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
|
Bhat S, Sarkar S, Zaffar D, Dandona P, Kalyani RR. Omega-3 Fatty Acids in Cardiovascular Disease and Diabetes: a Review of Recent Evidence. Curr Cardiol Rep 2023; 25:51-65. [PMID: 36729217 DOI: 10.1007/s11886-022-01831-0] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 11/08/2022] [Indexed: 02/03/2023]
Abstract
PURPOSE OF REVIEW Omega-3 fatty acids (n-3 FA) lower triglycerides, have anti-inflammatory properties, and improve metabolism. Clinical evidence of cardiovascular benefit with omega-3 fatty acids is mixed. We discuss mechanisms providing biological plausibility of benefit of omega-3 fatty acids in cardiovascular risk reduction and review clinical trials investigating the benefits of prescription omega-3 fatty acids in dyslipidemia, atherosclerotic cardiovascular disease (ASCVD), and diabetes. RECENT FINDINGS Although early trials showed no benefit of omega-3 fatty acids in ASCVD, the REDUCE-IT trial noted significant risk reduction in ASCVD events with highly purified EPA (icosapent ethyl) use which has changed the landscape for currently available therapeutic options. However, other large trials like STRENGTH and VITAL, which used different formulations of prescription omega-3 fatty acids, did not note significant cardiovascular risk reduction. Thus the effectiveness of omega-3 fatty acids for cardiovascular disease prevention is an ongoing topic of debate. A relative paucity of studies examining benefits for glycemic outcomes in persons with diabetes exists; however, few studies have suggested lack of benefit to date. Significant residual cardiovascular risk exists for individuals with hypertriglyceridemia. Prescription omega-3 fatty acids are more commonly used for CV risk reduction in these patients. Clinical guideline statements now recommend icosapent ethyl use for selected individuals with hypertriglyceridemia to reduce cardiovascular events given recent evidence from the REDUCE-IT trial. Nonetheless, data from other large scale trials has been mixed, and future research is needed to better understand how different preparations of omega-3 may differ in their cardiovascular and metabolic effects, and the mechanisms for their benefit.
Collapse
Affiliation(s)
- Salman Bhat
- Division of Endocrinology, Diabetes and Metabolism, Department of Medicine, The Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Sudipa Sarkar
- Division of Endocrinology, Diabetes and Metabolism, Department of Medicine, The Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Duha Zaffar
- Department of Internal Medicine, University of Maryland Midtown Campus, Baltimore, MD, USA
| | - Paresh Dandona
- Division of Endocrinology, Diabetes and Metabolism, University at Buffalo, Buffalo, NY, USA
| | - Rita R Kalyani
- Division of Endocrinology, Diabetes and Metabolism, Department of Medicine, The Johns Hopkins University School of Medicine, Baltimore, MD, USA.
| |
Collapse
|
3
|
Đidara M, Poljičak-Milas N, Novoselec J, Pavlić M, Đud D, Šperanda M. Effect of dietary supplementation with linseed and organic selenium on plasma metabolic parameters in Holstein dairy cows. JOURNAL OF APPLIED ANIMAL RESEARCH 2022. [DOI: 10.1080/09712119.2022.2110500] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/15/2022]
Affiliation(s)
- Mislav Đidara
- Department of Animal Production and Biotechnology, Faculty of Agrobiotechnical Sciences Osijek, Josip Juraj Strossmayer University of Osijek Osijek, Croatia
| | - Nina Poljičak-Milas
- Department of Pathophysiology, Faculty of Veterinary medicine, University of Zagreb, Zagreb, Croatia
| | - Josip Novoselec
- Department of Animal Production and Biotechnology, Faculty of Agrobiotechnical Sciences Osijek, Josip Juraj Strossmayer University of Osijek Osijek, Croatia
| | - Martina Pavlić
- Croatian Agency for Agriculture and Food, Osijek, Croatia
| | | | - Marcela Šperanda
- Department of Animal Production and Biotechnology, Faculty of Agrobiotechnical Sciences Osijek, Josip Juraj Strossmayer University of Osijek Osijek, Croatia
| |
Collapse
|
4
|
Heras A, Gomi R, Young M, Chang CL, Wasserman E, Sharma A, Wu W, Gu J, Balaji U, White R, Permaul P, Janahi I, Worgall TS, Worgall S. Dietary long-chain omega 3 fatty acids modify sphingolipid metabolism to facilitate airway hyperreactivity. Sci Rep 2022; 12:19735. [PMID: 36396956 PMCID: PMC9672127 DOI: 10.1038/s41598-022-21083-w] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2022] [Accepted: 09/22/2022] [Indexed: 11/19/2022] Open
Abstract
Omega-3 polyunsaturated fatty acids (n-3 PUFAs) are essential nutrients that can affect inflammatory responses. While n-3 PUFAs are generally considered beneficial for cardiovascular disease and obesity, the effects on asthma, the most common inflammatory lung disease are unclear. While prenatal dietary n-3 PUFAs decrease the risk for childhood wheezing, postnatal dietary n-3 PUFAs can worsen allergic airway inflammation. Sphingolipid metabolism is also affected by dietary n-3 PUFAs. Decreased sphingolipid synthesis leads to airway hyperreactivity, besides inflammation, a cardinal feature of asthma, and common genetic asthma risk alleles lead to lower sphingolipid synthesis. We investigated the effect of dietary n-3 PUFAs on sphingolipid metabolism and airway reactivity. Comparing a fish-oil diet with a high n-3 PUFA content (FO) to an isocaloric coconut oil-enriched diet (CO), we found an n-3 PUFA-dependent effect on increased airway reactivity, that was not accompanied by inflammation. Lung and whole blood content of dihydroceramides, ceramides, sphingomyelins, and glucosylceramides were lower in mice fed the n-3 PUFA enriched diet consistent with lower sphingolipid synthesis. In contrast, phosphorylated long chain bases such as sphingosine 1-phosphate were increased. These findings suggest that dietary n-3 PUFAs affect pulmonary sphingolipid composition to favor innate airway hyperreactivity, independent of inflammation, and point to an important role of n-3 PUFAs in sphingolipid metabolism.
