1
|
Chen Y, Touboul R, Chen Y, Chang CL. Strategic delivery of omega-3 fatty acids for modulating inflammatory neurodegenerative diseases. Front Aging Neurosci 2025; 17:1535094. [PMID: 40166615 PMCID: PMC11955621 DOI: 10.3389/fnagi.2025.1535094] [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: 11/26/2024] [Accepted: 02/28/2025] [Indexed: 04/02/2025] Open
Abstract
Objectives Early-life inflammatory events like infections and injuries may predispose the brain to Alzheimer's disease (AD) by disrupting neurodevelopment and raising vulnerability. The association between early neuroinflammation and subsequent neurodegeneration leading to dementia remains unclear. We hypothesize that omega-3 (n-3) fatty acids (FA), especially eicosapentaenoic acid (EPA) and docosahexaenoic acid (DHA), positively regulate neuro-immune cells, preserving their cell membrane structure and metabolic homeostasis. Our study examined whether strategic delivery of n-3 FA via injectable n-3 triglycerides (TG) can influence microglial lipid metabolism to prevent or delay AD progression. Methods and results We characterized n-3 treatment effects on modulating lipid and metabolic homeostasis in microglia during the critical window of brain development. Our preliminary studies on determining the effects of early n-3 treatment on brain cell homeostasis indicate that perinatal bolus n-3 TG injections suppressed activation of gliosis-associated markers in young mice predisposed to AD (5xFAD) and yielded sustained regulatory effects on the expression of inflammatory molecules, such as interleukin-6 (Il6) and tumor necrosis factor-alpha (Tnfα), in adult brains. A significant increase in high-frequency ultrasonic vocalizations (USV) was observed in P6 5xFAD mice that received perinatal n-3 compared to vehicle control, implicating enhanced active communication patterns. Improvement in behavior deficits was observed in n-3-treated adult AD mice. Perinatal n-3 TG treatment modified brain lipid composition in young offspring, increasing key membrane lipid species, such as phospholipids (PL) and lysophospholipids (lysoPL). Pro-inflammatory sphingolipids associated with neurodegeneration, including lactosylceramide, were significantly lower in mice treated with n-3 than those in saline-treated AD mice. Conclusion Our study establishes a proof of principle for targeting brain immune cell metabolism with injectable n-3 TG to mitigate neuroinflammation in AD pathogenesis, paving the way for future research into early treatments for related central nervous system (CNS) disorders.
Collapse
Affiliation(s)
- Yixin Chen
- Institute of Human Nutrition, Columbia University Vagelos College of Physicians and Surgeons, New York, NY, United States
| | - Roni Touboul
- Institute of Human Nutrition, Columbia University Vagelos College of Physicians and Surgeons, New York, NY, United States
| | - Yao Chen
- Institute of Human Nutrition, Columbia University Vagelos College of Physicians and Surgeons, New York, NY, United States
| | - Chuchun L. Chang
- Institute of Human Nutrition, Columbia University Vagelos College of Physicians and Surgeons, New York, NY, United States
- Division of Gastroenterology, Hepatology, and Nutrition, Department of Pediatrics, Columbia University Vagelos College of Physicians and Surgeons, New York, NY, United States
| |
Collapse
|
2
|
Kim JY, Kong SYJ, Jung E, Cho YS. Omega-3 Fatty Acids as Potential Predictors of Sudden Cardiac Death and Cardiovascular Mortality: A Systematic Review and Meta-Analysis. J Clin Med 2024; 14:26. [PMID: 39797109 PMCID: PMC11722046 DOI: 10.3390/jcm14010026] [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: 11/25/2024] [Revised: 12/12/2024] [Accepted: 12/14/2024] [Indexed: 01/13/2025] Open
Abstract
Background/Objectives: Sudden cardiac death (SCD) poses a significant burden on the modern-day public health system; however, while our understanding of the underlying pathophysiology is still evolving and may not be complete, many insights are known and applied every day. Targeted prevention methods are continually being developed and refined. We conducted a systemic review and meta-analysis to identify a blood nutritional biomarker that can predict and screen population groups at high risk for cardiovascular disease mortality (CVD mortality) or SCD. Methods: The literature search was conducted from November 2023 to 31 January 2024. Based on previous literature research, we studied the association between omega-3 fatty acids (n-3 FA; eicosapentaenoic acid [EPA], docosapentaenoic acid [DPA] and docosahexaenoic acid [DHA]) and SCD and/or CVD mortality individually and in combination. We evaluated and selected 10 prospective cohort studies out of 1789 related publications, with an average follow-up period of 8.7 years. A multivariate adjusted hazard ratio (HR) with 95% confidence interval (CI) was calculated and sub-analyzed to obtain a general trend of reduced risk of SCD in a high n-3 FA intake group from the general population. Results: Finally, we included 10 articles with a total sample size of 310,955 participants. We found an inverse association between circulating n-3 FA levels and SCD. The summary HR of SCD and CVD mortality for high versus low circulating n-3 FA levels (EPA + DHA + DPA) in serum plasma phospholipid was 0.55 (95% CI: 0.37-0.82) and that of EPA + DHA in RBC was 0.67 (95% CI: 0.45-0.99). Based on the sub-analysis, the HR of EPA (%) was 0.79 (95% CI: 0.60-0.82) and that of DHA (%) was 0.72 (95% CI: 0.60-0.87). Conclusions: Our results suggest a potential cardio-protective association between high EPA and DHA levels in blood and a reduced incidence of adverse cardiac events.
Collapse
Affiliation(s)
- Ji Young Kim
- Department of Medicine, Seoul National University, Seoul 08826, Republic of Korea;
| | | | - Eujene Jung
- Department of Emergency Medicine, Chonnam National University Hospital, Gwangju 61469, Republic of Korea
- Department of Emergency Medicine, Chonnam National University Medical School, Gwangju 61469, Republic of Korea
| | - Yong Soo Cho
- Department of Emergency Medicine, Chonnam National University Hospital, Gwangju 61469, Republic of Korea
- Department of Emergency Medicine, Chonnam National University Medical School, Gwangju 61469, Republic of Korea
| |
Collapse
|
3
|
Zirpoli H, Bernis ME, Sabir H, Manual Kollareth DJ, Hamilton JA, Huang N, Ng J, Sosunov SA, Gaebler B, Ten VS, Deckelbaum RJ. Omega-3 fatty acid diglyceride emulsions as a novel injectable acute therapeutic in neonatal hypoxic-ischemic brain injury. Biomed Pharmacother 2024; 175:116749. [PMID: 38761420 PMCID: PMC11156760 DOI: 10.1016/j.biopha.2024.116749] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2024] [Revised: 05/10/2024] [Accepted: 05/10/2024] [Indexed: 05/20/2024] Open
Abstract
Hypoxic-ischemic encephalopathy (HIE), resulting from a lack of blood flow and oxygen before or during newborn delivery, is a leading cause of cerebral palsy and neurological disability in children. Therapeutic hypothermia (TH), the current standard of care in HIE, is only beneficial in 1 of 7-8 cases. Therefore, there is a critical need for more efficient treatments. We have previously reported that omega-3 (n-3) fatty acids (FA) carried by triglyceride (TG) lipid emulsions provide neuroprotection after experimental hypoxic-ischemic (HI) injury in neonatal mice. Herein, we propose a novel acute therapeutic approach using an n-3 diglyceride (DG) lipid emulsions. Importantly, n-3 DG preparations had much smaller particle size compared to commercially available or lab-made n-3 TG emulsions. We showed that n-3 DG molecules have the advantage of incorporating at substantially higher levels than n-3 TG into an in vitro model of phospholipid membranes. We also observed that n-3 DG after parenteral administration in neonatal mice reaches the bloodstream more rapidly than n-3 TG. Using neonatal HI brain injury models in mice and rats, we found that n-3 DG emulsions provide superior neuroprotection than n-3 TG emulsions or TH in decreasing brain infarct size. Additionally, we found that n-3 DGs attenuate microgliosis and astrogliosis. Thus, n-3 DG emulsions are a superior, promising, and novel therapy for treating HIE.
Collapse
Affiliation(s)
- Hylde Zirpoli
- Institute of Human Nutrition, Columbia University Irving Medical Center, New York, NY 10032, USA; Department of Pediatrics, Vagelos College of Physicians and Surgeons, Columbia University Irving Medical Center, New York, NY 10032, USA.
| | - Maria Eugenia Bernis
- Department of Neonatology and Pediatric Intensive Care, Children's Hospital, University of Bonn, Germany; Deutsches Zentrum für Neurodegenerative Erkrankungen (DZNE), Bonn 53127, Germany
| | - Hemmen Sabir
- Department of Neonatology and Pediatric Intensive Care, Children's Hospital, University of Bonn, Germany; Deutsches Zentrum für Neurodegenerative Erkrankungen (DZNE), Bonn 53127, Germany
| | - Denny Joseph Manual Kollareth
- Department of Pediatrics, Vagelos College of Physicians and Surgeons, Columbia University Irving Medical Center, New York, NY 10032, USA
| | - James A Hamilton
- Department of Physiology & Biophysics, Department of Biomedical Engineering, Boston University School of Medicine, Boston, MA 02215, USA
| | - Nasi Huang
- Department of Physiology & Biophysics, Department of Biomedical Engineering, Boston University School of Medicine, Boston, MA 02215, USA
| | - Jesse Ng
- Department of Physiology & Biophysics, Department of Biomedical Engineering, Boston University School of Medicine, Boston, MA 02215, USA
| | - Sergey A Sosunov
- Department of Pediatrics, Vagelos College of Physicians and Surgeons, Columbia University Irving Medical Center, New York, NY 10032, USA; Division of Neonatology, Department of Pediatrics, Robert Wood Johnson Medical School, Rutgers University, New Brunswick, NJ 08901, USA
| | | | - Vadim S Ten
- Department of Pediatrics, Vagelos College of Physicians and Surgeons, Columbia University Irving Medical Center, New York, NY 10032, USA; Division of Neonatology, Department of Pediatrics, Robert Wood Johnson Medical School, Rutgers University, New Brunswick, NJ 08901, USA
| | - Richard J Deckelbaum
- Institute of Human Nutrition, Columbia University Irving Medical Center, New York, NY 10032, USA; Department of Pediatrics, Vagelos College of Physicians and Surgeons, Columbia University Irving Medical Center, New York, NY 10032, USA
| |
Collapse
|
4
|
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
|
5
|
Mosavi SS, Rabizadeh S, Yadegar A, Seifouri S, Mohammadi F, Qahremani R, Salehi SS, Rajab A, Esteghamati A, Nakhjavani M. Therapeutic effects of resveratrol and Omega-3 in mice atherosclerosis: focus on histopathological changes. BMC Complement Med Ther 2023; 23:81. [PMID: 36932392 PMCID: PMC10024363 DOI: 10.1186/s12906-023-03899-9] [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: 12/14/2022] [Accepted: 02/27/2023] [Indexed: 03/19/2023] Open
Abstract
BACKGROUND Resveratrol and omega-3 have been shown to prevent atherosclerosis. However, histopathological changes and their comparison have not been studied well. This study investigated the therapeutic effects of resveratrol and omega-3 in experimental atherosclerosis of mice. METHODS We divided sixty 6-week-old male C57BL/6 mice into six groups and followed for 10 weeks: (1) standard diet, (2) atherogenic diet, (3) atherogenic diet along with resveratrol from the start of the sixth week, (4) atherogenic diet along with omega-3 from the start of the sixth week, (5) standard diet along with resveratrol from the start of the sixth week, (6) standard diet along with omega-3 from the start of the sixth week. RESULTS The mice fed on an atherogenic diet had a larger fat area and a thicker aortic wall thickness than mice fed on a standard diet. The use of omega-3 and resveratrol in the mice with an atherogenic diet resulted in a significantly reduced fat area (p-value = 0.003), and resveratrol had a significantly higher effect. Omega-3 or resveratrol induced a significant reduction in aortic wall thickness in mice on an atherogenic diet, and there was no significant difference between them. Among the mice with a standard diet, this study did not observe any significant changes in the fat area or the aortic wall thickness with the consumption of omega-3 or resveratrol. CONCLUSIONS Resveratrol and omega-3 had a regressive and therapeutic role in atherosclerosis, with a more significant effect in favor of resveratrol.
Collapse
Affiliation(s)
- Shamsi Sadat Mosavi
- grid.414574.70000 0004 0369 3463Endocrinology and Metabolism Research Center (EMRC), Vali-Asr Hospital, Tehran University of Medical Sciences, Imam Khomeini Hospital Complex, Tohid Squre, P.O Box: 13145-784, Tehran, Iran
| | - Soghra Rabizadeh
- grid.414574.70000 0004 0369 3463Endocrinology and Metabolism Research Center (EMRC), Vali-Asr Hospital, Tehran University of Medical Sciences, Imam Khomeini Hospital Complex, Tohid Squre, P.O Box: 13145-784, Tehran, Iran
| | - Amirhossein Yadegar
- grid.414574.70000 0004 0369 3463Endocrinology and Metabolism Research Center (EMRC), Vali-Asr Hospital, Tehran University of Medical Sciences, Imam Khomeini Hospital Complex, Tohid Squre, P.O Box: 13145-784, Tehran, Iran
| | - Sara Seifouri
- grid.414574.70000 0004 0369 3463Endocrinology and Metabolism Research Center (EMRC), Vali-Asr Hospital, Tehran University of Medical Sciences, Imam Khomeini Hospital Complex, Tohid Squre, P.O Box: 13145-784, Tehran, Iran
| | - Fatemeh Mohammadi
- grid.414574.70000 0004 0369 3463Endocrinology and Metabolism Research Center (EMRC), Vali-Asr Hospital, Tehran University of Medical Sciences, Imam Khomeini Hospital Complex, Tohid Squre, P.O Box: 13145-784, Tehran, Iran
| | - Reihane Qahremani
- grid.414574.70000 0004 0369 3463Endocrinology and Metabolism Research Center (EMRC), Vali-Asr Hospital, Tehran University of Medical Sciences, Imam Khomeini Hospital Complex, Tohid Squre, P.O Box: 13145-784, Tehran, Iran
| | - Salome Sadat Salehi
- grid.414574.70000 0004 0369 3463Endocrinology and Metabolism Research Center (EMRC), Vali-Asr Hospital, Tehran University of Medical Sciences, Imam Khomeini Hospital Complex, Tohid Squre, P.O Box: 13145-784, Tehran, Iran
| | - Armin Rajab
- grid.414574.70000 0004 0369 3463Endocrinology and Metabolism Research Center (EMRC), Vali-Asr Hospital, Tehran University of Medical Sciences, Imam Khomeini Hospital Complex, Tohid Squre, P.O Box: 13145-784, Tehran, Iran
| | - Alireza Esteghamati
- grid.414574.70000 0004 0369 3463Endocrinology and Metabolism Research Center (EMRC), Vali-Asr Hospital, Tehran University of Medical Sciences, Imam Khomeini Hospital Complex, Tohid Squre, P.O Box: 13145-784, Tehran, Iran
| | - Manouchehr Nakhjavani
- grid.414574.70000 0004 0369 3463Endocrinology and Metabolism Research Center (EMRC), Vali-Asr Hospital, Tehran University of Medical Sciences, Imam Khomeini Hospital Complex, Tohid Squre, P.O Box: 13145-784, Tehran, Iran
| |
Collapse
|
6
|
Chen L, Yang CS, Chen SD, Zhou QX, Wang GQ, Cai SL, Li WH, Luo HZ. Multi-omics characterization of the unsaturated fatty acid biosynthesis pathway in colon cancer. Am J Cancer Res 2022; 12:3985-4000. [PMID: 36119831 PMCID: PMC9442000] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2022] [Accepted: 07/30/2022] [Indexed: 06/15/2023] Open
Abstract
The biosynthesis of unsaturated fatty acids is involved in the initiation and progression of colon adenocarcinoma (COAD). In this study, we aimed to investigate the multi-omics characteristics of unsaturated fatty acid biosynthesis-related genes and explore their prognostic value in colon cancer by analyzing the data from The Cancer Genome Atlas (TCGA) and Gene Expression Omnibus (GEO) databases. An unsaturated fatty acid biosynthesis pathway related-genes enrichment score (BUFAS) was constructed utilizing the single sample gene set enrichment analysis (ssGSEA). We discovered that a high BUFAS was associated with longer overall survival (OS) in both the training and the validation sets. Multivariable analysis including the clinical characteristics further verified the independent prognostic value of the BUFAS in both the TCGA-COAD and the GSE39582 datasets. In addition, GSEA analysis revealed that BUFAS was positively associated with several signaling pathways, including MTORC1, peroxisome, and pathways related to fatty acid metabolism, while was negatively associated with other signaling pathways, such as hedgehog, NOTCH, and Wnt/beta-catenin pathway. Furthermore, in the COAD cell lines of the Genomics of Drug Sensitivity in Cancer (GDSC) database, we found that BUFAS was positively correlated with the drug sensitivities of cisplatin, gemcitabine, camptothecin, lapatinib, and afatinib, while was negatively correlated with that of ponatinib. Moreover, in the COAD single-cell transcriptomic dataset (GSE146771), the BUFAS varied among different cell types and was enriched in mast cells and fibroblasts. Taken together, the BUFAS we constructed could be used as an independent prognostic signature in predicting the OS and drug resistance of colon cancer. Unsaturated fatty acid biosynthesis pathway might serve as potential therapeutic targets for cancer treatment.
Collapse
Affiliation(s)
- Ling Chen
- Department of Gastrointestinal Surgery, Xiangya HospitalNo. 87 Xiangya Road, Changsha, Hunan, China
| | - Chang-Shun Yang
- Department of Surgical Oncology, Shengli Clinical Medical College of Fujian Medical UniversityNo. 134 East Street, Fuzhou, Fujian, China
| | - Si-Dong Chen
- Burning Rock Biotech, Building 6, Phase 2, Standard Industrial Unit, No. 7 LuoXuan 4th Road, International Biotech IslandGuangzhou, Guangdong, China
| | - Qiao-Xia Zhou
- Burning Rock Biotech, Building 6, Phase 2, Standard Industrial Unit, No. 7 LuoXuan 4th Road, International Biotech IslandGuangzhou, Guangdong, China
| | - Guo-Qiang Wang
- Burning Rock Biotech, Building 6, Phase 2, Standard Industrial Unit, No. 7 LuoXuan 4th Road, International Biotech IslandGuangzhou, Guangdong, China
| | - Shang-Li Cai
- Burning Rock Biotech, Building 6, Phase 2, Standard Industrial Unit, No. 7 LuoXuan 4th Road, International Biotech IslandGuangzhou, Guangdong, China
| | - Wei-Hua Li
- Department of Surgical Oncology, Shengli Clinical Medical College of Fujian Medical UniversityNo. 134 East Street, Fuzhou, Fujian, China
| | - Hong-Zhi Luo
- Department of Tumor Surgery, Zhongshan City People’s HospitalNo. 2 Sunwen Middle Road, Zhongshan, Guangdong, China
| |
Collapse
|
7
|
Reilly NA, Lutgens E, Kuiper J, Heijmans BT, Jukema JW. Effects of fatty acids on T cell function: role in atherosclerosis. Nat Rev Cardiol 2021; 18:824-837. [PMID: 34253911 DOI: 10.1038/s41569-021-00582-9] [Citation(s) in RCA: 31] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 06/01/2021] [Indexed: 01/08/2023]
Abstract
T cells are among the most common cell types present in atherosclerotic plaques and are increasingly being recognized as a central mediator in atherosclerosis development and progression. At the same time, triglycerides and fatty acids have re-emerged as crucial risk factors for atherosclerosis. Triglycerides and fatty acids are important components of the milieu to which the T cell is exposed from the circulation to the plaque, and increasing evidence shows that fatty acids influence T cell function. In this Review, we discuss the effects of fatty acids on four components of the T cell response - metabolism, activation, proliferation and polarization - and the influence of these changes on the pathogenesis of atherosclerosis. We also discuss how quiescent T cells can undergo a type of metabolic reprogramming induced by exposure to fatty acids in the circulation that influences the subsequent functions of T cells after activation, such as in atherosclerotic plaques.
