ, Nair V, Lin Y, Lou CKL, Benetti AA, Yu X, Lim NJY, Tan MS, Lim HY, Lim SY, Thiam CH, Looi WD, Zharkova O, Chew NWS, Ng CH, Bonney GK, Muthiah M, Chen X, Pastorin G, Richards AM, Angeli V, Storm G, Wang JW. Injectable liposomal docosahexaenoic acid alleviates atherosclerosis progression and enhances plaque stability. J Control Release 2023; 360:344-364. [PMID: 37406819 DOI: 10.1016/j.jconrel.2023.06.035] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2022] [Revised: 06/12/2023] [Accepted: 06/27/2023] [Indexed: 07/07/2023]
Abstract
Atherosclerosis is a chronic inflammatory vascular disease that is characterized by the accumulation of lipids and immune cells in plaques built up inside artery walls. Docosahexaenoic acid (DHA, 22:6n-3), an omega-3 polyunsaturated fatty acid (PUFA), which exerts anti-inflammatory and antioxidant properties, has long been purported to be of therapeutic benefit to atherosclerosis patients. However, large clinical trials have yielded inconsistent data, likely due to variations in the formulation, dosage, and bioavailability of DHA following oral intake. To fully exploit its potential therapeutic effects, we have developed an injectable liposomal DHA formulation intended for intravenous administration as a plaque-targeted nanomedicine. The liposomal formulation protects DHA against chemical degradation and increases its local concentration within atherosclerotic lesions. Mechanistically, DHA liposomes are readily phagocytosed by activated macrophages, exert potent anti-inflammatory and antioxidant effects, and inhibit foam cell formation. Upon intravenous administration, DHA liposomes accumulate preferentially in atherosclerotic lesional macrophages and promote polarization of macrophages towards an anti-inflammatory M2 phenotype, resulting in attenuation of atherosclerosis progression in both ApoE-/- and Ldlr-/- experimental models. Plaque composition analysis demonstrates that liposomal DHA inhibits macrophage infiltration, reduces lipid deposition, and increases collagen content, thus improving the stability of atherosclerotic plaques against rupture. Matrix-assisted laser desorption/ionization mass spectrometry imaging (MALDI-MSI) further reveals that DHA liposomes can partly restore the complex lipid profile of the plaques to that of early-stage plaques. In conclusion, DHA liposomes offer a promising approach for applying DHA to stabilize atherosclerotic plaques and attenuate atherosclerosis progression, thereby preventing atherosclerosis-related cardiovascular events.
Collapse
Affiliation(s)
- Suet Yen Chong
- Department of Surgery, Yong Loo Lin School of Medicine, National University of Singapore, 119228 Singapore, Singapore; Cardiovascular Research Institute, Yong Loo Lin School of Medicine, National University of Singapore, 117599 Singapore, Singapore; Nanomedicine Translational Research Programme, Centre for NanoMedicine, Yong Loo Lin School of Medicine, National University of Singapore, 117609 Singapore, Singapore
| | - Xiaoyuan Wang
- Department of Surgery, Yong Loo Lin School of Medicine, National University of Singapore, 119228 Singapore, Singapore; Cardiovascular Research Institute, Yong Loo Lin School of Medicine, National University of Singapore, 117599 Singapore, Singapore
| | - Louis van Bloois
- Department of Pharmaceutics, Faculty of Science, Utrecht University, 3584 CG Utrecht, the Netherlands
| | - Chenyuan Huang
- Department of Surgery, Yong Loo Lin School of Medicine, National University of Singapore, 119228 Singapore, Singapore; Cardiovascular Research Institute, Yong Loo Lin School of Medicine, National University of Singapore, 117599 Singapore, Singapore; Nanomedicine Translational Research Programme, Centre for NanoMedicine, Yong Loo Lin School of Medicine, National University of Singapore, 117609 Singapore, Singapore
| | - Nilofer Sayed Syeda
- Department of Surgery, Yong Loo Lin School of Medicine, National University of Singapore, 119228 Singapore, Singapore; Nanomedicine Translational Research Programme, Centre for NanoMedicine, Yong Loo Lin School of Medicine, National University of Singapore, 117609 Singapore, Singapore
| | - Sitong Zhang
- Department of Surgery, Yong Loo Lin School of Medicine, National University of Singapore, 119228 Singapore, Singapore; Nanomedicine Translational Research Programme, Centre for NanoMedicine, Yong Loo Lin School of Medicine, National University of Singapore, 117609 Singapore, Singapore
| | - Hui Jun Ting
- Department of Surgery, Yong Loo Lin School of Medicine, National University of Singapore, 119228 Singapore, Singapore; Cardiovascular Research Institute, Yong Loo Lin School of Medicine, National University of Singapore, 117599 Singapore, Singapore; Nanomedicine Translational Research Programme, Centre for NanoMedicine, Yong Loo Lin School of Medicine, National University of Singapore, 117609 Singapore, Singapore
| | - Vaarsha Nair
- Department of Surgery, Yong Loo Lin School of Medicine, National University of Singapore, 119228 Singapore, Singapore; Nanomedicine Translational Research Programme, Centre for NanoMedicine, Yong Loo Lin School of Medicine, National University of Singapore, 117609 Singapore, Singapore
| | - Yuanzhe Lin
- Department of Biomedical Engineering, National University of Singapore, 117583 Singapore, Singapore
| | - Charles Kang Liang Lou
- Department of Surgery, Yong Loo Lin School of Medicine, National University of Singapore, 119228 Singapore, Singapore; Nanomedicine Translational Research Programme, Centre for NanoMedicine, Yong Loo Lin School of Medicine, National University of Singapore, 117609 Singapore, Singapore
| | - Ayca Altay Benetti
- Department of Pharmacy, Faculty of Science, National University of Singapore, 117543 Singapore, Singapore
| | - Xiaodong Yu
- Department of Surgery, Yong Loo Lin School of Medicine, National University of Singapore, 119228 Singapore, Singapore; Nanomedicine Translational Research Programme, Centre for NanoMedicine, Yong Loo Lin School of Medicine, National University of Singapore, 117609 Singapore, Singapore
| | - Nicole Jia Ying Lim
- Yong Loo Lin School of Medicine, National University of Singapore, 117599 Singapore, Singapore
| | - Michelle Siying Tan
- Department of Surgery, Yong Loo Lin School of Medicine, National University of Singapore, 119228 Singapore, Singapore; Nanomedicine Translational Research Programme, Centre for NanoMedicine, Yong Loo Lin School of Medicine, National University of Singapore, 117609 Singapore, Singapore
| | - Hwee Ying Lim
- Immunology Translational Research Programme, Department of Microbiology and Immunology, Yong Loo Lin School of Medicine, National University of Singapore, 117456 Singapore, Singapore; Immunology Programme, Life Sciences Institute, National University of Singapore, 117456 Singapore, Singapore
| | - Sheau Yng Lim
- Immunology Translational Research Programme, Department of Microbiology and Immunology, Yong Loo Lin School of Medicine, National University of Singapore, 117456 Singapore, Singapore; Immunology Programme, Life Sciences Institute, National University of Singapore, 117456 Singapore, Singapore
| | - Chung Hwee Thiam
- Immunology Translational Research Programme, Department of Microbiology and Immunology, Yong Loo Lin School of Medicine, National University of Singapore, 117456 Singapore, Singapore; Immunology Programme, Life Sciences Institute, National University of Singapore, 117456 Singapore, Singapore
| | - Wen Donq Looi
- Bruker Daltonics, Bruker Singapore Pte. Ltd., 138671 Singapore, Singapore
| | - Olga Zharkova
- Department of Surgery, Yong Loo Lin School of Medicine, National University of Singapore, 119228 Singapore, Singapore; Cardiovascular Research Institute, Yong Loo Lin School of Medicine, National University of Singapore, 117599 Singapore, Singapore
| | - Nicholas W S Chew
- Yong Loo Lin School of Medicine, National University of Singapore, 117599 Singapore, Singapore; Department of Cardiology, National University Heart Centre, National University Hospital, 119074 Singapore, Singapore
| | - Cheng Han Ng
- Yong Loo Lin School of Medicine, National University of Singapore, 117599 Singapore, Singapore
| | - Glenn Kunnath Bonney
- Department of Surgery, Yong Loo Lin School of Medicine, National University of Singapore, 119228 Singapore, Singapore; Division of Hepatobiliary and Pancreatic Surgery, Department of Surgery, National University Hospital, 119074 Singapore, Singapore
| | - Mark Muthiah
- Yong Loo Lin School of Medicine, National University of Singapore, 117599 Singapore, Singapore; Division of Gastroenterology and Hepatology, Department of Medicine, National University Hospital, 119074 Singapore, Singapore; National University Centre for Organ Transplantation, National University Health System, 119074 Singapore, Singapore
| | - Xiaoyuan Chen
- Department of Surgery, Yong Loo Lin School of Medicine, National University of Singapore, 119228 Singapore, Singapore; Nanomedicine Translational Research Programme, Centre for NanoMedicine, Yong Loo Lin School of Medicine, National University of Singapore, 117609 Singapore, Singapore; Department of Diagnostic Radiology, Yong Loo Lin School of Medicine, National University of Singapore, 119074 Singapore, Singapore; Departments of Chemical and Biomolecular Engineering, and Biomedical Engineering, Faculty of Engineering, National University of Singapore, 117575 Singapore, Singapore; Clinical Imaging Research Centre, Centre for Translational Medicine, Yong Loo Lin School of Medicine, National University of Singapore, 117599 Singapore, Singapore
| | - Giorgia Pastorin
- Department of Pharmacy, Faculty of Science, National University of Singapore, 117543 Singapore, Singapore
| | - A Mark Richards
- Cardiovascular Research Institute, Yong Loo Lin School of Medicine, National University of Singapore, 117599 Singapore, Singapore
| | - Veronique Angeli
- Immunology Translational Research Programme, Department of Microbiology and Immunology, Yong Loo Lin School of Medicine, National University of Singapore, 117456 Singapore, Singapore; Immunology Programme, Life Sciences Institute, National University of Singapore, 117456 Singapore, Singapore
| | - Gert Storm
- Department of Surgery, Yong Loo Lin School of Medicine, National University of Singapore, 119228 Singapore, Singapore; Nanomedicine Translational Research Programme, Centre for NanoMedicine, Yong Loo Lin School of Medicine, National University of Singapore, 117609 Singapore, Singapore; Department of Pharmaceutics, Faculty of Science, Utrecht University, 3584 CG Utrecht, the Netherlands; Department of Biomaterials, Science and Technology, Faculty of Science and Technology, University of Twente, 7522 NB Enschede, the Netherlands.
| | - Jiong-Wei Wang
- Department of Surgery, Yong Loo Lin School of Medicine, National University of Singapore, 119228 Singapore, Singapore; Cardiovascular Research Institute, Yong Loo Lin School of Medicine, National University of Singapore, 117599 Singapore, Singapore; Nanomedicine Translational Research Programme, Centre for NanoMedicine, Yong Loo Lin School of Medicine, National University of Singapore, 117609 Singapore, Singapore; Department of Physiology, National University of Singapore, 117593 Singapore, Singapore.
| |
Collapse
|
68
|
Nshimiyimana R, Libreros S, Simard M, Chiang N, Rodriguez AR, Spur BW, Haeggström JZ, Serhan CN. Stereochemistry and functions of the new cysteinyl-resolvin, 4S,5R-RCTR1, in efferocytosis and erythrophagocytosis of human senescent erythrocytes. Am J Hematol 2023; 98:1000-1016. [PMID: 37139907 PMCID: PMC10429686 DOI: 10.1002/ajh.26932] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2023] [Revised: 04/03/2023] [Accepted: 04/05/2023] [Indexed: 05/05/2023]
Abstract
Specialized pro-resolving lipid mediators play key functions in the resolution of the acute inflammatory response. Herein, we elucidate the stereochemical structure of the new 4S,5R-RCTR1, a cysteinyl-resolvin, recently uncovered in human leukocytes incubated with a 4S,5S-epoxy-resolvin intermediate, using liquid chromatography-tandem mass spectrometry (LC-MS/MS) and ultra-violet (UV) spectrophotometry. With this approach, the physical properties of the new mediator prepared by total organic synthesis were matched to enzymatically produced biogenic material. In addition, we confirmed the potent biological actions of 4S,5R-RCTR1 with human M2-like macrophage phagocytosis of live bacteria, efferocytosis of apoptotic neutrophils, and erythrophagocytosis of senescent human red blood cells in a concentration-dependent manner from 0.1 to 10 nM. Taken together, these results establish the complete stereochemistry of 4S,5R-RCTR1 as 5R-glutathionyl-4S,17S-dihydroxy-6E,8E,10Z,13Z,15E,19Z-docosahexaenoic acid and give evidence of its novel bioactivities in human phagocyte responses. Moreover, they confirm and extend the stereoselective functions of the 4S,5R-RCTR1 with isolated human phagocytes of interest in the resolution of inflammation.
Collapse
Affiliation(s)
- Robert Nshimiyimana
- Center for Experimental Therapeutics and Reperfusion Injury, Department of Anesthesiology, Perioperative and Pain Medicine, Brigham and Women’s Hospital and Harvard Medical School, Boston, Massachusetts 02115, USA
| | - Stephania Libreros
- Center for Experimental Therapeutics and Reperfusion Injury, Department of Anesthesiology, Perioperative and Pain Medicine, Brigham and Women’s Hospital and Harvard Medical School, Boston, Massachusetts 02115, USA
| | - Mélissa Simard
- Center for Experimental Therapeutics and Reperfusion Injury, Department of Anesthesiology, Perioperative and Pain Medicine, Brigham and Women’s Hospital and Harvard Medical School, Boston, Massachusetts 02115, USA
| | - Nan Chiang
- Center for Experimental Therapeutics and Reperfusion Injury, Department of Anesthesiology, Perioperative and Pain Medicine, Brigham and Women’s Hospital and Harvard Medical School, Boston, Massachusetts 02115, USA
| | - Ana R. Rodriguez
- Department of Cell Biology, Rowan University–School of Medicine, Stratford, New Jersey 08084, USA
| | - Bernd W. Spur
- Department of Cell Biology, Rowan University–School of Medicine, Stratford, New Jersey 08084, USA
| | - Jesper Z. Haeggström
- Division of Physiological Chemistry II, Department of Medical Biochemistry and Biophysics, Karolinska institute, S-171 77 Stockholm, Sweden
| | - Charles N. Serhan
- Center for Experimental Therapeutics and Reperfusion Injury, Department of Anesthesiology, Perioperative and Pain Medicine, Brigham and Women’s Hospital and Harvard Medical School, Boston, Massachusetts 02115, USA
| |
Collapse
|
69
|
Bae JH, Lim H, Lim S. The Potential Cardiometabolic Effects of Long-Chain ω-3 Polyunsaturated Fatty Acids: Recent Updates and Controversies. Adv Nutr 2023; 14:612-628. [PMID: 37031750 PMCID: PMC10334139 DOI: 10.1016/j.advnut.2023.03.014] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2022] [Revised: 03/09/2023] [Accepted: 03/30/2023] [Indexed: 04/11/2023] Open
Abstract
Various health-related effects of long-chain (LC) ω-3 PUFAs, EPA, and DHA have been suggested. LC ω-3 PUFAs reduce TG concentrations and have anti-inflammatory, immunomodulatory, antiplatelet, and vascular protective effects. Controversially, they might help in restoring glucose homeostasis via the gut microbiota. However, previous studies have not shown the clear benefits of LC ω-3 PUFAs for CVDs. REDUCE-IT and STRENGTH-representative randomized controlled trials (RCTs) that examined whether LC ω-3 PUFAs would prevent major adverse cardiovascular (CV) events (MACE)-showed conflicting results with differences in the types, doses, or comparators of LC ω-3 PUFAs and study populations. Therefore, we performed a meta-analysis using major RCTs to address this inconsistency and assess the clinical and biological effects of LC ω-3 PUFAs. We included RCTs that involved ≥500 participants with ≥1 y follow-up. Of 17 studies involving 143,410 people, LC ω-3 PUFA supplementation showed beneficial effects on CV death (RR: 0.94; 95% CI: 0.88, 0.99; P = 0.029) and fatal or nonfatal MI (RR: 0.83; 95% CI: 0.72, 0.95; P = 0.010). RCTs on EPA alone showed better results for 3-point MACE, CV death, and fatal or nonfatal MI. However, the benefits were not found for fatal or nonfatal stroke, all-cause mortality, and hospitalization for heart failure. Of note, studies of both the EPA/DHA combination and EPA alone showed a significant increase in risk of new-onset atrial fibrillation. Thus, well-designed studies are needed to investigate the underlying mechanisms involved in the distinct effects of EPA compared with DHA on cardiometabolic diseases. This review discusses the potential benefits and safety of LC ω-3 PUFAs from a cardiometabolic perspective focusing on recent updates and controversies.