Collapse
Affiliation(s)
- Andrea Heras
- Department of Pediatrics, Weill Cornell Medicine, 413 East 69th Street, Room 1200, New York, NY, 10021, USA
| | - Rika Gomi
- Department of Pediatrics, Weill Cornell Medicine, 413 East 69th Street, Room 1200, New York, NY, 10021, USA
| | - Madeline Young
- Department of Pediatrics, Weill Cornell Medicine, 413 East 69th Street, Room 1200, New York, NY, 10021, USA
| | - Chuchun L Chang
- Institute of Human Nutrition/Department of Pediatrics, Columbia University Medical Center, New York, NY, USA
| | - Emily Wasserman
- Department of Pediatrics, Weill Cornell Medicine, 413 East 69th Street, Room 1200, New York, NY, 10021, USA
- Weill Cornell Medicine, Drukier Institute for Children's Health, New York, USA
| | - Anurag Sharma
- Department of Pediatrics, Weill Cornell Medicine, 413 East 69th Street, Room 1200, New York, NY, 10021, USA
| | - Wenzhu Wu
- Department of Pediatrics, Weill Cornell Medicine, 413 East 69th Street, Room 1200, New York, NY, 10021, USA
| | - Jinghua Gu
- Weill Cornell Medicine, Drukier Institute for Children's Health, New York, USA
| | - Uthra Balaji
- Weill Cornell Medicine, Drukier Institute for Children's Health, New York, USA
| | - Rachel White
- Department of Pediatrics, Weill Cornell Medicine, 413 East 69th Street, Room 1200, New York, NY, 10021, USA
| | - Perdita Permaul
- Department of Pediatrics, Weill Cornell Medicine, 413 East 69th Street, Room 1200, New York, NY, 10021, USA
| | | | - Tilla S Worgall
- Department of Pathology and Cell Biology, Columbia University Medical Center, New York, USA
| | - Stefan Worgall
- Department of Pediatrics, Weill Cornell Medicine, 413 East 69th Street, Room 1200, New York, NY, 10021, USA.
- Weill Cornell Medicine, Drukier Institute for Children's Health, New York, USA.
- Department of Genetic Medicine, Weill Cornell Medicine, New York, USA.
| |
Collapse
|
5
|
Shi H, Zhou X, He X, Wang R, Zhou W. Camellia oil Enhances Plasma Antioxidant Metabolism and Improves Plasma Lipid Metabolism in High-fat Diet-fed Rats. Nat Prod Commun 2022. [DOI: 10.1177/1934578x221081368] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Abstract
Living on a high-fat, high-calorie, and high-protein diet for a long period may compromise human immunity due to the long-term accumulation of free radicals and plasma lipids. The antioxidant and lipid-lowering compounds (ie polyphenols and vitamin E) in Camellia oil help to decrease the risk of numerous ailments, including cardiovascular disease (CVD), and obesity. The aims of this study were to study the hypolipidemic and antioxidant effects of Camellia oil in high-fat-fed rats and to promote the high-value use of camellia resources. The high-fat-fed rats were administrated with 2.5, 7.5, and 15 mL/kg BW Camellia oil (Camellia oil group), and 10 mg/kg BW atorvastatin (atorvastatin group), respectively, and compared with a model group (only fed with high fat) and a control group (fed with basal diet). Several parameters were measured, including (1) body weight (BW), liver-to-BW ratio; (2) plasma total cholesterol (TC), triglyceride (TG), high-density lipoprotein cholesterol (HDL-C), and low-density lipoprotein cholesterol (LDL-C); and (3) alanine aminotransferase (ALT), alanine aminotransferase (AST), superoxide dismutase (SOD), glutathione peroxidase (GSH-Px) activity, model driven architecture (MDA) content, lipid metabolism-related genes, and antioxidant-related genes in liver tissue. Compared with the model group, the high-fat-fed rats in the Camellia oil and atorvastatin group had significantly lower BW and liver-to-BW ratio ( P < .01), plasma TC, TG, and LDL-C levels and ALT and AST activities, but higher HDL-C levels. The relative expressions of ACAT1, DGAT2, FAS, and SREBP genes were significantly reduced in the Camellia oil and atorvastatin groups, while the relative expressions of LCAT, UCP2, MCD, and CPT-1 genes were significantly increased. The rats in the Camellia oil group showed significantly higher SOD and GSH-Px activities, significantly lower MDA content, and significantly higher relative expression of antioxidant genes (eg SOD1, GPx1, CAT, and Gclm). Thus, atorvastatin and Camellia oil exhibited significant hypolipidemic and antioxidant effects, which were better at a dose of 7.5 mL/kg (BW) of Camellia oil. Therefore, Camellia oil becomes a potential new natural resource for future research and development of antioxidant and hypolipidemic drugs, nutraceuticals, and additives.
Collapse
Affiliation(s)
- Hao Shi
- Central South University of Forestry and Technology, Changsha, People’s Republic of China
- Hunan Applied technology University, Changde, People’s Republic of China
| | | | - Xiaoe He
- Hunan Applied technology University, Changde, People’s Republic of China
| | - Rencai Wang
- Hunan Applied technology University, Changde, People’s Republic of China
- Hunan Agricultural University, Changsha, People’s Republic of China
| | - Wenhua Zhou
- Central South University of Forestry and Technology, Changsha, People’s Republic of China
- Hunan Applied technology University, Changde, People’s Republic of China
| |
Collapse
|
6
|
Bandara EMS, Edirisinghe DIU, Wanniarachchi DDCDS, Peiris H, Perera PPR, Jayakrishan AG, Waikar HD, Sharma SK, Abeysuriya V, Chandrasena LG. A comparative study on fatty acid profile in selected vessels of coronary artery bypass graft (CABG). PLoS One 2022; 17:e0260780. [PMID: 35061685 PMCID: PMC8782383 DOI: 10.1371/journal.pone.0260780] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2021] [Accepted: 10/22/2021] [Indexed: 11/19/2022] Open
Abstract
Atherosclerosis is one of the leading non-communicable diseases in Sri Lanka. Analysis of fatty acid composition in blood vessels is important in understanding the development of atherosclerosis. Here, analyses of fatty acid profiles in major arteries which are commonly used in Coronary Artery Bypass Graft surgery (CABG) were subjected to investigation. Patients (n = 27) undergoing elective CABG were enrolled in the study. A small biopsy segment of the saphenous vein (SV), radial artery (RA), and left internal mammary artery (LIMA) of patients was obtained during the surgery. The fatty acid (FA) profile of tissue samples was analyzed using Gas Chromatography-Mass Spectroscopy (GCMS). Among the different arteries tested, palmitic acid and stearic acid were the predominant fatty acids. As far as monounsaturated FA (MUFA) are concerned, oleic acid was found to be the most abundant MUFA in vessels. The FA profile of LIMA samples had a higher SFA percentage and lower unsaturated FA percentage compared to other vessels. Furthermore, the vessel samples of RA indicated the highest percentage of pro-inflammatory ω -6 polyunsaturated fatty acids (PUFA) as well as a higher percentage ratio between ω -6: ω -3 PUFA. The fatty acid composition and ω -6: ω -3 PUFA ratio suggests that LIMA graft is preferred for CABG over RA and SV.