Collapse
Affiliation(s)
- Nathalie A Reilly
- Molecular Epidemiology, Department of Biomedical Data Sciences, Leiden University Medical Centre, Leiden, Netherlands
- Department of Cardiology, Leiden University Medical Centre, Leiden, Netherlands
| | - Esther Lutgens
- Department of Medical Biochemistry, Amsterdam University Medical Centre, Amsterdam, Netherlands
- Institute for Cardiovascular Prevention (IPEK), Ludwig-Maximilians Universität, Munich, Germany
- German Centre for Cardiovascular Research (DZHK), partner site Munich Heart Alliance, Munich, Germany
| | - Johan Kuiper
- Leiden Academic Centre for Drug Research, Division of Biotherapeutics, Leiden University, Leiden, Netherlands
| | - Bastiaan T Heijmans
- Molecular Epidemiology, Department of Biomedical Data Sciences, Leiden University Medical Centre, Leiden, Netherlands
| | - J Wouter Jukema
- Department of Cardiology, Leiden University Medical Centre, Leiden, Netherlands.
- Netherlands Heart Institute, Utrecht, Netherlands.
| |
Collapse
|
8
|
Bersch-Ferreira AC, Hall WL, Santos RHN, Torreglosa CR, Sampaio G, Tereza da Silva J, Alves R, Ross MB, Gehringer MO, Kovacs C, Marcadenti A, Magnoni D, Weber B, Rogero MM. The effect of the a regional cardioprotective nutritional program on inflammatory biomarkers and metabolic risk factors in secondary prevention for cardiovascular disease, a randomised trial. Clin Nutr 2021; 40:3828-3835. [PMID: 34130029 DOI: 10.1016/j.clnu.2021.04.035] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2020] [Revised: 01/28/2021] [Accepted: 04/19/2021] [Indexed: 11/18/2022]
Abstract
BACKGROUND & AIMS To evaluate the effect of the Brazilian Cardioprotective Diet Program (BALANCE Program) on inflammatory biomarkers, involved in the pathophysiology of the atherosclerosis, on inflammatory biomarkers, cardiovascular risk factors, and on plasma fatty acids in cardiovascular disease secondary prevention patients. METHODS In this substudy of the BALANCE Program randomized clinical trial, a total of 369 patients aged 45 years or older, who have experienced cardiovascular disease in the previous 10 years, were included. These patients were randomized into two groups and followed up for six months: BALANCE Program group and control group (conventional nutrition advice). In the initial and six-month final visits, anthropometry (body weight, height and waist circumference), food intake evaluation by 24-h dietary recall, plasma inflammatory biomarkers (IL-6, IL-8, IL-10, IL-12, tumor necrosis factor-α, adiponectin, and C-reactive protein levels), blood pressure, glycemia, insulinemia, lipid profile, and plasma fatty acids levels were evaluated. RESULTS The BALANCE Program group showed increased plasma alpha-linolenic acid levels (P = 0.008), reduction in waist circumference (P = 0.049) and BMI (P = 0.032). No difference was observed among plasma inflammatory biomarkers and clinical data. CONCLUSION After six months of follow-up, BALANCE Program led to a significant reduction on BMI and waist circumference in individuals in secondary prevention for cardiovascular disease. Although plasmatic alpha-linolenic acid has increased, there was no impact on plasma inflammatory biomarkers. CLINICAL TRIAL REGISTRATION NCT01620398.
Collapse
Affiliation(s)
- Angela C Bersch-Ferreira
- Research Institute-HCor, São Paulo, Brazil; Department of Nutrition, School of Public Health, University of São Paulo, São Paulo, Brazil
| | - Wendy L Hall
- Department of Nutritional Sciences, Faculty of Life Sciences & Medicine, Kings College, London, England
| | | | - Camila R Torreglosa
- Research Institute-HCor, São Paulo, Brazil; Program in Global Health and Sustainability, University of São Paulo, São Paulo, SP, Brazil
| | - Geni Sampaio
- Department of Nutrition, School of Public Health, University of São Paulo, São Paulo, Brazil
| | | | - Renata Alves
- Department of Nutrition, School of Public Health, University of São Paulo, São Paulo, Brazil; Department of Clinical Nutrition at Instituto Dante Pazzanese de Cardiologia, São Paulo, Brazil
| | - Maria Beatriz Ross
- Department of Nutrition, School of Public Health, University of São Paulo, São Paulo, Brazil
| | | | - Cristiane Kovacs
- Department of Clinical Nutrition at Instituto Dante Pazzanese de Cardiologia, São Paulo, Brazil
| | - Aline Marcadenti
- Research Institute-HCor, São Paulo, Brazil; Department of Nutrition, School of Public Health, University of São Paulo, São Paulo, Brazil
| | - Daniel Magnoni
- Department of Clinical Nutrition at Instituto Dante Pazzanese de Cardiologia, São Paulo, Brazil
| | | | - Marcelo Macedo Rogero
- Department of Nutrition, School of Public Health, University of São Paulo, São Paulo, Brazil.
| |
Collapse
|
9
|
Mariamenatu AH, Abdu EM. Overconsumption of Omega-6 Polyunsaturated Fatty Acids (PUFAs) versus Deficiency of Omega-3 PUFAs in Modern-Day Diets: The Disturbing Factor for Their "Balanced Antagonistic Metabolic Functions" in the Human Body. J Lipids 2021; 2021:8848161. [PMID: 33815845 PMCID: PMC7990530 DOI: 10.1155/2021/8848161] [Citation(s) in RCA: 121] [Impact Index Per Article: 30.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2020] [Revised: 02/01/2021] [Accepted: 03/01/2021] [Indexed: 12/19/2022] Open
Abstract
Polyunsaturated fatty acids (PUFAs) contain ≥2 double-bond desaturations within the acyl chain. Omega-3 (n-3) and Omega-6 (n-6) PUFAs are the two known important families in human health and nutrition. In both Omega families, many forms of PUFAs exist: α-linolenic acid (ALA), eicosapentaenoic acid (EPA), and docosahexaenoic acid (DHA) from the n-3 family and linoleic acid (LA), dihomo-γ-linolenic acid (DGLA), and arachidonic acid (AA) from the n-6 family are the important PUFAs for human health. Omega-3 and Omega-6 PUFAs are competitively metabolized by the same set of desaturation, elongation, and oxygenase enzymes. The lipid mediators produced from their oxidative metabolism perform opposing (antagonistic) functions in the human body. Except for DGLA, n-6 PUFA-derived lipid mediators enhance inflammation, platelet aggregation, and vasoconstriction, while those of n-3 inhibit inflammation and platelet aggregation and enhance vasodilation. Overconsumption of n-6 PUFAs with low intake of n-3 PUFAs is highly associated with the pathogenesis of many modern diet-related chronic diseases. The volume of n-6 PUFAs is largely exceeding the volume of n-3PUFAs. The current n-6/n-3 ratio is 20-50/1. Due to higher ratios of n-6/n-3 in modern diets, larger quantities of LA- and AA-derived lipid mediators are produced, becoming the main causes of the formation of thrombus and atheroma, the allergic and inflammatory disorders, and the proliferation of cells, as well as the hyperactive endocannabinoid system. Therefore, in order to reduce all of these risks which are due to overconsumption of n-6 PUFAs, individuals are required to take both PUFAs in the highly recommended n-6/n-3 ratio which is 4-5/1.
Collapse
Affiliation(s)
- Abeba Haile Mariamenatu
- Department of Biotechnology, College of Natural and Computational Science, Debre Berhan University, P.O. Box 445, Debre Berhan, Ethiopia
| | - Emebet Mohammed Abdu
- Department of Biology, College of Natural and Computational Science, Debre Berhan University, P.O. Box 445, Debre Berhan, Ethiopia
| |
Collapse
|
10
|
X. S. Oliveira M, Palma ASV, Reis BR, Franco CSR, Marconi APS, Shiozaki FA, G. Reis L, Salles MSV, Netto AS. Inclusion of soybean and linseed oils in the diet of lactating dairy cows makes the milk fatty acid profile nutritionally healthier for the human diet. PLoS One 2021; 16:e0246357. [PMID: 33561133 PMCID: PMC7872270 DOI: 10.1371/journal.pone.0246357] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2020] [Accepted: 01/15/2021] [Indexed: 01/09/2023] Open
Abstract
Fluid milk and its derivatives are important dietary ingredients that contribute to daily nutrient intake of the modern Homo sapiens. To produce milk that is healthier for human consumption, the present study evaluated the effect of adding soybean oil and linseed oil in the diet of lactating cows. The fatty acid profile of milk, milk composition, and the blood parameters of cows were evaluated. Eighteen Holstein cows were distributed in a replicated Latin square design and distributed according to the following treatments: 1) Control (CC): traditional dairy cow diet, without addition of oil; 2) Soybean oil (SO): 2.5% addition of soybean oil to the traditional diet, as a source of omega-6; 3) Linseed oil (LO): 2.5% addition of linseed oil in the diet as a source of omega-3. Milk production was not affected, but oil supplementation decreased feed intake by 1.93 kg/cow/day. The milk fat percentage was significantly lower when cows were supplemented with vegetable oil (3.37, 2.75 and 2.89% for CC, SO and LO, respectively). However, both soybean and linseed oils decreased the concentration of saturated fatty acids (66.89, 56.52 and 56.60 g/100g for CC, SO and LO respectively), increased the amount of unsaturated fatty acids in milk (33.05, 43.39, and 43.35 g/100g for CC, SO and LO respectively) and decreased the ratio between saturated/unsaturated fatty acids (2.12, 1.34, and 1.36 for CC, SO and LO respectively). Furthermore, SO and LO increased significantly the concentration of monounsaturated fatty acids (29.58, 39.55 and 39.47 g/100g for CC, SO and LO respectively), though it did not significantly alter the level of polyunsaturated fatty acids in milk fat (3.57, 3.93 and 3.98 g/100g for CC, SO and LO respectively). Supplementation with LO enhanced the concentration of omega-3 fatty acids on milk (0.32, 0.36, and 1.02 for CC, SO and LO respectively). Blood variables aspartate aminotransferase, gamma glutamyl transferase, urea, albumin, creatinine and total proteins were not altered. On the other hand, total cholesterol, HDL and LDL were greater in the group supplemented with vegetable oils. Supplementation with vegetable oils reduced the dry matter intake of cows, the fat content of milk, and improved saturated/unsaturated fatty acid ratio of milk fat. Compared to the SO treatment, animals fed LO produced milk with greater content of omega-3, and a more desirable omega-6/omega-3 ratio on a human nutrition perspective. Thus, the inclusion of SO and LO in the diet of lactating dairy cows makes the milk fatty acid profile nutritionally healthier for the human consumption.
Collapse
Affiliation(s)
- Mauricio X. S. Oliveira
- Faculty of Animal Science and Food Engineering, University of São Paulo, Pirassununga, São Paulo, Brazil
| | - Andre S. V. Palma
- Faculty of Animal Science and Food Engineering, University of São Paulo, Pirassununga, São Paulo, Brazil
| | - Barbara R. Reis
- Faculty of Animal Science and Food Engineering, University of São Paulo, Pirassununga, São Paulo, Brazil
| | - Camila S. R. Franco
- Faculty of Animal Science and Food Engineering, University of São Paulo, Pirassununga, São Paulo, Brazil
| | - Alessandra P. S. Marconi
- Faculty of Animal Science and Food Engineering, University of São Paulo, Pirassununga, São Paulo, Brazil
| | - Fabiana A. Shiozaki
- Faculty of Animal Science and Food Engineering, University of São Paulo, Pirassununga, São Paulo, Brazil
| | - Leriana G. Reis
- Faculty of Animal Science and Food Engineering, University of São Paulo, Pirassununga, São Paulo, Brazil
| | | | - Arlindo S. Netto
- Faculty of Animal Science and Food Engineering, University of São Paulo, Pirassununga, São Paulo, Brazil
| |
Collapse
|
11
|
Martelli A, Citi V, Calderone V. Recent efforts in drug discovery on vascular inflammation and consequent atherosclerosis. Expert Opin Drug Discov 2020; 16:411-427. [PMID: 33256484 DOI: 10.1080/17460441.2021.1850688] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
Introduction: Preservation of vascular endothelium integrity and maintenance of its full functionality are fundamental aspects in order to avoid both cardiovascular and non-cardiovascular diseases.Areas covered: Although a massive endothelial disruption is a rare condition, caused by acute and uncontrolled inflammatory responses (e.g. the cytokine storm induced by SARS-CoV-2 infection), more frequently the vascular tree is the first target of slowly progressive inflammatory processes which affect the integrity of endothelium and its 'barrier' function, supporting the onset of atherosclerotic plaque and spreading inflammation. This endothelial dysfunction leads to decrease NO biosynthesis, impaired regulation of vascular tone, and increased platelet aggregation. Such chronic subclinic inflammation leads to macrophage infiltration in atherosclerotic lesions. Therefore, many efforts should be addressed to find useful approaches to preserve vascular endothelium from inflammation. In this review, the authors have evaluated the most recent strategies to counteract this pathological condition.Expert opinion: The therapeutic and nutraceutical approaches represent useful tools to treat or prevent different phases of vascular inflammation. In particular, the pharmacological approach should be used in advanced phases characterized by clinical signs of vascular disease, whilst the nutraceutical approach may represent a promising preventive strategy to preserve the integrity of the endothelial tissue.
Collapse
Affiliation(s)
- Alma Martelli
- Department of Pharmacy, University of Pisa, Pisa, Italy.,Interdepartmental Research Centre "Nutraceuticals and Food for Health (NUTRAFOOD)", University of Pisa, Pisa, Italy.,Interdepartmental Research Centre of Ageing, Biology and Pathology, University of Pisa, Pisa, Italy
| | | | - Vincenzo Calderone
- Department of Pharmacy, University of Pisa, Pisa, Italy.,Interdepartmental Research Centre "Nutraceuticals and Food for Health (NUTRAFOOD)", University of Pisa, Pisa, Italy.,Interdepartmental Research Centre of Ageing, Biology and Pathology, University of Pisa, Pisa, Italy
| |
Collapse
|
12
|
Jalili M, Hekmatdoost A. Dietary ω-3 fatty acids and their influence on inflammation via Toll-like receptor pathways. Nutrition 2020; 85:111070. [PMID: 33545546 DOI: 10.1016/j.nut.2020.111070] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2020] [Revised: 10/05/2020] [Accepted: 11/02/2020] [Indexed: 12/17/2022]
Abstract
Dietary intake of long-chain, highly unsaturated ω-3 fatty acids (FAs) is considered indispensable for humans. The ω-3 FAs have been known to be anti-inflammatory and immunomodulatory dietary factors; however, the modes of action on pathogen recognition receptors (PRRs) and downstream signaling pathways have not been fully elucidated. Dietary sources contain various amounts of ω-3 long-chain fatty acids (LCFAs) of different lengths and the association between intake of these polyunsaturated fatty acids (PUFAs) with underlying mechanisms of various immune-related disorders can be of great interest. The potential anti-inflammatory role for ω-3 LCFAs can be explained by modification of lipid rafts, modulation of inflammatory mediators such as cytokines and PRRs. Toll-like receptors (TLRs) are a group of PRRs that play an important role in the recognition of bacterial infections and ω-3 FAs have been implicated in the modulation of downstream signaling of TLR-4, an important receptor for recognition of gram-negative bacteria. The ω-3 FAs docosahexaenoic acid and eicosapentaenoic acid have been investigated in vivo and in vitro for their effects on the nuclear factor-κB activation pathway. Identification of the effects of ω-3 FAs on other key molecular factors like prostaglandins and leukotrienes and their signals may help the recognition and development of medicines to suppress the main mediators and turn on the expression of anti-inflammatory cytokines and nuclear receptors.
Collapse
Affiliation(s)
- Mahsa Jalili
- Cell, Molecular Biology Group, Department of Biology, Faculty of Natural Sciences, Norwegian University of Science and Technology, Trondheim, Norway.
| | - Azita Hekmatdoost
- Department of Clinical Nutrition, Faculty of Nutrition and Food Sciences, Shahid Beheshti University of Medical Sciences, National Nutrition and Food Technology Research Institute, Tehran, Iran
| |
Collapse
|
13
|
Zirpoli H, Chang CL, Carpentier YA, Michael-Titus AT, Ten VS, Deckelbaum RJ. Novel Approaches for Omega-3 Fatty Acid Therapeutics: Chronic Versus Acute Administration to Protect Heart, Brain, and Spinal Cord. Annu Rev Nutr 2020; 40:161-187. [PMID: 32966188 DOI: 10.1146/annurev-nutr-082018-124539] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
This article reviews novel approaches for omega-3 fatty acid (FA) therapeutics and the linked molecular mechanisms in cardiovascular and central nervous system (CNS) diseases. In vitro and in vivo research studies indicate that omega-3 FAs affect synergic mechanisms that include modulation of cell membrane fluidity, regulation of intracellular signaling pathways, and production of bioactive mediators. We compare how chronic and acute treatments with omega-3 FAs differentially trigger pathways of protection in heart, brain, and spinal cord injuries. We also summarize recent omega-3 FA randomized clinical trials and meta-analyses and discuss possible reasons for controversial results, with suggestions on improving the study design for future clinical trials. Acute treatment with omega-3 FAs offers a novel approach for preserving cardiac and neurological functions, and the combinations of acute treatment with chronic administration of omega-3 FAs might represent an additional therapeutic strategy for ameliorating adverse cardiovascular and CNS outcomes.
Collapse
Affiliation(s)
- Hylde Zirpoli
- Institute of Human Nutrition, Vagelos College of Physicians and Surgeons, Columbia University Irving Medical Center, New York, NY 10032, USA;
| | - Chuchun L Chang
- Institute of Human Nutrition, Vagelos College of Physicians and Surgeons, Columbia University Irving Medical Center, New York, NY 10032, USA;
| | - Yvon A Carpentier
- Clinical Nutrition Unit, Université Libre de Bruxelles, 1050 Brussels, Belgium.,Nutrition Lipid Developments, SPRL, 1050 Brussels, Belgium
| | - Adina T Michael-Titus
- Center for Neuroscience, Surgery, and Trauma, Queen Mary University of London, London E1 4NS, United Kingdom
| | - Vadim S Ten
- Department of Pediatrics, Columbia University Irving Medical Center, New York, NY 10032, USA
| | - Richard J Deckelbaum
- Institute of Human Nutrition, Vagelos College of Physicians and Surgeons, Columbia University Irving Medical Center, New York, NY 10032, USA; .,Department of Pediatrics, Columbia University Irving Medical Center, New York, NY 10032, USA
| |
Collapse
|
14
|
Ayee MAA, Bunker BC, De Groot JL. Membrane modulatory effects of omega-3 fatty acids: Analysis of molecular level interactions. CURRENT TOPICS IN MEMBRANES 2020; 86:57-81. [PMID: 33837698 DOI: 10.1016/bs.ctm.2020.08.001] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Bioactive omega-3 polyunsaturated fatty acids have been shown to reduce the risk of death in patients with cardiovascular disease and alleviate the symptoms of other inflammatory diseases. However, the mechanisms of action of these effects remain unclear. It has been postulated that omega-3 polyunsaturated fatty acids modify cell membranes by incorporation into the membrane and altering the signaling properties of cellular receptors. In this chapter, we explore the effects of omega-3 polyunsaturated fatty acids on cell membrane structure and function. We present a review of the current evidence for the health benefits of these compounds and explore the molecular mechanisms through which omega-3 polyunsaturated fatty acids interact with membrane lipids and modulate bilayer structure. Using computational models of multicomponent phospholipid bilayers, we assess the consequences of incorporation of these fatty acids on membrane lipid packing, water permeation, and membrane structure.