Collapse
Affiliation(s)
- Jae Hyun Bae
- Department of Internal Medicine, Korea University Anam Hospital, Korea University College of Medicine, Seoul, Republic of Korea
| | - Hyunjung Lim
- Department of Medical Nutrition, Research Institute of Medical Nutrition, Graduate School of East-West Medical Science, Kyung Hee University, Yongin, Republic of Korea
| | - Soo Lim
- Department of Internal Medicine, Seoul National University Bundang Hospital, Seoul National University College of Medicine, Seongnam, Republic of Korea.
| |
Collapse
|
70
|
Kirkpatrick CF, Sikand G, Petersen KS, Anderson CAM, Aspry KE, Bolick JP, Kris-Etherton PM, Maki KC. Nutrition interventions for adults with dyslipidemia: A Clinical Perspective from the National Lipid Association. J Clin Lipidol 2023; 17:428-451. [PMID: 37271600 DOI: 10.1016/j.jacl.2023.05.099] [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: 05/18/2023] [Accepted: 05/19/2023] [Indexed: 06/06/2023]
Abstract
Lifestyle habits can have a profound impact on atherosclerotic cardiovascular disease (ASCVD) risk. The National Lipid Association previously published recommendations for lifestyle therapies to manage dyslipidemia. This Clinical Perspective provides an update with a focus on nutrition interventions for the three most common dyslipidemias in adults: 1) low-density lipoprotein cholesterol (LDL-C) elevation; 2) triglyceride (TG) elevation, including severe hypertriglyceridemia with chylomicronemia; and 3) combined dyslipidemia, with elevations in both LDL-C and TG levels. Lowering LDL-C and non-high-density lipoprotein cholesterol are the primary objectives for reducing ASCVD risk. With severe TG elevation (≥500 mg/dL), the primary objective is to prevent pancreatitis and ASCVD risk reduction is secondary. Nutrition interventions that lower LDL-C levels include reducing cholesterol-raising fatty acids and dietary cholesterol, as well as increasing intakes of unsaturated fatty acids, plant proteins, viscous fibers, and reducing adiposity for patients with overweight or obesity. Selected dietary supplements may be employed as dietary adjuncts. Nutrition interventions for all patients with elevated TG levels include restricting intakes of alcohol, added sugars, and refined starches. Additional lifestyle factors that reduce TG levels are participating in daily physical activity and reducing adiposity in patients with overweight or obesity. For patients with severe hypertriglyceridemia, an individualized approach is essential. Nutrition interventions for addressing concurrent elevations in LDL-C and TG include a combination of the strategies described for lowering LDL-C and TG. A multidisciplinary approach is recommended to facilitate success in making and sustaining dietary changes and the assistance of a registered dietitian nutritionist is highly recommended.
Collapse
Affiliation(s)
- Carol F Kirkpatrick
- Midwest Biomedical Research, Addison, IL, USA; Kasiska Division of Health Sciences, Idaho State University, Pocatello, ID, USA
| | - Geeta Sikand
- University of California Irvine Heart Disease Prevention Program, Irvine, CA, USA
| | | | - Cheryl A M Anderson
- Herbert Wertheim School of Public Health and Human Longevity Science, University of California San Diego, La Jolla, CA, USA
| | - Karen E Aspry
- Lifespan Cardiovascular Institute, and Alpert Medical School, Brown University, Providence, RI, USA
| | | | - Penny M Kris-Etherton
- Department of Nutritional Sciences, The Pennsylvania State University, University Park, PA, USA
| | - Kevin C Maki
- Midwest Biomedical Research, Addison, IL, USA; Indiana University School of Public Health-Bloomington, Bloomington, IN, USA.
| |
Collapse
|
71
|
Gora AH, Rehman S, Dias J, Fernandes JMO, Olsvik PA, Sørensen M, Kiron V. Protective mechanisms of a microbial oil against hypercholesterolemia: evidence from a zebrafish model. Front Nutr 2023; 10:1161119. [PMID: 37435570 PMCID: PMC10332275 DOI: 10.3389/fnut.2023.1161119] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2023] [Accepted: 04/20/2023] [Indexed: 07/13/2023] Open
Abstract
A Western diet elevates the circulating lipoprotein and triglyceride levels which are the major risk factors in cardiovascular disease (CVD) development. Consumption of long-chain omega-3 fatty acids can stall the disease progression. Although these fatty acids can significantly impact the intestine under a hypercholesterolemic condition, the associated changes have not been studied in detail. Therefore, we investigated the alterations in the intestinal transcriptome along with the deviations in the plasma lipids and liver histomorphology of zebrafish offered DHA- and EPA-rich oil. Fish were allocated to 4 dietary treatments: a control group, a high cholesterol group and microbial oil groups with low (3.3%) and high (6.6%) inclusion levels. We quantified the total cholesterol, lipoprotein and triglyceride levels in the plasma. In addition, we assessed the liver histology, intestinal transcriptome and plasma lipidomic profiles of the study groups. The results suggested that higher levels of dietary microbial oil could control the CVD risk factor indices in zebrafish plasma. Furthermore, microbial oil-fed fish had fewer liver vacuoles and higher mRNA levels of genes involved in β-oxidation and HDL maturation. Analyses of the intestine transcriptome revealed that microbial oil supplementation could influence the expression of genes altered by a hypercholesterolemic diet. The plasma lipidomic profiles revealed that the higher level of microbial oil tested could elevate the long-chain poly-unsaturated fatty acid content of triglyceride species and lower the concentration of several lysophosphatidylcholine and diacylglycerol molecules. Our study provides insights into the effectiveness of microbial oil against dyslipidemia in zebrafish.
Collapse
Affiliation(s)
- Adnan H. Gora
- Faculty of Biosciences and Aquaculture, Nord University, Bodø, Norway
| | - Saima Rehman
- Faculty of Biosciences and Aquaculture, Nord University, Bodø, Norway
| | | | | | - Pål A. Olsvik
- Faculty of Biosciences and Aquaculture, Nord University, Bodø, Norway
| | - Mette Sørensen
- Faculty of Biosciences and Aquaculture, Nord University, Bodø, Norway
| | - Viswanath Kiron
- Faculty of Biosciences and Aquaculture, Nord University, Bodø, Norway
| |
Collapse
|
72
|
Wang Z, Zhang X, Qu Y, Zhang S, Chen Y, Chen X, Qi X, Liu P, Liu S, Jiang S, Man R, He L, Wu L, Li Z, Shang Y, Qiu Z, Liu F, Xu C, Lai C, Ge J. Icosapent ethyl therapy for very high triglyceride levels: a 12-week, multi-center, placebo-controlled, randomized, double-blinded, phase III clinical trial in China. Lipids Health Dis 2023; 22:71. [PMID: 37301827 DOI: 10.1186/s12944-023-01838-8] [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: 01/06/2023] [Accepted: 05/29/2023] [Indexed: 06/12/2023] Open
Abstract
OBJECTIVES Eicosapentaenoic acid in its ethyl ester form is the single active component of icosapent ethyl (IPE). This study was a phase III, multi-center trial assessing the safety and efficiency of IPE for treating very high triglyceride (TG) in a Chinese cohort. METHODS Patients having TG levels (5.6-22.6 mmol/L) were enrolled and randomly assigned to receive a treatment of oral intake of 4 g or 2 g/day of IPE, or placebo. Before and after 12 weeks of treatment, TG levels were assessed and the median was calculated to determine the change between the baseline and week 12. In addition to examining TG levels, the impact of such treatments on other lipid changes was also investigated. The official Drug Clinical Trial Information Management Platform has registered this study (CTR20170362). RESULTS Random assignments were performed on 373 patients (mean age 48.9 years; 75.1% male). IPE (4 g/day) lowered TG levels by an average of 28.4% from baseline and by an average of 19.9% after correction for placebo (95% CI: 29.8%-10.0%, P < 0.001). In addition, plasma concentration of non-high-density lipoprotein cholesterol (non-HDL-C), very low-density lipoprotein (VLDL) cholesterol, and VLDL-TG remarkedly reduced after IPE (4 g/day) treatment by a median of 14.6%, 27.9%, and 25.2%, respectively compared with participants in placebo group. Compared to the placebo, neither 4 nor 2 g of IPE daily elevated LDL-C levels with statistical significance. IPE was well tolerated by all the treatment groups. CONCLUSIONS IPE at 4 g/day dramatically lowered other atherogenic lipids without a noticeable increase in LDL-C, thereby decreasing TG levels in an exceptionally high-TG Chinese population.
Collapse
Affiliation(s)
- Zhen Wang
- Department of Cardiology, Shanghai Institute of Cardiovascular Diseases, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Xin Zhang
- Department of Cardiac Function, The First Affiliated Hospital of Baotou Medical College, Inner Mongolia University of Science and Technology, Baotou, Inner Mongolia, China
| | - Yanling Qu
- Department of Cardiology, Yuncheng Central Hospital, Yuncheng, Shanxi, China
| | - Shuyang Zhang
- Department of Cardiology, Peking Union Medical College Hospital, Peking Union Medical College & Chinese Academy of Medical Sciences, Beijing, China
| | - Yundai Chen
- Department of Cardiology, Chinese PLA General Hospital, Beijing, China
| | - Xiaoping Chen
- Department of Cardiology, West China Hospital, Sichuan University, Chengdu, China
| | - Xin Qi
- Department of Cardiology, Tianjin Union Medical Center, Tianjin, China
| | - Peijing Liu
- Department of Cardiology, Affiliated Hospital of Jiangsu University, Zhenjiang, Jiangsu, China
| | - Shuqin Liu
- Department of Cardiology, Liaocheng People's Hospital and Liaocheng Clinical School of Taishan Medical University, Liaocheng, Shandong, China
| | - Shan Jiang
- Department of Cardiology, Jinzhou Central Hospital, Jinzhou, Liaoning, China
| | - Ronghai Man
- Department of Cardiology, Chifeng Municipal Hospital, Chifeng, Inner Mongolia, China
| | - Liping He
- Department of Cardiology, Inner Mongolia People's Hospital, Huhhot, China
| | - Ling Wu
- Department of Cardiology, General Hospital of Ningxia Medical University, Yinchuan, China
| | - Zhanquan Li
- Department of Cardiology, Liaoning Provincial People's Hospital, Shenyang, China
| | - Yijun Shang
- Department of Cardiology, Jilin Central General Hospital, Jilin, China
| | - Zhaohui Qiu
- Department of Cardiology, Shanghai Tongren Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Feng Liu
- Departments of Cardiology, Suzhou Kowloon Hospital, School of Medicine Shanghai Jiaotong University, Suzhou, Jiangsu, China
| | - Chenhong Xu
- Department of Cardiology, Jingzhou Central Hospital, Jingzhou, Hubei, China
| | - Chunlin Lai
- Department of Cardiology, Shanxi Provincial People's Hospital, Taiyuan, China
| | - Junbo Ge
- Department of Cardiology, Shanghai Institute of Cardiovascular Diseases, Zhongshan Hospital, Fudan University, Shanghai, China.
| |
Collapse
|
73
|
Hu C, Zhou Y, Wu X, Jia X, Zhu Y, Zheng R, Wang S, Lin L, Qi H, Lin H, Li M, Wang T, Zhao Z, Xu M, Xu Y, Chen Y, Ning G, Borges MC, Wang W, Zheng J, Bi Y, Lu J. Evaluating the distinct pleiotropic effects of omega-3 fatty acids on type 2 diabetes mellitus: a mendelian randomization study. J Transl Med 2023; 21:370. [PMID: 37286992 DOI: 10.1186/s12967-023-04202-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2023] [Accepted: 05/14/2023] [Indexed: 06/09/2023] Open
Abstract
BACKGROUND Observational studies and conventional Mendelian randomization (MR) studies showed inconclusive evidence to support the association between omega-3 fatty acids and type 2 diabetes. We aim to evaluate the causal effect of omega-3 fatty acids on type 2 diabetes mellitus (T2DM), and the distinct intermediate phenotypes linking the two. METHODS Two-sample MR was performed using genetic instruments derived from a recent genome-wide association study (GWAS) of omega-3 fatty acids (N = 114,999) from UK Biobank and outcome data obtained from a large-scale T2DM GWAS (62,892 cases and 596,424 controls) in European ancestry. MR-Clust was applied to determine clustered genetic instruments of omega-3 fatty acids that influences T2DM. Two-step MR analysis was used to identify potential intermediate phenotypes (e.g. glycemic traits) that linking omega-3 fatty acids with T2DM. RESULTS Univariate MR showed heterogenous effect of omega-3 fatty acids on T2DM. At least two pleiotropic effects between omega-3 fatty acids and T2DM were identified using MR-Clust. For cluster 1 with seven instruments, increasing omega-3 fatty acids reduced T2DM risk (OR: 0.52, 95%CI 0.45-0.59), and decreased HOMA-IR (β = - 0.13, SE = 0.05, P = 0.02). On the contrary, MR analysis using 10 instruments in cluster 2 showed that increasing omega-3 fatty acids increased T2DM risk (OR:1.10; 95%CI 1.06-1.15), and decreased HOMA-B (β = - 0.04, SE = 0.01, P = 4.52 × 10-5). Two-step MR indicated that increasing omega-3 fatty acid levels decreased T2DM risk via decreasing HOMA-IR in cluster 1, while increased T2DM risk via decreasing HOMA-B in cluster 2. CONCLUSIONS This study provides evidence to support two distinct pleiotropic effects of omega-3 fatty acids on T2DM risk influenced by different gene clusters, which could be partially explained by distinct effects of omega-3 fatty acids on insulin resistance and beta cell dysfunction. The pleiotropic feature of omega-3 fatty acids variants and its complex relationships with T2DM need to be carefully considered in future genetic and clinical studies.