Collapse
Affiliation(s)
- E. M. S. Bandara
- Department of Medical Laboratory Science, Faculty of Allied Health Sciences, University of Sri Jayewardenepura, Nugegoda, Sri Lanka
| | - D. I. U. Edirisinghe
- Instrument Center, Faculty of Applied Sciences, University of Sri Jayewardenepura, Nugegoda, Sri Lanka
| | | | - H. Peiris
- Department of Biochemistry, Faculty of Medical Sciences, University of Sri Jayewardenepura, Nugegoda, Sri Lanka
| | - P. P. R. Perera
- Department of Biochemistry, Faculty of Medical Sciences, University of Sri Jayewardenepura, Nugegoda, Sri Lanka
| | - A. G. Jayakrishan
- Nawaloka Hospital Research and Education Foundation, Nawaloka Hospitals PLC, Colombo, Sri Lanka
| | - H. D. Waikar
- Nawaloka Hospital Research and Education Foundation, Nawaloka Hospitals PLC, Colombo, Sri Lanka
| | - S. K. Sharma
- Nawaloka Hospital Research and Education Foundation, Nawaloka Hospitals PLC, Colombo, Sri Lanka
| | - V. Abeysuriya
- Nawaloka Hospital Research and Education Foundation, Nawaloka Hospitals PLC, Colombo, Sri Lanka
| | - L. G. Chandrasena
- Nawaloka Hospital Research and Education Foundation, Nawaloka Hospitals PLC, Colombo, Sri Lanka
| |
Collapse
|
7
|
Demina EP, Smutova V, Pan X, Fougerat A, Guo T, Zou C, Chakraberty R, Snarr BD, Shiao TC, Roy R, Orekhov AN, Miyagi T, Laffargue M, Sheppard DC, Cairo CW, Pshezhetsky AV. Neuraminidases 1 and 3 Trigger Atherosclerosis by Desialylating Low-Density Lipoproteins and Increasing Their Uptake by Macrophages. J Am Heart Assoc 2021; 10:e018756. [PMID: 33554615 PMCID: PMC7955353 DOI: 10.1161/jaha.120.018756] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Background Chronic vascular disease atherosclerosis starts with an uptake of atherogenic modified low-density lipoproteins (LDLs) by resident macrophages, resulting in formation of arterial fatty streaks and eventually atheromatous plaques. Increased plasma sialic acid levels, increased neuraminidase activity, and reduced sialic acid LDL content have been previously associated with atherosclerosis and coronary artery disease in human patients, but the mechanism underlying this association has not been explored. Methods and Results We tested the hypothesis that neuraminidases contribute to development of atherosclerosis by removing sialic acid residues from glycan chains of the LDL glycoprotein and glycolipids. Atherosclerosis progression was investigated in apolipoprotein E and LDL receptor knockout mice with genetic deficiency of neuraminidases 1, 3, and 4 or those treated with specific neuraminidase inhibitors. We show that desialylation of the LDL glycoprotein, apolipoprotein B 100, by human neuraminidases 1 and 3 increases the uptake of human LDL by human cultured macrophages and by macrophages in aortic root lesions in Apoe-/- mice via asialoglycoprotein receptor 1. Genetic inactivation or pharmacological inhibition of neuraminidases 1 and 3 significantly delays formation of fatty streaks in the aortic root without affecting the plasma cholesterol and LDL levels in Apoe-/- and Ldlr-/- mouse models of atherosclerosis. Conclusions Together, our results suggest that neuraminidases 1 and 3 trigger the initial phase of atherosclerosis and formation of aortic fatty streaks by desialylating LDL and increasing their uptake by resident macrophages.
Collapse
Affiliation(s)
- Ekaterina P Demina
- Departments of Pediatrics and Biochemistry Sainte-Justine University Hospital Research CenterUniversity of Montreal Quebec Canada
| | - Victoria Smutova
- Departments of Pediatrics and Biochemistry Sainte-Justine University Hospital Research CenterUniversity of Montreal Quebec Canada
| | - Xuefang Pan
- Departments of Pediatrics and Biochemistry Sainte-Justine University Hospital Research CenterUniversity of Montreal Quebec Canada
| | - Anne Fougerat
- Departments of Pediatrics and Biochemistry Sainte-Justine University Hospital Research CenterUniversity of Montreal Quebec Canada
| | - Tianlin Guo
- Department of Chemistry University of Alberta Edmonton Alberta Canada
| | - Chunxia Zou
- Department of Chemistry University of Alberta Edmonton Alberta Canada
| | | | - Brendan D Snarr
- Departments of Medicine, Microbiology and Immunology McGill University Montreal Quebec Canada
| | - Tze C Shiao
- Department of Chemistry Université du Québec à Montréal Montreal Quebec Canada
| | - Rene Roy
- Department of Chemistry Université du Québec à Montréal Montreal Quebec Canada
| | | | - Taeko Miyagi
- Miyagi Cancer Center Research Institute Natori Miyagi Japan
| | - Muriel Laffargue
- Institut National de la Santé et de la Recherche MédicaleUMR 1048Institute of Metabolic and Cardiovascular Diseases Toulouse France
| | - Donald C Sheppard
- Departments of Medicine, Microbiology and Immunology McGill University Montreal Quebec Canada
| | | | - Alexey V Pshezhetsky
- Departments of Pediatrics and Biochemistry Sainte-Justine University Hospital Research CenterUniversity of Montreal Quebec Canada
| |
Collapse
|
8
|
Oppedisano F, Macrì R, Gliozzi M, Musolino V, Carresi C, Maiuolo J, Bosco F, Nucera S, Caterina Zito M, Guarnieri L, Scarano F, Nicita C, Coppoletta AR, Ruga S, Scicchitano M, Mollace R, Palma E, Mollace V. The Anti-Inflammatory and Antioxidant Properties of n-3 PUFAs: Their Role in Cardiovascular Protection. Biomedicines 2020; 8:biomedicines8090306. [PMID: 32854210 PMCID: PMC7554783 DOI: 10.3390/biomedicines8090306] [Citation(s) in RCA: 142] [Impact Index Per Article: 28.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2020] [Revised: 08/22/2020] [Accepted: 08/24/2020] [Indexed: 12/15/2022] Open
Abstract
Polyunsaturated fatty acids (n-3 PUFAs) are long-chain polyunsaturated fatty acids with 18, 20 or 22 carbon atoms, which have been found able to counteract cardiovascular diseases. Eicosapentaenoic acid (EPA) and docosahexaenoic acid (DHA), in particular, have been found to produce both vaso- and cardio-protective response via modulation of membrane phospholipids thereby improving cardiac mitochondrial functions and energy production. However, antioxidant properties of n-3 PUFAs, along with their anti-inflammatory effect in both blood vessels and cardiac cells, seem to exert beneficial effects in cardiovascular impairment. In fact, dietary supplementation with n-3 PUFAs has been demonstrated to reduce oxidative stress-related mitochondrial dysfunction and endothelial cell apoptosis, an effect occurring via an increased activity of endogenous antioxidant enzymes. On the other hand, n-3 PUFAs have been shown to counteract the release of pro-inflammatory cytokines in both vascular tissues and in the myocardium, thereby restoring vascular reactivity and myocardial performance. Here we summarize the molecular mechanisms underlying the anti-oxidant and anti-inflammatory effect of n-3 PUFAs in vascular and cardiac tissues and their implication in the prevention and treatment of cardiovascular disease.