Collapse
Affiliation(s)
- Manuela A A Ayee
- Department of Engineering, Dordt University, Sioux Center, IA, United States.
| | - Brendan C Bunker
- Department of Engineering, Dordt University, Sioux Center, IA, United States
| | - Jordan L De Groot
- Department of Engineering, Dordt University, Sioux Center, IA, United States
| |
Collapse
|
15
|
Lombardi M, Chiabrando JG, Vescovo GM, Bressi E, Del Buono MG, Carbone S, Koenig RA, Van Tassell BW, Abbate A, Biondi-Zoccai G, Dixon DL. Impact of Different Doses of Omega-3 Fatty Acids on Cardiovascular Outcomes: a Pairwise and Network Meta-analysis. Curr Atheroscler Rep 2020; 22:45. [PMID: 32671519 DOI: 10.1007/s11883-020-00865-5] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
PURPOSE OF REVIEW Omega-3 fatty acid (O3FA) supplementation has shown conflicting evidence regarding its benefit in cardiovascular events. We performed a pairwise and network meta-analysis to elucidate the benefit of different doses of O3FA supplementation in cardiovascular prevention. RECENT FINDINGS Fourteen studies were identified providing data on 125,763 patients. A prespecified cut-off value of < 1 g per day was set for low-dose (LD) O3FA and > 1 g per day for high-dose (HD) O3FA. The efficacy outcomes of interest were total death, cardiac death, sudden cardiac death, myocardial infarction, stroke, coronary revascularization, unstable angina, and major vascular events. Safety outcomes of interest were bleeding, gastrointestinal disturbances, and atrial fibrillation events. HD treatment was associated with a lower risk of cardiac death (IRR 0.79, 95% CI [0.65-0.96], p = 0.03 versus control), myocardial infarction (0.71 [0.62-0.82], p < 0.0001 versus control and 0.79 [0.67-0.92], p = 0.003 versus LD), coronary revascularization (0.74 [0.66-0.83], p < 0.0001 versus control and 0.74 [0.66-0.84], p < 0.0001 versus LD), unstable angina (0.73 [0.62-0.86], p = 0.0001 versus control and 0.74 [0.62-0.89], p = 0.002 versus LD), and major vascular events (0.78 [0.71-0.85], p < 0.0001 versus control and 0.79 [0.72-0.88], p < 0.0001 versus LD). HD treatment was associated with increased risk for bleeding events (1.49 [1.2-1.84], p = 0.0002 versus control and 1.63 [1.16-2.3], p = 0.005 versus LD) and increased atrial fibrillation events compared to control (1.35 [1.1-1.66], p = 0.004). HD O3FA treatment was associated with lower cardiovascular events compared to LD and to control, but increased risk for bleeding and atrial fibrillation events.
Collapse
Affiliation(s)
- Marco Lombardi
- VCU Pauley Heart Center, Virginia Commonwealth University, Richmond, VA, USA
- Department of Cardiovascular and Thoracic Sciences, Catholic University of the Sacred Heart, Rome, Italy
| | - Juan G Chiabrando
- Interventional Cardiology Service, Hospital Italiano de Buenos Aires, Buenos Aires, Argentina
- Health Science Statistics Applied Laboratory (LEACS), Pharmacology and Toxicology Department, School of Medicine, University of Buenos Aires, Buenos Aires, Argentina
| | - Giovanni M Vescovo
- Department of Cardiac Thoracic, Vascular Sciences and Public Health, University of Padua, Padua, Italy
| | - Edoardo Bressi
- VCU Pauley Heart Center, Virginia Commonwealth University, Richmond, VA, USA
- Division of Cardiology, Policlinico Casilino, Rome, Italy
| | - Marco Giuseppe Del Buono
- VCU Pauley Heart Center, Virginia Commonwealth University, Richmond, VA, USA
- Department of Cardiovascular and Thoracic Sciences, Catholic University of the Sacred Heart, Rome, Italy
| | - Salvatore Carbone
- VCU Pauley Heart Center, Virginia Commonwealth University, Richmond, VA, USA
- Department of Kinesiology & Health Sciences, College of Humanities & Sciences, Virginia Commonwealth University, Richmond, VA, USA
| | - Rachel A Koenig
- Tompkins-McCaw Library for the Health Sciences, VCU Libraries Virginia Commonwealth University, Richmond, VA, USA
| | - Benjamin W Van Tassell
- VCU Pauley Heart Center, Virginia Commonwealth University, Richmond, VA, USA
- Department of Pharmacotherapy & Outcomes Science, Virginia Commonwealth University, Richmond, VA, USA
| | - Antonio Abbate
- VCU Pauley Heart Center, Virginia Commonwealth University, Richmond, VA, USA
| | - Giuseppe Biondi-Zoccai
- Department of Medical-Surgical Sciences and Biotechnologies, Sapienza University of Rome, Latina, Italy
- Mediterranea Cardiocentro, Napoli, Italy
| | - Dave L Dixon
- VCU Pauley Heart Center, Virginia Commonwealth University, Richmond, VA, USA.
- Department of Pharmacotherapy & Outcomes Science, Virginia Commonwealth University, Richmond, VA, USA.
| |
Collapse
|
16
|
Blood clearance kinetics and organ delivery of medium-chain triglyceride and fish oil-containing lipid emulsions: Comparing different animal species. Clin Nutr 2020; 40:987-996. [PMID: 32753350 DOI: 10.1016/j.clnu.2020.07.002] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2020] [Revised: 07/01/2020] [Accepted: 07/05/2020] [Indexed: 12/11/2022]
Abstract
BACKGROUND & AIMS Medium-chain triglycerides (TG) (MCT) and fish oil (FO) TG are incorporated as the core TG component into intravenous (IV) lipid emulsions for infusion in parenteral nutrition. Bolus injections of IV emulsions, on the other hand, have emerged as a novel therapeutic approach to treat various acute disorders. However, intravascular metabolism and organ delivery of acute IV injection of emulsions containing both MCT and FO are not fully defined, nor have they been characterized across common experimental animal models. We characterized and compared blood clearance kinetics and organ distribution of bolus injections of MCT/FO emulsions among different animal species. We also examined whether sex differences or feeding status can affect catabolic properties of MCT/FO lipid emulsions. DESIGN Blood clearance rates of lipid emulsions with specific TG composition were compared in rats IV injected with [3H]cholesteryl hexadecyl ether labeled pure n-6 long-chain (LCT) and n-3 FO TG lipid emulsions, or emulsions containing MCT and FO at different ratios (wt/wt), which include 8:2 (80% MCT: 20% FO), 5:4:1 (50% MCT: 40% LCT: 10% FO) and SMOF (30% LCT: 30% MCT: 25% olive oil: 10% FO). Dose-response effects (0.016 mg-1.6 mg TG/g body weight) of the MCT/FO 8:2 emulsions on blood clearance properties and organ delivery were determined in both mice and rats. Blood clearance kinetics and organ uptake of MCT/FO 8:2 emulsions were compared between male and female rats and between fed and fasted rats. Changes in plasma lipid profiles after acute injections of MCT/FO 8:2 lipid emulsion at different doses (0.043, 0.133, and 0.4 mg TG/g body weight) were characterized in non-human primates (Cynomolgus monkeys). RESULTS MCT/FO 8:2 emulsion was cleared faster in rats when compared with other emulsions with different TG contents. Mice had faster blood clearance and higher fractional catabolic rates (FCR) when compared with the rats injected with MCT/FO 8:2 emulsions regardless of the injected doses. Mice and rats had similar plasma TG and free fatty acid (FFA) levels after low- or high-dose injections of the MCT/FO emulsion. Tissue distribution of the MCT/FO 8:2 lipid emulsion are comparable between mice and rats, where liver had the highest uptake per recovered dose among all organs (>60%). Feeding status and sex differences did not alter the blood clearance rate of the MCT/FO 8:2 emulsion in rats. In a nonhuman primate model, dose-response increases in plasma TG and FFA were observed after IV injection of MCT/FO 8:2 emulsions within the 1st 10 min. CONCLUSION A lipid emulsion containing both MCT and FO TG is cleared rapidly in blood and readily available for organ uptake in rodent and primate animal models. Characterization of the blood clearance properties of the MCT/FO 8:2 emulsion administered in various animal models may provide further insight into the safety and efficacy profiles for future therapeutic use of bolus injections of MCT/FO emulsions in humans.
Collapse
|
17
|
Gupta J, Gupta R. Nutraceutical Status and Scientific Strategies for Enhancing Production of Omega-3 Fatty Acids from Microalgae and their Role in Healthcare. Curr Pharm Biotechnol 2020; 21:1616-1631. [PMID: 32619166 DOI: 10.2174/1389201021666200703201014] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2020] [Revised: 04/28/2020] [Accepted: 06/16/2020] [Indexed: 11/22/2022]
Abstract
Adherence to Omega-3 fatty acids (O3FAs) as Nutraceuticals for medicinal applications provides health improvement. The prevention and treatment of diseases with O3FAs hold promise in clinical therapy and significantly reduces the risk of chronic disorders. Polyunsaturated fatty acids (PUFA) O3FAs have beneficial effects in the treatment of cardiovascular disorders, diabetic disease, foetal development, Alzheimer's disease, retinal problem, growth and brain development of infants and antitumor effects. Association to current analysis promotes the application of algal biomass for production of O3FAs, mode of action, fate, weight management, immune functions, pharmaceutical and therapeutic applications serving potent sources in healthcare management. A search of the literature was conducted in the databases of WHO website, Sci.org, PubMed, academics and Google. The authors performed search strategies and current scenario of O3FAs in health associated disorders. Promising outcomes and future strategies towards O3FAs may play a pivotal role in Nutraceutical industries in the cure of human health in the future.
Collapse
Affiliation(s)
- Jitendra Gupta
- Institute of Pharmaceutical Research, GLA University, Mathura-281406, Uttar Pradesh, India
| | - Reena Gupta
- Institute of Pharmaceutical Research, GLA University, Mathura-281406, Uttar Pradesh, India
| |
Collapse
|
18
|
Zakirov FH, Zhang D, Grechko AV, Wu WK, Poznyak AV, Orekhov AN. Lipid-based gene delivery to macrophage mitochondria for atherosclerosis therapy. Pharmacol Res Perspect 2020; 8:e00584. [PMID: 32237116 PMCID: PMC7111069 DOI: 10.1002/prp2.584] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2019] [Revised: 01/17/2020] [Accepted: 01/18/2020] [Indexed: 12/12/2022] Open
Abstract
Atherosclerosis with associated cardiovascular diseases remains one of the main causes of disability and death worldwide, requiring development of new solutions for prevention and treatment. Macrophages are the key effectors of a series of events involved in atherogenesis, such as inflammation, plaque formation, and changes in lipid metabolism. Some of these events were shown to be associated with mitochondrial dysfunction and excessive mitochondrial DNA (mtDNA) damage. Moreover, macrophages represent a promising target for novel therapeutic approaches that are based on the expression of various receptors and nanoparticle uptake. Lipid-based gene delivery to mitochondria is considered to be an interesting strategy for mtDNA damage correction. To date, several nanocarriers and their modifications have been developed that demonstrate high transfection efficiency and low cytotoxicity. This review discusses the possibilities of lipid-based gene delivery to macrophage mitochondria for atherosclerosis therapy.
Collapse
Affiliation(s)
- Felix H Zakirov
- I. M. Sechenov First Moscow State Medical University (Sechenov University), Moscow, Russian Federation
| | - Dongwei Zhang
- Diabetes Research Center, Traditional Chinese Medicine School, Beijing University of Chinese Medicine, Beijing, China
| | - Andrey V Grechko
- Federal Research and Clinical Center of Intensive Care Medicine and Rehabilitology, Moscow, Russian Federation
| | - Wei-Kai Wu
- Department of Internal Medicine, National Taiwan University Hospital, Taipei, Taiwan
| | - Anastasia V Poznyak
- Institute for Atherosclerosis Research, Skolkovo Innovative Center, Moscow, Russia
| | - Alexander N Orekhov
- Institute of Human Morphology, Moscow, Russia
- Laboratory of Angiopathology, Institute of General Pathology and Pathophysiology, Moscow, Russia
| |
Collapse
|
19
|
Abdelhamid AS, Brown TJ, Brainard JS, Biswas P, Thorpe GC, Moore HJ, Deane KHO, Summerbell CD, Worthington HV, Song F, Hooper L. Omega-3 fatty acids for the primary and secondary prevention of cardiovascular disease. Cochrane Database Syst Rev 2020; 3:CD003177. [PMID: 32114706 PMCID: PMC7049091 DOI: 10.1002/14651858.cd003177.pub5] [Citation(s) in RCA: 120] [Impact Index Per Article: 24.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
BACKGROUND Omega-3 polyunsaturated fatty acids from oily fish (long-chain omega-3 (LCn3)), including eicosapentaenoic acid (EPA) and docosahexaenoic acid (DHA)), as well as from plants (alpha-linolenic acid (ALA)) may benefit cardiovascular health. Guidelines recommend increasing omega-3-rich foods, and sometimes supplementation, but recent trials have not confirmed this. OBJECTIVES To assess the effects of increased intake of fish- and plant-based omega-3 fats for all-cause mortality, cardiovascular events, adiposity and lipids. SEARCH METHODS We searched CENTRAL, MEDLINE and Embase to February 2019, plus ClinicalTrials.gov and World Health Organization International Clinical Trials Registry to August 2019, with no language restrictions. We handsearched systematic review references and bibliographies and contacted trial authors. SELECTION CRITERIA We included randomised controlled trials (RCTs) that lasted at least 12 months and compared supplementation or advice to increase LCn3 or ALA intake, or both, versus usual or lower intake. DATA COLLECTION AND ANALYSIS Two review authors independently assessed trials for inclusion, extracted data and assessed validity. We performed separate random-effects meta-analysis for ALA and LCn3 interventions, and assessed dose-response relationships through meta-regression. MAIN RESULTS We included 86 RCTs (162,796 participants) in this review update and found that 28 were at low summary risk of bias. Trials were of 12 to 88 months' duration and included adults at varying cardiovascular risk, mainly in high-income countries. Most trials assessed LCn3 supplementation with capsules, but some used LCn3- or ALA-rich or enriched foods or dietary advice compared to placebo or usual diet. LCn3 doses ranged from 0.5 g a day to more than 5 g a day (19 RCTs gave at least 3 g LCn3 daily). Meta-analysis and sensitivity analyses suggested little or no effect of increasing LCn3 on all-cause mortality (risk ratio (RR) 0.97, 95% confidence interval (CI) 0.93 to 1.01; 143,693 participants; 11,297 deaths in 45 RCTs; high-certainty evidence), cardiovascular mortality (RR 0.92, 95% CI 0.86 to 0.99; 117,837 participants; 5658 deaths in 29 RCTs; moderate-certainty evidence), cardiovascular events (RR 0.96, 95% CI 0.92 to 1.01; 140,482 participants; 17,619 people experienced events in 43 RCTs; high-certainty evidence), stroke (RR 1.02, 95% CI 0.94 to 1.12; 138,888 participants; 2850 strokes in 31 RCTs; moderate-certainty evidence) or arrhythmia (RR 0.99, 95% CI 0.92 to 1.06; 77,990 participants; 4586 people experienced arrhythmia in 30 RCTs; low-certainty evidence). Increasing LCn3 may slightly reduce coronary heart disease mortality (number needed to treat for an additional beneficial outcome (NNTB) 334, RR 0.90, 95% CI 0.81 to 1.00; 127,378 participants; 3598 coronary heart disease deaths in 24 RCTs, low-certainty evidence) and coronary heart disease events (NNTB 167, RR 0.91, 95% CI 0.85 to 0.97; 134,116 participants; 8791 people experienced coronary heart disease events in 32 RCTs, low-certainty evidence). Overall, effects did not differ by trial duration or LCn3 dose in pre-planned subgrouping or meta-regression. There is little evidence of effects of eating fish. Increasing ALA intake probably makes little or no difference to all-cause mortality (RR 1.01, 95% CI 0.84 to 1.20; 19,327 participants; 459 deaths in 5 RCTs, moderate-certainty evidence),cardiovascular mortality (RR 0.96, 95% CI 0.74 to 1.25; 18,619 participants; 219 cardiovascular deaths in 4 RCTs; moderate-certainty evidence), coronary heart disease mortality (RR 0.95, 95% CI 0.72 to 1.26; 18,353 participants; 193 coronary heart disease deaths in 3 RCTs; moderate-certainty evidence) and coronary heart disease events (RR 1.00, 95% CI 0.82 to 1.22; 19,061 participants; 397 coronary heart disease events in 4 RCTs; low-certainty evidence). However, increased ALA may slightly reduce risk of cardiovascular disease events (NNTB 500, RR 0.95, 95% CI 0.83 to 1.07; but RR 0.91, 95% CI 0.79 to 1.04 in RCTs at low summary risk of bias; 19,327 participants; 884 cardiovascular disease events in 5 RCTs; low-certainty evidence), and probably slightly reduces risk of arrhythmia (NNTB 91, RR 0.73, 95% CI 0.55 to 0.97; 4912 participants; 173 events in 2 RCTs; moderate-certainty evidence). Effects on stroke are unclear. Increasing LCn3 and ALA had little or no effect on serious adverse events, adiposity, lipids and blood pressure, except increasing LCn3 reduced triglycerides by ˜15% in a dose-dependent way (high-certainty evidence). AUTHORS' CONCLUSIONS This is the most extensive systematic assessment of effects of omega-3 fats on cardiovascular health to date. Moderate- and low-certainty evidence suggests that increasing LCn3 slightly reduces risk of coronary heart disease mortality and events, and reduces serum triglycerides (evidence mainly from supplement trials). Increasing ALA slightly reduces risk of cardiovascular events and arrhythmia.
Collapse
Affiliation(s)
- Asmaa S Abdelhamid
- University of East AngliaNorwich Medical SchoolNorwich Research ParkNorwichNorfolkUKNR4 7TJ
| | - Tracey J Brown
- University of East AngliaNorwich Medical SchoolNorwich Research ParkNorwichNorfolkUKNR4 7TJ
| | - Julii S Brainard
- University of East AngliaNorwich Medical SchoolNorwich Research ParkNorwichNorfolkUKNR4 7TJ
| | - Priti Biswas
- University of East AngliaMED/HSCNorwich Research ParkNorwichUKNR4 7TJ
| | - Gabrielle C Thorpe
- University of East AngliaSchool of Health SciencesEarlham RoadNorwichUKNR4 7TJ
| | - Helen J Moore
- Teesside UniversitySchool of Social Sciences, Humanities and LawMiddlesboroughUKTS1 3BA
| | - Katherine HO Deane
- University of East AngliaSchool of Health SciencesEarlham RoadNorwichUKNR4 7TJ
| | - Carolyn D Summerbell
- Durham UniversityDepartment of Sport and Exercise Sciences42 Old ElvetDurhamUKDH13HN
| | - Helen V Worthington
- Division of Dentistry, School of Medical Sciences, Faculty of Biology, Medicine and Health, The University of ManchesterCochrane Oral HealthCoupland Building 3Oxford RoadManchesterUKM13 9PL
| | - Fujian Song
- University of East AngliaNorwich Medical SchoolNorwich Research ParkNorwichNorfolkUKNR4 7TJ
| | - Lee Hooper
- University of East AngliaNorwich Medical SchoolNorwich Research ParkNorwichNorfolkUKNR4 7TJ
| | | |
Collapse
|
20
|
Bork CS, Lasota AN, Lundbye-Christensen S, Jakobsen MU, Tjønneland A, Overvad K, Schmidt EB. Adipose tissue content of alpha-linolenic acid and development of peripheral artery disease: a Danish case-cohort study. Eur J Nutr 2019; 59:3191-3200. [PMID: 31832750 DOI: 10.1007/s00394-019-02159-2] [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: 05/15/2019] [Accepted: 12/04/2019] [Indexed: 11/26/2022]
Abstract
PURPOSE The aim of this study was to investigate the association between adipose tissue content of the plant-derived n-3 fatty acid, alpha-linolenic acid, and the rate of incident peripheral artery disease (PAD). METHODS We conducted a case-cohort study nested within the Danish Diet, Cancer and Health cohort (n = 57,053), which was established between 1993 and 1997. Potential PAD cases were identified using linkage with The Danish National Patient Register and all potential cases were validated. Adipose tissue samples from the buttock were collected at baseline and fatty acid composition was determined in cases and in a random sample (n = 3500) from the cohort by gas chromatography. Statistical analyses were performed using weighted Cox regression allowing for different baseline hazards among sexes. RESULTS During a median of 13.5 years of follow-up, we identified 863 PAD cases with complete information. The median adipose tissue content of ALA in the sub-cohort (n = 3197) was 0.84% (interquartile range 0.73-0.94%) of total fatty acids. In multivariate analyses including adjustment for established risk factors, we observed a U-shaped association between ALA in adipose tissue and rate of PAD, but the association was not statistically significant (P = 0.131). Similar pattern of associations were observed between ALA content in adipose tissue and the rate of PAD among men and women. CONCLUSIONS We found indications of a U-shaped association between adipose tissue content of ALA and the rate of PAD, but the association was not statistically significant.