Collapse
Affiliation(s)
- Chunyan Hu
- Department of Endocrine and Metabolic Diseases, Shanghai Institute of Endocrine and Metabolic Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, 197 Rui Jin 2nd Road, Shanghai, 200025, China
- Shanghai National Clinical Research Center for Metabolic Diseases, Key Laboratory for Endocrine and Metabolic Diseases of the National Health Commission of the PR China, Shanghai Key Laboratory for Endocrine Tumor, State Key Laboratory of Medical Genomics, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yulin Zhou
- Department of Endocrine and Metabolic Diseases, Shanghai Institute of Endocrine and Metabolic Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, 197 Rui Jin 2nd Road, Shanghai, 200025, China
- Shanghai National Clinical Research Center for Metabolic Diseases, Key Laboratory for Endocrine and Metabolic Diseases of the National Health Commission of the PR China, Shanghai Key Laboratory for Endocrine Tumor, State Key Laboratory of Medical Genomics, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Xueyan Wu
- Department of Endocrine and Metabolic Diseases, Shanghai Institute of Endocrine and Metabolic Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, 197 Rui Jin 2nd Road, Shanghai, 200025, China
- Shanghai National Clinical Research Center for Metabolic Diseases, Key Laboratory for Endocrine and Metabolic Diseases of the National Health Commission of the PR China, Shanghai Key Laboratory for Endocrine Tumor, State Key Laboratory of Medical Genomics, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Xiaojing Jia
- Department of Endocrine and Metabolic Diseases, Shanghai Institute of Endocrine and Metabolic Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, 197 Rui Jin 2nd Road, Shanghai, 200025, China
- Shanghai National Clinical Research Center for Metabolic Diseases, Key Laboratory for Endocrine and Metabolic Diseases of the National Health Commission of the PR China, Shanghai Key Laboratory for Endocrine Tumor, State Key Laboratory of Medical Genomics, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yuanyue Zhu
- Department of Endocrine and Metabolic Diseases, Shanghai Institute of Endocrine and Metabolic Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, 197 Rui Jin 2nd Road, Shanghai, 200025, China
- Shanghai National Clinical Research Center for Metabolic Diseases, Key Laboratory for Endocrine and Metabolic Diseases of the National Health Commission of the PR China, Shanghai Key Laboratory for Endocrine Tumor, State Key Laboratory of Medical Genomics, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Ruizhi Zheng
- Department of Endocrine and Metabolic Diseases, Shanghai Institute of Endocrine and Metabolic Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, 197 Rui Jin 2nd Road, Shanghai, 200025, China
- Shanghai National Clinical Research Center for Metabolic Diseases, Key Laboratory for Endocrine and Metabolic Diseases of the National Health Commission of the PR China, Shanghai Key Laboratory for Endocrine Tumor, State Key Laboratory of Medical Genomics, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Shuangyuan Wang
- Department of Endocrine and Metabolic Diseases, Shanghai Institute of Endocrine and Metabolic Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, 197 Rui Jin 2nd Road, Shanghai, 200025, China
- Shanghai National Clinical Research Center for Metabolic Diseases, Key Laboratory for Endocrine and Metabolic Diseases of the National Health Commission of the PR China, Shanghai Key Laboratory for Endocrine Tumor, State Key Laboratory of Medical Genomics, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Lin Lin
- Department of Endocrine and Metabolic Diseases, Shanghai Institute of Endocrine and Metabolic Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, 197 Rui Jin 2nd Road, Shanghai, 200025, China
- Shanghai National Clinical Research Center for Metabolic Diseases, Key Laboratory for Endocrine and Metabolic Diseases of the National Health Commission of the PR China, Shanghai Key Laboratory for Endocrine Tumor, State Key Laboratory of Medical Genomics, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Hongyan Qi
- Department of Endocrine and Metabolic Diseases, Shanghai Institute of Endocrine and Metabolic Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, 197 Rui Jin 2nd Road, Shanghai, 200025, China
- Shanghai National Clinical Research Center for Metabolic Diseases, Key Laboratory for Endocrine and Metabolic Diseases of the National Health Commission of the PR China, Shanghai Key Laboratory for Endocrine Tumor, State Key Laboratory of Medical Genomics, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Hong Lin
- Department of Endocrine and Metabolic Diseases, Shanghai Institute of Endocrine and Metabolic Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, 197 Rui Jin 2nd Road, Shanghai, 200025, China
- Shanghai National Clinical Research Center for Metabolic Diseases, Key Laboratory for Endocrine and Metabolic Diseases of the National Health Commission of the PR China, Shanghai Key Laboratory for Endocrine Tumor, State Key Laboratory of Medical Genomics, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Mian Li
- Department of Endocrine and Metabolic Diseases, Shanghai Institute of Endocrine and Metabolic Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, 197 Rui Jin 2nd Road, Shanghai, 200025, China
- Shanghai National Clinical Research Center for Metabolic Diseases, Key Laboratory for Endocrine and Metabolic Diseases of the National Health Commission of the PR China, Shanghai Key Laboratory for Endocrine Tumor, State Key Laboratory of Medical Genomics, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Tiange Wang
- Department of Endocrine and Metabolic Diseases, Shanghai Institute of Endocrine and Metabolic Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, 197 Rui Jin 2nd Road, Shanghai, 200025, China
- Shanghai National Clinical Research Center for Metabolic Diseases, Key Laboratory for Endocrine and Metabolic Diseases of the National Health Commission of the PR China, Shanghai Key Laboratory for Endocrine Tumor, State Key Laboratory of Medical Genomics, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Zhiyun Zhao
- Department of Endocrine and Metabolic Diseases, Shanghai Institute of Endocrine and Metabolic Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, 197 Rui Jin 2nd Road, Shanghai, 200025, China
- Shanghai National Clinical Research Center for Metabolic Diseases, Key Laboratory for Endocrine and Metabolic Diseases of the National Health Commission of the PR China, Shanghai Key Laboratory for Endocrine Tumor, State Key Laboratory of Medical Genomics, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Min Xu
- Department of Endocrine and Metabolic Diseases, Shanghai Institute of Endocrine and Metabolic Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, 197 Rui Jin 2nd Road, Shanghai, 200025, China
- Shanghai National Clinical Research Center for Metabolic Diseases, Key Laboratory for Endocrine and Metabolic Diseases of the National Health Commission of the PR China, Shanghai Key Laboratory for Endocrine Tumor, State Key Laboratory of Medical Genomics, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yu Xu
- Department of Endocrine and Metabolic Diseases, Shanghai Institute of Endocrine and Metabolic Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, 197 Rui Jin 2nd Road, Shanghai, 200025, China
- Shanghai National Clinical Research Center for Metabolic Diseases, Key Laboratory for Endocrine and Metabolic Diseases of the National Health Commission of the PR China, Shanghai Key Laboratory for Endocrine Tumor, State Key Laboratory of Medical Genomics, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yuhong Chen
- Department of Endocrine and Metabolic Diseases, Shanghai Institute of Endocrine and Metabolic Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, 197 Rui Jin 2nd Road, Shanghai, 200025, China
- Shanghai National Clinical Research Center for Metabolic Diseases, Key Laboratory for Endocrine and Metabolic Diseases of the National Health Commission of the PR China, Shanghai Key Laboratory for Endocrine Tumor, State Key Laboratory of Medical Genomics, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Guang Ning
- Department of Endocrine and Metabolic Diseases, Shanghai Institute of Endocrine and Metabolic Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, 197 Rui Jin 2nd Road, Shanghai, 200025, China
- Shanghai National Clinical Research Center for Metabolic Diseases, Key Laboratory for Endocrine and Metabolic Diseases of the National Health Commission of the PR China, Shanghai Key Laboratory for Endocrine Tumor, State Key Laboratory of Medical Genomics, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Maria-Carolina Borges
- MRC Integrative Epidemiology Unit (IEU), Bristol Medical School, University of Bristol, Oakfield House, Oakfield Grove, Bristol, BS8 2BN, UK
- Population Health Sciences, Bristol Medical School, University of Bristol, Bristol, UK
| | - Weiqing Wang
- Department of Endocrine and Metabolic Diseases, Shanghai Institute of Endocrine and Metabolic Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, 197 Rui Jin 2nd Road, Shanghai, 200025, China.
- Shanghai National Clinical Research Center for Metabolic Diseases, Key Laboratory for Endocrine and Metabolic Diseases of the National Health Commission of the PR China, Shanghai Key Laboratory for Endocrine Tumor, State Key Laboratory of Medical Genomics, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China.
| | - Jie Zheng
- Department of Endocrine and Metabolic Diseases, Shanghai Institute of Endocrine and Metabolic Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, 197 Rui Jin 2nd Road, Shanghai, 200025, China.
- Shanghai National Clinical Research Center for Metabolic Diseases, Key Laboratory for Endocrine and Metabolic Diseases of the National Health Commission of the PR China, Shanghai Key Laboratory for Endocrine Tumor, State Key Laboratory of Medical Genomics, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China.
- MRC Integrative Epidemiology Unit (IEU), Bristol Medical School, University of Bristol, Oakfield House, Oakfield Grove, Bristol, BS8 2BN, UK.
| | - Yufang Bi
- Department of Endocrine and Metabolic Diseases, Shanghai Institute of Endocrine and Metabolic Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, 197 Rui Jin 2nd Road, Shanghai, 200025, China.
- Shanghai National Clinical Research Center for Metabolic Diseases, Key Laboratory for Endocrine and Metabolic Diseases of the National Health Commission of the PR China, Shanghai Key Laboratory for Endocrine Tumor, State Key Laboratory of Medical Genomics, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China.
| | - Jieli Lu
- Department of Endocrine and Metabolic Diseases, Shanghai Institute of Endocrine and Metabolic Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, 197 Rui Jin 2nd Road, Shanghai, 200025, China.
- Shanghai National Clinical Research Center for Metabolic Diseases, Key Laboratory for Endocrine and Metabolic Diseases of the National Health Commission of the PR China, Shanghai Key Laboratory for Endocrine Tumor, State Key Laboratory of Medical Genomics, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China.
| |
Collapse
|
74
|
Laranjeira FS, Neves NM, Raimundo A, Horta AB. Severe Hypertriglyceridemia: A 10-Year Review in a Portuguese Hospital. Cureus 2023; 15:e41239. [PMID: 37529514 PMCID: PMC10387820 DOI: 10.7759/cureus.41239] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/30/2023] [Indexed: 08/03/2023] Open
Abstract
INTRODUCTION Severe hypertriglyceridemia (SHTG) is a rare condition associated with serious complications, such as acute pancreatitis (AP), and the best treatment is still a matter of discussion. The aim of this study is to outline the demographics, management, and outcomes (recurrence and mortality) of complications in patients with SHTG. MATERIAL AND METHODS A retrospective, observational, and analytical study was carried out by obtaining clinical data from the electronic health records of patients with SHTG admitted to the Internal and Intensive Medicine units from the 1st of January 2009 to the 31st of December 2020 in a university hospital. RESULTS The cohort included 17 patients. The most common complication was AP (13/17 = 76.5%). Admission to the intensive care unit (ICU) was observed in 84.2%. Among patients with AP, the most commonly administered therapies were insulin (82.4%) and fibrates (76.5%). Plasmapheresis was used in 58.8%, and the criteria for using this technique were mainly based on clinical and laboratory abnormalities. There were no deaths. The readmission rate at 30 days was 36.3%. CONCLUSION This study shows the morbidity profile associated with SHTG, with a high level of ICU admissions and also a high level of the use of plasmapheresis. In our population, this approach had good results, and this should be highlighted as there are no clear international guidelines for this intervention. Distinguishing between patients with familial chylomicronemia syndrome or with multifactorial chylomicronemia is important as recent specific therapy for lipoprotein lipase (LPL) genetic deficit is available. In the near future, the performance of a genetic study should be considered in patients with SHTG as an attempt to avoid the high recurrence rate of complications of this disease.
Collapse
Affiliation(s)
| | - Nuno M Neves
- Internal Medicine Department, Hospital da Luz Lisboa, Lisboa, PRT
| | - Anabela Raimundo
- Internal Medicine Department, Hospital da Luz Lisboa, Lisboa, PRT
| | | |
Collapse
|
75
|
Gupta A, Alkhalil M. The Emerging Role of Icosapent Ethyl in Patients with Cardiovascular Disease: Mechanistic Insights and Future Applications. J Clin Med 2023; 12:3758. [PMID: 37297952 PMCID: PMC10253987 DOI: 10.3390/jcm12113758] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2023] [Revised: 05/21/2023] [Accepted: 05/23/2023] [Indexed: 06/12/2023] Open
Abstract
Omega-3 polyunsaturated fatty acids (PUFAs) were early established as therapeutic option for patients with high triglyceride levels. Their effects on lipoprotein particles, including a reduction in very low-density lipoprotein and a shift from small to large low-density lipoprotein, is increasingly recognised. This is coupled with their ability to be incorporated within the cellular membrane, leading to plaque stability and anti-inflammatory effects. Nonetheless, recent clinical trials have not been consistent in demonstrating the potential cardioprotective effects of omega-3 fatty acids. This is despite the circumstantial evidence from imaging studies illustrating the stabilising effects on atherosclerotic plaques and slowing of plaque progression. In this article, we will review the effects of omega-3 fatty acids, eicosapentaenoic acid (EPA) and docosahexaenoic acid (DHA), on lipid biomarkers, atherosclerotic plaque features, and clinical outcome studies and provide a mechanistic role in managing residual risk of atherosclerosis. This will provide better insight into the inconsistency of the recently reported clinical outcome studies.
Collapse
Affiliation(s)
- Ashish Gupta
- Cardiothoracic Centre, Freeman Hospital, Newcastle upon Tyne NE7 7DN, UK;
| | - Mohammad Alkhalil
- Cardiothoracic Centre, Freeman Hospital, Newcastle upon Tyne NE7 7DN, UK;
- Translational and Clinical Research Institute, Newcastle University, Newcastle upon Tyne NE1 7RU, UK
- Department of Cardiothoracic Services, Freeman Hospital, Freeman Road, Newcastle upon Tyne NE7 7DN, UK
| |
Collapse
|
76
|
Liu J, Meng Q, Zheng L, Yu P, Hu H, Zhuang R, Ge X, Liu Z, Liang X, Zhou X. Effect of n-3 PUFA on left ventricular remodelling in chronic heart failure: a systematic review and meta-analysis. Br J Nutr 2023; 129:1500-1509. [PMID: 35241186 DOI: 10.1017/s0007114521004979] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
Accumulating evidence suggests that supplementation of n-3 PUFA was associated with reduction in risk of major cardiovascular events. This meta-analysis was to systematically evaluate whether daily supplementation and accumulated intake of n-3 PUFA are associated with improved left ventricular (LV) remodelling in patients with chronic heart failure (CHF). Articles were obtained from Pubmed, Clinical key and Web of Science from inception to January 1 in 2021, and a total of twelve trials involving 2162 participants were eligible for inclusion. The sources of study heterogeneity were explained by I2 statistic and subgroup analysis. Compared with placebo groups, n-3 PUFA supplementation improved LV ejection fraction (LVEF) (eleven trials, 2112 participants, weighted mean difference (WMD) = 2·52, 95 % CI 1·25, 3·80, I2 = 87·8 %) and decreased LV end systolic volume (five studies, 905 participants, WMD = -3·22, 95 % CI 3·67, -2·77, I2 = 0·0 %) using the continuous variables analysis. Notably, the high accumulated n-3 PUFA dosage groups (≥ 600 g) presented a prominent improvement in LVEF, while the low and middle accumulated dosage (≤ 300 and 300-600 g) showed no effects on LVEF. In addition, n-3 PUFA supplementation decreased the levels of pro-inflammatory mediators including TNF-α, IL-6 (IL-6) and hypersensitive c-reactive protein. Therefore, the present meta-analysis demonstrated that n-3 PUFA consumption was associated with a substantial improvement of LV function and remodelling in patients subjected to CHF. The accumulated dosage of n-3 PUFA intake is vital for its cardiac protective role.