Collapse
Affiliation(s)
- Francesca Oppedisano
- Institute of Research for Food Safety and Health (IRC-FSH), Department of Health Sciences, University “Magna Graecia” of Catanzaro, 88100 Catanzaro, Italy; (F.O.); (R.M.); (M.G.); (V.M.); (C.C.); (J.M.); (F.B.); (S.N.); (M.C.Z.); (L.G.); (F.S.); (C.N.); (A.R.C.); (S.R.); (M.S.); (R.M.); (E.P.)
| | - Roberta Macrì
- Institute of Research for Food Safety and Health (IRC-FSH), Department of Health Sciences, University “Magna Graecia” of Catanzaro, 88100 Catanzaro, Italy; (F.O.); (R.M.); (M.G.); (V.M.); (C.C.); (J.M.); (F.B.); (S.N.); (M.C.Z.); (L.G.); (F.S.); (C.N.); (A.R.C.); (S.R.); (M.S.); (R.M.); (E.P.)
| | - Micaela Gliozzi
- Institute of Research for Food Safety and Health (IRC-FSH), Department of Health Sciences, University “Magna Graecia” of Catanzaro, 88100 Catanzaro, Italy; (F.O.); (R.M.); (M.G.); (V.M.); (C.C.); (J.M.); (F.B.); (S.N.); (M.C.Z.); (L.G.); (F.S.); (C.N.); (A.R.C.); (S.R.); (M.S.); (R.M.); (E.P.)
| | - Vincenzo Musolino
- Institute of Research for Food Safety and Health (IRC-FSH), Department of Health Sciences, University “Magna Graecia” of Catanzaro, 88100 Catanzaro, Italy; (F.O.); (R.M.); (M.G.); (V.M.); (C.C.); (J.M.); (F.B.); (S.N.); (M.C.Z.); (L.G.); (F.S.); (C.N.); (A.R.C.); (S.R.); (M.S.); (R.M.); (E.P.)
| | - Cristina Carresi
- Institute of Research for Food Safety and Health (IRC-FSH), Department of Health Sciences, University “Magna Graecia” of Catanzaro, 88100 Catanzaro, Italy; (F.O.); (R.M.); (M.G.); (V.M.); (C.C.); (J.M.); (F.B.); (S.N.); (M.C.Z.); (L.G.); (F.S.); (C.N.); (A.R.C.); (S.R.); (M.S.); (R.M.); (E.P.)
| | - Jessica Maiuolo
- Institute of Research for Food Safety and Health (IRC-FSH), Department of Health Sciences, University “Magna Graecia” of Catanzaro, 88100 Catanzaro, Italy; (F.O.); (R.M.); (M.G.); (V.M.); (C.C.); (J.M.); (F.B.); (S.N.); (M.C.Z.); (L.G.); (F.S.); (C.N.); (A.R.C.); (S.R.); (M.S.); (R.M.); (E.P.)
| | - Francesca Bosco
- Institute of Research for Food Safety and Health (IRC-FSH), Department of Health Sciences, University “Magna Graecia” of Catanzaro, 88100 Catanzaro, Italy; (F.O.); (R.M.); (M.G.); (V.M.); (C.C.); (J.M.); (F.B.); (S.N.); (M.C.Z.); (L.G.); (F.S.); (C.N.); (A.R.C.); (S.R.); (M.S.); (R.M.); (E.P.)
| | - Saverio Nucera
- Institute of Research for Food Safety and Health (IRC-FSH), Department of Health Sciences, University “Magna Graecia” of Catanzaro, 88100 Catanzaro, Italy; (F.O.); (R.M.); (M.G.); (V.M.); (C.C.); (J.M.); (F.B.); (S.N.); (M.C.Z.); (L.G.); (F.S.); (C.N.); (A.R.C.); (S.R.); (M.S.); (R.M.); (E.P.)
| | - Maria Caterina Zito
- Institute of Research for Food Safety and Health (IRC-FSH), Department of Health Sciences, University “Magna Graecia” of Catanzaro, 88100 Catanzaro, Italy; (F.O.); (R.M.); (M.G.); (V.M.); (C.C.); (J.M.); (F.B.); (S.N.); (M.C.Z.); (L.G.); (F.S.); (C.N.); (A.R.C.); (S.R.); (M.S.); (R.M.); (E.P.)
| | - Lorenza Guarnieri
- Institute of Research for Food Safety and Health (IRC-FSH), Department of Health Sciences, University “Magna Graecia” of Catanzaro, 88100 Catanzaro, Italy; (F.O.); (R.M.); (M.G.); (V.M.); (C.C.); (J.M.); (F.B.); (S.N.); (M.C.Z.); (L.G.); (F.S.); (C.N.); (A.R.C.); (S.R.); (M.S.); (R.M.); (E.P.)