Collapse
Affiliation(s)
- Christian S Bork
- Department of Cardiology, Aalborg University Hospital, Søndre Skovvej 15, 9000, Aalborg, Denmark.
| | - Anne N Lasota
- Department of Vascular Surgery, Aalborg University Hospital, Aalborg, Denmark
| | | | - Marianne U Jakobsen
- Division of Diet, Disease Prevention and Toxicology, National Food Institute, Technical University of Denmark, Kgs. Lyngby, Denmark
| | - Anne Tjønneland
- Danish Cancer Society Research Center, Copenhagen, Denmark
- Department of Public Health, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Kim Overvad
- Department of Cardiology, Aalborg University Hospital, Søndre Skovvej 15, 9000, Aalborg, Denmark
- Department of Public Health, Aarhus University, Aarhus, Denmark
| | - Erik B Schmidt
- Department of Cardiology, Aalborg University Hospital, Søndre Skovvej 15, 9000, Aalborg, Denmark
- Department of Clinical Medicine, Aalborg University, Aalborg, Denmark
| |
Collapse
|
21
|
Simonetto M, Infante M, Sacco RL, Rundek T, Della-Morte D. A Novel Anti-Inflammatory Role of Omega-3 PUFAs in Prevention and Treatment of Atherosclerosis and Vascular Cognitive Impairment and Dementia. Nutrients 2019; 11:2279. [PMID: 31547601 PMCID: PMC6835717 DOI: 10.3390/nu11102279] [Citation(s) in RCA: 93] [Impact Index Per Article: 15.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2019] [Revised: 09/11/2019] [Accepted: 09/19/2019] [Indexed: 12/13/2022] Open
Abstract
Atherosclerosis is an inflammatory chronic disease affecting arterial vessels and leading to vascular diseases, such as stroke and myocardial infarction. The relationship between atherosclerosis and risk of neurodegeneration has been established, in particular with vascular cognitive impairment and dementia (VCID). Systemic atherosclerosis increases the risk of VCID by inducing cerebral infarction, or through systemic or local inflammatory factors that underlie both atherosclerosis and cognition. Omega-3 and omega-6 polyunsaturated fatty acids (PUFAs) are involved in inflammatory processes, but with opposite roles. Specifically, omega-3 PUFAs exert anti-inflammatory properties by competing with omega-6 PUFAs and displacing arachidonic acid in membrane phospholipids, decreasing the production of pro-inflammatory eicosanoids. Experimental studies and some clinical trials have demonstrated that omega-3 PUFA supplementation may reduce the risk of different phenotypes of atherosclerosis and cardiovascular disease. This review describes the link between atherosclerosis, VCID and inflammation, as well as how omega-3 PUFA supplementation may be useful to prevent and treat inflammatory-related diseases.
Collapse
Affiliation(s)
- Marialaura Simonetto
- Department of Neurology and The Evelyn F. McKnight Brain Institute, University of Miami Miller School of Medicine, Miami, FL 33136, USA.
| | - Marco Infante
- Diabetes Research Institute (DRI) and Clinical Cell Transplant Program, University of Miami Miller School of Medicine, Miami, FL 33136, USA.
- Department of Systems Medicine, University of Rome "Tor Vergata", 00133 Rome, Italy.
| | - Ralph L Sacco
- Department of Neurology and The Evelyn F. McKnight Brain Institute, University of Miami Miller School of Medicine, Miami, FL 33136, USA.
| | - Tatjana Rundek
- Department of Neurology and The Evelyn F. McKnight Brain Institute, University of Miami Miller School of Medicine, Miami, FL 33136, USA.
| | - David Della-Morte
- Department of Neurology and The Evelyn F. McKnight Brain Institute, University of Miami Miller School of Medicine, Miami, FL 33136, USA.
- Department of Systems Medicine, University of Rome "Tor Vergata", 00133 Rome, Italy.
- San Raffaele Roma Open University, 00166 Rome, Italy.
| |
Collapse
|
22
|
Intake of α-linolenic acid is not consistently associated with a lower risk of peripheral artery disease: results from a Danish cohort study. Br J Nutr 2019; 122:86-92. [PMID: 31006418 DOI: 10.1017/s0007114519000874] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Intake of the plant-derived n-3 fatty acid α-linolenic acid (ALA) has been associated with anti-atherosclerotic properties. However, information on the association between ALA intake and development of peripheral artery disease (PAD) is lacking. In this follow-up study, we investigated the association between dietary intake of ALA and the rate of PAD among middle-aged Danish men and women enrolled into the Danish Diet, Cancer and Health cohort between 1993 and 1997. Incident PAD cases were identified through the Danish National Patient Register. Intake of ALA was assessed using a validated FFQ. Statistical analyses were performed using Cox proportional hazard regression allowing for separate baseline hazards among sexes and adjusted for established risk factors for PAD. During a median of 13·6 years of follow-up, we identified 950 valid cases of PAD with complete information on covariates. The median energy-adjusted ALA intake within the cohort was 1·76 g/d (95 % central range: 0·94-3·28). In multivariable analyses, we found no statistically significant association between intake of ALA and the rate of PAD (P = 0·339). Also, no statistically significant associations were observed in analyses including additional adjustment for co-morbidities and in sex-specific analyses. In supplemental analyses with additional adjustment for potential dietary risk factors, we found a weak inverse association of PAD with ALA intake above the median, but the association was not statistically significant (P = 0·314). In conclusion, dietary intake of ALA was not consistently associated with decreased risk of PAD.
Collapse
|
23
|
Rousseau-Ralliard D, Valentino SA, Aubrière MC, Dahirel M, Lallemand MS, Archilla C, Jouneau L, Fournier N, Richard C, Aioun J, Vitorino Carvalho A, Jérôme L, Slama R, Duranthon V, Cassee FR, Chavatte-Palmer P, Couturier-Tarrade A. Effects of first-generation in utero exposure to diesel engine exhaust on second-generation placental function, fatty acid profiles and foetal metabolism in rabbits: preliminary results. Sci Rep 2019; 9:9710. [PMID: 31273257 PMCID: PMC6609606 DOI: 10.1038/s41598-019-46130-x] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2018] [Accepted: 06/19/2019] [Indexed: 12/19/2022] Open
Abstract
Atmospheric pollution has major health effects on directly exposed subjects but intergenerational consequences are poorly characterized. We previously reported that diesel engine exhaust (DE) could lead to structural changes in the placenta of in utero exposed rabbits (first generation, F1). The effects of maternal exposure to DE were further studied on second-generation (F2) rabbits. Pregnant F0 females were exposed to filtered, diluted DE (1 mg/m3, median particle diameter: 69 nm) or clean filtered air (controls) for 2 h/day, 5 days/week by nose-only exposure during days 3–27 post-conception (dpc). Adult female offspring (F1) were mated to control males: F1 tissues and F2 foeto-placental units were collected at 28 dpc and placental structure and gene expression (microarray) analysed. Fatty acid profiles were determined in foetal and maternal plasma, maternal liver and placenta. In F1, compared to controls, hepatic neutral lipid contents were increased in exposed animals without change in the blood biochemistry. In F2, the placental lipid contents were higher, with higher monounsaturated fatty acids and reduced pro-inflammatory arachidonic acid (AA), without placental structural changes. Conversely, the proportion of anti-inflammatory n-3 polyunsaturated fatty acids in F2 plasma was increased while that of AA was decreased. Gene set enrichment analyses (GSEA) of F2 placenta transcriptomic data identified that the proteasome complex and ubiquitin pathways genes were over-represented and ion channel function and inflammation pathways genes were under-represented in exposed animals. These preliminary results demonstrate that diesel engine exhaust exposure and in utero indirect exposure should be considered as a programming factor within the context of the DOHaD (Developmental Origins of Health and Disease) with a probable intergenerational transmission.
Collapse
Affiliation(s)
- Delphine Rousseau-Ralliard
- UMR BDR, INRA, ENVA, Université Paris Saclay, Jouy en Josas, France. .,PremUp Foundation, Paris, France.
| | - Sarah A Valentino
- UMR BDR, INRA, ENVA, Université Paris Saclay, Jouy en Josas, France.,PremUp Foundation, Paris, France
| | - Marie-Christine Aubrière
- UMR BDR, INRA, ENVA, Université Paris Saclay, Jouy en Josas, France.,PremUp Foundation, Paris, France
| | - Michèle Dahirel
- UMR BDR, INRA, ENVA, Université Paris Saclay, Jouy en Josas, France.,PremUp Foundation, Paris, France
| | - Marie-Sylvie Lallemand
- UMR BDR, INRA, ENVA, Université Paris Saclay, Jouy en Josas, France.,PremUp Foundation, Paris, France
| | | | - Luc Jouneau
- UMR BDR, INRA, ENVA, Université Paris Saclay, Jouy en Josas, France
| | - Natalie Fournier
- University Paris-Sud, EA 4041/4529 Lip (Sys)2, UFR de Pharmacie, Châtenay-Malabry, France.,Hôpital Européen Georges Pompidou (AP-HP), Laboratoire de Biochimie, UF Cardio-Vasculaire, Paris, France
| | - Christophe Richard
- UMR BDR, INRA, ENVA, Université Paris Saclay, Jouy en Josas, France.,PremUp Foundation, Paris, France
| | - Josiane Aioun
- UMR BDR, INRA, ENVA, Université Paris Saclay, Jouy en Josas, France.,PremUp Foundation, Paris, France
| | | | | | - Rémy Slama
- Inserm, Univ. Grenoble Alpes, CNRS, IAB joint Research Center, Team of Environmental Epidemiology Applied to Reproduction and Respiratory Health, Grenoble, France
| | | | - Flemming R Cassee
- Centre for Sustainability, Environment and Health, National Institute for Public Health and the Environment, Bilthoven, Netherlands.,Institute of Risk Assessment Sciences, Utrecht University, Utrecht, Netherlands
| | - Pascale Chavatte-Palmer
- UMR BDR, INRA, ENVA, Université Paris Saclay, Jouy en Josas, France.,PremUp Foundation, Paris, France
| | - Anne Couturier-Tarrade
- UMR BDR, INRA, ENVA, Université Paris Saclay, Jouy en Josas, France. .,PremUp Foundation, Paris, France.
| |
Collapse
|
24
|
Guan L, Geng X, Stone C, Cosky EEP, Ji Y, Du H, Zhang K, Sun Q, Ding Y. PM 2.5 exposure induces systemic inflammation and oxidative stress in an intracranial atherosclerosis rat model. ENVIRONMENTAL TOXICOLOGY 2019; 34:530-538. [PMID: 30672636 DOI: 10.1002/tox.22707] [Citation(s) in RCA: 87] [Impact Index Per Article: 14.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/11/2018] [Revised: 12/22/2018] [Accepted: 12/26/2018] [Indexed: 06/09/2023]
Abstract
OBJECTIVES Exposure to airborne particle (PM2.5 ) is a risk factor for intracranial atherosclerosis (ICA). Because of the established role of systemic inflammation and oxidative stress by PM2.5 , we determined whether these processes account for PM2.5 -mediated ICA, and also whether omega-3 fatty acid (O3FA) dietary supplementation could attenuate them. METHODS Adult Sprague-Dawley rats were exposed to filtered air (FA) or PM2.5 and fed either a normal chow diet (NCD) or a high-cholesterol diet (HCD), administered with or without O3FA (5 mg/kg/day by gavage) for 12 weeks. The lumen and thickness of the middle cerebral artery (MCA) were assessed. Serum tumor necrosis factor alpha (TNF-α), interleukin 6 (IL-6), interleukin-1β (IL-1β), and interferon gamma (IFN-γ) were detected by ELISA. Reactive oxygen species (ROS), malondialdehyde (MDA), superoxide dismutase (SOD) activity, nicotinamide adenine dinucleotide phosphate (NADPH) oxidase (NOX) activity, mRNA levels of Nrf2, HO-1, NQO-1, and protein level of NOX subunit gp91 were quantified to determine the oxidative profile of brain vessels. RESULTS PM2.5 increased (P < .05) ICA, especially in the HCD group; elevated serum TNF-α, IL-6, IL-1β, and IFN-γ; increased cerebrovascular ROS, MDA, NOX activity, and gp91 protein levels; and decreased cerebrovascular SOD activity. Nrf2, HO-1, and NQO-1 mRNA levels were upregulated (P < .05) by PM2.5 exposure, especially in the HCD group. O3FA attenuated (P < .05) PM2.5 -induced systemic inflammation, vascular oxidative injury, and ICA. CONCLUSIONS PM2.5 exposure induced systemic inflammation, cerebrovascular oxidative injury, and ICA in rats with HCD. O3FA prevented ICA development, and may therefore exert a protective effect against the atherogenic potential of PM2.5 .
Collapse
Affiliation(s)
- Longfei Guan
- China-America Institute of Neuroscience, Beijing Luhe Hospital, Capital Medical University, Beijing, China
- Department of Neurosurgery, Wayne State University School of Medicine, Detroit, Michigan
| | - Xiaokun Geng
- China-America Institute of Neuroscience, Beijing Luhe Hospital, Capital Medical University, Beijing, China
- Department of Neurosurgery, Wayne State University School of Medicine, Detroit, Michigan
- Department of Neurology, Beijing Luhe Hospital, Capital Medical University, Beijing, China
| | - Christopher Stone
- Department of Neurosurgery, Wayne State University School of Medicine, Detroit, Michigan
| | - Eric E P Cosky
- Department of Neurosurgery, Wayne State University School of Medicine, Detroit, Michigan
| | - Yu Ji
- Department of General Surgery, Beijing Luhe Hospital, Capital Medical University, Beijing, China
| | - Huishan Du
- China-America Institute of Neuroscience, Beijing Luhe Hospital, Capital Medical University, Beijing, China
- Department of Neurology, Beijing Luhe Hospital, Capital Medical University, Beijing, China
| | - Kezhong Zhang
- Center for Molecular Medicine and Genetics, Wayne State University School of Medicine, Detroit, Michigan
| | - Qinghua Sun
- Division of Environmental Health Sciences, College of Public Health, Ohio State University, Columbus, Ohio
| | - Yuchuan Ding
- Department of Neurosurgery, Wayne State University School of Medicine, Detroit, Michigan
- Department of Research & Development Center, John D. Dingell VA Medical Center, Detroit, Michigan
| |
Collapse
|
25
|
Abdelhamid AS, Brown TJ, Brainard JS, Biswas P, Thorpe GC, Moore HJ, Deane KHO, AlAbdulghafoor FK, Summerbell CD, Worthington HV, Song F, Hooper L. Omega-3 fatty acids for the primary and secondary prevention of cardiovascular disease. Cochrane Database Syst Rev 2018; 11:CD003177. [PMID: 30521670 PMCID: PMC6517311 DOI: 10.1002/14651858.cd003177.pub4] [Citation(s) in RCA: 95] [Impact Index Per Article: 13.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
BACKGROUND Researchers have suggested that omega-3 polyunsaturated fatty acids from oily fish (long-chain omega-3 (LCn3), including eicosapentaenoic acid (EPA) and docosahexaenoic acid (DHA)), as well as from plants (alpha-linolenic acid (ALA)) benefit cardiovascular health. Guidelines recommend increasing omega-3-rich foods, and sometimes supplementation, but recent trials have not confirmed this. OBJECTIVES To assess effects of increased intake of fish- and plant-based omega-3 for all-cause mortality, cardiovascular (CVD) events, adiposity and lipids. SEARCH METHODS We searched CENTRAL, MEDLINE and Embase to April 2017, plus ClinicalTrials.gov and World Health Organization International Clinical Trials Registry to September 2016, with no language restrictions. We handsearched systematic review references and bibliographies and contacted authors. SELECTION CRITERIA We included randomised controlled trials (RCTs) that lasted at least 12 months and compared supplementation and/or advice to increase LCn3 or ALA intake versus usual or lower intake. DATA COLLECTION AND ANALYSIS Two review authors independently assessed studies for inclusion, extracted data and assessed validity. We performed separate random-effects meta-analysis for ALA and LCn3 interventions, and assessed dose-response relationships through meta-regression. MAIN RESULTS We included 79 RCTs (112,059 participants) in this review update and found that 25 were at low summary risk of bias. Trials were of 12 to 72 months' duration and included adults at varying cardiovascular risk, mainly in high-income countries. Most studies assessed LCn3 supplementation with capsules, but some used LCn3- or ALA-rich or enriched foods or dietary advice compared to placebo or usual diet. LCn3 doses ranged from 0.5g/d LCn3 to > 5 g/d (16 RCTs gave at least 3g/d LCn3).Meta-analysis and sensitivity analyses suggested little or no effect of increasing LCn3 on all-cause mortality (RR 0.98, 95% CI 0.90 to 1.03, 92,653 participants; 8189 deaths in 39 trials, high-quality evidence), cardiovascular mortality (RR 0.95, 95% CI 0.87 to 1.03, 67,772 participants; 4544 CVD deaths in 25 RCTs), cardiovascular events (RR 0.99, 95% CI 0.94 to 1.04, 90,378 participants; 14,737 people experienced events in 38 trials, high-quality evidence), coronary heart disease (CHD) mortality (RR 0.93, 95% CI 0.79 to 1.09, 73,491 participants; 1596 CHD deaths in 21 RCTs), stroke (RR 1.06, 95% CI 0.96 to 1.16, 89,358 participants; 1822 strokes in 28 trials) or arrhythmia (RR 0.97, 95% CI 0.90 to 1.05, 53,796 participants; 3788 people experienced arrhythmia in 28 RCTs). There was a suggestion that LCn3 reduced CHD events (RR 0.93, 95% CI 0.88 to 0.97, 84,301 participants; 5469 people experienced CHD events in 28 RCTs); however, this was not maintained in sensitivity analyses - LCn3 probably makes little or no difference to CHD event risk. All evidence was of moderate GRADE quality, except as noted.Increasing ALA intake probably makes little or no difference to all-cause mortality (RR 1.01, 95% CI 0.84 to 1.20, 19,327 participants; 459 deaths, 5 RCTs),cardiovascular mortality (RR 0.96, 95% CI 0.74 to 1.25, 18,619 participants; 219 cardiovascular deaths, 4 RCTs), and CHD mortality (1.1% to 1.0%, RR 0.95, 95% CI 0.72 to 1.26, 18,353 participants; 193 CHD deaths, 3 RCTs) and ALA may make little or no difference to CHD events (RR 1.00, 95% CI 0.80 to 1.22, 19,061 participants, 397 CHD events, 4 RCTs, low-quality evidence). However, increased ALA may slightly reduce risk of cardiovascular events (from 4.8% to 4.7%, RR 0.95, 95% CI 0.83 to 1.07, 19,327 participants; 884 CVD events, 5 RCTs, low-quality evidence with greater effects in trials at low summary risk of bias), and probably reduces risk of arrhythmia (3.3% to 2.6%, RR 0.79, 95% CI 0.57 to 1.10, 4,837 participants; 141 events, 1 RCT). Effects on stroke are unclear.Sensitivity analysis retaining only trials at low summary risk of bias moved effect sizes towards the null (RR 1.0) for all LCn3 primary outcomes except arrhythmias, but for most ALA outcomes, effect sizes moved to suggest protection. LCn3 funnel plots suggested that adding in missing studies/results would move effect sizes towards null for most primary outcomes. There were no dose or duration effects in subgrouping or meta-regression.There was no evidence that increasing LCn3 or ALA altered serious adverse events, adiposity or lipids, except LCn3 reduced triglycerides by ˜15% in a dose-dependant way (high-quality evidence). AUTHORS' CONCLUSIONS This is the most extensive systematic assessment of effects of omega-3 fats on cardiovascular health to date. Moderate- and high-quality evidence suggests that increasing EPA and DHA has little or no effect on mortality or cardiovascular health (evidence mainly from supplement trials). Previous suggestions of benefits from EPA and DHA supplements appear to spring from trials with higher risk of bias. Low-quality evidence suggests ALA may slightly reduce CVD event and arrhythmia risk.