Collapse
Affiliation(s)
- Jing Liu
- Research Center for Translational Medicine, Shanghai East Hospital, Tongji University School of Medicine, Shanghai200120, People's Republic of China
- Department of Burn & Plastic Surgery, Beijing Children's Hospital, Capital Medical University, National Center for Children's Health, Beijing100045, People's Republic of China
| | - Qingshu Meng
- Research Center for Translational Medicine, Shanghai East Hospital, Tongji University School of Medicine, Shanghai200120, People's Republic of China
- Institute of Integrated Traditional Chinese and Western Medicine for Cardiovascular Chronic Diseases, Tongji University School of Medicine, Shanghai200120, People's Republic of China
- Shanghai Heart Failure Research Center, Shanghai East Hospital, Tongji University School of Medicine, Shanghai200120, People's Republic of China
| | - Liang Zheng
- Research Center for Translational Medicine, Shanghai East Hospital, Tongji University School of Medicine, Shanghai200120, People's Republic of China
- Institute of Integrated Traditional Chinese and Western Medicine for Cardiovascular Chronic Diseases, Tongji University School of Medicine, Shanghai200120, People's Republic of China
- Shanghai Heart Failure Research Center, Shanghai East Hospital, Tongji University School of Medicine, Shanghai200120, People's Republic of China
| | - Ping Yu
- Department of Heart Failure, Shanghai East Hospital, Tongji University School of Medicine, Shanghai200120, People's Republic of China
| | - Hao Hu
- Department of Heart Failure, Shanghai East Hospital, Tongji University School of Medicine, Shanghai200120, People's Republic of China
| | - Rulin Zhuang
- Research Center for Translational Medicine, Shanghai East Hospital, Tongji University School of Medicine, Shanghai200120, People's Republic of China
- Department of Cardiovascular Surgery, Shanghai East Hospital, Tongji University School of Medicine, Shanghai200120, People's Republic of China
| | - Xinyu Ge
- Research Center for Translational Medicine, Shanghai East Hospital, Tongji University School of Medicine, Shanghai200120, People's Republic of China
- Department of Cardiovascular Surgery, Shanghai East Hospital, Tongji University School of Medicine, Shanghai200120, People's Republic of China
| | - Zhongmin Liu
- Research Center for Translational Medicine, Shanghai East Hospital, Tongji University School of Medicine, Shanghai200120, People's Republic of China
- Shanghai Heart Failure Research Center, Shanghai East Hospital, Tongji University School of Medicine, Shanghai200120, People's Republic of China
- Department of Cardiovascular Surgery, Shanghai East Hospital, Tongji University School of Medicine, Shanghai200120, People's Republic of China
| | - Xiaoting Liang
- Research Center for Translational Medicine, Shanghai East Hospital, Tongji University School of Medicine, Shanghai200120, People's Republic of China
- Institute of Integrated Traditional Chinese and Western Medicine for Cardiovascular Chronic Diseases, Tongji University School of Medicine, Shanghai200120, People's Republic of China
- Shanghai Heart Failure Research Center, Shanghai East Hospital, Tongji University School of Medicine, Shanghai200120, People's Republic of China
- Institute for Regenerative Medicine, Shanghai East Hospital, School of Life Sciences and Technology, Tongji University, Shanghai200120, People's Republic of China
| | - Xiaohui Zhou
- Research Center for Translational Medicine, Shanghai East Hospital, Tongji University School of Medicine, Shanghai200120, People's Republic of China
- Institute of Integrated Traditional Chinese and Western Medicine for Cardiovascular Chronic Diseases, Tongji University School of Medicine, Shanghai200120, People's Republic of China
- Shanghai Heart Failure Research Center, Shanghai East Hospital, Tongji University School of Medicine, Shanghai200120, People's Republic of China
| |
Collapse
|
77
|
Yap CX, Henders AK, Alvares GA, Giles C, Huynh K, Nguyen A, Wallace L, McLaren T, Yang Y, Hernandez LM, Gandal MJ, Hansell NK, Cleary D, Grove R, Hafekost C, Harun A, Holdsworth H, Jellett R, Khan F, Lawson LP, Leslie J, Levis Frenk M, Masi A, Mathew NE, Muniandy M, Nothard M, Miller JL, Nunn L, Strike LT, Cadby G, Moses EK, de Zubicaray GI, Thompson PM, McMahon KL, Wright MJ, Visscher PM, Dawson PA, Dissanayake C, Eapen V, Heussler HS, Whitehouse AJO, Meikle PJ, Wray NR, Gratten J. Interactions between the lipidome and genetic and environmental factors in autism. Nat Med 2023; 29:936-949. [PMID: 37076741 PMCID: PMC10115648 DOI: 10.1038/s41591-023-02271-1] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2022] [Accepted: 02/22/2023] [Indexed: 04/21/2023]
Abstract
Autism omics research has historically been reductionist and diagnosis centric, with little attention paid to common co-occurring conditions (for example, sleep and feeding disorders) and the complex interplay between molecular profiles and neurodevelopment, genetics, environmental factors and health. Here we explored the plasma lipidome (783 lipid species) in 765 children (485 diagnosed with autism spectrum disorder (ASD)) within the Australian Autism Biobank. We identified lipids associated with ASD diagnosis (n = 8), sleep disturbances (n = 20) and cognitive function (n = 8) and found that long-chain polyunsaturated fatty acids may causally contribute to sleep disturbances mediated by the FADS gene cluster. We explored the interplay of environmental factors with neurodevelopment and the lipidome, finding that sleep disturbances and unhealthy diet have a convergent lipidome profile (with potential mediation by the microbiome) that is also independently associated with poorer adaptive function. In contrast, ASD lipidome differences were accounted for by dietary differences and sleep disturbances. We identified a large chr19p13.2 copy number variant genetic deletion spanning the LDLR gene and two high-confidence ASD genes (ELAVL3 and SMARCA4) in one child with an ASD diagnosis and widespread low-density lipoprotein-related lipidome derangements. Lipidomics captures the complexity of neurodevelopment, as well as the biological effects of conditions that commonly affect quality of life among autistic people.
Collapse
Affiliation(s)
- Chloe X Yap
- Mater Research Institute, The University of Queensland, Brisbane, Queensland, Australia.
- Institute for Molecular Bioscience, The University of Queensland, Brisbane, Queensland, Australia.
- Cooperative Research Centre for Living with Autism, Long Pocket, Queensland, Australia.
| | - Anjali K Henders
- Institute for Molecular Bioscience, The University of Queensland, Brisbane, Queensland, Australia
- Cooperative Research Centre for Living with Autism, Long Pocket, Queensland, Australia
| | - Gail A Alvares
- Cooperative Research Centre for Living with Autism, Long Pocket, Queensland, Australia
- Telethon Kids Institute, The University of Western Australia, Perth, Western Australia, Australia
| | - Corey Giles
- Baker Heart and Diabetes Institute, Melbourne, Victoria, Australia
- Baker Department of Cardiometabolic Health, The University of Melbourne, Melbourne, Victoria, Australia
| | - Kevin Huynh
- Baker Heart and Diabetes Institute, Melbourne, Victoria, Australia
- Baker Department of Cardiometabolic Health, The University of Melbourne, Melbourne, Victoria, Australia
| | - Anh Nguyen
- Baker Heart and Diabetes Institute, Melbourne, Victoria, Australia
- Baker Department of Cardiometabolic Health, The University of Melbourne, Melbourne, Victoria, Australia
| | - Leanne Wallace
- Institute for Molecular Bioscience, The University of Queensland, Brisbane, Queensland, Australia
- Cooperative Research Centre for Living with Autism, Long Pocket, Queensland, Australia
| | - Tiana McLaren
- Institute for Molecular Bioscience, The University of Queensland, Brisbane, Queensland, Australia
- Cooperative Research Centre for Living with Autism, Long Pocket, Queensland, Australia
| | - Yuanhao Yang
- Institute for Molecular Bioscience, The University of Queensland, Brisbane, Queensland, Australia
- Cooperative Research Centre for Living with Autism, Long Pocket, Queensland, Australia
| | - Leanna M Hernandez
- Department of Psychiatry and Biobehavioral Sciences, Semel Institute, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA, USA
| | - Michael J Gandal
- Department of Psychiatry and Biobehavioral Sciences, Semel Institute, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA, USA
- Lifespan Brain Institute at Penn Medicine and The Children's Hospital of Philadelphia, Department of Psychiatry, University of Pennsylvania, Philadelphia, PA, USA
- Program in Neurobehavioral Genetics, Semel Institute, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA, USA
- Department of Human Genetics, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA, USA
| | - Narelle K Hansell
- Queensland Brain Institute, The University of Queensland, Brisbane, Queensland, Australia
| | - Dominique Cleary
- Cooperative Research Centre for Living with Autism, Long Pocket, Queensland, Australia
- Telethon Kids Institute, The University of Western Australia, Perth, Western Australia, Australia
| | - Rachel Grove
- Cooperative Research Centre for Living with Autism, Long Pocket, Queensland, Australia
- Faculty of Health, University of Technology Sydney, Sydney, New South Wales, Australia
- School of Psychiatry, Faculty of Medicine, University of New South Wales, Sydney, New South Wales, Australia
| | - Claire Hafekost
- Cooperative Research Centre for Living with Autism, Long Pocket, Queensland, Australia
- Telethon Kids Institute, The University of Western Australia, Perth, Western Australia, Australia
| | - Alexis Harun
- Cooperative Research Centre for Living with Autism, Long Pocket, Queensland, Australia
- Telethon Kids Institute, The University of Western Australia, Perth, Western Australia, Australia
| | - Helen Holdsworth
- Mater Research Institute, The University of Queensland, Brisbane, Queensland, Australia
- Cooperative Research Centre for Living with Autism, Long Pocket, Queensland, Australia
- Child Health Research Centre, The University of Queensland, Brisbane, Queensland, Australia
| | - Rachel Jellett
- Cooperative Research Centre for Living with Autism, Long Pocket, Queensland, Australia
- Olga Tennison Autism Research Centre, La Trobe University, Melbourne, Victoria, Australia
| | - Feroza Khan
- Cooperative Research Centre for Living with Autism, Long Pocket, Queensland, Australia
- School of Psychiatry, Faculty of Medicine, University of New South Wales, Sydney, New South Wales, Australia
| | - Lauren P Lawson
- Cooperative Research Centre for Living with Autism, Long Pocket, Queensland, Australia
- Department of Psychology, Counselling and Therapy, La Trobe University, Melbourne, Victoria, Australia
| | - Jodie Leslie
- Cooperative Research Centre for Living with Autism, Long Pocket, Queensland, Australia
- Telethon Kids Institute, The University of Western Australia, Perth, Western Australia, Australia
| | - Mira Levis Frenk
- Mater Research Institute, The University of Queensland, Brisbane, Queensland, Australia
- Cooperative Research Centre for Living with Autism, Long Pocket, Queensland, Australia
- Child Health Research Centre, The University of Queensland, Brisbane, Queensland, Australia
| | - Anne Masi
- Cooperative Research Centre for Living with Autism, Long Pocket, Queensland, Australia
- School of Psychiatry, Faculty of Medicine, University of New South Wales, Sydney, New South Wales, Australia
| | - Nisha E Mathew
- Cooperative Research Centre for Living with Autism, Long Pocket, Queensland, Australia
- School of Psychiatry, Faculty of Medicine, University of New South Wales, Sydney, New South Wales, Australia
| | - Melanie Muniandy
- Cooperative Research Centre for Living with Autism, Long Pocket, Queensland, Australia
- Olga Tennison Autism Research Centre, La Trobe University, Melbourne, Victoria, Australia
| | - Michaela Nothard
- Mater Research Institute, The University of Queensland, Brisbane, Queensland, Australia
- Cooperative Research Centre for Living with Autism, Long Pocket, Queensland, Australia
- Olga Tennison Autism Research Centre, La Trobe University, Melbourne, Victoria, Australia
| | - Jessica L Miller
- Queensland Brain Institute, The University of Queensland, Brisbane, Queensland, Australia
| | - Lorelle Nunn
- Institute for Molecular Bioscience, The University of Queensland, Brisbane, Queensland, Australia
| | - Lachlan T Strike
- Queensland Brain Institute, The University of Queensland, Brisbane, Queensland, Australia
| | - Gemma Cadby
- School of Population and Global Health, The University of Western Australia, Perth, Western Australia, Australia
| | - Eric K Moses
- Menzies Institute for Medical Research, University of Tasmania, Hobart, Tasmania, Australia
- School of Biomedical Sciences, The University of Western Australia, Perth, Western Australia, Australia
| | - Greig I de Zubicaray
- School of Psychology and Counselling, Faculty of Health, Queensland University of Technology, Brisbane, Queensland, Australia
| | - Paul M Thompson
- Imaging Genetics Center, Mark and Mary Stevens Neuroimaging and Informatics Institute, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| | - Katie L McMahon
- School of Clinical Sciences, Centre for Biomedical Technologies, Queensland University of Technology, Brisbane, Queensland, Australia
| | - Margaret J Wright
- Queensland Brain Institute, The University of Queensland, Brisbane, Queensland, Australia
- Centre for Advanced Imaging, The University of Queensland, Brisbane, Queensland, Australia
| | - Peter M Visscher
- Institute for Molecular Bioscience, The University of Queensland, Brisbane, Queensland, Australia
| | - Paul A Dawson
- Mater Research Institute, The University of Queensland, Brisbane, Queensland, Australia
- Cooperative Research Centre for Living with Autism, Long Pocket, Queensland, Australia
| | - Cheryl Dissanayake
- Cooperative Research Centre for Living with Autism, Long Pocket, Queensland, Australia
- Olga Tennison Autism Research Centre, La Trobe University, Melbourne, Victoria, Australia
| | - Valsamma Eapen
- Cooperative Research Centre for Living with Autism, Long Pocket, Queensland, Australia
- School of Psychiatry, Faculty of Medicine, University of New South Wales, Sydney, New South Wales, Australia
- Academic Unit of Child Psychiatry South West Sydney, Ingham Institute for Applied Medical Research, Liverpool Hospital, Sydney, New South Wales, Australia
| | - Helen S Heussler
- Cooperative Research Centre for Living with Autism, Long Pocket, Queensland, Australia
- Child Health Research Centre, The University of Queensland, Brisbane, Queensland, Australia
- Child Development Program, Children's Health Queensland, Brisbane, Queensland, Australia
| | - Andrew J O Whitehouse
- Cooperative Research Centre for Living with Autism, Long Pocket, Queensland, Australia
- Telethon Kids Institute, The University of Western Australia, Perth, Western Australia, Australia
| | - Peter J Meikle
- Baker Heart and Diabetes Institute, Melbourne, Victoria, Australia
- Baker Department of Cardiometabolic Health, The University of Melbourne, Melbourne, Victoria, Australia
- Baker Department of Cardiovascular Research, Translation and Implementation, La Trobe University, Melbourne, Victoria, Australia
| | - Naomi R Wray
- Institute for Molecular Bioscience, The University of Queensland, Brisbane, Queensland, Australia
- Cooperative Research Centre for Living with Autism, Long Pocket, Queensland, Australia
- Queensland Brain Institute, The University of Queensland, Brisbane, Queensland, Australia
| | - Jacob Gratten
- Mater Research Institute, The University of Queensland, Brisbane, Queensland, Australia.