| | - Federica Scarano
- Institute of Research for Food Safety and Health (IRC-FSH), Department of Health Sciences, University “Magna Graecia” of Catanzaro, 88100 Catanzaro, Italy; (F.O.); (R.M.); (M.G.); (V.M.); (C.C.); (J.M.); (F.B.); (S.N.); (M.C.Z.); (L.G.); (F.S.); (C.N.); (A.R.C.); (S.R.); (M.S.); (R.M.); (E.P.)
| | - Caterina Nicita
- Institute of Research for Food Safety and Health (IRC-FSH), Department of Health Sciences, University “Magna Graecia” of Catanzaro, 88100 Catanzaro, Italy; (F.O.); (R.M.); (M.G.); (V.M.); (C.C.); (J.M.); (F.B.); (S.N.); (M.C.Z.); (L.G.); (F.S.); (C.N.); (A.R.C.); (S.R.); (M.S.); (R.M.); (E.P.)
| | - Anna Rita Coppoletta
- Institute of Research for Food Safety and Health (IRC-FSH), Department of Health Sciences, University “Magna Graecia” of Catanzaro, 88100 Catanzaro, Italy; (F.O.); (R.M.); (M.G.); (V.M.); (C.C.); (J.M.); (F.B.); (S.N.); (M.C.Z.); (L.G.); (F.S.); (C.N.); (A.R.C.); (S.R.); (M.S.); (R.M.); (E.P.)
| | - Stefano Ruga
- Institute of Research for Food Safety and Health (IRC-FSH), Department of Health Sciences, University “Magna Graecia” of Catanzaro, 88100 Catanzaro, Italy; (F.O.); (R.M.); (M.G.); (V.M.); (C.C.); (J.M.); (F.B.); (S.N.); (M.C.Z.); (L.G.); (F.S.); (C.N.); (A.R.C.); (S.R.); (M.S.); (R.M.); (E.P.)
| | - Miriam Scicchitano
- Institute of Research for Food Safety and Health (IRC-FSH), Department of Health Sciences, University “Magna Graecia” of Catanzaro, 88100 Catanzaro, Italy; (F.O.); (R.M.); (M.G.); (V.M.); (C.C.); (J.M.); (F.B.); (S.N.); (M.C.Z.); (L.G.); (F.S.); (C.N.); (A.R.C.); (S.R.); (M.S.); (R.M.); (E.P.)
| | - Rocco Mollace
- Institute of Research for Food Safety and Health (IRC-FSH), Department of Health Sciences, University “Magna Graecia” of Catanzaro, 88100 Catanzaro, Italy; (F.O.); (R.M.); (M.G.); (V.M.); (C.C.); (J.M.); (F.B.); (S.N.); (M.C.Z.); (L.G.); (F.S.); (C.N.); (A.R.C.); (S.R.); (M.S.); (R.M.); (E.P.)
- Division of Cardiology, University Hospital Policlinico Tor Vergata, 00133 Rome, Italy
| | - Ernesto Palma
- Institute of Research for Food Safety and Health (IRC-FSH), Department of Health Sciences, University “Magna Graecia” of Catanzaro, 88100 Catanzaro, Italy; (F.O.); (R.M.); (M.G.); (V.M.); (C.C.); (J.M.); (F.B.); (S.N.); (M.C.Z.); (L.G.); (F.S.); (C.N.); (A.R.C.); (S.R.); (M.S.); (R.M.); (E.P.)
| | - Vincenzo Mollace
- Institute of Research for Food Safety and Health (IRC-FSH), Department of Health Sciences, University “Magna Graecia” of Catanzaro, 88100 Catanzaro, Italy; (F.O.); (R.M.); (M.G.); (V.M.); (C.C.); (J.M.); (F.B.); (S.N.); (M.C.Z.); (L.G.); (F.S.); (C.N.); (A.R.C.); (S.R.); (M.S.); (R.M.); (E.P.)
- IRCCS San Raffaele Pisana, 00163 Roma, Italy
- Correspondence:
| |
Collapse
|
9
|
Abstract
PURPOSE OF REVIEW Lipoprotein lipase (LpL) is well known for its lipolytic action in blood lipoprotein triglyceride catabolism. This article summarizes the recent mechanistic and molecular studies on elucidating the 'unconventional' roles of LpL in mediating biological events related to immune cell response and lipid transport in the pathogenesis of cardiovascular disease (CVD) and tissue degenerative disorders. RECENT FINDINGS Several approaches to inactivate the inhibitors that block LpL enzymatic activity have reestablished the importance of systemic LpL activity in reducing CVD risk. On the other hand, increasing evidence suggests that focal arterial expression of LpL relates to aortic macrophage levels and inflammatory processes. In the hematopoietic origin, LpL also plays a role in modulating hematopoietic stem cell proliferation and circulating blood cell levels and phenotypes. Finally, building upon the strong genetic evidence on the association with assorted brain disorders, a new era in exploring the mechanistic insights into the functions and activity of LpL in brain that impacts central nerve systems has begun. SUMMARY A better understanding of the molecular action of LpL will help to devise novel strategies for intervention of a number of diseases, including blood cell or metabolic disorders, as well to inhibit pathways related to CVD and tissue degenerative processes.
Collapse
Affiliation(s)
- Chuchun L Chang
- Institute of Human Nutrition, College of Physicians and Surgeons, Columbia University, New York, New York, USA
| |
Collapse
|
10
|
|
11
|
Chaudhry R, Viljoen A, Wierzbicki AS. Pharmacological treatment options for severe hypertriglyceridemia and familial chylomicronemia syndrome. Expert Rev Clin Pharmacol 2018; 11:589-598. [PMID: 29842811 DOI: 10.1080/17512433.2018.1480368] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
INTRODUCTION A spectrum of disorders, ranging from rare severe cases of homozygous null lipoprotein lipase deficiency (LPLD)-familial chylomicronemia syndrome (FCS) to heterozygous missense LPLD or polygenic causes, result in hypertriglyceridemia and pancreatitis. The effects of mutations are exacerbated by environmental factors such as diet, pregnancy, and insulin resistance. Areas covered: In this review, authors discuss chronic treatment of FCS by ultra-low fat diets allied with the use of fibrates, omega-3 fatty acids, niacin, statins, and insulin-sensitizing therapies depending on the extent of residual lipoprotein lipase (LPL) activity; novel therapies in development target triglyceride (TG)-rich lipoprotein particle clearance. Previously, a gene therapy approach to LPL-alipogene tiparvovec showed that direct targeting of LPL function reduced pancreatitis events. An antisense oligonucleotide to apolipoprotein-C3, volanesorsen has been shown to decrease TGs by 70-80% and possibly to reduce rates of pancreatitis admissions. Studies are underway to validate its long-term efficacy and safety. Other approaches investigating the role of LPL modulating proteins such as angiopoietin-like petide-3 (ANGPTL3) are under consideration. Expert opinion: Current therapeutic options are not sufficient for management of many cases of FCS. The availability of antisense anti-apoC3 therapies and, in the future, ANGPTL3 therapies may remedy this.