Collapse
Affiliation(s)
- Asmaa S Abdelhamid
- University of East AngliaNorwich Medical SchoolNorwich Research ParkNorwichNorfolkUKNR4 7TJ
| | - Tracey J Brown
- University of East AngliaNorwich Medical SchoolNorwich Research ParkNorwichNorfolkUKNR4 7TJ
| | - Julii S Brainard
- University of East AngliaNorwich Medical SchoolNorwich Research ParkNorwichNorfolkUKNR4 7TJ
| | - Priti Biswas
- University of East AngliaMED/HSCNorwich Research ParkNorwichUKNR4 7TJ
| | - Gabrielle C Thorpe
- University of East AngliaSchool of Health SciencesEarlham RoadNorwichUKNR4 7TJ
| | - Helen J Moore
- Durham UniversityWolfson Research InstituteDurhamUKDH1 3LE
| | - Katherine HO Deane
- University of East AngliaSchool of Health SciencesEarlham RoadNorwichUKNR4 7TJ
| | - Fai K AlAbdulghafoor
- University of East AngliaNorwich Medical SchoolNorwich Research ParkNorwichNorfolkUKNR4 7TJ
| | - Carolyn D Summerbell
- Durham UniversityDepartment of Sport and Exercise Science42 Old ElvetDurhamUKDH13HN
| | - Helen V Worthington
- Division of Dentistry, School of Medical Sciences, Faculty of Biology, Medicine and Health, The University of ManchesterCochrane Oral HealthJR Moore BuildingOxford RoadManchesterUKM13 9PL
| | - Fujian Song
- University of East AngliaNorwich Medical SchoolNorwich Research ParkNorwichNorfolkUKNR4 7TJ
| | - Lee Hooper
- University of East AngliaNorwich Medical SchoolNorwich Research ParkNorwichNorfolkUKNR4 7TJ
| | | |
Collapse
|
26
|
Estrada-Luna D, Ortiz-Rodriguez MA, Medina-Briseño L, Carreón-Torres E, Izquierdo-Vega JA, Sharma A, Cancino-Díaz JC, Pérez-Méndez O, Belefant-Miller H, Betanzos-Cabrera G. Current Therapies Focused on High-Density Lipoproteins Associated with Cardiovascular Disease. Molecules 2018; 23:molecules23112730. [PMID: 30360466 PMCID: PMC6278283 DOI: 10.3390/molecules23112730] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2018] [Revised: 10/20/2018] [Accepted: 10/21/2018] [Indexed: 02/06/2023] Open
Abstract
High-density lipoproteins (HDL) comprise a heterogeneous family of lipoprotein particles divided into subclasses that are determined by density, size and surface charge as well as protein composition. Epidemiological studies have suggested an inverse correlation between High-density lipoprotein-cholesterol (HDL-C) levels and the risk of cardiovascular diseases and atherosclerosis. HDLs promote reverse cholesterol transport (RCT) and have several atheroprotective functions such as anti-inflammation, anti-thrombosis, and anti-oxidation. HDLs are considered to be atheroprotective because they are associated in serum with paraoxonases (PONs) which protect HDL from oxidation. Polyphenol consumption reduces the risk of chronic diseases in humans. Polyphenols increase the binding of HDL to PON1, increasing the catalytic activity of PON1. This review summarizes the evidence currently available regarding pharmacological and alternative treatments aimed at improving the functionality of HDL-C. Information on the effectiveness of the treatments has contributed to the understanding of the molecular mechanisms that regulate plasma levels of HDL-C, thereby promoting the development of more effective treatment of cardiovascular diseases. For that purpose, Scopus and Medline databases were searched to identify the publications investigating the impact of current therapies focused on high-density lipoproteins.
Collapse
Affiliation(s)
- Diego Estrada-Luna
- Instituto Nacional de Cardiología "Ignacio Chávez" Juan Badiano No. 1, Belisario Domínguez Sección 16, 14080 Tlalpan, Mexico City, Mexico.
| | - María Araceli Ortiz-Rodriguez
- Facultad de Nutrición, Universidad Autónoma del Estado de Morelos, UAEM, Calle Río Iztaccihuatl S/N, Vista Hermosa, 62350 Cuernavaca, Morelos, Mexico.
| | - Lizett Medina-Briseño
- Universidad de la Sierra Sur, UNSIS, Miahuatlán de Porfirio Díaz, 70800 Oaxaca, Mexico.
| | - Elizabeth Carreón-Torres
- Instituto Nacional de Cardiología "Ignacio Chávez" Juan Badiano No. 1, Belisario Domínguez Sección 16, 14080 Tlalpan, Mexico City, Mexico.
| | - Jeannett Alejandra Izquierdo-Vega
- Área Académica de Medicina, Instituto de Ciencias de la Salud, Universidad Autónoma del Estado de Hidalgo, Carretera Actopan-Tilcuautla, Ex-Hacienda La Concepción S/N, San Agustín Tlaxiaca, 42160 Hidalgo, Mexico.
| | - Ashutosh Sharma
- Tecnologico de Monterrey, School of Engineering and Sciences, Campus Queretaro, Epigmenio Gonzalez 500, 76130 Queretaro, Mexico.
| | - Juan Carlos Cancino-Díaz
- Departamento de Microbiología, Escuela Nacional de Ciencias Biológicas del Instituto Politécnico Nacional, 11340 Ciudad de México, Mexico.
| | - Oscar Pérez-Méndez
- Instituto Nacional de Cardiología "Ignacio Chávez" Juan Badiano No. 1, Belisario Domínguez Sección 16, 14080 Tlalpan, Mexico City, Mexico.
| | | | - Gabriel Betanzos-Cabrera
- Área Académica de Medicina, Instituto de Ciencias de la Salud, Universidad Autónoma del Estado de Hidalgo, Carretera Actopan-Tilcuautla, Ex-Hacienda La Concepción S/N, San Agustín Tlaxiaca, 42160 Hidalgo, Mexico.
| |
Collapse
|
27
|
Mortazavi A, Nematipoor E, Djalali M, Keshavarz SA, Samavat S, Zarei M, Javanbakht MH. The Effect of Omega-3 Fatty Acids on Serum Apelin Levels in Cardiovascular Disease: A Randomized, Double-Blind, Placebo-Controlled Trial. Rep Biochem Mol Biol 2018; 7:59-66. [PMID: 30324119 PMCID: PMC6175591] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2017] [Accepted: 09/23/2017] [Indexed: 06/08/2023]
Abstract
BACKGROUND Cardiovascular disease (CVD) is the leading cause of mortality worldwide. Omega-3 fatty acids have been shown to have both anti-atherogenic and anti-inflammatory effects through inducing the expression and production of adipokines. Adipokines such as apelin, have been observed to play a protective role in the incidence and progression of CVD. The aim of this study was to assess the influence of omega-3 fatty acids supplementation on the serum apelin levels in patients with cardiovascular disease. METHODS Forty-six male patients with CVD participated in the study. Patients were randomly allocated into two groups receiving either omega-3 fatty acids or a placebo. Participants received 4 g of omega-3 fatty acids (EPA: 720 mg, DHA: 480 mg) or a placebo (edible paraffin) for 8 weeks. Serum apelin levels, high sensitive C-reactive protein (hs-CRP), and lipid profiles were measured. Dietary intake, anthropometric parameters, body composition, systolic and diastolic blood pressure were evaluated before and after the 8 weeks of intervention. Statistical analyses were performed using SPSS version 22. RESULTS Two participants from the placebo group withdrew from the study. Prior to the intervention, no significant differences were present between the two groups in age, body mass index, body composition, dietary intakes, lipid profiles and blood pressure. Compared to placebo, the intake of omega-3 fatty acids increased serum apelin levels (p= 0.018), decreased the levels of LDL cholesterol, and decreased serum hs-CRP concentrations (p= 0.007, p= 0.011 respectively). Additionally, the concentrations of VLDL, TG and hs-CRP (p= 0.037, p= 0.037 and p= 0.016 respectively) declined compared to baseline and final values in the omega-3 fatty acids group. CONCLUSION Omega-3 fatty acid supplementation increases serum apelin and HDL concentrations, while decreasing serum LDL-C and hs-CRP levels.
Collapse
Affiliation(s)
- Akramsadat Mortazavi
- Department of cellular and molecular nutrition, School of nutritional sciences and dietetics, International Campus, Tehran University of Medical Sciences, Tehran, Iran.
| | - Ebrahim Nematipoor
- Department of cardiology, Tehran University of Medical Sciences, Tehran, Iran.
| | - Mahmoud Djalali
- Department of cellular and molecular nutrition, School of nutritional sciences and dietetics, Tehran University of Medical Sciences, Tehran, Iran.
| | - Seyed Ali Keshavarz
- Department of clinical nutrition, School of nutritional sciences and dietetics, Tehran University of Medical Sciences, Tehran, Iran.
| | - Simin Samavat
- Department of cellular and molecular nutrition, School of nutritional sciences and dietetics, Tehran University of Medical Sciences, Tehran, Iran.
| | - Mahnaz Zarei
- Department of cellular and molecular nutrition, School of nutritional sciences and dietetics, Tehran University of Medical Sciences, Tehran, Iran.
| | - Mohammad Hassan Javanbakht
- Department of cellular and molecular nutrition, School of nutritional sciences and dietetics, International Campus, Tehran University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
28
|
Kain V, Halade GV. Immune responsive resolvin D1 programs peritoneal macrophages and cardiac fibroblast phenotypes in diversified metabolic microenvironment. J Cell Physiol 2018; 234:3910-3920. [PMID: 30191990 DOI: 10.1002/jcp.27165] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2017] [Accepted: 07/11/2018] [Indexed: 12/20/2022]
Abstract
Bioactive lipid mediators derived from n-3 and n-6 fatty acids are known to modulate leukocytes. Metabolic transformation of essential fatty acids to endogenous bioactive molecules plays a major role in human health. Here we tested the potential of substrates; linoleic acid (LA) and docosahexaenoic acid (DHA) and their bioactive products; resolvin D1 (RvD1) and 12- S-hydroxyeicosatetraenoic acids (HETE) to modulate macrophage plasticity and cardiac fibroblast phenotype in presence or absence of lipid metabolizing enzyme 12/15-lipoxygenase (LOX). Peritoneal macrophages and cardiac fibroblasts were isolated from wild-type (C57BL/6J) and 12/15LOX -/- mice and treated with DHA, LA, 12(S)-HETE, and RvD1 for 4, 8, 12, and 24 hr. LA, DHA, 12(S)-HETE, and RvD1 elicited mRNA expression of proinflammatory markers; tumor necrosis factor-α ( Tnf-α), interleukin 6 ( IL-6), chemokine (C-C motif) ligand 2 (Ccl2), and IL-1β in wild type (WT) and in 12/15LOX -/- macrophages at early time point (4 hr). Bioactive immunoresolvent RvD1 lowered the levels of Tnf-α, IL-6, and IL-1β at 24 hr time point. Both DHA and RvD1 stimulated the proresolving markers such as arginase 1 ( Arg-1), chitinase-like protein 3 ( Ym-1), and mannose receptor C-type 1 in WT macrophage. RvD1 induced proresolving phenotype Arg-1 expression in both WT 12/15LOX -/- macrophages even in presence of 12(S)-HETE. RvD1 peaked 5LOX expression in both WT and 12/15LOX -/- at 24 hr time point compared with DHA. RvD1 diminished cyclooxygenase-2 but upregulated 5LOX expression in fibroblast compared with DHA. In summary, the feed-forward enzymatic interaction with fatty acids substrates and direct mediators (RvD1 and 12(S)-HETE) are responsive in determining macrophages phenotype and cardiac fibroblast plasticity. Particularly, macrophages and fibroblast phenotypes are responsive to milieu and RvD1 governs the milieu-dependent chemokine signaling in presence or absence of 12/15LOX enzyme to resolve inflammation.
Collapse
Affiliation(s)
- Vasundhara Kain
- Division of Cardiovascular Disease, Department of Medicine, The University of Alabama at Birmingham, Birmingham, Alabama
| | - Ganesh V Halade
- Division of Cardiovascular Disease, Department of Medicine, The University of Alabama at Birmingham, Birmingham, Alabama
| |
Collapse
|
29
|
Abdelhamid AS, Brown TJ, Brainard JS, Biswas P, Thorpe GC, Moore HJ, Deane KHO, AlAbdulghafoor FK, Summerbell CD, Worthington HV, Song F, Hooper L. Omega-3 fatty acids for the primary and secondary prevention of cardiovascular disease. Cochrane Database Syst Rev 2018; 7:CD003177. [PMID: 30019766 PMCID: PMC6513557 DOI: 10.1002/14651858.cd003177.pub3] [Citation(s) in RCA: 122] [Impact Index Per Article: 17.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
BACKGROUND Researchers have suggested that omega-3 polyunsaturated fatty acids from oily fish (long-chain omega-3 (LCn3), including eicosapentaenoic acid (EPA) and docosahexaenoic acid (DHA)), as well as from plants (alpha-linolenic acid (ALA)) benefit cardiovascular health. Guidelines recommend increasing omega-3-rich foods, and sometimes supplementation, but recent trials have not confirmed this. OBJECTIVES To assess effects of increased intake of fish- and plant-based omega-3 for all-cause mortality, cardiovascular (CVD) events, adiposity and lipids. SEARCH METHODS We searched CENTRAL, MEDLINE and Embase to April 2017, plus ClinicalTrials.gov and World Health Organization International Clinical Trials Registry to September 2016, with no language restrictions. We handsearched systematic review references and bibliographies and contacted authors. SELECTION CRITERIA We included randomised controlled trials (RCTs) that lasted at least 12 months and compared supplementation and/or advice to increase LCn3 or ALA intake versus usual or lower intake. DATA COLLECTION AND ANALYSIS Two review authors independently assessed studies for inclusion, extracted data and assessed validity. We performed separate random-effects meta-analysis for ALA and LCn3 interventions, and assessed dose-response relationships through meta-regression. MAIN RESULTS We included 79 RCTs (112,059 participants) in this review update and found that 25 were at low summary risk of bias. Trials were of 12 to 72 months' duration and included adults at varying cardiovascular risk, mainly in high-income countries. Most studies assessed LCn3 supplementation with capsules, but some used LCn3- or ALA-rich or enriched foods or dietary advice compared to placebo or usual diet.Meta-analysis and sensitivity analyses suggested little or no effect of increasing LCn3 on all-cause mortality (RR 0.98, 95% CI 0.90 to 1.03, 92,653 participants; 8189 deaths in 39 trials, high-quality evidence), cardiovascular mortality (RR 0.95, 95% CI 0.87 to 1.03, 67,772 participants; 4544 CVD deaths in 25 RCTs), cardiovascular events (RR 0.99, 95% CI 0.94 to 1.04, 90,378 participants; 14,737 people experienced events in 38 trials, high-quality evidence), coronary heart disease (CHD) mortality (RR 0.93, 95% CI 0.79 to 1.09, 73,491 participants; 1596 CHD deaths in 21 RCTs), stroke (RR 1.06, 95% CI 0.96 to 1.16, 89,358 participants; 1822 strokes in 28 trials) or arrhythmia (RR 0.97, 95% CI 0.90 to 1.05, 53,796 participants; 3788 people experienced arrhythmia in 28 RCTs). There was a suggestion that LCn3 reduced CHD events (RR 0.93, 95% CI 0.88 to 0.97, 84,301 participants; 5469 people experienced CHD events in 28 RCTs); however, this was not maintained in sensitivity analyses - LCn3 probably makes little or no difference to CHD event risk. All evidence was of moderate GRADE quality, except as noted.Increasing ALA intake probably makes little or no difference to all-cause mortality (RR 1.01, 95% CI 0.84 to 1.20, 19,327 participants; 459 deaths, 5 RCTs),cardiovascular mortality (RR 0.96, 95% CI 0.74 to 1.25, 18,619 participants; 219 cardiovascular deaths, 4 RCTs), and it may make little or no difference to CHD events (RR 1.00, 95% CI 0.80 to 1.22, 19,061 participants, 397 CHD events, 4 RCTs, low-quality evidence). However, increased ALA may slightly reduce risk of cardiovascular events (from 4.8% to 4.7%, RR 0.95, 95% CI 0.83 to 1.07, 19,327 participants; 884 CVD events, 5 RCTs, low-quality evidence), and probably reduces risk of CHD mortality (1.1% to 1.0%, RR 0.95, 95% CI 0.72 to 1.26, 18,353 participants; 193 CHD deaths, 3 RCTs), and arrhythmia (3.3% to 2.6%, RR 0.79, 95% CI 0.57 to 1.10, 4,837 participants; 141 events, 1 RCT). Effects on stroke are unclear.Sensitivity analysis retaining only trials at low summary risk of bias moved effect sizes towards the null (RR 1.0) for all LCn3 primary outcomes except arrhythmias, but for most ALA outcomes, effect sizes moved to suggest protection. LCn3 funnel plots suggested that adding in missing studies/results would move effect sizes towards null for most primary outcomes. There were no dose or duration effects in subgrouping or meta-regression.There was no evidence that increasing LCn3 or ALA altered serious adverse events, adiposity or lipids, although LCn3 slightly reduced triglycerides and increased HDL. ALA probably reduces HDL (high- or moderate-quality evidence). AUTHORS' CONCLUSIONS This is the most extensive systematic assessment of effects of omega-3 fats on cardiovascular health to date. Moderate- and high-quality evidence suggests that increasing EPA and DHA has little or no effect on mortality or cardiovascular health (evidence mainly from supplement trials). Previous suggestions of benefits from EPA and DHA supplements appear to spring from trials with higher risk of bias. Low-quality evidence suggests ALA may slightly reduce CVD event risk, CHD mortality and arrhythmia.
Collapse
Affiliation(s)
- Asmaa S Abdelhamid
- University of East AngliaNorwich Medical SchoolNorwich Research ParkNorwichUKNR4 7TJ
| | - Tracey J Brown
- University of East AngliaNorwich Medical SchoolNorwich Research ParkNorwichUKNR4 7TJ
| | - Julii S Brainard
- University of East AngliaNorwich Medical SchoolNorwich Research ParkNorwichUKNR4 7TJ
| | - Priti Biswas
- University of East AngliaMED/HSCNorwich Research ParkNorwichUKNR4 7TJ
| | - Gabrielle C Thorpe
- University of East AngliaSchool of Health SciencesEarlham RoadNorwichUKNR4 7TJ
| | - Helen J Moore
- Durham UniversityWolfson Research InstituteDurhamUKDH1 3LE
| | - Katherine HO Deane
- University of East AngliaSchool of Health SciencesEarlham RoadNorwichUKNR4 7TJ
| | - Fai K AlAbdulghafoor
- University of East AngliaNorwich Medical SchoolNorwich Research ParkNorwichUKNR4 7TJ
| | - Carolyn D Summerbell
- Durham UniversityDepartment of Sport and Exercise Science42 Old ElvetDurhamUKDH13HN
| | - Helen V Worthington
- Division of Dentistry, School of Medical Sciences, Faculty of Biology, Medicine and Health, The University of ManchesterCochrane Oral HealthJR Moore BuildingOxford RoadManchesterUKM13 9PL
| | - Fujian Song
- University of East AngliaNorwich Medical SchoolNorwich Research ParkNorwichUKNR4 7TJ
| | - Lee Hooper
- University of East AngliaNorwich Medical SchoolNorwich Research ParkNorwichUKNR4 7TJ
| |
Collapse
|
30
|
Jung TW, Park HS, Choi GH, Kim D, Ahn SH, Kim DS, Lee T, Jeong JH. Maresin 1 attenuates pro-inflammatory reactions and ER stress in HUVECs via PPARα-mediated pathway. Mol Cell Biochem 2018; 448:335-347. [DOI: 10.1007/s11010-018-3392-y] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2018] [Accepted: 06/26/2018] [Indexed: 12/11/2022]
|
31
|
Wang K, Zha Y, Lei H, Xu X. MRI Study on the Changes of Bone Marrow Microvascular Permeability and Fat Content after Total-Body X-Ray Irradiation. Radiat Res 2017; 189:205-212. [PMID: 29251550 DOI: 10.1667/rr14865.1] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/03/2022]
Abstract
In this study, we investigated microvascular perfusion status, changes to fat content and fatty acid composition in the bone marrow of rat femurs after total-body irradiation by quantitative permeability parameters of dynamic contrast-enhanced magnetic resonance imaging (DCE-MRI) and ex vivo high-resolution magic angle spinning (HRMAS) 1H nuclear magnetic resonance spectroscopy (NMRS). Thirty-six Sprague-Dawley rats were randomly assigned to either an irradiated or nonirradiated control group. Permeability imaging using DCE-MRI and HRMAS 1H NMRS was performed before irradiation, as well as at days 4 and 7 postirradiation. The volume transfer constant (Ktrans) values increased to 2.219 ± 0.418/min ( P < 0.01) at day 4 and to 2.760 ± 0.217/min at day 7 ( P < 0.01) postirradiation. The plasma fraction (vp) values gradually decreased. The proportion of (n-6) polyunsaturated fatty acids (PUFA) gradually reached a peak at day 7, the proportion of (n-3) PUFA gradually decreased and the proportion of saturated fatty acids gradually increased. After irradiation, Ktrans at different times showed significant negative correlation with (n-3) PUFA ( r = -0.6393, P < 0.01) and significant positive correlation with (n-6) PUFA ( r = 0.6841, P < 0.05). These findings indicate that bone marrow microcirculation perfusion and vascular permeability correlated with fat content at an early time point after irradiation. A pathophysiological mechanism may exist based on fat-vascular permeability in the case of injury to bone marrow microcirculation.