- Institute for Molecular Bioscience, The University of Queensland, Brisbane, Queensland, Australia.
- Cooperative Research Centre for Living with Autism, Long Pocket, Queensland, Australia.
| |
Collapse
|
78
|
Omega-3-Rich Fish-Oil-Influenced Mouse Gut Microbiome Shaped by Intermittent Consumption of Beef. Curr Microbiol 2023; 80:119. [PMID: 36855004 DOI: 10.1007/s00284-023-03223-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2022] [Accepted: 02/11/2023] [Indexed: 03/02/2023]
Abstract
Beef consumption can provide various amino acids, lipids, vitamins, and minerals; however, excessive intake causes metabolic disorders and increases the probability of obesity, atherosclerosis, and colorectal cancer. The intake of omega-3 fatty acids can ameliorate metabolic disorders by lowering blood glucose and triglyceride levels. In the present study, we investigated the effect of omega-3-rich fish oil on body performance and the gut microbiome in a beef-rich diet. Four-week-old C57BL/6 mice were distributed into four groups (chow diet [Chow], chow with beef diet [Beef], chow with omega-3 diet [Cw3], and chow with beef and omega-3 diet [Bw3]). We observed that body weight was unaltered between groups, and serum triglyceride levels were reduced in the omega-3 supplemented groups. The beta diversity indices, unweighted UniFrac distance (P = 0.001), and Jaccard distance (P = 0.001) showed statistically significant differences, and the principal coordinates analysis plot showed a clear separation between groups. In addition, the taxonomic comparison revealed that beef consumption increased numerous potentially pathogenic bacteria, including Escherichia-Shigella, Mucispirillum, Helicobacter, and Desulfovibrio, which were decreased following omega-3 supplementation. Metabolic comparison based on 16S rRNA revealed that energy and glucose metabolism were higher in omega-3 supplemented groups. Our findings suggest that the omega-3 supplementation under intermittent beef consumption contributes to changes in the gut microbiome and microbial metabolic pathways.
Collapse
|
79
|
Damaiyanti DW, Tsai ZY, Masbuchin AN, Huang CY, Liu PY. Interplay between fish oil, obesity and cardiometabolic diabetes. J Formos Med Assoc 2023:S0929-6646(23)00098-0. [DOI: 10.1016/j.jfma.2023.03.013] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2022] [Revised: 02/24/2023] [Accepted: 03/14/2023] [Indexed: 03/31/2023] Open
|
80
|
Dadkhah Tehrani S, Shojaei M, Bagherniya M, Pirro M, Sahebkar A. The effects of phytochemicals on serum triglycerides in subjects with hypertriglyceridemia: A systematic review of randomized controlled trials. Phytother Res 2023; 37:1640-1662. [PMID: 36756995 DOI: 10.1002/ptr.7763] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2022] [Revised: 12/02/2022] [Accepted: 01/22/2023] [Indexed: 02/10/2023]
Abstract
This systematic review aimed to evaluate the efficacy of phytochemicals on lipid parameters in patients with hypertriglyceridemia (HTG). A comprehensive search was performed in PubMed/Medline, Scopus, ISI Web of Science, and Google Scholar from inception up to October 2021 to recognize randomized controlled trials (RCTs) assessing the effects of phytochemicals on lipid profiles in patients with HTG. Forty-eight RCTs including 53 arms and comprising 3,478 HTG patients met the eligibility criteria. Phytochemicals significantly reduced the serum levels of triglycerides in 32 out 53 arms, total cholesterol in 22 out of 51, low-density lipoprotein cholesterol in 21 out of 48, very low-density lipoprotein cholesterol in 1 out of 5, apolipoprotein B in 2 out of 4, and lipoprotein(a) levels in 2 out of 4 arms. Furthermore, phytochemicals supplementation increased the levels of high-density lipoprotein cholesterol in 15 out of 48 arms. In brief, phytochemicals supplementation might have beneficial effects on HTG. In most of the studies, phytochemicals had a favorable effect on at least one of the lipid parameters.
Collapse
Affiliation(s)
- Sahar Dadkhah Tehrani
- Student Research Committee, Department of Community Nutrition, School of Nutrition and Food Science, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Mehrnaz Shojaei
- Student Research Committee, Department of Community Nutrition, School of Nutrition and Food Science, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Mohammad Bagherniya
- Food Security Research Center and Department of Community Nutrition, School of Nutrition and Food Science, Isfahan University of Medical Sciences, Isfahan, Iran.,Anesthesia and Critical Care Research Center, Isfahan University of Medical Sciences, fahan, Iran
| | - Matteo Pirro
- Unit of Internal Medicine, Angiology and Arteriosclerosis Diseases, Department of Medicine, University of Perugia, Perugia, Italy
| | - Amirhossein Sahebkar
- Applied Biomedical Research Center, Mashhad University of Medical Sciences, Mashhad, Iran.,Biotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran.,Department of Biotechnology, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran
| |
Collapse
|
81
|
Xu J, Huang X, Guo Y, Ma X, Li P, Zhou S, Zhang C, Chen R, Van Halm-Lutterodt N, Yuan L. Discrepant modulating effects of dietary docosahexaenoic acid on cerebral lipids, fatty acid transporter expression and soluble beta-amyloid levels in ApoE -/- and C57BL/6J mice. Neuropathol Appl Neurobiol 2023; 49:e12855. [PMID: 36259948 DOI: 10.1111/nan.12855] [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: 04/01/2022] [Revised: 08/03/2022] [Accepted: 08/21/2022] [Indexed: 11/30/2022]
Abstract
AIMS The study was designed to explore the role of apolipoprotein E (ApoE) deficiency concomitant with dietary docosahexaenoic acid (DHA) treatment on brain β-amyloid (Aβ) and lipid levels. METHOD A 5-month dietary DHA intervention was conducted in ApoE-deficient (ApoE-/- ) mice and wild-type C57BL/6J (C57 wt) mice. The Morris water maze test was performed to assess the behaviour of the animals. The cortical contents of soluble Aβ1-40 and Aβ1-42 were detected by enzyme-linked immunosorbent assay (ELISA). Cortical fatty acid levels were detected by gas chromatography. Gene and protein expression of molecules associated with cerebral Aβ and lipid metabolism were measured using real-time polymerase chain reaction (PCR), Western blot and histological methods. RESULTS DHA treatment increased the content of cortical DHA and n-3 polyunsaturated fatty acids (n-3 PUFAs) but decreased the ratio of n-6/n-3 PUFAs in ApoE-/- mice; whereas the content of cortical DHA and n-3 PUFAs in C57 wt mice remained unchanged after DHA treatment. Cerebral Fabp5 and Cd36 gene expression were significantly downregulated in DHA-fed C57 wt mice; cerebral Cd36 and Scarb1 gene expression were significantly upregulated, whereas Fabp5 gene expression was downregulated in DHA-fed ApoE-/- mice. In comparison with C57 wt mice, the content of cortical soluble Aβ1-42 , total cholesterol (TC) and low-density lipoprotein cholesterol (LDL-C) increased, whereas the level of high-density lipoprotein cholesterol (HDL-C) decreased in ApoE-/- mice. Interestingly, these differences were significantly reversed by DHA dietary treatment. CONCLUSION DHA intervention has discrepant impacts on cerebral lipids, fatty acid transporter expression and soluble Aβ levels in ApoE-/- and C57 wt mice, suggesting the modifying role of ApoE status on the responses of cerebral lipids and Aβ metabolism to DHA treatment.
Collapse
Affiliation(s)
- Jingjing Xu
- School of Public Health, Capital Medical University, Beijing, China.,Beijing Key Laboratory of Environmental Toxicology, Capital Medical University, Beijing, China
| | - Xiaochen Huang
- School of Public Health, Capital Medical University, Beijing, China.,Beijing Key Laboratory of Environmental Toxicology, Capital Medical University, Beijing, China.,Nutrition Department, Beijing Jishuitan Hospital, Beijing, China
| | - Yujie Guo
- School of Public Health, Capital Medical University, Beijing, China.,Beijing Key Laboratory of Environmental Toxicology, Capital Medical University, Beijing, China
| | - Xiaojun Ma
- School of Public Health, Capital Medical University, Beijing, China.,Beijing Key Laboratory of Environmental Toxicology, Capital Medical University, Beijing, China
| | - Pengfei Li
- School of Public Health, Capital Medical University, Beijing, China.,Beijing Key Laboratory of Environmental Toxicology, Capital Medical University, Beijing, China
| | - Shaobo Zhou
- School of Science, Faculty of Engineering and Science, University of Greenwich, Chatham, UK
| | - Chi Zhang
- School of Biological Sciences, University of Nebraska-Lincoln, Lincoln, Nebraska, USA
| | - Rui Chen
- School of Public Health, Capital Medical University, Beijing, China.,Beijing Key Laboratory of Environmental Toxicology, Capital Medical University, Beijing, China
| | | | - Linhong Yuan
- School of Public Health, Capital Medical University, Beijing, China.,Beijing Key Laboratory of Environmental Toxicology, Capital Medical University, Beijing, China
| |
Collapse
|
82
|
Bansal N, Kumar S, Brar PC. Update on management of paediatric dyslipidaemia. Curr Opin Endocrinol Diabetes Obes 2023; 30:52-64. [PMID: 36541082 DOI: 10.1097/med.0000000000000794] [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: 12/24/2022]
Abstract
PURPOSE OF REVIEW Atherosclerosis and associated cardiovascular risk factors originate in childhood; hence, early management of dyslipidaemia is vital. However, hypercholesterolemia remains untreated or undertreated in many youths. We review current therapies, drugs under investigation and consider potential future directions for the management of paediatric dyslipidaemia to highlight the recent evidence and new therapeutic options for future use. RECENT FINDINGS Cardiovascular disease (CVD) risk factors in childhood, including dyslipidaemia, are associated with CVD risk and clinical CVD events in adulthood. Recent data show that initiation of statin therapy in childhood in children with familial hypercholesterolemia reduces the risk of CVD in adulthood. Several well tolerated and efficacious treatment options have become available in recent times for the management of dyslipidaemia in youth. Many new lipid-lowering drugs are under investigation to widen the available choices. Some of these drugs are now available for use in paediatrics, while some remain targets for future use. SUMMARY We review available treatment options for paediatric dyslipidaemia management, discuss potential limitations and propose future directions. We also acknowledge the need for continued research in paediatrics for optimal paediatric dyslipidaemia management.
Collapse
Affiliation(s)
- Nidhi Bansal
- Division of Pediatric Endocrinology, Department of Pediatrics, Baylor College of Medicine, Texas Children's Hospital, Houston, Texas
| | - Seema Kumar
- Division of Pediatric Endocrinology, Department of Pediatric and Adolescent Medicine, Mayo Clinic, Rochester, Minnesota
| | - Preneet Cheema Brar
- Division of Pediatric Endocrinology and Diabetes, Department of Pediatrics, New York University Grossman School of Medicine, New York, New York, USA
| |
Collapse
|
83
|
Takeda Y, Sakuma I, Hiramitsu S, Okada M, Ueda S, Sakurai M. The effects of pemafibrate and omega-3 fatty acid ethyl on apoB-48 in dyslipidemic patients treated with statin: A prospective, multicenter, open-label, randomized, parallel group trial in Japan (PROUD48 study). Front Cardiovasc Med 2023; 10:1094100. [PMID: 36760560 PMCID: PMC9905248 DOI: 10.3389/fcvm.2023.1094100] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2022] [Accepted: 01/06/2023] [Indexed: 01/27/2023] Open
Abstract
Background We compared the lowering effects of pemafibrate and omega-3 fatty acid ethyl on fasting apolipoprotein (apo) B-48 (apoB-48), a marker that reflects postprandial hypertriglyceridemia, which is one of the residual risks for atherosclerotic cardiovascular disease (ASCVD) with statin treatment. Methods This prospective, multicenter, open-label, randomized, parallel group trial was conducted at 4 medical institutions between April 2020 and May 2022. A total of 126 ambulatory patients with dyslipidemia receiving statin treatment for more than 4 weeks, aged 20-79 years with fasting triglyceride (TG) levels of ≥177 mg/dl were randomly assigned to 16-week pemafibrate 0.4 mg per day treatment group (PEMA, n = 63) or omega-3 fatty acid ethyl 4 g per day treatment group (OMEGA-3, n = 63). The primary endpoint was the percentage change in fasting apoB-48 from baseline to week 16. Results The percentage changes in fasting apoB-48 in PEMA and OMEGA-3 were -50.8% (interquartile range -62.9 to -30.3%) and -17.5% (-38.3 to 15.3%) (P < 0.001), respectively. As the secondary endpoints, the changes in fasting apoB-48 in PEMA and OMEGA-3 were -3.10 μg/ml (-5.63 to -1.87) and -0.90 μg/ml (-2.95 to 0.65) (P < 0.001), respectively. Greater decreases with significant differences in the percentage changes in TG, remnant lipoprotein cholesterol, apoC-III, fasting plasma glucose, alanine aminotransferase, gamma-glutamyl transpeptidase, and alkaline phosphatase were observed in PEMA, compared with OMEGA-3. Greater increases with significant differences in those in high-density lipoprotein (HDL) cholesterol, apoA-I, and apoA-II were observed in PEMA, compared with OMEGA-3. PEMA showed anti-atherosclerotic lipoprotein profiles in gel-permeation high-performance liquid chromatography analyses, compared with OMEGA-3. Although adverse events occurred in 9 of 63 (14.3%) patients in PEMA and 3 of 63 (4.8%) patients in OMEGA-3, no serious adverse events associated with drug were observed in either group. Conclusions This is the first randomized trial to compare the lowering effects of pemafibrate and omega-3 fatty acid ethyl on fasting apoB-48. We concluded that pemafibrate was superior to omega-3 fatty acid ethyl in lowering effect of fasting apoB-48. Pemafibrate is expected to reduce the residual risk for ASCVD with statin treatment. Clinical trial registration https://rctportal.niph.go.jp/en, identifier jRCTs071200011.
Collapse
Affiliation(s)
- Yasutaka Takeda
- Division of Metabolism and Biosystemic Science, Gastroenterology, and Hematology/Oncology, Department of Medicine, Asahikawa Medical University, Asahikawa, Japan,*Correspondence: Yasutaka Takeda,
| | - Ichiro Sakuma
- Caress Sapporo Hokko Memorial Clinic, Sapporo, Japan
| | | | | | - Shinichiro Ueda
- Department of Clinical Pharmacology and Therapeutics, University of the Ryukyus, Nishihara, Japan
| | - Masaru Sakurai
- Department of Social and Environmental Medicine, Kanazawa Medical University, Uchinada, Japan
| |
Collapse
|
84
|
Djuricic I, Calder PC. Pros and Cons of Long-Chain Omega-3 Polyunsaturated Fatty Acids in Cardiovascular Health. Annu Rev Pharmacol Toxicol 2023; 63:383-406. [PMID: 36662586 DOI: 10.1146/annurev-pharmtox-051921-090208] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023]
Abstract
The long-chain omega-3 fatty acids, eicosapentaenoic acid (EPA) and docosahexaenoic acid (DHA), are found in seafood, supplements, and concentrated pharmaceutical preparations. Prospective cohort studies demonstrate an association between higher intakes of EPA+DHA or higher levels of EPA and DHA in the body and lower risk of developing cardiovascular disease (CVD), especially coronary heart disease and myocardial infarction, and of cardiovascular mortality in the general population. The cardioprotective effect of EPA and DHA is due to the beneficial modulation of a number of risk factors for CVD. Some large trials support the use of EPA+DHA (or EPA alone) in high-risk patients, although the evidence is inconsistent. This review presents key studies of EPA and DHA in the primary and secondary prevention of CVD, briefly describes potential mechanisms of action, and discusses recently published RCTs and meta-analyses. Potential adverse aspects of long-chain omega-3 fatty acids in relation to CVD are discussed.