Collapse
Affiliation(s)
- Rabia Chaudhry
- a Specialist Registrar Chemical Pathology/Metabolic Medicine , Lister Hospital , Stevenage , Hertfordshire , UK
| | - Adie Viljoen
- b Consultant in Metabolic Medicine/Chemical Pathology , East Hertfordshire Hospitals, Lister Hospital , Stevenage , Hertfordshire , UK
| | - Anthony S Wierzbicki
- c Department of Metabolic Medicine/Chemical Pathology , Guy's and St Thomas' Hospitals , London , UK
| |
Collapse
|
12
|
Chang CL, Garcia-Arcos I, Nyrén R, Olivecrona G, Kim JY, Hu Y, Agrawal RR, Murphy AJ, Goldberg IJ, Deckelbaum RJ. Lipoprotein Lipase Deficiency Impairs Bone Marrow Myelopoiesis and Reduces Circulating Monocyte Levels. Arterioscler Thromb Vasc Biol 2018; 38:509-519. [PMID: 29371243 DOI: 10.1161/atvbaha.117.310607] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2017] [Accepted: 01/10/2018] [Indexed: 11/16/2022]
Abstract
OBJECTIVE Tissue macrophages induce and perpetuate proinflammatory responses, thereby promoting metabolic and cardiovascular disease. Lipoprotein lipase (LpL), the rate-limiting enzyme in blood triglyceride catabolism, is expressed by macrophages in atherosclerotic plaques. We questioned whether LpL, which is also expressed in the bone marrow (BM), affects circulating white blood cells and BM proliferation and modulates macrophage retention within the artery. APPROACH AND RESULTS We characterized blood and tissue leukocytes and inflammatory molecules in transgenic LpL knockout mice rescued from lethal hypertriglyceridemia within 18 hours of life by muscle-specific LpL expression (MCKL0 mice). LpL-deficient mice had ≈40% reduction in blood white blood cell, neutrophils, and total and inflammatory monocytes (Ly6C/Ghi). LpL deficiency also significantly decreased expression of BM macrophage-associated markers (F4/80 and TNF-α [tumor necrosis factor α]), master transcription factors (PU.1 and C/EBPα), and colony-stimulating factors (CSFs) and their receptors, which are required for monocyte and monocyte precursor proliferation and differentiation. As a result, differentiation of macrophages from BM-derived monocyte progenitors and monocytes was decreased in MCKL0 mice. Furthermore, although LpL deficiency was associated with reduced BM uptake and accumulation of triglyceride-rich particles and macrophage CSF-macrophage CSF receptor binding, triglyceride lipolysis products (eg, linoleic acid) stimulated expression of macrophage CSF and macrophage CSF receptor in BM-derived macrophage precursor cells. Arterial macrophage numbers decreased after heparin-mediated LpL cell dissociation and by genetic knockout of arterial LpL. Reconstitution of LpL-expressing BM replenished aortic macrophage density. CONCLUSIONS LpL regulates peripheral leukocyte levels and affects BM monocyte progenitor differentiation and aortic macrophage accumulation.
Collapse
Affiliation(s)
- Chuchun L Chang
- From Institute of Human Nutrition (C.L.C., J.Y.K., R.R.A., R.J.D.), Division of Preventive Medicine and Nutrition, Department of Medicine (I.G.-A.), Division of Molecular Medicine, Department of Medicine (Y.H., A.J.M., I.J.G.), and Department of Pediatrics (R.J.D.), College of Physicians and Surgeons, Columbia University, New York; Department of Medical Biosciences/Physiological Chemistry, Umeå University, Sweden (R.N., G.O.); Division of Endocrinology, Diabetes, and Metabolism, New York University School of Medicine, New York (Y.H., I.J.G.); Haematopoiesis and Leukocyte Biology, Baker Heart and Diabetes Institute, Melbourne, Victoria, Australia (A.J.M.); and Department of Immunology, Monash University, Melbourne, Victoria, Australia (A.J.M.)
| | - Itsaso Garcia-Arcos
- From Institute of Human Nutrition (C.L.C., J.Y.K., R.R.A., R.J.D.), Division of Preventive Medicine and Nutrition, Department of Medicine (I.G.-A.), Division of Molecular Medicine, Department of Medicine (Y.H., A.J.M., I.J.G.), and Department of Pediatrics (R.J.D.), College of Physicians and Surgeons, Columbia University, New York; Department of Medical Biosciences/Physiological Chemistry, Umeå University, Sweden (R.N., G.O.); Division of Endocrinology, Diabetes, and Metabolism, New York University School of Medicine, New York (Y.H., I.J.G.); Haematopoiesis and Leukocyte Biology, Baker Heart and Diabetes Institute, Melbourne, Victoria, Australia (A.J.M.); and Department of Immunology, Monash University, Melbourne, Victoria, Australia (A.J.M.)
| | - Rakel Nyrén
- From Institute of Human Nutrition (C.L.C., J.Y.K., R.R.A., R.J.D.), Division of Preventive Medicine and Nutrition, Department of Medicine (I.G.-A.), Division of Molecular Medicine, Department of Medicine (Y.H., A.J.M., I.J.G.), and Department of Pediatrics (R.J.D.), College of Physicians and Surgeons, Columbia University, New York; Department of Medical Biosciences/Physiological Chemistry, Umeå University, Sweden (R.N., G.O.); Division of Endocrinology, Diabetes, and Metabolism, New York University School of Medicine, New York (Y.H., I.J.G.); Haematopoiesis and Leukocyte Biology, Baker Heart and Diabetes Institute, Melbourne, Victoria, Australia (A.J.M.); and Department of Immunology, Monash University, Melbourne, Victoria, Australia (A.J.M.)