Collapse
Affiliation(s)
- Kejun Wang
- a Department of Radiology Renmin Hospital of Wuhan University, Wuhan, China
| | - Yunfei Zha
- a Department of Radiology Renmin Hospital of Wuhan University, Wuhan, China
| | - Hao Lei
- b Wuhan Institute of Physics and Mathematics, Chinese Academy of Sciences, Wuhan, China; and
| | - Xiao Xu
- c Life Science, GE Healthcare, Shanghai, China
| |
Collapse
|
32
|
Zock PL, Blom WAM, Nettleton JA, Hornstra G. Progressing Insights into the Role of Dietary Fats in the Prevention of Cardiovascular Disease. Curr Cardiol Rep 2017; 18:111. [PMID: 27650783 PMCID: PMC5030225 DOI: 10.1007/s11886-016-0793-y] [Citation(s) in RCA: 48] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
Dietary fats have important effects on the risk of cardiovascular disease (CVD). Abundant evidence shows that partial replacement of saturated fatty acids (SAFA) with unsaturated fatty acids improves the blood lipid and lipoprotein profile and reduces the risk of coronary heart disease (CHD). Low-fat diets high in refined carbohydrates and sugar are not effective. Very long-chain polyunsaturated n-3 or omega-3 fatty acids (n-3 VLCPUFA) present in fish have multiple beneficial metabolic effects, and regular intake of fatty fish is associated with lower risks of fatal CHD and stroke. Food-based guidelines on dietary fats recommend limiting the consumption of animal fats high in SAFA, using vegetable oils high in monounsaturated (MUFA) and polyunsaturated fatty acids (PUFA), and eating fatty fish. These recommendations are part of a healthy eating pattern that also includes ample intake of plant-based foods rich in fiber and limited sugar and salt.
Collapse
Affiliation(s)
- Peter L Zock
- Unilever R&D Vlaardingen, Olivier van Noortlaan 120, 3133 AT, Vlaardingen, The Netherlands.
| | - Wendy A M Blom
- Unilever R&D Vlaardingen, Olivier van Noortlaan 120, 3133 AT, Vlaardingen, The Netherlands
| | - Joyce A Nettleton
- Member of the IUNS-associated International Expert Movement to Improve Dietary Fat Quality, ScienceVoice Consulting, 2931 Race Street, Denver, CO, 80205, USA
| | - Gerard Hornstra
- Member of the IUNS-associated International Expert Movement to Improve Dietary Fat Quality, Maastricht University, Brikkenoven 14, 6247 BG, Gronsveld, Netherlands
| |
Collapse
|
33
|
Momtazi AA, Banach M, Pirro M, Katsiki N, Sahebkar A. Regulation of PCSK9 by nutraceuticals. Pharmacol Res 2017; 120:157-169. [PMID: 28363723 DOI: 10.1016/j.phrs.2017.03.023] [Citation(s) in RCA: 101] [Impact Index Per Article: 12.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/26/2017] [Accepted: 03/27/2017] [Indexed: 12/19/2022]
Abstract
PCSK9 (proprotein convertase subtilisin kexin type 9) is a liver secretory enzyme that regulates plasma low-density lipoprotein (LDL) cholesterol (LDL-C) levels through modulation of LDL receptor (LDLR) density on the surface of hepatocytes. Inhibition of PCSK9 using monoclonal antibodies can efficiently lower plasma LDL-C, non-high-density lipoprotein cholesterol and lipoprotein (a). PCSK9 inhibition is also an effective adjunct to statin therapy; however, the cost-effectiveness of currently available PCSK9 inhibitors is under question. Nutraceuticals offer a safe and cost-effective option for PCSK9 inhibition. Several nutraceuticals have been reported to modulate PCSK9 levels and exert LDL-lowering activity. Mechanistically, those nutraceuticals that inhibit PCSK9 through a SREBP (sterol-responsive element binding protein)-independent pathway can be more effective in lowering plasma LDL-C levels compared with those inhibiting PCSK9 through the SREBP pathway. The present review aims to collect available data on the nutraceuticals with PCSK9-inhibitory effect and the underlying mechanisms.
Collapse
Affiliation(s)
- Amir Abbas Momtazi
- Nanotechnology Research Center, Department of Medical Biotechnology, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran; Student Research Committee, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Maciej Banach
- Department of Hypertension, WAM University Hospital in Lodz, Medical University of Lodz, Zeromskiego 113, Lodz, Poland; Polish Mother's Memorial Hospital Research Institute (PMMHRI), Lodz, Poland
| | - Matteo Pirro
- Unit of Internal Medicine, Angiology and Arteriosclerosis Diseases, Department of Medicine, University of Perugia, Perugia, Italy
| | - Niki Katsiki
- Second Propedeutic Department of Internal Medicine, Medical School, Aristotle University of Thessaloniki, Hippocration Hospital, Thessaloniki, Greece
| | - Amirhossein Sahebkar
- Biotechnology Research Center, Mashhad University of Medical Sciences, Mashhad 9177948564, Iran; Metabolic Research Centre, Royal Perth Hospital, School of Medicine and Pharmacology, University of Western Australia, Perth, Australia.
| |
Collapse
|
34
|
Eicosapentaenoic acid-enriched phospholipids improve atherosclerosis by mediating cholesterol metabolism. J Funct Foods 2017. [DOI: 10.1016/j.jff.2017.02.020] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
|
35
|
Association between polyunsaturated fatty acids and inflammatory markers in patients in secondary prevention of cardiovascular disease. Nutrition 2017; 37:30-36. [DOI: 10.1016/j.nut.2016.12.006] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2016] [Revised: 11/19/2016] [Accepted: 12/14/2016] [Indexed: 12/21/2022]
|
36
|
Moreno-Perez S, Luna P, Señorans J, Rocha-Martin J, Guisan JM, Fernandez-Lorente G. Enzymatic transesterification in a solvent-free system: synthesis of sn-2 docosahexaenoyl monoacylglycerol. BIOCATAL BIOTRANSFOR 2017. [DOI: 10.1080/10242422.2017.1319823] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/19/2022]
Affiliation(s)
| | - Pilar Luna
- Departamento de Química Física Aplicada, Universidad Autónoma, Madrid, UK
| | - Javier Señorans
- Departamento de Química Física Aplicada, Universidad Autónoma, Madrid, UK
| | | | - Jose M. Guisan
- Instituto de Catálisis CSIC, Campus UAM-CSIC, Madrid, UK
| | - Gloria Fernandez-Lorente
- Instituto de Catálisis CSIC, Campus UAM-CSIC, Madrid, UK
- Instituto de Investigación en Ciencias de la Alimentación (CIAL) CSIC-UAM, Madrid, UK
| |
Collapse
|
37
|
Moreno-Perez S, Turati DFM, Borges JP, Luna P, Señorans FJ, Guisan JM, Fernandez-Lorente G. Critical Role of Different Immobilized Biocatalysts of a Given Lipase in the Selective Ethanolysis of Sardine Oil. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2017; 65:117-122. [PMID: 27973785 DOI: 10.1021/acs.jafc.6b05243] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/06/2023]
Abstract
Different immobilized derivatives of two lipases were tested as catalysts of the synthesis of ethyl esters of omega-3 fatty acids during the ethanolysis of sardine oil in solvent-free systems at 25 °C. Lipases from Thermomyces lanuginosus (TLL) and Lecitase Ultra (a phospholipase with lipolytic activity) were studied. Lipases were adsorbed on hydrophobic Sepabeads C18 through the open active center and on an anion-exchanger Duolite with the active center exposed to the reaction medium. TLL-Sepabeads derivatives exhibit a high activity of 9 UI/mg of immobilized enzyme, and they are 20-fold more active than TLL-Duolite derivatives and almost 1000-fold more active than Lipozyme TL IM (the commercial derivative from Novozymes). Lecitase-Sepabeads exhibit a high selectivity for the synthesis of the ethyl ester of EPA that is 43-fold faster than the synthesis of the ethyl ester of DHA.
Collapse
Affiliation(s)
| | - Daniela Flavia Machado Turati
- Department of Biochemistry and Microbiology, Univ Estadual Paulista at Rio Claro - 8 UNESP , Rio Claro, SP 13506-900, Brazil
| | - Janaina Pires Borges
- Departamento de Quı́mica e Tecnologia, Instituto de Química - UNESP , 14800-069, 10 Araraquara, SP, Brazil
| | - Pilar Luna
- Departamento de Quı́mica Fı́sica Aplicada, Universidad Autónoma , 28049 Madrid, Sapin
| | | | - Jose M Guisan
- Instituto de Catálisis, CSIC , Campus UAM-CSIC, 28049 Madrid, Spain
| | | |
Collapse
|
38
|
Takashima A, Fukuda D, Tanaka K, Higashikuni Y, Hirata Y, Nishimoto S, Yagi S, Yamada H, Soeki T, Wakatsuki T, Taketani Y, Shimabukuro M, Sata M. Combination of n-3 polyunsaturated fatty acids reduces atherogenesis in apolipoprotein E-deficient mice by inhibiting macrophage activation. Atherosclerosis 2016; 254:142-150. [DOI: 10.1016/j.atherosclerosis.2016.10.002] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/14/2016] [Revised: 10/04/2016] [Accepted: 10/04/2016] [Indexed: 11/28/2022]
|
39
|
Alshehry ZH, Mundra PA, Barlow CK, Mellett NA, Wong G, McConville MJ, Simes J, Tonkin AM, Sullivan DR, Barnes EH, Nestel PJ, Kingwell BA, Marre M, Neal B, Poulter NR, Rodgers A, Williams B, Zoungas S, Hillis GS, Chalmers J, Woodward M, Meikle PJ. Plasma Lipidomic Profiles Improve on Traditional Risk Factors for the Prediction of Cardiovascular Events in Type 2 Diabetes Mellitus. Circulation 2016; 134:1637-1650. [PMID: 27756783 DOI: 10.1161/circulationaha.116.023233] [Citation(s) in RCA: 198] [Impact Index Per Article: 22.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/27/2016] [Accepted: 09/29/2016] [Indexed: 12/14/2022]
Abstract
BACKGROUND Clinical lipid measurements do not show the full complexity of the altered lipid metabolism associated with diabetes mellitus or cardiovascular disease. Lipidomics enables the assessment of hundreds of lipid species as potential markers for disease risk. METHODS Plasma lipid species (310) were measured by a targeted lipidomic analysis with liquid chromatography electrospray ionization-tandem mass spectrometry on a case-cohort (n=3779) subset from the ADVANCE trial (Action in Diabetes and Vascular Disease: Preterax and Diamicron-MR Controlled Evaluation). The case-cohort was 61% male with a mean age of 67 years. All participants had type 2 diabetes mellitus with ≥1 additional cardiovascular risk factors, and 35% had a history of macrovascular disease. Weighted Cox regression was used to identify lipid species associated with future cardiovascular events (nonfatal myocardial infarction, nonfatal stroke, and cardiovascular death) and cardiovascular death during a 5-year follow-up period. Multivariable models combining traditional risk factors with lipid species were optimized with the Akaike information criteria. C statistics and NRIs were calculated within a 5-fold cross-validation framework. RESULTS Sphingolipids, phospholipids (including lyso- and ether- species), cholesteryl esters, and glycerolipids were associated with future cardiovascular events and cardiovascular death. The addition of 7 lipid species to a base model (14 traditional risk factors and medications) to predict cardiovascular events increased the C statistic from 0.680 (95% confidence interval [CI], 0.678-0.682) to 0.700 (95% CI, 0.698-0.702; P<0.0001) with a corresponding continuous NRI of 0.227 (95% CI, 0.219-0.235). The prediction of cardiovascular death was improved with the incorporation of 4 lipid species into the base model, showing an increase in the C statistic from 0.740 (95% CI, 0.738-0.742) to 0.760 (95% CI, 0.757-0.762; P<0.0001) and a continuous net reclassification index of 0.328 (95% CI, 0.317-0.339). The results were validated in a subcohort with type 2 diabetes mellitus (n=511) from the LIPID trial (Long-Term Intervention With Pravastatin in Ischemic Disease). CONCLUSIONS The improvement in the prediction of cardiovascular events, above traditional risk factors, demonstrates the potential of plasma lipid species as biomarkers for cardiovascular risk stratification in diabetes mellitus. CLINICAL TRIAL REGISTRATION URL: https://clinicaltrials.gov. Unique identifier: NCT00145925.
Collapse
Affiliation(s)
- Zahir H Alshehry
- From Baker IDI Heart and Diabetes Institute, Melbourne, VIC, Australia (Z.H.A., P.A.M., C.K.B., N.A.M., G.W., P.J.N., B.A.K., P.J.M.); King Fahad Medical City, Riyadh, Saudi Arabia (Z.H.A.); Department of Biochemistry and Molecular Biology, University of Melbourne, Melbourne, VIC, Australia (Z.H.A., M.J.M., P.J.M.); NHMRC Clinical Trials Centre, University of Sydney, Sydney, NSW, Australia (J.S., E.H.B.); School of Public Health and Preventive Medicine, Monash University, Melbourne, VIC, Australia (A.M.T., S.Z.); Royal Prince Alfred Hospital, Sydney, NSW, Australia (D.R.S.); Hópital Bichat-Claude Bernard and Université Paris 7, Paris, France (M.M.); George Institute for Global Health, Sydney, NSW, Australia (B.N., N.R.P., S.Z., G.S.H., J.C., M.W.); University College London and National Institute for Health Research, University College London Hospitals, Biomedical Research Centre, London, UK (B.W.); Department of Cardiology, Royal Perth Hospital/University of Western Australia, Perth, WA, Australia (G.S.H.); George Institute for Global Health, University of Oxford, Oxford, UK (M.W.); and Department of Epidemiology, Johns Hopkins University, Baltimore, MD (M.W.)
| | - Piyushkumar A Mundra
- From Baker IDI Heart and Diabetes Institute, Melbourne, VIC, Australia (Z.H.A., P.A.M., C.K.B., N.A.M., G.W., P.J.N., B.A.K., P.J.M.); King Fahad Medical City, Riyadh, Saudi Arabia (Z.H.A.); Department of Biochemistry and Molecular Biology, University of Melbourne, Melbourne, VIC, Australia (Z.H.A., M.J.M., P.J.M.); NHMRC Clinical Trials Centre, University of Sydney, Sydney, NSW, Australia (J.S., E.H.B.); School of Public Health and Preventive Medicine, Monash University, Melbourne, VIC, Australia (A.M.T., S.Z.); Royal Prince Alfred Hospital, Sydney, NSW, Australia (D.R.S.); Hópital Bichat-Claude Bernard and Université Paris 7, Paris, France (M.M.); George Institute for Global Health, Sydney, NSW, Australia (B.N., N.R.P., S.Z., G.S.H., J.C., M.W.); University College London and National Institute for Health Research, University College London Hospitals, Biomedical Research Centre, London, UK (B.W.); Department of Cardiology, Royal Perth Hospital/University of Western Australia, Perth, WA, Australia (G.S.H.); George Institute for Global Health, University of Oxford, Oxford, UK (M.W.); and Department of Epidemiology, Johns Hopkins University, Baltimore, MD (M.W.)
| | - Christopher K Barlow
- From Baker IDI Heart and Diabetes Institute, Melbourne, VIC, Australia (Z.H.A., P.A.M., C.K.B., N.A.M., G.W., P.J.N., B.A.K., P.J.M.); King Fahad Medical City, Riyadh, Saudi Arabia (Z.H.A.); Department of Biochemistry and Molecular Biology, University of Melbourne, Melbourne, VIC, Australia (Z.H.A., M.J.M., P.J.M.); NHMRC Clinical Trials Centre, University of Sydney, Sydney, NSW, Australia (J.S., E.H.B.); School of Public Health and Preventive Medicine, Monash University, Melbourne, VIC, Australia (A.M.T., S.Z.); Royal Prince Alfred Hospital, Sydney, NSW, Australia (D.R.S.); Hópital Bichat-Claude Bernard and Université Paris 7, Paris, France (M.M.); George Institute for Global Health, Sydney, NSW, Australia (B.N., N.R.P., S.Z., G.S.H., J.C., M.W.); University College London and National Institute for Health Research, University College London Hospitals, Biomedical Research Centre, London, UK (B.W.); Department of Cardiology, Royal Perth Hospital/University of Western Australia, Perth, WA, Australia (G.S.H.); George Institute for Global Health, University of Oxford, Oxford, UK (M.W.); and Department of Epidemiology, Johns Hopkins University, Baltimore, MD (M.W.)
| | - Natalie A Mellett
- From Baker IDI Heart and Diabetes Institute, Melbourne, VIC, Australia (Z.H.A., P.A.M., C.K.B., N.A.M., G.W., P.J.N., B.A.K., P.J.M.); King Fahad Medical City, Riyadh, Saudi Arabia (Z.H.A.); Department of Biochemistry and Molecular Biology, University of Melbourne, Melbourne, VIC, Australia (Z.H.A., M.J.M., P.J.M.); NHMRC Clinical Trials Centre, University of Sydney, Sydney, NSW, Australia (J.S., E.H.B.); School of Public Health and Preventive Medicine, Monash University, Melbourne, VIC, Australia (A.M.T., S.Z.); Royal Prince Alfred Hospital, Sydney, NSW, Australia (D.R.S.); Hópital Bichat-Claude Bernard and Université Paris 7, Paris, France (M.M.); George Institute for Global Health, Sydney, NSW, Australia (B.N., N.R.P., S.Z., G.S.H., J.C., M.W.); University College London and National Institute for Health Research, University College London Hospitals, Biomedical Research Centre, London, UK (B.W.); Department of Cardiology, Royal Perth Hospital/University of Western Australia, Perth, WA, Australia (G.S.H.); George Institute for Global Health, University of Oxford, Oxford, UK (M.W.); and Department of Epidemiology, Johns Hopkins University, Baltimore, MD (M.W.)
| | - Gerard Wong
- From Baker IDI Heart and Diabetes Institute, Melbourne, VIC, Australia (Z.H.A., P.A.M., C.K.B., N.A.M., G.W., P.J.N., B.A.K., P.J.M.); King Fahad Medical City, Riyadh, Saudi Arabia (Z.H.A.); Department of Biochemistry and Molecular Biology, University of Melbourne, Melbourne, VIC, Australia (Z.H.A., M.J.M., P.J.M.); NHMRC Clinical Trials Centre, University of Sydney, Sydney, NSW, Australia (J.S., E.H.B.); School of Public Health and Preventive Medicine, Monash University, Melbourne, VIC, Australia (A.M.T., S.Z.); Royal Prince Alfred Hospital, Sydney, NSW, Australia (D.R.S.); Hópital Bichat-Claude Bernard and Université Paris 7, Paris, France (M.M.); George Institute for Global Health, Sydney, NSW, Australia (B.N., N.R.P., S.Z., G.S.H., J.C., M.W.); University College London and National Institute for Health Research, University College London Hospitals, Biomedical Research Centre, London, UK (B.W.); Department of Cardiology, Royal Perth Hospital/University of Western Australia, Perth, WA, Australia (G.S.H.); George Institute for Global Health, University of Oxford, Oxford, UK (M.W.); and Department of Epidemiology, Johns Hopkins University, Baltimore, MD (M.W.)