Collapse
Affiliation(s)
- Ivana Djuricic
- Department of Bromatology, Faculty of Pharmacy, University of Belgrade, Belgrade, Serbia
| | - Philip C Calder
- School of Human Development and Health, Faculty of Medicine, University of Southampton, Southampton, United Kingdom;
- NIHR Southampton Biomedical Research Centre, University Hospital Southampton NHS Foundation Trust and University of Southampton, Southampton, United Kingdom
| |
Collapse
|
85
|
Monteiro JP, Ferreira HB, Melo T, Flanagan C, Urbani N, Neves J, Domingues P, Domingues MR. The plasma phospholipidome of the bottlenose dolphin ( Tursiops truncatus) is modulated by both sex and developmental stage. Mol Omics 2023; 19:35-47. [PMID: 36314173 DOI: 10.1039/d2mo00202g] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Lipidomics represent a valid complementary tool to the biochemical analysis of plasma in humans. However, in cetaceans, these tools have been unexplored. Here, we evaluated how the plasma lipid composition of Tursiops truncatus is modulated by developmental stage and sex, aiming at a potential use of lipidomics in integrated strategies to monitor cetacean health. We characterized the fatty acid profile and detected a total of 26 fatty acids in T. truncatus plasma. The most abundant fatty acids were palmitic acid (C16:0), stearic acid (C18:0) and oleic acid (C18:1n-9). Interestingly, there are consistent differences between the fatty acid profile of mature female and mature male specimens. Phospholipidome analysis identified 320 different lipid species belonging to phosphatidylcholine (PC, 105 lipid species), lysophosphatidylcholine (42), phosphatidylethanolamine (PE, 67), lysophosphatidylethanolamine (18), phosphatidylglycerol (14), lysophosphatidylglycerol (8), phosphatidylinositol (14), lysophosphatidylinositol (2), phosphatidylserine (3), sphingomyelin (45) and ceramides (2) classes. The statistical analysis of the phospholipidome showed that its composition allows discriminating mature animals between sexes and mature males from immature males. Notably, discrimination between sexes is mainly determined by the contents of PE plasmalogens and lysophospholipids (LPC and LPE), while the differences between mature and immature male animals were mainly determined by the levels of PC lipids. This is the first time that a correlation between developmental stage and sex and the lipid composition of the plasma has been established in cetaceans. Being able to discern between age and sex-related changes is an encouraging step towards using these tools to also detect differences related to disease/dysfunction processes.
Collapse
Affiliation(s)
- João P Monteiro
- Mass Spectrometry Centre, LAQV-REQUIMTE, Department of Chemistry, University of Aveiro, Aveiro, Portugal. .,CESAM-Centre for Environmental and Marine Studies, Department of Chemistry, University of Aveiro, Aveiro, Portugal
| | - Helena B Ferreira
- Mass Spectrometry Centre, LAQV-REQUIMTE, Department of Chemistry, University of Aveiro, Aveiro, Portugal. .,CESAM-Centre for Environmental and Marine Studies, Department of Chemistry, University of Aveiro, Aveiro, Portugal
| | - Tânia Melo
- Mass Spectrometry Centre, LAQV-REQUIMTE, Department of Chemistry, University of Aveiro, Aveiro, Portugal. .,CESAM-Centre for Environmental and Marine Studies, Department of Chemistry, University of Aveiro, Aveiro, Portugal
| | | | | | | | - Pedro Domingues
- Mass Spectrometry Centre, LAQV-REQUIMTE, Department of Chemistry, University of Aveiro, Aveiro, Portugal. .,CESAM-Centre for Environmental and Marine Studies, Department of Chemistry, University of Aveiro, Aveiro, Portugal
| | - M Rosário Domingues
- Mass Spectrometry Centre, LAQV-REQUIMTE, Department of Chemistry, University of Aveiro, Aveiro, Portugal. .,CESAM-Centre for Environmental and Marine Studies, Department of Chemistry, University of Aveiro, Aveiro, Portugal
| |
Collapse
|
86
|
Aryankalayil MJ, Bylicky MA, Martello S, Chopra S, Sproull M, May JM, Shankardass A, MacMillan L, Vanpouille-Box C, Dalo J, Scott KMK, Norman Coleman C. Microarray analysis identifies coding and non-coding RNA markers of liver injury in whole body irradiated mice. Sci Rep 2023; 13:200. [PMID: 36604457 PMCID: PMC9814510 DOI: 10.1038/s41598-022-26784-w] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2022] [Accepted: 12/20/2022] [Indexed: 01/06/2023] Open
Abstract
Radiation injury from medical, accidental, or intentional sources can induce acute and long-term hepatic dysregulation, fibrosis, and cancer. This long-term hepatic dysregulation decreases quality of life and may lead to death. Our goal in this study is to determine acute changes in biological pathways and discover potential RNA biomarkers predictive of radiation injury. We performed whole transcriptome microarray analysis of mouse liver tissue (C57BL/6 J) 48 h after whole-body irradiation with 1, 2, 4, 8, and 12 Gray to identify significant expression changes in mRNAs, lncRNAs, and miRNAs, We also validated changes in specific RNAs through qRT-PCR. We used Ingenuity Pathway Analysis (IPA) to identify pathways associated with gene expression changes. We observed significant dysregulation of multiple mRNAs across all doses. In contrast, miRNA dysregulation was observed upwards of 2 Gray. The most significantly upregulated mRNAs function as tumor suppressors: Cdkn1a, Phlda3, and Eda2r. The most significantly downregulated mRNAs were involved in hemoglobin synthesis, inflammation, and mitochondrial function including multiple members of Hbb and Hba. The most significantly upregulated miRNA included: miR-34a-5p, miR-3102-5p, and miR-3960, while miR-342-3p, miR-142a-3p, and miR-223-3p were most significantly downregulated. IPA predicted activation of cell cycle checkpoint control pathways and inhibition of pathways relevant to inflammation and erythropoietin. Clarifying expression of mRNA, miRNA and lncRNA at a short time point (48 h) offers insight into potential biomarkers, including radiation markers shared across organs and animal models. This information, once validated in human models, can aid in development of bio-dosimetry biomarkers, and furthers our understanding of acute pathway dysregulation.
Collapse
Affiliation(s)
- Molykutty J. Aryankalayil
- grid.48336.3a0000 0004 1936 8075Radiation Oncology Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, 10 Center Drive, Room B3B406, Bethesda, MD 20892 USA
| | - Michelle A. Bylicky
- grid.48336.3a0000 0004 1936 8075Radiation Oncology Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, 10 Center Drive, Room B3B406, Bethesda, MD 20892 USA
| | - Shannon Martello
- grid.48336.3a0000 0004 1936 8075Radiation Oncology Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, 10 Center Drive, Room B3B406, Bethesda, MD 20892 USA
| | - Sunita Chopra
- grid.48336.3a0000 0004 1936 8075Radiation Oncology Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, 10 Center Drive, Room B3B406, Bethesda, MD 20892 USA
| | - Mary Sproull
- grid.48336.3a0000 0004 1936 8075Radiation Oncology Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, 10 Center Drive, Room B3B406, Bethesda, MD 20892 USA
| | - Jared M. May
- grid.48336.3a0000 0004 1936 8075Radiation Oncology Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, 10 Center Drive, Room B3B406, Bethesda, MD 20892 USA
| | - Aman Shankardass
- grid.48336.3a0000 0004 1936 8075Radiation Oncology Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, 10 Center Drive, Room B3B406, Bethesda, MD 20892 USA
| | - Laurel MacMillan
- grid.420517.50000 0004 0490 0428Gryphon Scientific, Takoma Park, MD 20912 USA
| | - Claire Vanpouille-Box
- grid.5386.8000000041936877XDepartment of Radiation Oncology, Weill Cornell Medicine, New York, NY 10065 USA
| | - Juan Dalo
- grid.48336.3a0000 0004 1936 8075Radiation Oncology Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, 10 Center Drive, Room B3B406, Bethesda, MD 20892 USA
| | - Kevin M. K. Scott
- grid.48336.3a0000 0004 1936 8075Radiation Oncology Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, 10 Center Drive, Room B3B406, Bethesda, MD 20892 USA
| | - C. Norman Coleman
- grid.48336.3a0000 0004 1936 8075Radiation Oncology Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, 10 Center Drive, Room B3B406, Bethesda, MD 20892 USA ,grid.48336.3a0000 0004 1936 8075Radiation Research Program, National Cancer Institute, National Institutes of Health, Rockville, MD 20850 USA
| |
Collapse
|
87
|
Santos HO, May TL, Bueno AA. Eating more sardines instead of fish oil supplementation: Beyond omega-3 polyunsaturated fatty acids, a matrix of nutrients with cardiovascular benefits. Front Nutr 2023; 10:1107475. [PMID: 37143475 PMCID: PMC10153001 DOI: 10.3389/fnut.2023.1107475] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2022] [Accepted: 03/27/2023] [Indexed: 05/06/2023] Open
Abstract
Omega-3 polyunsaturated fatty acids (n-3 PUFA) play a significant role in the prevention and management of cardiometabolic diseases associated with a mild chronic pro-inflammatory background, including type 2 diabetes, hypertension, hypertriglyceridaemia, and fatty liver disease. The effects of n-3 PUFA supplements specifically, remain controversial regarding reducing risks of cardiovascular events. n-3 PUFA supplements come at a cost for the consumer and can result in polypharmacy for patients on pharmacotherapy. Sardines are a well-known, inexpensive source of n-3 PUFA and their consumption could reduce the need for n-3 PUFA supplementation. Moreover, sardines contain other cardioprotective nutrients, although further insights are crucial to translate a recommendation for sardine consumption into clinical practice. The present review discusses the matrix of nutrients contained in sardines which confer health benefits for cardiometabolism, beyond n-3 PUFA. Sardines contain calcium, potassium, magnesium, zinc, iron, taurine, arginine and other nutrients which together modulate mild inflammation and exacerbated oxidative stress observed in cardiovascular disease and in haemodynamic dysfunction. In a common serving of sardines, calcium, potassium, and magnesium are the minerals at higher amounts to elicit clinical benefits, whilst other nutrients are present in lower but valuable amounts. A pragmatic approach towards the consumption of such nutrients in the clinical scenario should be adopted to consider the dose-response relationship effects on physiological interactions. As most recommendations currently available are based on an indirect rationale of the physiological actions of the nutrients found in sardines, randomised clinical trials are warranted to expand the evidence on the benefits of sardine consumption.
Collapse
Affiliation(s)
- Heitor O. Santos
- School of Medicine, Federal University of Uberlandia (UFU), Uberlandia, Minas Gerais, Brazil
- *Correspondence: Heitor O. Santos,
| | - Theresa L. May
- School of Science and the Environment, University of Worcester, Worcester, United Kingdom
| | - Allain A. Bueno
- School of Science and the Environment, University of Worcester, Worcester, United Kingdom
| |
Collapse
|
88
|
Teramoto T, Ogawa H, Ueshima H, Okada Y, Haze K, Matsui S, Fujikawa K, Hashimoto T, Sakui S, Nishimura K, Kajita M, Horimoto A, Fernandez J. Effect of omega-3 fatty acids on cardiovascular events in high-risk patients with hypertriglyceridemia in Japan: a 3-year post-marketing surveillance study (OCEAN3 survey). Expert Opin Drug Saf 2023; 22:81-90. [PMID: 35772177 DOI: 10.1080/14740338.2022.2094914] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022]
Abstract
BACKGROUND Studies on the efficacy of prescription omega-3 polyunsaturated fatty acids to reduce cardiovascular events have produced conflicting results. RESEARCH DESIGN AND METHODS This 3-year prospective post-marketing surveillance study evaluated the effect of omega-3-acid ethyl esters (O3AEE; usual dosage 2 g/day) on cardiovascular events in high-risk statin-treated Japanese patients with hypertriglyceridemia. Statin-treated patients not receiving O3AEE were included as a reference cohort. The composite primary endpoint was cardiovascular death, myocardial infarction, stroke, angina requiring coronary revascularization, or peripheral arterial disease requiring surgery or peripheral arterial intervention. RESULTS At 3 years, Kaplan-Meier estimated cumulative incidence of the primary endpoint was 2.5% (95% confidence interval, 2.1%-2.9%) in O3AEE-treated patients (N = 6,580) and 2.7% (2.4%-3.1%) in non-O3AEE-treated patients (N = 7,784; hazard ratio, 0.99; 95% confidence interval, 0.79-1.23). Incidence of heart failure requiring hospitalization was 0.4% with O3AEE versus 0.8% in non-O3AEE-treated patients (hazard ratio, 0.47; 95% confidence interval, 0.28-0.78; P < 0.05). CONCLUSIONS Among patients receiving statins, cardiovascular event incidence did not differ significantly between O3AEE-treated patients and non-O3AEE-treated patients. Further studies are required before definitive conclusions can be drawn on the effect of O3AEE on cardiovascular event incidence in high-risk patients with hypertriglyceridemia. TRIAL REGISTRATION ClinicalTrials.gov, NCT02285166.
Collapse
Affiliation(s)
- Tamio Teramoto
- Teikyo Academic Research Center (TARC), Teikyo University, Tokyo, Japan
| | | | - Hirotsugu Ueshima
- NCD Epidemiology Research Center, Shiga University of Medical Science, Shiga, Japan
| | - Yasushi Okada
- Department of Cerebrovascular Medicine and Neurology, National Hospital Organization Kyushu Medical Center, Fukuoka, Japan
| | - Kazuo Haze
- Department of Cardiology, Kashiwara Municipal Hospital, Kashiwara, Japan
| | | | - Keita Fujikawa
- Japan Medical Office, Takeda Pharmaceutical Company, Tokyo, Japan
| | - Takamasa Hashimoto
- Statistical and Quantitative Sciences, Takeda Pharmaceutical Company, Osaka, Japan
| | - Sho Sakui
- Statistical and Quantitative Sciences, Takeda Pharmaceutical Company, Osaka, Japan
| | | | - Mika Kajita
- Japan Medical Office, Takeda Pharmaceutical Company, Tokyo, Japan
| | | | | |
Collapse
|
89
|
Huh JH, Jo SH. Omega-3 fatty acids and atrial fibrillation. Korean J Intern Med 2022; 38:282-289. [PMID: 36514212 PMCID: PMC10175873 DOI: 10.3904/kjim.2022.266] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/11/2022] [Accepted: 11/21/2022] [Indexed: 12/15/2022] Open
Abstract
Although some clinical trials have demonstrated reduced incidence of cardiovascular disease with the use of omega-3 fatty acids, others have found an increased risk of atrial fibrillation (AF). AF is the most common sustained cardiac arrhythmia worldwide. It is associated with high morbidity and mortality rates and significant public health burden. Previous studies of the effect of omega-3 fatty acids on AF occurrence have reported contradictory results. Here we reviewed the effect of omega-3 fatty acids on the risk of AF.