| | - Gunilla Olivecrona
- From Institute of Human Nutrition (C.L.C., J.Y.K., R.R.A., R.J.D.), Division of Preventive Medicine and Nutrition, Department of Medicine (I.G.-A.), Division of Molecular Medicine, Department of Medicine (Y.H., A.J.M., I.J.G.), and Department of Pediatrics (R.J.D.), College of Physicians and Surgeons, Columbia University, New York; Department of Medical Biosciences/Physiological Chemistry, Umeå University, Sweden (R.N., G.O.); Division of Endocrinology, Diabetes, and Metabolism, New York University School of Medicine, New York (Y.H., I.J.G.); Haematopoiesis and Leukocyte Biology, Baker Heart and Diabetes Institute, Melbourne, Victoria, Australia (A.J.M.); and Department of Immunology, Monash University, Melbourne, Victoria, Australia (A.J.M.)
| | - Ji Young Kim
- From Institute of Human Nutrition (C.L.C., J.Y.K., R.R.A., R.J.D.), Division of Preventive Medicine and Nutrition, Department of Medicine (I.G.-A.), Division of Molecular Medicine, Department of Medicine (Y.H., A.J.M., I.J.G.), and Department of Pediatrics (R.J.D.), College of Physicians and Surgeons, Columbia University, New York; Department of Medical Biosciences/Physiological Chemistry, Umeå University, Sweden (R.N., G.O.); Division of Endocrinology, Diabetes, and Metabolism, New York University School of Medicine, New York (Y.H., I.J.G.); Haematopoiesis and Leukocyte Biology, Baker Heart and Diabetes Institute, Melbourne, Victoria, Australia (A.J.M.); and Department of Immunology, Monash University, Melbourne, Victoria, Australia (A.J.M.)
| | - Yunying Hu
- From Institute of Human Nutrition (C.L.C., J.Y.K., R.R.A., R.J.D.), Division of Preventive Medicine and Nutrition, Department of Medicine (I.G.-A.), Division of Molecular Medicine, Department of Medicine (Y.H., A.J.M., I.J.G.), and Department of Pediatrics (R.J.D.), College of Physicians and Surgeons, Columbia University, New York; Department of Medical Biosciences/Physiological Chemistry, Umeå University, Sweden (R.N., G.O.); Division of Endocrinology, Diabetes, and Metabolism, New York University School of Medicine, New York (Y.H., I.J.G.); Haematopoiesis and Leukocyte Biology, Baker Heart and Diabetes Institute, Melbourne, Victoria, Australia (A.J.M.); and Department of Immunology, Monash University, Melbourne, Victoria, Australia (A.J.M.)
| | - Rishi R Agrawal
- From Institute of Human Nutrition (C.L.C., J.Y.K., R.R.A., R.J.D.), Division of Preventive Medicine and Nutrition, Department of Medicine (I.G.-A.), Division of Molecular Medicine, Department of Medicine (Y.H., A.J.M., I.J.G.), and Department of Pediatrics (R.J.D.), College of Physicians and Surgeons, Columbia University, New York; Department of Medical Biosciences/Physiological Chemistry, Umeå University, Sweden (R.N., G.O.); Division of Endocrinology, Diabetes, and Metabolism, New York University School of Medicine, New York (Y.H., I.J.G.); Haematopoiesis and Leukocyte Biology, Baker Heart and Diabetes Institute, Melbourne, Victoria, Australia (A.J.M.); and Department of Immunology, Monash University, Melbourne, Victoria, Australia (A.J.M.)
| | - Andrew J Murphy
- From Institute of Human Nutrition (C.L.C., J.Y.K., R.R.A., R.J.D.), Division of Preventive Medicine and Nutrition, Department of Medicine (I.G.-A.), Division of Molecular Medicine, Department of Medicine (Y.H., A.J.M., I.J.G.), and Department of Pediatrics (R.J.D.), College of Physicians and Surgeons, Columbia University, New York; Department of Medical Biosciences/Physiological Chemistry, Umeå University, Sweden (R.N., G.O.); Division of Endocrinology, Diabetes, and Metabolism, New York University School of Medicine, New York (Y.H., I.J.G.); Haematopoiesis and Leukocyte Biology, Baker Heart and Diabetes Institute, Melbourne, Victoria, Australia (A.J.M.); and Department of Immunology, Monash University, Melbourne, Victoria, Australia (A.J.M.)
| | - Ira J Goldberg
- From Institute of Human Nutrition (C.L.C., J.Y.K., R.R.A., R.J.D.), Division of Preventive Medicine and Nutrition, Department of Medicine (I.G.-A.), Division of Molecular Medicine, Department of Medicine (Y.H., A.J.M., I.J.G.), and Department of Pediatrics (R.J.D.), College of Physicians and Surgeons, Columbia University, New York; Department of Medical Biosciences/Physiological Chemistry, Umeå University, Sweden (R.N., G.O.); Division of Endocrinology, Diabetes, and Metabolism, New York University School of Medicine, New York (Y.H., I.J.G.); Haematopoiesis and Leukocyte Biology, Baker Heart and Diabetes Institute, Melbourne, Victoria, Australia (A.J.M.); and Department of Immunology, Monash University, Melbourne, Victoria, Australia (A.J.M.).
| | - Richard J Deckelbaum
- From Institute of Human Nutrition (C.L.C., J.Y.K., R.R.A., R.J.D.), Division of Preventive Medicine and Nutrition, Department of Medicine (I.G.-A.), Division of Molecular Medicine, Department of Medicine (Y.H., A.J.M., I.J.G.), and Department of Pediatrics (R.J.D.), College of Physicians and Surgeons, Columbia University, New York; Department of Medical Biosciences/Physiological Chemistry, Umeå University, Sweden (R.N., G.O.); Division of Endocrinology, Diabetes, and Metabolism, New York University School of Medicine, New York (Y.H., I.J.G.); Haematopoiesis and Leukocyte Biology, Baker Heart and Diabetes Institute, Melbourne, Victoria, Australia (A.J.M.); and Department of Immunology, Monash University, Melbourne, Victoria, Australia (A.J.M.).