| | - Malcolm J McConville
- From Baker IDI Heart and Diabetes Institute, Melbourne, VIC, Australia (Z.H.A., P.A.M., C.K.B., N.A.M., G.W., P.J.N., B.A.K., P.J.M.); King Fahad Medical City, Riyadh, Saudi Arabia (Z.H.A.); Department of Biochemistry and Molecular Biology, University of Melbourne, Melbourne, VIC, Australia (Z.H.A., M.J.M., P.J.M.); NHMRC Clinical Trials Centre, University of Sydney, Sydney, NSW, Australia (J.S., E.H.B.); School of Public Health and Preventive Medicine, Monash University, Melbourne, VIC, Australia (A.M.T., S.Z.); Royal Prince Alfred Hospital, Sydney, NSW, Australia (D.R.S.); Hópital Bichat-Claude Bernard and Université Paris 7, Paris, France (M.M.); George Institute for Global Health, Sydney, NSW, Australia (B.N., N.R.P., S.Z., G.S.H., J.C., M.W.); University College London and National Institute for Health Research, University College London Hospitals, Biomedical Research Centre, London, UK (B.W.); Department of Cardiology, Royal Perth Hospital/University of Western Australia, Perth, WA, Australia (G.S.H.); George Institute for Global Health, University of Oxford, Oxford, UK (M.W.); and Department of Epidemiology, Johns Hopkins University, Baltimore, MD (M.W.)
| | - John Simes
- From Baker IDI Heart and Diabetes Institute, Melbourne, VIC, Australia (Z.H.A., P.A.M., C.K.B., N.A.M., G.W., P.J.N., B.A.K., P.J.M.); King Fahad Medical City, Riyadh, Saudi Arabia (Z.H.A.); Department of Biochemistry and Molecular Biology, University of Melbourne, Melbourne, VIC, Australia (Z.H.A., M.J.M., P.J.M.); NHMRC Clinical Trials Centre, University of Sydney, Sydney, NSW, Australia (J.S., E.H.B.); School of Public Health and Preventive Medicine, Monash University, Melbourne, VIC, Australia (A.M.T., S.Z.); Royal Prince Alfred Hospital, Sydney, NSW, Australia (D.R.S.); Hópital Bichat-Claude Bernard and Université Paris 7, Paris, France (M.M.); George Institute for Global Health, Sydney, NSW, Australia (B.N., N.R.P., S.Z., G.S.H., J.C., M.W.); University College London and National Institute for Health Research, University College London Hospitals, Biomedical Research Centre, London, UK (B.W.); Department of Cardiology, Royal Perth Hospital/University of Western Australia, Perth, WA, Australia (G.S.H.); George Institute for Global Health, University of Oxford, Oxford, UK (M.W.); and Department of Epidemiology, Johns Hopkins University, Baltimore, MD (M.W.)
| | - Andrew M Tonkin
- From Baker IDI Heart and Diabetes Institute, Melbourne, VIC, Australia (Z.H.A., P.A.M., C.K.B., N.A.M., G.W., P.J.N., B.A.K., P.J.M.); King Fahad Medical City, Riyadh, Saudi Arabia (Z.H.A.); Department of Biochemistry and Molecular Biology, University of Melbourne, Melbourne, VIC, Australia (Z.H.A., M.J.M., P.J.M.); NHMRC Clinical Trials Centre, University of Sydney, Sydney, NSW, Australia (J.S., E.H.B.); School of Public Health and Preventive Medicine, Monash University, Melbourne, VIC, Australia (A.M.T., S.Z.); Royal Prince Alfred Hospital, Sydney, NSW, Australia (D.R.S.); Hópital Bichat-Claude Bernard and Université Paris 7, Paris, France (M.M.); George Institute for Global Health, Sydney, NSW, Australia (B.N., N.R.P., S.Z., G.S.H., J.C., M.W.); University College London and National Institute for Health Research, University College London Hospitals, Biomedical Research Centre, London, UK (B.W.); Department of Cardiology, Royal Perth Hospital/University of Western Australia, Perth, WA, Australia (G.S.H.); George Institute for Global Health, University of Oxford, Oxford, UK (M.W.); and Department of Epidemiology, Johns Hopkins University, Baltimore, MD (M.W.)
| | - David R Sullivan
- From Baker IDI Heart and Diabetes Institute, Melbourne, VIC, Australia (Z.H.A., P.A.M., C.K.B., N.A.M., G.W., P.J.N., B.A.K., P.J.M.); King Fahad Medical City, Riyadh, Saudi Arabia (Z.H.A.); Department of Biochemistry and Molecular Biology, University of Melbourne, Melbourne, VIC, Australia (Z.H.A., M.J.M., P.J.M.); NHMRC Clinical Trials Centre, University of Sydney, Sydney, NSW, Australia (J.S., E.H.B.); School of Public Health and Preventive Medicine, Monash University, Melbourne, VIC, Australia (A.M.T., S.Z.); Royal Prince Alfred Hospital, Sydney, NSW, Australia (D.R.S.); Hópital Bichat-Claude Bernard and Université Paris 7, Paris, France (M.M.); George Institute for Global Health, Sydney, NSW, Australia (B.N., N.R.P., S.Z., G.S.H., J.C., M.W.); University College London and National Institute for Health Research, University College London Hospitals, Biomedical Research Centre, London, UK (B.W.); Department of Cardiology, Royal Perth Hospital/University of Western Australia, Perth, WA, Australia (G.S.H.); George Institute for Global Health, University of Oxford, Oxford, UK (M.W.); and Department of Epidemiology, Johns Hopkins University, Baltimore, MD (M.W.)
| | - Elizabeth H Barnes
- From Baker IDI Heart and Diabetes Institute, Melbourne, VIC, Australia (Z.H.A., P.A.M., C.K.B., N.A.M., G.W., P.J.N., B.A.K., P.J.M.); King Fahad Medical City, Riyadh, Saudi Arabia (Z.H.A.); Department of Biochemistry and Molecular Biology, University of Melbourne, Melbourne, VIC, Australia (Z.H.A., M.J.M., P.J.M.); NHMRC Clinical Trials Centre, University of Sydney, Sydney, NSW, Australia (J.S., E.H.B.); School of Public Health and Preventive Medicine, Monash University, Melbourne, VIC, Australia (A.M.T., S.Z.); Royal Prince Alfred Hospital, Sydney, NSW, Australia (D.R.S.); Hópital Bichat-Claude Bernard and Université Paris 7, Paris, France (M.M.); George Institute for Global Health, Sydney, NSW, Australia (B.N., N.R.P., S.Z., G.S.H., J.C., M.W.); University College London and National Institute for Health Research, University College London Hospitals, Biomedical Research Centre, London, UK (B.W.); Department of Cardiology, Royal Perth Hospital/University of Western Australia, Perth, WA, Australia (G.S.H.); George Institute for Global Health, University of Oxford, Oxford, UK (M.W.); and Department of Epidemiology, Johns Hopkins University, Baltimore, MD (M.W.)
| | - Paul J Nestel
- From Baker IDI Heart and Diabetes Institute, Melbourne, VIC, Australia (Z.H.A., P.A.M., C.K.B., N.A.M., G.W., P.J.N., B.A.K., P.J.M.); King Fahad Medical City, Riyadh, Saudi Arabia (Z.H.A.); Department of Biochemistry and Molecular Biology, University of Melbourne, Melbourne, VIC, Australia (Z.H.A., M.J.M., P.J.M.); NHMRC Clinical Trials Centre, University of Sydney, Sydney, NSW, Australia (J.S., E.H.B.); School of Public Health and Preventive Medicine, Monash University, Melbourne, VIC, Australia (A.M.T., S.Z.); Royal Prince Alfred Hospital, Sydney, NSW, Australia (D.R.S.); Hópital Bichat-Claude Bernard and Université Paris 7, Paris, France (M.M.); George Institute for Global Health, Sydney, NSW, Australia (B.N., N.R.P., S.Z., G.S.H., J.C., M.W.); University College London and National Institute for Health Research, University College London Hospitals, Biomedical Research Centre, London, UK (B.W.); Department of Cardiology, Royal Perth Hospital/University of Western Australia, Perth, WA, Australia (G.S.H.); George Institute for Global Health, University of Oxford, Oxford, UK (M.W.); and Department of Epidemiology, Johns Hopkins University, Baltimore, MD (M.W.)
| | - Bronwyn A Kingwell
- From Baker IDI Heart and Diabetes Institute, Melbourne, VIC, Australia (Z.H.A., P.A.M., C.K.B., N.A.M., G.W., P.J.N., B.A.K., P.J.M.); King Fahad Medical City, Riyadh, Saudi Arabia (Z.H.A.); Department of Biochemistry and Molecular Biology, University of Melbourne, Melbourne, VIC, Australia (Z.H.A., M.J.M., P.J.M.); NHMRC Clinical Trials Centre, University of Sydney, Sydney, NSW, Australia (J.S., E.H.B.); School of Public Health and Preventive Medicine, Monash University, Melbourne, VIC, Australia (A.M.T., S.Z.); Royal Prince Alfred Hospital, Sydney, NSW, Australia (D.R.S.); Hópital Bichat-Claude Bernard and Université Paris 7, Paris, France (M.M.); George Institute for Global Health, Sydney, NSW, Australia (B.N., N.R.P., S.Z., G.S.H., J.C., M.W.); University College London and National Institute for Health Research, University College London Hospitals, Biomedical Research Centre, London, UK (B.W.); Department of Cardiology, Royal Perth Hospital/University of Western Australia, Perth, WA, Australia (G.S.H.); George Institute for Global Health, University of Oxford, Oxford, UK (M.W.); and Department of Epidemiology, Johns Hopkins University, Baltimore, MD (M.W.)
| | - Michel Marre
- From Baker IDI Heart and Diabetes Institute, Melbourne, VIC, Australia (Z.H.A., P.A.M., C.K.B., N.A.M., G.W., P.J.N., B.A.K., P.J.M.); King Fahad Medical City, Riyadh, Saudi Arabia (Z.H.A.); Department of Biochemistry and Molecular Biology, University of Melbourne, Melbourne, VIC, Australia (Z.H.A., M.J.M., P.J.M.); NHMRC Clinical Trials Centre, University of Sydney, Sydney, NSW, Australia (J.S., E.H.B.); School of Public Health and Preventive Medicine, Monash University, Melbourne, VIC, Australia (A.M.T., S.Z.); Royal Prince Alfred Hospital, Sydney, NSW, Australia (D.R.S.); Hópital Bichat-Claude Bernard and Université Paris 7, Paris, France (M.M.); George Institute for Global Health, Sydney, NSW, Australia (B.N., N.R.P., S.Z., G.S.H., J.C., M.W.); University College London and National Institute for Health Research, University College London Hospitals, Biomedical Research Centre, London, UK (B.W.); Department of Cardiology, Royal Perth Hospital/University of Western Australia, Perth, WA, Australia (G.S.H.); George Institute for Global Health, University of Oxford, Oxford, UK (M.W.); and Department of Epidemiology, Johns Hopkins University, Baltimore, MD (M.W.)
| | - Bruce Neal
- From Baker IDI Heart and Diabetes Institute, Melbourne, VIC, Australia (Z.H.A., P.A.M., C.K.B., N.A.M., G.W., P.J.N., B.A.K., P.J.M.); King Fahad Medical City, Riyadh, Saudi Arabia (Z.H.A.); Department of Biochemistry and Molecular Biology, University of Melbourne, Melbourne, VIC, Australia (Z.H.A., M.J.M., P.J.M.); NHMRC Clinical Trials Centre, University of Sydney, Sydney, NSW, Australia (J.S., E.H.B.); School of Public Health and Preventive Medicine, Monash University, Melbourne, VIC, Australia (A.M.T., S.Z.); Royal Prince Alfred Hospital, Sydney, NSW, Australia (D.R.S.); Hópital Bichat-Claude Bernard and Université Paris 7, Paris, France (M.M.); George Institute for Global Health, Sydney, NSW, Australia (B.N., N.R.P., S.Z., G.S.H., J.C., M.W.); University College London and National Institute for Health Research, University College London Hospitals, Biomedical Research Centre, London, UK (B.W.); Department of Cardiology, Royal Perth Hospital/University of Western Australia, Perth, WA, Australia (G.S.H.); George Institute for Global Health, University of Oxford, Oxford, UK (M.W.); and Department of Epidemiology, Johns Hopkins University, Baltimore, MD (M.W.)
| | - Neil R Poulter
- From Baker IDI Heart and Diabetes Institute, Melbourne, VIC, Australia (Z.H.A., P.A.M., C.K.B., N.A.M., G.W., P.J.N., B.A.K., P.J.M.); King Fahad Medical City, Riyadh, Saudi Arabia (Z.H.A.); Department of Biochemistry and Molecular Biology, University of Melbourne, Melbourne, VIC, Australia (Z.H.A., M.J.M., P.J.M.); NHMRC Clinical Trials Centre, University of Sydney, Sydney, NSW, Australia (J.S., E.H.B.); School of Public Health and Preventive Medicine, Monash University, Melbourne, VIC, Australia (A.M.T., S.Z.); Royal Prince Alfred Hospital, Sydney, NSW, Australia (D.R.S.); Hópital Bichat-Claude Bernard and Université Paris 7, Paris, France (M.M.); George Institute for Global Health, Sydney, NSW, Australia (B.N., N.R.P., S.Z., G.S.H., J.C., M.W.); University College London and National Institute for Health Research, University College London Hospitals, Biomedical Research Centre, London, UK (B.W.); Department of Cardiology, Royal Perth Hospital/University of Western Australia, Perth, WA, Australia (G.S.H.); George Institute for Global Health, University of Oxford, Oxford, UK (M.W.); and Department of Epidemiology, Johns Hopkins University, Baltimore, MD (M.W.)
| | - Anthony Rodgers
- From Baker IDI Heart and Diabetes Institute, Melbourne, VIC, Australia (Z.H.A., P.A.M., C.K.B., N.A.M., G.W., P.J.N., B.A.K., P.J.M.); King Fahad Medical City, Riyadh, Saudi Arabia (Z.H.A.); Department of Biochemistry and Molecular Biology, University of Melbourne, Melbourne, VIC, Australia (Z.H.A., M.J.M., P.J.M.); NHMRC Clinical Trials Centre, University of Sydney, Sydney, NSW, Australia (J.S., E.H.B.); School of Public Health and Preventive Medicine, Monash University, Melbourne, VIC, Australia (A.M.T., S.Z.); Royal Prince Alfred Hospital, Sydney, NSW, Australia (D.R.S.); Hópital Bichat-Claude Bernard and Université Paris 7, Paris, France (M.M.); George Institute for Global Health, Sydney, NSW, Australia (B.N., N.R.P., S.Z., G.S.H., J.C., M.W.); University College London and National Institute for Health Research, University College London Hospitals, Biomedical Research Centre, London, UK (B.W.); Department of Cardiology, Royal Perth Hospital/University of Western Australia, Perth, WA, Australia (G.S.H.); George Institute for Global Health, University of Oxford, Oxford, UK (M.W.); and Department of Epidemiology, Johns Hopkins University, Baltimore, MD (M.W.)
| | - Bryan Williams
- From Baker IDI Heart and Diabetes Institute, Melbourne, VIC, Australia (Z.H.A., P.A.M., C.K.B., N.A.M., G.W., P.J.N., B.A.K., P.J.M.); King Fahad Medical City, Riyadh, Saudi Arabia (Z.H.A.); Department of Biochemistry and Molecular Biology, University of Melbourne, Melbourne, VIC, Australia (Z.H.A., M.J.M., P.J.M.); NHMRC Clinical Trials Centre, University of Sydney, Sydney, NSW, Australia (J.S., E.H.B.); School of Public Health and Preventive Medicine, Monash University, Melbourne, VIC, Australia (A.M.T., S.Z.); Royal Prince Alfred Hospital, Sydney, NSW, Australia (D.R.S.); Hópital Bichat-Claude Bernard and Université Paris 7, Paris, France (M.M.); George Institute for Global Health, Sydney, NSW, Australia (B.N., N.R.P., S.Z., G.S.H., J.C., M.W.); University College London and National Institute for Health Research, University College London Hospitals, Biomedical Research Centre, London, UK (B.W.); Department of Cardiology, Royal Perth Hospital/University of Western Australia, Perth, WA, Australia (G.S.H.); George Institute for Global Health, University of Oxford, Oxford, UK (M.W.); and Department of Epidemiology, Johns Hopkins University, Baltimore, MD (M.W.)
| | - Sophia Zoungas
- From Baker IDI Heart and Diabetes Institute, Melbourne, VIC, Australia (Z.H.A., P.A.M., C.K.B., N.A.M., G.W., P.J.N., B.A.K., P.J.M.); King Fahad Medical City, Riyadh, Saudi Arabia (Z.H.A.); Department of Biochemistry and Molecular Biology, University of Melbourne, Melbourne, VIC, Australia (Z.H.A., M.J.M., P.J.M.); NHMRC Clinical Trials Centre, University of Sydney, Sydney, NSW, Australia (J.S., E.H.B.); School of Public Health and Preventive Medicine, Monash University, Melbourne, VIC, Australia (A.M.T., S.Z.); Royal Prince Alfred Hospital, Sydney, NSW, Australia (D.R.S.); Hópital Bichat-Claude Bernard and Université Paris 7, Paris, France (M.M.); George Institute for Global Health, Sydney, NSW, Australia (B.N., N.R.P., S.Z., G.S.H., J.C., M.W.); University College London and National Institute for Health Research, University College London Hospitals, Biomedical Research Centre, London, UK (B.W.); Department of Cardiology, Royal Perth Hospital/University of Western Australia, Perth, WA, Australia (G.S.H.); George Institute for Global Health, University of Oxford, Oxford, UK (M.W.); and Department of Epidemiology, Johns Hopkins University, Baltimore, MD (M.W.)
| | - Graham S Hillis
- From Baker IDI Heart and Diabetes Institute, Melbourne, VIC, Australia (Z.H.A., P.A.M., C.K.B., N.A.M., G.W., P.J.N., B.A.K., P.J.M.); King Fahad Medical City, Riyadh, Saudi Arabia (Z.H.A.); Department of Biochemistry and Molecular Biology, University of Melbourne, Melbourne, VIC, Australia (Z.H.A., M.J.M., P.J.M.); NHMRC Clinical Trials Centre, University of Sydney, Sydney, NSW, Australia (J.S., E.H.B.); School of Public Health and Preventive Medicine, Monash University, Melbourne, VIC, Australia (A.M.T., S.Z.); Royal Prince Alfred Hospital, Sydney, NSW, Australia (D.R.S.); Hópital Bichat-Claude Bernard and Université Paris 7, Paris, France (M.M.); George Institute for Global Health, Sydney, NSW, Australia (B.N., N.R.P., S.Z., G.S.H., J.C., M.W.); University College London and National Institute for Health Research, University College London Hospitals, Biomedical Research Centre, London, UK (B.W.); Department of Cardiology, Royal Perth Hospital/University of Western Australia, Perth, WA, Australia (G.S.H.); George Institute for Global Health, University of Oxford, Oxford, UK (M.W.); and Department of Epidemiology, Johns Hopkins University, Baltimore, MD (M.W.)
| | - John Chalmers
- From Baker IDI Heart and Diabetes Institute, Melbourne, VIC, Australia (Z.H.A., P.A.M., C.K.B., N.A.M., G.W., P.J.N., B.A.K., P.J.M.); King Fahad Medical City, Riyadh, Saudi Arabia (Z.H.A.); Department of Biochemistry and Molecular Biology, University of Melbourne, Melbourne, VIC, Australia (Z.H.A., M.J.M., P.J.M.); NHMRC Clinical Trials Centre, University of Sydney, Sydney, NSW, Australia (J.S., E.H.B.); School of Public Health and Preventive Medicine, Monash University, Melbourne, VIC, Australia (A.M.T., S.Z.); Royal Prince Alfred Hospital, Sydney, NSW, Australia (D.R.S.); Hópital Bichat-Claude Bernard and Université Paris 7, Paris, France (M.M.); George Institute for Global Health, Sydney, NSW, Australia (B.N., N.R.P., S.Z., G.S.H., J.C., M.W.); University College London and National Institute for Health Research, University College London Hospitals, Biomedical Research Centre, London, UK (B.W.); Department of Cardiology, Royal Perth Hospital/University of Western Australia, Perth, WA, Australia (G.S.H.); George Institute for Global Health, University of Oxford, Oxford, UK (M.W.); and Department of Epidemiology, Johns Hopkins University, Baltimore, MD (M.W.)