Collapse
Affiliation(s)
- Ji Hye Huh
- Division of Endocrinology, Department of Internal Medicine, Hallym University Sacred Heart Hospital, Anyang, Korea
| | - Sang-Ho Jo
- Division of Cardiology, Department of Internal Medicine, Hallym University Sacred Heart Hospital, Anyang, Korea
| |
Collapse
|
90
|
Li JJ, Dou KF, Zhou ZG, Zhao D, Ye P, Zhao JJ, Guo LX. Role of omega-3 fatty acids in the prevention and treatment of cardiovascular Diseases: A consensus statement from the Experts' Committee Of National Society Of Cardiometabolic Medicine. Front Pharmacol 2022; 13:1069992. [PMID: 36578548 PMCID: PMC9791266 DOI: 10.3389/fphar.2022.1069992] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2022] [Accepted: 11/28/2022] [Indexed: 12/14/2022] Open
Abstract
Low-density lipoprotein cholesterol (LDL-C) has been considered as the primary target for the prevention and treatment of atherosclerotic cardiovascular disease (ASCVD). However, there are still residual cardiovascular risks in some patients even if LDL-C achieves the target level. Emerging evidence suggestes that elevated triglyceride (TG) level or triglyceride-rich lipoprotein (TRL) cholesterol (TRL-C) is one of the important components of the residual cardiovascular risks. Omega-3 fatty acids have been shown to be one of the effective drugs for reducing TG. However, its efficacy in reducing the risk of ASCVD is inconsistent in large randomized clinical trials. There is lack of consensus among Experts regarding the application of omega-3 fatty acids in cardiovascular diseases including heart failure, arrhythmia, cardiomyopathy, hypertension, and sudden death. Hence, the current consensus will comprehensively and scientifically present the detailed knowledge about the omega-3 fatty acids from a variety of aspects to provide a reference for its management of omega-3 fatty acids application in the Chinese population.
Collapse
Affiliation(s)
- Jian-Jun Li
- Cardiometabolic Center, Fuwai Hospital, Chinese Academy of Medical Sciences, Beijing, China
| | - Ke-Fei Dou
- Cardiometabolic Center, Fuwai Hospital, Chinese Academy of Medical Sciences, Beijing, China
| | - Zhi-Guang Zhou
- Department of Metabolism and Endocrinology, The Second Xiangya Hospital of Central South University, Changsha, Hunan, China
| | - Dong Zhao
- Department of Epidemiology, Beijing Anzhen Hospital Affiliated to Capital Medical University, Beijing, China
| | - Ping Ye
- Department of Cardiology of the First Medical Center, Chinese PLA General Hospital, Beijing, China
| | - Jia-Jun Zhao
- Endocrine Department, Shandong Provincial Hospital, Jinan, Shandong, China
| | - Li-Xin Guo
- Endocrine Department, Beijing Hospital, Beijing, China
| |
Collapse
|
91
|
Bale BF, Doneen AL, Leimgruber PP, Vigerust DJ. The critical issue linking lipids and inflammation: Clinical utility of stopping oxidative stress. Front Cardiovasc Med 2022; 9:1042729. [PMID: 36439997 PMCID: PMC9682196 DOI: 10.3389/fcvm.2022.1042729] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2022] [Accepted: 10/24/2022] [Indexed: 07/30/2023] Open
Abstract
The formation of an atheroma begins when lipoproteins become trapped in the intima. Entrapped lipoproteins become oxidized and activate the innate immune system. This immunity represents the primary association between lipids and inflammation. When the trapping continues, the link between lipids and inflammation becomes chronic and detrimental, resulting in atherosclerosis. When entrapment ceases, the association between lipids and inflammation is temporary and healthy, and the atherogenic process halts. Therefore, the link between lipids and inflammation depends upon lipoprotein retention in the intima. The entrapment is due to electrostatic forces uniting apolipoprotein B to polysaccharide chains on intimal proteoglycans. The genetic transformation of contractile smooth muscle cells in the media into migratory secretory smooth muscle cells produces the intimal proteoglycans. The protein, platelet-derived growth factor produced by activated platelets, is the primary stimulus for this genetic change. Oxidative stress is the main stimulus to activate platelets. Therefore, minimizing oxidative stress would significantly reduce the retention of lipoproteins. Less entrapment decreases the association between lipids and inflammation. More importantly, it would halt atherogenesis. This review will analyze oxidative stress as the critical link between lipids, inflammation, and the pathogenesis of atherosclerosis. Through this perspective, we will discuss stopping oxidative stress to disrupt a harmful association between lipids and inflammation. Numerous therapeutic options will be discussed to mitigate oxidative stress. This paper will add a new meaning to the Morse code distress signal SOS-stopping oxidative stress.
Collapse
Affiliation(s)
- Bradley Field Bale
- Department of Medical Education and Clinical Sciences, Washington State University College of Medicine, Spokane, WA, United States
| | - Amy Lynn Doneen
- Department of Medical Education and Clinical Sciences, Washington State University College of Medicine, Spokane, WA, United States
| | - Pierre P. Leimgruber
- Department of Medical Education and Clinical Sciences, Washington State University College of Medicine, Spokane, WA, United States
- Department of Medical Education and Clinical Sciences, University of Washington School of Medicine, Seattle, WA, United States
| | - David John Vigerust
- Department of Neurological Surgery, Vanderbilt University School of Medicine, Nashville, TN, United States
| |
Collapse
|
92
|
Abstract
PURPOSE OF REVIEW This review aims to discuss the potential roles of omega-3 (ω-3) and omega-6 (ω-6) polyunsaturated fatty acids (PUFAs) in the prevention and treatment of metabolic diseases, to provide the latest evidence from epidemiological and clinical studies, and to highlight novel insights into this field. RECENT FINDINGS Higher dietary or circulating ω-3 PUFA levels are related to a lower risk of metabolic syndrome. Novel findings in obesity indicate higher proportions of ω-6 and ω-3 PUFAs, a modulated oxylipin profile and an altered transcriptome in subcutaneous white adipose tissue, that seem resistant to the effects of ω-3 PUFAs compared with what occurs in normal weight individuals. ω-3 PUFAs may improve the blood lipid profile and glycemic outcomes in patients with type 2 diabetes mellitus and reduce liver fat in nonalcoholic fatty liver disease (NAFLD); the findings of several recent meta-analyses support these effects. Genetic background affects inter-individual variability in the insulin sensitivity response to ω-3 PUFA supplementation. ω-3 PUFAs have prebiotic effects, altering the gut microbiota. SUMMARY Although evidence for health benefits of ω-3 PUFAs is strong, recent findings suggest a more personalized approach to ω-3 PUFA intake for individuals at high risk for metabolic diseases.
Collapse
Affiliation(s)
- Ivana Djuricic
- Department of Bromatology, Faculty of Pharmacy, University of Belgrade, Belgrade, Serbia
| | - Philip C Calder
- School of Human Development and Health, Faculty of Medicine, University of Southampton
- NIHR Southampton Biomedical Research Centre, University Hospital Southampton NHS Foundation Trust and University of Southampton, Southampton, UK
| |
Collapse
|
93
|
Affiliation(s)
- Joseph J Saseen
- Department of Clinical Pharmacy, University of Colorado Anschutz Medical Campus, Skaggs School of Pharmacy; Aurora, CO, USA; Department of Family Medicine, University of Colorado Anschutz Medical Campus, School of Medicine; Aurora, CO, USA.
| | - Salim S Virani
- Section of Cardiology and Cardiovascular Research, Department of Medicine, Baylor College of Medicine, Houston, TX, USA; Health Policy, Quality & Informatics Program, Health Services Research and Development Center for Innovations in Quality, Effectiveness, and Safety (IQuESt), Michael E. DeBakey Veterans Affairs Medical Center, Houston, TX, USA; Section of Cardiology, Michael E. DeBakey Veterans Affairs Medical Center, Houston, TX, USA
| |
Collapse
|
94
|
Gharraee N, Wang Z, Pflum A, Medina-Hernandez D, Herrington D, Zhu X, Meléndez GC. Eicosapentaenoic Acid Ameliorates Cardiac Fibrosis and Tissue Inflammation in Spontaneously Hypertensive Rats. J Lipid Res 2022; 63:100292. [PMID: 36206854 PMCID: PMC9643491 DOI: 10.1016/j.jlr.2022.100292] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2022] [Revised: 09/21/2022] [Accepted: 09/22/2022] [Indexed: 11/06/2022] Open
Abstract
Hypertension affects 1 in 3 adults in the United States and leads to left ventricular (LV) concentric hypertrophy, interstitial fibrosis, and increased stiffness. The treatment of cardiac fibrosis remains challenging and empiric. Eicosapentaenoic acid (EPA) is an omega-3 polyunsaturated fatty acid that is highly effective in reducing cardiovascular events in patients and cardiac fibrosis and hypertrophy in animals when administered before pressure overload by promoting the increase of anti-inflammatory M1 macrophages. In this study, we investigated whether EPA mitigates the exacerbation of cardiac remodeling and fibrosis induced by established hypertension, a situation that closely recapitulates a clinical scenario. Twelve-week-old spontaneously hypertensive rats were randomized to eat an EPA-enriched or control diet for 20 weeks. We report that rats eating the EPA-enriched diet exhibited a reduction of interstitial cardiac fibrosis and ameliorated LV diastolic dysfunction despite the continuous increase in blood pressure. However, we found that EPA did not have an impact on cardiac hypertrophy. Interestingly, the EPA diet increased mRNA expression of M2 macrophage marker Mrc1 and interleukin-10 in cardiac tissue. These findings indicated that the antifibrotic effects of EPA are mediated in part by phenotypic polarization of macrophages toward anti-inflammatory M2 macrophages and increases of the anti-inflammatory cytokine, interleukin-10. In summary, EPA prevents the exacerbation of cardiac fibrosis and LV diastolic dysfunction during sustained pressure overload. EPA could represent a novel treatment strategy for hypertensive cardiomyopathy.
Collapse
Affiliation(s)
- Nazli Gharraee
- Department of Internal Medicine, Section on Cardiology, Wake Forest School of Medicine, Winston-Salem, North Carolina, USA
| | - Zhan Wang
- Department of Internal Medicine, Section on Molecular Medicine, Wake Forest School of Medicine, Winston-Salem, North Carolina, USA
| | - Adam Pflum
- Department of Internal Medicine, Section on Cardiology, Wake Forest School of Medicine, Winston-Salem, North Carolina, USA
| | - Danielle Medina-Hernandez
- Department of Internal Medicine, Section on Cardiology, Wake Forest School of Medicine, Winston-Salem, North Carolina, USA
| | - David Herrington
- Department of Internal Medicine, Section on Cardiology, Wake Forest School of Medicine, Winston-Salem, North Carolina, USA
| | - Xuewei Zhu
- Department of Internal Medicine, Section on Molecular Medicine, Wake Forest School of Medicine, Winston-Salem, North Carolina, USA
| | - Giselle C Meléndez
- Department of Internal Medicine, Section on Cardiology, Wake Forest School of Medicine, Winston-Salem, North Carolina, USA; Department of Pathology, Section on Comparative Medicine, Wake Forest School of Medicine, Winston-Salem, North Carolina, USA.
| |
Collapse
|
95
|
Yang B, Tseng PT, Hu X, Zeng BY, Chang JPC, Liu Y, Chu WJ, Zhang SS, Zhou ZL, Chu CS, Chang CH, Tu YK, Wu YC, Stubbs B, Carvalho AF, Lin PY, Matsuoka YJ, Suen MW, Su KP. Comparative efficacy of omega-3 polyunsaturated fatty acids on major cardiovascular events: A network meta-analysis of randomized controlled trials. Prog Lipid Res 2022; 88:101196. [PMID: 36341839 DOI: 10.1016/j.plipres.2022.101196] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2022] [Revised: 10/23/2022] [Accepted: 10/24/2022] [Indexed: 11/06/2022]
Abstract
The role of omega-3 polyunsaturated fatty acids (PUFAs) in primary and secondary prevention on major cardiovascular events (MCE) is inconclusive due to the potential heterogeneity in study designs of formulas, dosages, and ratios of eicosapentaenoic acid (EPA) and docosahexaenoic acid (DHA) from the findings of previous randomized controlled trials (RCTs). Here we conducted a comprehensive narrative review of pre-clinical studies and updated a network meta-analysis (NMA) to determine the comparative efficacy against MCE with different EPA/DHA dosages and formulas. We found that pure EPA was ranked the best option in the secondary prevention (hazard ratio: 0.72, 95% confidence interval: 0.65 to 0.81) from the NMA of 39 RCTs with 88,359 participants. There was no evidence of omega-3 PUFAs' efficacy in primary prevention. The mechanisms of omega-3 PUFAs' cardiovascular protection might link to the effects of anti-inflammation and stabilization of endothelial function from PUFA's derivatives including eicosanoids and the special pre-resolving mediators (SPMs).
Collapse
Affiliation(s)
- Bo Yang
- School of Public Health and Management & Institute of Lipids Medicine, Wenzhou Medical University, Wenzhou, China; Department of Psychiatry & Mind-Body Interface Laboratory (MBI-Lab), China Medical University Hospital, Taichung, Taiwan
| | - Ping-Tao Tseng
- Department of Psychology, College of Medical and Health Science, Asia University, Taichung, Taiwan; Prospect Clinic for Otorhinolaryngology & Neurology, Kaohsiung City, Taiwan; Institute of Biomedical Sciences, National Sun Yat-sen University, Kaohsiung, Taiwan; Institute of Precision Medicine, National Sun Yat-sen University, Kaohsiung, Taiwan
| | - Xiang Hu
- The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Bing-Yan Zeng
- Institute of Biomedical Sciences, National Sun Yat-sen University, Kaohsiung, Taiwan; Department of Internal Medicine, E-Da Dachang Hospital, Kaohsiung, Taiwan
| | - Jane Pei-Chen Chang
- Department of Psychiatry & Mind-Body Interface Laboratory (MBI-Lab), China Medical University Hospital, Taichung, Taiwan; Department of Psychological Medicine, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, UK; College of Medicine, China Medical University, Taichung, Taiwan
| | - Yang Liu
- School of Public Health and Management & Institute of Lipids Medicine, Wenzhou Medical University, Wenzhou, China
| | - Wei-Jie Chu
- School of Public Health and Management & Institute of Lipids Medicine, Wenzhou Medical University, Wenzhou, China
| | - Shuang-Shuang Zhang
- School of Public Health and Management & Institute of Lipids Medicine, Wenzhou Medical University, Wenzhou, China
| | - Zhi-Liang Zhou
- The 2nd Affiliated Hospital and Yuying Children's Hospital, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Chih-Sheng Chu
- Center of Lipid Bioscience, Kaohsiung Medical University Hospital, Kaohsiung, Taiwan
| | - Cheng-Ho Chang
- Department of Psychiatry, Kaohsiung Veterans General Hospital, Kaohsiung, Taiwan; Department of Food Science, National Pingtung University of Science and Technology, Pingtung, Taiwan
| | - Yu-Kang Tu
- Institute of Epidemiology & Preventive Medicine, College of Public Health, National Taiwan University, Taipei, Taiwan; Department of Dentistry, National Taiwan University Hospital, Taipei, Taiwan
| | - Yi-Cheng Wu
- Department of Sports Medicine, Landseed International Hospital, Taoyuan, Taiwan
| | - Brendon Stubbs
- Department of Psychological Medicine, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, UK; Physiotherapy Department, South London and Maudsley NHS Foundation Trust, London, UK; Positive Ageing Research Institute (PARI), Faculty of Health, Social Care, Medicine and Education, Anglia Ruskin University, Chelmsford, UK
| | - Andre F Carvalho
- Innovation in Mental and Physical Health and Clinical Treatment (IMPACT) Strategic Research Centre, School of Medicine, Barwon Health, Deakin University, Geelong, VIC, Australia
| | - Pao-Yen Lin
- Department of Psychiatry, Kaohsiung Chang Gung Memorial Hospital and Chang Gung University College of Medicine, Kaohsiung, Taiwan; Institute for Translational Research in Biomedical Sciences, Kaohsiung Chang Gung Memorial Hospital, Kaohsiung, Taiwan
| | - Yutaka J Matsuoka
- Former Division Chief of Health Care Research, National Cancer Centre Japan, Japan
| | - Mein-Woei Suen
- Department of Psychology, College of Medical and Health Science, Asia University, Taichung, Taiwan; Gender Equality Education and Research Center, Asia University, Taichung, Taiwan; Department of Medical Research, Asia University Hospital, Asia University, Taichung, Taiwan; Department of Medical Research, China Medical University Hospital, China Medical University, Taichung, Taiwan
| | - Kuan-Pin Su
- School of Public Health and Management & Institute of Lipids Medicine, Wenzhou Medical University, Wenzhou, China; Department of Psychiatry & Mind-Body Interface Laboratory (MBI-Lab), China Medical University Hospital, Taichung, Taiwan; College of Medicine, China Medical University, Taichung, Taiwan; An-Nan Hospital, China Medical University, Tainan, Taiwan.