| | - Mark Woodward
- From Baker IDI Heart and Diabetes Institute, Melbourne, VIC, Australia (Z.H.A., P.A.M., C.K.B., N.A.M., G.W., P.J.N., B.A.K., P.J.M.); King Fahad Medical City, Riyadh, Saudi Arabia (Z.H.A.); Department of Biochemistry and Molecular Biology, University of Melbourne, Melbourne, VIC, Australia (Z.H.A., M.J.M., P.J.M.); NHMRC Clinical Trials Centre, University of Sydney, Sydney, NSW, Australia (J.S., E.H.B.); School of Public Health and Preventive Medicine, Monash University, Melbourne, VIC, Australia (A.M.T., S.Z.); Royal Prince Alfred Hospital, Sydney, NSW, Australia (D.R.S.); Hópital Bichat-Claude Bernard and Université Paris 7, Paris, France (M.M.); George Institute for Global Health, Sydney, NSW, Australia (B.N., N.R.P., S.Z., G.S.H., J.C., M.W.); University College London and National Institute for Health Research, University College London Hospitals, Biomedical Research Centre, London, UK (B.W.); Department of Cardiology, Royal Perth Hospital/University of Western Australia, Perth, WA, Australia (G.S.H.); George Institute for Global Health, University of Oxford, Oxford, UK (M.W.); and Department of Epidemiology, Johns Hopkins University, Baltimore, MD (M.W.)
| | - Peter J Meikle
- From Baker IDI Heart and Diabetes Institute, Melbourne, VIC, Australia (Z.H.A., P.A.M., C.K.B., N.A.M., G.W., P.J.N., B.A.K., P.J.M.); King Fahad Medical City, Riyadh, Saudi Arabia (Z.H.A.); Department of Biochemistry and Molecular Biology, University of Melbourne, Melbourne, VIC, Australia (Z.H.A., M.J.M., P.J.M.); NHMRC Clinical Trials Centre, University of Sydney, Sydney, NSW, Australia (J.S., E.H.B.); School of Public Health and Preventive Medicine, Monash University, Melbourne, VIC, Australia (A.M.T., S.Z.); Royal Prince Alfred Hospital, Sydney, NSW, Australia (D.R.S.); Hópital Bichat-Claude Bernard and Université Paris 7, Paris, France (M.M.); George Institute for Global Health, Sydney, NSW, Australia (B.N., N.R.P., S.Z., G.S.H., J.C., M.W.); University College London and National Institute for Health Research, University College London Hospitals, Biomedical Research Centre, London, UK (B.W.); Department of Cardiology, Royal Perth Hospital/University of Western Australia, Perth, WA, Australia (G.S.H.); George Institute for Global Health, University of Oxford, Oxford, UK (M.W.); and Department of Epidemiology, Johns Hopkins University, Baltimore, MD (M.W.).
| |
Collapse
|
40
|
Liu L, Hu Q, Wu H, Xue Y, Cai L, Fang M, Liu Z, Yao P, Wu Y, Gong Z. Protective role of n6/n3 PUFA supplementation with varying DHA/EPA ratios against atherosclerosis in mice. J Nutr Biochem 2016; 32:171-80. [DOI: 10.1016/j.jnutbio.2016.02.010] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2015] [Revised: 01/16/2016] [Accepted: 02/08/2016] [Indexed: 10/22/2022]
|
41
|
Abstract
PURPOSE OF REVIEW A major step in energy metabolism is hydrolysis of triacylglycerol-rich lipoproteins (TRLs) to release fatty acids that can be used or stored. This is accomplished by lipoprotein lipase (LPL) at 'binding lipolysis sites' at the vascular endothelium. A multitude of interactions are involved in this seemingly simple reaction. Recent advances in the understanding of some of these factors will be discussed in an attempt to build a comprehensive picture. RECENT FINDINGS The first event in catabolism of TRLs is that they dock at the vascular endothelium. This requires LPL and GPIHBP1, the endothelial transporter of LPL.Kinetic studies in rats with labeled chylomicrons showed that once a chylomicron has docked in the heart it stays for minutes and a large number of triacylglycerol molecules are split. The distribution of binding between tissues reflects the amount of LPL, as evident from studies with mutant mice.Clearance of TRLs is often slowed down in metabolic disease, as was demonstrated both in mice and men. In mice, this was directly connected to decreased amounts of endothelial LPL. SUMMARY The LPL system is central in energy metabolism and results from interplay between several factors. Rapid and exciting progress is being made.
Collapse
Affiliation(s)
- Gunilla Olivecrona
- Department of Medical Biosciences/Physiological Chemistry, Umeå University, Umeå, Sweden
| |
Collapse
|
42
|
Hernández-Angeles C, Castelo-Branco C. Cardiovascular risk in climacteric women: focus on diet. Climacteric 2016; 19:215-21. [DOI: 10.3109/13697137.2016.1173025] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023]
|
43
|
Moreno-Perez S, Luna P, Señorans F, Guisan J, Fernandez-Lorente G. Enzymatic synthesis of triacylglycerols of docosahexaenoic acid: Transesterification of its ethyl esters with glycerol. Food Chem 2015; 187:225-9. [DOI: 10.1016/j.foodchem.2015.04.095] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2014] [Revised: 03/10/2015] [Accepted: 04/22/2015] [Indexed: 11/27/2022]
|
44
|
Torres N, Guevara-Cruz M, Velázquez-Villegas LA, Tovar AR. Nutrition and Atherosclerosis. Arch Med Res 2015; 46:408-26. [DOI: 10.1016/j.arcmed.2015.05.010] [Citation(s) in RCA: 151] [Impact Index Per Article: 15.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2015] [Accepted: 05/12/2015] [Indexed: 12/15/2022]
|
45
|
Altered concentrate to forage ratio in cows ration enhanced bioproduction of specific size subpopulation of milk fat globules. Food Chem 2015; 179:199-205. [DOI: 10.1016/j.foodchem.2015.01.138] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2014] [Revised: 01/18/2015] [Accepted: 01/31/2015] [Indexed: 01/22/2023]
|
46
|
Liu F, Li Z, Lv X, Ma J. Dietary n-3 polyunsaturated fatty acid intakes modify the effect of genetic variation in fatty acid desaturase 1 on coronary artery disease. PLoS One 2015; 10:e0121255. [PMID: 25849351 PMCID: PMC4388373 DOI: 10.1371/journal.pone.0121255] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2014] [Accepted: 01/29/2015] [Indexed: 01/07/2023] Open
Abstract
BACKGROUND Previous studies suggested that dietary fatty acids could affect blood lipids by interacting with genetic variations in fatty acid desaturase 1 (FADS1). However, little is known about their direct effects on coronary artery disease (CAD). The aim of this study was to evaluate whether dietary n-3 long-chain polyunsaturated fatty acids (LCPUFAs)-eicosapentaenoic acid (EPA) and docosahexaenoic acid (DHA) could modulate the effect of FADS1 rs174547 polymorphism on CAD. METHODS FADS1 single-nucleotide polymorphisms rs174547 genotypes were measured in 440 CAD patients and 838 healthy controls. Dietary EPA and DHA intakes were assessed with a validated quantitative frequency food questionnaire. The association between FADS1 rs174547 and CAD was estimated using logistic regression under both dominant and additive genetic models. The interactions between rs174547 polymorphism and LCPUFAs were analyzed by using multiple logistic regression and the "genotype × n-3 LCPUFAs" interaction term was included into the model. RESULTS We found that the minor T allele of FADS1 rs174547 increased CAD risk (OR = 1.36, 95%CIs 1.03-1.80), and observed significant interaction between rs174547 and dietary EPA intakes on CAD (P-interaction = 0.028). The T-allele was only associated with higher CAD risk among individuals with lower dietary EPA intakes, but not in those with higher EPA intakes. Similarly, significant interaction was also observed between rs174547 and dietary DHA intakes on CAD (P-interaction = 0.020). CONCLUSIONS Dietary n-3 LCPUFA intakes could modulate the association between FADS1 rs174547 polymorphism and CAD. High dietary n-3 LCPUFA intakes could negate the unfavorable effect of genetic variation in FADS1 on CAD in middle-aged and elderly Chinese population.
Collapse
Affiliation(s)
- Fengqiong Liu
- Department of Epidemiology and health statistics, School of Public Health, Fujian Medical University, Fuzhou, Fujian, China
| | - Zhongxia Li
- Department of Nutrition, School of Public Health, Sun Yat-Sen University, Guangzhou, Guangdong, China
| | - Xiaofei Lv
- Department of Nutrition, School of Public Health, Sun Yat-Sen University, Guangzhou, Guangdong, China
| | - Jing Ma
- Department of Nutrition, School of Public Health, Sun Yat-Sen University, Guangzhou, Guangdong, China
| |
Collapse
|
47
|
Zirpoli H, Abdillahi M, Quadri N, Ananthakrishnan R, Wang L, Rosario R, Zhu Z, Deckelbaum RJ, Ramasamy R. Acute administration of n-3 rich triglyceride emulsions provides cardioprotection in murine models after ischemia-reperfusion. PLoS One 2015; 10:e0116274. [PMID: 25559887 PMCID: PMC4283969 DOI: 10.1371/journal.pone.0116274] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2014] [Accepted: 12/08/2014] [Indexed: 11/19/2022] Open
Abstract
Dietary n-3 fatty acids (FAs) may reduce cardiovascular disease risk. We questioned whether acute administration of n-3 rich triglyceride (TG) emulsions could preserve cardiac function and decrease injury after ischemia/reperfusion (I/R) insult. We used two different experimental models: in vivo, C57BL/6 mice were exposed to acute occlusion of the left anterior descending coronary artery (LAD), and ex-vivo, C57BL/6 murine hearts were perfused using Langendorff technique (LT). In the LAD model, mice treated with n-3 TG emulsion (1.5g/kg body weight), immediately after ischemia and 1h later during reperfusion, significantly reduced infarct size and maintained cardiac function (p<0.05). In the LT model, administration of n-3 TG emulsion (300mgTG/100ml) during reperfusion significantly improved functional recovery (p<0.05). In both models, lactate dehydrogenase (LDH) levels, as a marker of injury, were significantly reduced by n-3 TG emulsion. To investigate the mechanisms by which n-3 FAs protects hearts from I/R injury, we investigated changes in key pathways linked to cardioprotection. In the ex-vivo model, we showed that n-3 FAs increased phosphorylation of AKT and GSK3β proteins (p<0.05). Acute n-3 TG emulsion treatment also increased Bcl-2 protein level and reduced an autophagy marker, Beclin-1 (p<0.05). Additionally, cardioprotection by n-3 TG emulsion was linked to changes in PPARγ protein expression (p<0.05). Rosiglitazone and p-AKT inhibitor counteracted the positive effect of n-3 TG; GSK3β inhibitor plus n-3 TG significantly inhibited LDH release. We conclude that acute n-3 TG injection during reperfusion provides cardioprotection. This may prove to be a novel acute adjunctive reperfusion therapy after treating patients with myocardial infarction.
Collapse
Affiliation(s)
- Hylde Zirpoli
- Institute of Human Nutrition, College of Physicians and Surgeons, Columbia University, New York, New York, United States of America
- Department of Pediatrics, College of Physicians and Surgeons, Columbia University, New York, New York, United States of America
| | - Mariane Abdillahi
- Institute of Human Nutrition, College of Physicians and Surgeons, Columbia University, New York, New York, United States of America
- Department of Medicine, New York University School of Medicine, New York, New York, United States of America
| | - Nosirudeen Quadri
- Department of Medicine, New York University School of Medicine, New York, New York, United States of America
| | - Radha Ananthakrishnan
- Department of Medicine, New York University School of Medicine, New York, New York, United States of America
| | - Lingjie Wang
- Department of Medicine, New York University School of Medicine, New York, New York, United States of America
| | - Rosa Rosario
- Department of Medicine, New York University School of Medicine, New York, New York, United States of America
| | - Zhengbin Zhu
- Department of Medicine, New York University School of Medicine, New York, New York, United States of America
| | - Richard J. Deckelbaum
- Institute of Human Nutrition, College of Physicians and Surgeons, Columbia University, New York, New York, United States of America
- Department of Pediatrics, College of Physicians and Surgeons, Columbia University, New York, New York, United States of America
- * E-mail: (R. Ramasamy); (RJD)
| | - Ravichandran Ramasamy
- Department of Medicine, New York University School of Medicine, New York, New York, United States of America
- * E-mail: (R. Ramasamy); (RJD)
| |
Collapse
|
48
|
Li J, Li FR, Wei D, Jia W, Kang JX, Stefanovic-Racic M, Dai Y, Zhao AZ. Endogenous ω-3 polyunsaturated fatty acid production confers resistance to obesity, dyslipidemia, and diabetes in mice. Mol Endocrinol 2014; 28:1316-28. [PMID: 24978197 DOI: 10.1210/me.2014-1011] [Citation(s) in RCA: 47] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023] Open
Abstract
Despite the well-documented health benefits of ω-3 polyunsaturated fatty acids (PUFAs), their use in clinical management of hyperglycemia and obesity has shown little success. To better define the mechanisms of ω-3 PUFAs in regulating energy balance and insulin sensitivity, we deployed a transgenic mouse model capable of endogenously producing ω-3 PUFAs while reducing ω-6 PUFAs owing to the expression of a Caenorhabditis elegans fat-1 gene encoding an ω-3 fatty acid desaturase. When challenged with high-fat diets, fat-1 mice strongly resisted obesity, diabetes, hypercholesterolemia, and hepatic steatosis. Endogenous elevation of ω-3 PUFAs and reduction of ω-6 PUFAs did not alter the amount of food intake but led to increased energy expenditure in the fat-1 mice. The requirements for the levels of ω-3 PUFAs as well as the ω-6/ω-3 ratios in controlling blood glucose and obesity are much more stringent than those in lipid metabolism. These metabolic phenotypes were accompanied by attenuation of the inflammatory state because tissue levels of prostaglandin E2, leukotriene B4, monocyte chemoattractant protein-1, and TNF-α were significantly decreased. TNF-α-induced nuclear factor-κB signaling was almost completely abolished. Consistent with the reduction in chronic inflammation and a significant increase in peroxisome proliferator-activated receptor-γ activity in the fat-1 liver tissue, hepatic insulin signaling was sharply elevated. The activities of prolipogenic regulators, such as liver X receptor, stearoyl-CoA desaturase-1, and sterol regulatory element binding protein-1 were sharply decreased, whereas the activity of peroxisome proliferator-activated receptor-α, a nuclear receptor that facilitates lipid β-oxidation, was markedly increased. Thus, endogenous conversion of ω-6 to ω-3 PUFAs via fat-1 strongly protects against obesity, diabetes, inflammation, and dyslipidemia and may represent a novel therapeutic modality to treat these prevalent disorders.
Collapse
Affiliation(s)
- Jie Li
- Department of Cell Biology and Physiology (J.L.) and Division of Endocrinology and Metabolism (M.S.-R.), University of Pittsburgh, Pittsburgh, Pennsylvania 15261; Department of Pathology (F.R.L), The First Affiliated Hospital of Nanjing Medical University, Nanjing 210029, China; Department of Endocrinology (D.W.), The Second People's Hospital of Chengdu, Chengdu 610017, China; Surgery (W.J.), West China Hospital, Sichuan University, Chengdu 610041, China; Department of Medicine (J.X.K.), Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts 02114; and State Key Laboratory of Reproductive Medicine (F.R.L., Y.D., A.Z.Z.), The Center of Metabolic Disease Research, Nanjing Medical University, Nanjing, Jiangsu Province 210029, China
| | | | | | | | | | | | | | | |
Collapse
|
49
|
Le Goff W. A new piece in the puzzling effect of n-3 fatty acids on atherosclerosis? Atherosclerosis 2014; 235:358-62. [PMID: 24926537 DOI: 10.1016/j.atherosclerosis.2014.03.038] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/14/2014] [Accepted: 03/14/2014] [Indexed: 01/05/2023]
Abstract
Omega-3 fatty acids (n-3) FA are reported to be protective against cardiovascular disease (CVD), notably through their beneficial action on atherosclerosis development. In this context dietary intake of long-chain marine eicosapentaenoic acid (EPA) and docosahexaenoic acid (DHA) is recommended and randomised trials largely support that EPA and DHA intake is associated with a reduction of CVD. However, mechanisms governing the atheroprotective action of n-3 FA are still unclear and numerous studies using mouse models conducted so far do not allow to reach a precise view of the cellular and molecular effects of n-3 FA on atherosclerosis. In the current issue of Atherosclerosis, Chang et al. provide important new information on the anti-atherogenic properties of n-3 FA by analysing the incremental replacement of saturated FA by pure fish oil as a source of EPA and DHA in Ldlr(-/-) mice fed a high fat/high cholesterol diet.
Collapse
Affiliation(s)
- Wilfried Le Goff
- INSERM UMR_S1166, Team 4: Integrative Biology of Atherosclerosis, F-75013 Paris, France; Université Pierre et Marie Curie-Paris6, F-75005 Paris, France; Institute of Cardiometabolism and Nutrition (ICAN), Pitié-Salpêtrière Hospital, F-75013 Paris, France.
| |
Collapse
|
50
|
Gelsomino G, Corsetto PA, Campia I, Montorfano G, Kopecka J, Castella B, Gazzano E, Ghigo D, Rizzo AM, Riganti C. Omega 3 fatty acids chemosensitize multidrug resistant colon cancer cells by down-regulating cholesterol synthesis and altering detergent resistant membranes composition. Mol Cancer 2013; 12:137. [PMID: 24225025 PMCID: PMC4225767 DOI: 10.1186/1476-4598-12-137] [Citation(s) in RCA: 73] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2013] [Accepted: 11/11/2013] [Indexed: 01/17/2023] Open
Abstract
Background The activity of P-glycoprotein (Pgp) and multidrug resistance related protein 1 (MRP1), two membrane transporters involved in multidrug resistance of colon cancer, is increased by high amounts of cholesterol in plasma membrane and detergent resistant membranes (DRMs). It has never been investigated whether omega 3 polyunsatured fatty acids (PUFAs), which modulate cholesterol homeostasis in dyslipidemic syndromes and have chemopreventive effects in colon cancer, may affect the response to chemotherapy in multidrug resistant (MDR) tumors. Methods We studied the effect of omega 3 PUFAs docosahexaenoic acid (DHA) and eicosapentaenoic acid (EPA) in human chemosensitive colon cancer HT29 cells and in their MDR counterpart, HT29-dx cells. Results MDR cells, which overexpressed Pgp and MRP1, had a dysregulated cholesterol metabolism, due to the lower expression of ubiquitin E3 ligase Trc8: this produced lower ubiquitination rate of 3-hydroxy-3-methylglutaryl-coenzyme A reductase (HMGCoAR), higher cholesterol synthesis, higher cholesterol content in MDR cells. We found that DHA and EPA re-activated Trc8 E3 ligase in MDR cells, restored the ubiquitination rate of HMGCoAR to levels comparable with chemosensitive cells, reduced the cholesterol synthesis and incorporation in DRMs. Omega 3 PUFAs were incorporated in whole lipids as well as in DRMs of MDR cells, and altered the lipid composition of these compartments. They reduced the amount of Pgp and MRP1 contained in DRMs, decreased the transporters activity, restored the antitumor effects of different chemotherapeutic drugs, restored a proper tumor-immune system recognition in response to chemotherapy in MDR cells. Conclusions Our work describes a new biochemical effect of omega 3 PUFAs, which can be useful to overcome chemoresistance in MDR colon cancer cells.
Collapse
Affiliation(s)
- Giada Gelsomino
- Department of Oncology, University of Torino, via Santena 5/bis, 10126 Torino, Italy.
| | | | | | | | | | | | | | | | | | | |
Collapse
|