| |
Collapse
|
96
|
Schefelker JM, Peterson AL. Screening and Management of Dyslipidemia in Children and Adolescents. J Clin Med 2022; 11:6479. [PMID: 36362707 PMCID: PMC9656613 DOI: 10.3390/jcm11216479] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2022] [Revised: 10/24/2022] [Accepted: 10/29/2022] [Indexed: 07/30/2023] Open
Abstract
This review provides an overview of pediatric dyslipidemia emphasizing screening and treatment recommendations. The presence of risk factors for cardiovascular disease in childhood poses significant risk for the development of atherosclerotic cardiovascular disease and cardiovascular events in adulthood. While atherogenic dyslipidemia is the most common dyslipidemia seen in children and can be suspected based on the presence of risk factors (such as obesity), familial hypercholesterolemia can be found in children with no risk factors. As such, universal cholesterol screening is recommended to identify children with these disorders in order to initiate treatment and reduce the risk of future cardiovascular disease. Treatment of pediatric dyslipidemia begins with lifestyle modifications, but primary genetic dyslipidemias may require medications such as statins. As pediatric lipid disorders often have genetic or familial components, it is important that all physicians are aware that cardiovascular risk begins in childhood, and can both identify these disorders in pediatric patients and counsel their adult patients with dyslipidemia to have their children screened.
Collapse
Affiliation(s)
| | - Amy L. Peterson
- Department of Pediatrics, Division of Pediatric Cardiology, School of Medicine and Public Health, University of Wisconsin, Madison, WI 53792, USA
| |
Collapse
|
97
|
Yang Y, Deng W, Wang Y, Li T, Chen Y, Long C, Wen Q, Wu Y, Chen Q. The effect of omega-3 fatty acids and its combination with statins on lipid profile in patients with hypertriglyceridemia: A systematic review and meta-analysis of randomized controlled trials. Front Nutr 2022; 9:1039056. [PMID: 36313109 PMCID: PMC9609787 DOI: 10.3389/fnut.2022.1039056] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2022] [Accepted: 09/27/2022] [Indexed: 11/13/2022] Open
Abstract
Background/Aim Omega-3 fatty acids (OM3-FA), a promising treatment for high triglycerides, have gradually attracted public attention. However, some studies showed that their application presented tricky problems, like increasing low-density lipoprotein cholesterol (LDL-C) levels. This study aimed to systematically evaluate the effect of OM3-FA or their combination with statins on the lipid profile in patients with hypertriglyceridemia. Materials and methods This study followed the preferred reporting items for systematic reviews and meta-analyses (PRISMA 2020) guidelines. PubMed, Embase, Web of science, and Cochrane library were searched up to May 15, 2022. The random-effects model was applied to calculate the mean difference (MD) and associated 95% confidence intervals (CI). Results This meta-analysis included 32 studies with 15,903 subjects. When OM3-FA was used as monotherapy compared with placebo, it significantly decreased TG (MD: -39.81, 95% CI: -54.94 to -24.69; p < 0.001), TC (MD: -2.98, 95% CI: -5.72 to -0.25, p = 0.03), very low-density lipoprotein cholesterol (VLDL-C) (MD: -25.12, 95% CI: -37.09 to -13.14; p < 0.001), and non-high-density lipoprotein cholesterol (non-HDL-C) levels (MD: -5.42, 95% CI: -8.06 to-2.78; p < 0.001), and greatly increased LDL-C (MD: 9.10, 95% CI: 4.27 to 13.94; p < 0.001) and HDL levels (MD: 1.60, 95% CI: 0.06 to 3.15; p = 0.04). Regarding apolipoprotein B (Apo-B) and apolipoprotein AI (Apo-AI), no significant effect was identified. When OM3-FA was combined with statins, significant reductions were observed in the concentrations of TG (MD: -29.63, 95% CI: -36.24 to -23.02; p < 0.001), TC (MD: -6.87, 95% CI: -9.30 to -4.45, p < 0.001), VLDL-C (-20.13, 95% CI: -24.76 to -15.50; p < 0.001), non-HDL-C (MD: -8.71, 95% CI: -11.45 to -5.98; p < 0.001), Apo-B (MD: -3.50, 95% CI: -5.37 to -1.64; p < 0.001), and Apo-AI (MD: -2.01, 95% CI: -3.07 to -0.95; p < 0.001). However, the combined therapy did not exert significant changes on the levels of high-density lipoprotein cholesterol (HDL-C) and LDL-C compared to control group. Conclusion The use of OM3-FA either as monotherapy or in combination with statins may potentially reduce the levels of TG, TC, VLDL-C, non-HDL-C, Apo-B, and Apo-AI while increasing the levels of LDL-C and HDL-C. Nevertheless, the effects of OM3-FA observed in this review should be interpreted with caution due to the high heterogeneity between the included studies. Systematic review registration [https://www.crd.york.ac.uk/prospero/], identifier [CRD42022329552].
Collapse
Affiliation(s)
- Yunjiao Yang
- Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, China,School of Clinical Medicine, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Wen Deng
- Mianyang Attached Hospital of Chengdu University of Traditional Chinese Medicine, Mianyang, China
| | - Yanmei Wang
- Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, China,School of Clinical Medicine, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Tongyi Li
- Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, China,School of Clinical Medicine, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Yiding Chen
- Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, China,School of Clinical Medicine, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Cong Long
- Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, China,School of Clinical Medicine, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Qing Wen
- Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, China,School of Clinical Medicine, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Yue Wu
- Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, China,School of Clinical Medicine, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Qiu Chen
- Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, China,*Correspondence: Qiu Chen,
| |
Collapse
|
98
|
Sala-Vila A, Fleming J, Kris-Etherton P, Ros E. Impact of α-Linolenic Acid, the Vegetable ω-3 Fatty Acid, on Cardiovascular Disease and Cognition. Adv Nutr 2022; 13:1584-1602. [PMID: 35170723 PMCID: PMC9526859 DOI: 10.1093/advances/nmac016] [Citation(s) in RCA: 41] [Impact Index Per Article: 13.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2021] [Revised: 12/31/2021] [Accepted: 02/11/2022] [Indexed: 01/28/2023] Open
Abstract
Given the evidence of the health benefits of plant-based diets and long-chain n-3 (ω-3) fatty acids, there is keen interest in better understanding the role of α-linolenic acid (ALA), a plant-derived n-3 fatty acid, on cardiometabolic diseases and cognition. There is increasing evidence for ALA largely based on its major food sources (i.e., walnuts and flaxseed); however, this lags behind our understanding of long-chain n-3 fatty acids. Meta-analyses of observational studies have shown that increasing dietary ALA is associated with a 10% lower risk of total cardiovascular disease and a 20% reduced risk of fatal coronary heart disease. Three randomized controlled trials (RCTs) [AlphaOmega trial, Prevención con Dieta Mediterránea (PREDIMED) trial, and Lyon Diet Heart Study] all showed benefits of diets high in ALA on cardiovascular-related outcomes, but the AlphaOmega trial, designed to specifically evaluate ALA effects, only showed a trend for benefit. RCTs have shown that dietary ALA reduced total cholesterol, LDL cholesterol, triglycerides, and blood pressure, and epidemiologic studies and some trials also have shown an anti-inflammatory effect of ALA, which collectively account for, in part, the cardiovascular benefits of ALA. A meta-analysis reported a trend toward diabetes risk reduction with both dietary and biomarker ALA. For metabolic syndrome and obesity, the evidence for ALA benefits is inconclusive. The role of ALA in cognition is in the early stages but shows promising evidence of counteracting cognitive impairment. Much has been learned about the health benefits of ALA and with additional research we will be better positioned to make strong evidence-based dietary recommendations for the reduction of many chronic diseases.
Collapse
Affiliation(s)
- Aleix Sala-Vila
- Fatty Acid Research Institute, Sioux Falls, SD, USA
- Cardiovascular Risk and Nutrition, Hospital del Mar Medical Research Institute (IMIM), Barcelona, Spain
| | - Jennifer Fleming
- Department of Nutritional Sciences, College of Health and Human Development, Pennsylvania State University, University Park, PA, USA
| | - Penny Kris-Etherton
- Department of Nutritional Sciences, College of Health and Human Development, Pennsylvania State University, University Park, PA, USA
| | - Emilio Ros
- Lipid Clinic, Endocrinology and Nutrition Service, Hospital Clínic, August Pi i Sunyer Biomedical Research Institute (IDIBAPS), Barcelona, Spain
- CIBER Physiopathology of Obesity and Nutrition (CIBEROBN), Instituto de Salud Carlos III (ISCIII), Madrid, Spain
| |
Collapse
|
99
|
Jiang M, Ding H, Huang Y, Wang L. Shear Stress and Metabolic Disorders-Two Sides of the Same Plaque. Antioxid Redox Signal 2022; 37:820-841. [PMID: 34148374 DOI: 10.1089/ars.2021.0126] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Significance: Shear stress and metabolic disorder are the two sides of the same atherosclerotic coin. Atherosclerotic lesions are prone to develop at branches and curvatures of arteries, which are exposed to oscillatory and low shear stress exerted by blood flow. Meanwhile, metabolic disorders are pivotal contributors to the formation and advancement of atherosclerotic plaques. Recent Advances: Accumulated evidence has provided insight into the impact and mechanisms of biomechanical forces and metabolic disorder on atherogenesis, in association with mechanotransduction, epigenetic regulation, and so on. Moreover, recent studies have shed light on the cross talk between the two drivers of atherosclerosis. Critical Issues: There are extensive cross talk and interactions between shear stress and metabolic disorder during the pathogenesis of atherosclerosis. The communications may amplify the proatherogenic effects through increasing oxidative stress and inflammation. Nonetheless, the precise mechanisms underlying such interactions remain to be fully elucidated as the cross talk network is considerably complex. Future Directions: A better understanding of the cross talk network may confer benefits for a more comprehensive clinical management of atherosclerosis. Critical mediators of the cross talk may serve as promising therapeutic targets for atherosclerotic vascular diseases, as they can inhibit effects from both sides of the plaque. Hence, further in-depth investigations with advanced omics approaches are required to develop novel and effective therapeutic strategies against atherosclerosis. Antioxid. Redox Signal. 37, 820-841.
Collapse
Affiliation(s)
- Minchun Jiang
- Heart and Vascular Institute, School of Biomedical Sciences, The Chinese University of Hong Kong, Hong Kong SAR, China.,Shenzhen Research Institute, Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Hong Kong SAR, China
| | - Huanyu Ding
- Heart and Vascular Institute, School of Biomedical Sciences, The Chinese University of Hong Kong, Hong Kong SAR, China.,Shenzhen Research Institute, Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Hong Kong SAR, China
| | - Yu Huang
- Heart and Vascular Institute, School of Biomedical Sciences, The Chinese University of Hong Kong, Hong Kong SAR, China.,Shenzhen Research Institute, Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Hong Kong SAR, China
| | - Li Wang
- Heart and Vascular Institute, School of Biomedical Sciences, The Chinese University of Hong Kong, Hong Kong SAR, China.,Shenzhen Research Institute, Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Hong Kong SAR, China
| |
Collapse
|
100
|
Huang Y, Deng Y, Zhang P, Lin J, Guo D, Yang L, Liu D, Xu B, Huang C, Zhang H. Associations of fish oil supplementation with incident dementia: Evidence from the UK Biobank cohort study. Front Neurosci 2022; 16:910977. [PMID: 36161159 PMCID: PMC9489907 DOI: 10.3389/fnins.2022.910977] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2022] [Accepted: 08/17/2022] [Indexed: 11/13/2022] Open
Abstract
Background Although numerous studies have investigated the association of dietary intake of omega-3 fatty acids with cognitive function and the risks of dementia, the relationship between fish oil supplementation and incident dementia in a large population-based cohort study has not yet well studied. Materials and methods A total of 211,094 community-dwelling older persons over 60 years from the UK Biobank cohorts enrolled between 2006 and 2010 that reported regularly taking fish oil and had no dementia at baseline, was included in the present study. All participants completed an electronic questionnaire regarding habitual use of supplements including fish oil. Results Overall, 83,283 (39.5%) participants reported regularly taking fish oil at baseline. Of 211,094 participants with the median age was 64.1 years, 5,274 participants developed dementia events during a median follow-up of 11.7 years, with 3,290 individuals derived from fish oil non-users. In the multivariable adjusted models, the adjusted hazard ratios (HRs) associated with fish oil supplementation for all-cause dementia, vascular dementia, frontotemporal dementia, and other dementia were 0.91 [CI = 0.84-0.97], 0.83 [CI = 0.71-0.97], 0.43 [CI = 0.26-0.72], 0.90 [CI = 0.82-0.98], respectively (all P < 0.05). However, no significant association between fish oil supplementation and Alzheimer's disease was found (HR = 1.00 [CI = 0.89-1.12], P = 0.977). In the subgroup analyses, the associations between use of fish oil and the risk of all-cause dementia (P for interaction = 0.007) and vascular dementia were stronger among men (P for interaction = 0.026). Conclusion Among older adults, regular fish oil supplementation was significantly associated with a lower risks of incident all-cause dementia, as well as vascular dementia, frontotemporal dementia and other dementia but not Alzheimer's disease. These findings support that habitual use of fish oils may be beneficial for the prevention of dementia in clinical practice.
Collapse
Affiliation(s)
- Yan Huang
- Department of Endocrinology and Metabolism, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Yajuan Deng
- Department of Endocrinology and Metabolism, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Peizhen Zhang
- Department of Endocrinology and Metabolism, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Jiayang Lin
- Department of Endocrinology and Metabolism, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Dan Guo
- Department of Endocrinology and Metabolism, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Linjie Yang
- Department of Endocrinology and Metabolism, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Deying Liu
- Department of Endocrinology and Metabolism, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Bingyan Xu
- Department of Endocrinology and Metabolism, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Chensihan Huang
- Department of Endocrinology and Metabolism, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Huijie Zhang
- Department of Endocrinology and Metabolism, Nanfang Hospital, Southern Medical University, Guangzhou, China
- Guangdong Provincial Key Laboratory of Shock and Microcirculation, Guangzhou, China
- Department of Food Safety and Health Research Center, School of Public Health, Southern Medical University, Guangzhou, Guangdong, China
| |
Collapse
|