| |
Collapse
|
13
|
Radiolabeled cholesteryl ethers: A need to analyze for biological stability before use. Biochem Biophys Rep 2017; 13:1-6. [PMID: 29188234 PMCID: PMC5697731 DOI: 10.1016/j.bbrep.2017.10.007] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2017] [Accepted: 10/23/2017] [Indexed: 11/22/2022] Open
Abstract
Radiolabeled cholesteryl ethers are widely used as non-metabolizable tracers for lipoproteins and lipid emulsions in a variety of in vitro and in vivo experiments. Since cholesteryl ethers do not leave cells after uptake and are not hydrolyzed by mammalian cellular enzymes, these compounds can act as markers for cumulative cell uptakes of labeled particles. We have employed [3H]cholesteryl oleoyl ether to study the uptake and distribution of triglyceride-rich emulsion particles on animal models. However, questionable unexpected results compelled us to analyze the stability of these ethers. We tested the stability of two commercially available radiolabeled cholesteryl ethers - [3H]cholesteryl oleoyl ether and [3H]cholesteryl hexadecyl ether from different suppliers, employing in vitro, in vivo and chemical model systems. Our results show that, among the two cholesteryl ethers tested, one ether was hydrolyzed to free cholesterol in vitro, in vivo and chemically under alkaline hydrolyzing agent. Free cholesterol, unlike cholesteryl ether, can then re-enter the circulation leading to confounding results. The other ether was not hydrolyzed to free cholesterol and remained as a stable ether. Hence, radiolabeled cholesteryl ethers should be analyzed for biological stability before utilizing them for in vitro or in vivo experiments. Tested stability of two commercially available radiolabeled cholesteryl ethers. One ether was hydrolyzed to free cholesterol (FC) in vitro and in vivo. FC, re-entered circulation giving questionable unexpected results in experiments. The other ether was unhydrolyzed in all model systems. Radiolabeled cholesteryl ethers should be analyzed for stability before use.
Collapse
|
14
|
Liu Y, Fang X, Zhang X, Huang J, He J, Peng L, Ye C, Wang Y, Xue F, Ai D, Li D, Zhu Y. Metabolic profiling of murine plasma reveals eicosapentaenoic acid metabolites protecting against endothelial activation and atherosclerosis. Br J Pharmacol 2017; 175:1190-1204. [PMID: 28771708 DOI: 10.1111/bph.13971] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2017] [Revised: 07/22/2017] [Accepted: 07/24/2017] [Indexed: 12/13/2022] Open
Abstract
BACKGROUND AND PURPOSE Atherosclerosis results from a maladaptive inflammatory response initiated by the intramural retention of LDL in susceptible areas of the arterial vasculature. The ω-3 polyunsaturated fatty acids (ω-3) have protective effects in atherosclerosis; however, their molecular mechanism is still largely unknown. The present study used a metabolomic approach to reveal the atheroprotective metabolites of ω-3 and investigate the underlying mechanisms. EXPERIMENTAL APPROACH We evaluated the development of atherosclerosis in LDL receptor-deficient mice (LDLR-/- ) fed a Western-type diet (WTD) plus ω-3 and also LDLR-/- and fat-1 transgenic (LDLR-/- -fat-1tg ) mice fed a WTD. The profiles of ω-3 in the plasma were screened by LC-MS/MS using unbiased systematic metabolomics analysis. We also studied the effect of metabolites of eicosapentaenoic acid (EPA) on endothelial activation in vitro. KEY RESULTS The ω-3 diet and fat-1 transgene decreased monocyte infiltration, inhibited the expression of pro-inflammatory genes and significantly attenuated atherosclerotic plaque formation and enhanced plaque stability in LDLR-/- mice. The content of 18-hydroxy-eicosapentaenoic acid (18-HEPE) and 17,18-epoxy-eicosatetraenoic acid (17,18-EEQ), from the cytochrome P450 pathway of EPA, was significantly higher in plasma from both ω-3-treated LDLR-/- and LDLR-/- -fat-1tg mice as compared with WTD-fed LDLR-/- mice. In vitro in endothelial cells, 18-HEPE or 17,18-EEQ decreased inflammatory gene expression induced by TNFα via NF-κB signalling and thereby inhibited monocyte adhesion to endothelial cells. CONCLUSIONS AND IMPLICATIONS EPA protected against the development of atherosclerosis in atheroprone mice via the metabolites 18-HEPE and/or 17,18-EEQ, which reduced endothelial activation. These compounds may have therapeutic implications in atherosclerosis. LINKED ARTICLES This article is part of a themed section on Spotlight on Small Molecules in Cardiovascular Diseases. To view the other articles in this section visit http://onlinelibrary.wiley.com/doi/10.1111/bph.v175.8/issuetoc.
Collapse
Affiliation(s)
- Yajin Liu
- Tianjin Key Laboratory of Metabolic Diseases and Department of Physiology and Pathophysiology, Tianjin Medical University, Tianjin, China
| | - Xuan Fang
- Department of Physiology and Pathophysiology, Peking University Health Science Center, Beijing, China
| | - Xu Zhang
- Tianjin Key Laboratory of Metabolic Diseases and Department of Physiology and Pathophysiology, Tianjin Medical University, Tianjin, China
| | - Jing Huang
- Tianjin Key Laboratory of Metabolic Diseases and Department of Physiology and Pathophysiology, Tianjin Medical University, Tianjin, China
| | - Jinlong He
- Tianjin Key Laboratory of Metabolic Diseases and Department of Physiology and Pathophysiology, Tianjin Medical University, Tianjin, China
| | - Liyuan Peng
- Tianjin Key Laboratory of Metabolic Diseases and Department of Physiology and Pathophysiology, Tianjin Medical University, Tianjin, China
| | - Chenji Ye
- Tianjin Key Laboratory of Metabolic Diseases and Department of Physiology and Pathophysiology, Tianjin Medical University, Tianjin, China
| | - Yingmei Wang
- Department of Gynecology and Obstetrics, Tianjin Medical University General Hospital, Tianjin, China
| | - Fengxia Xue
- Department of Gynecology and Obstetrics, Tianjin Medical University General Hospital, Tianjin, China
| | - Ding Ai
- Tianjin Key Laboratory of Metabolic Diseases and Department of Physiology and Pathophysiology, Tianjin Medical University, Tianjin, China
| | - Dan Li
- Department of Physiology and Pathophysiology, Peking University Health Science Center, Beijing, China
| | - Yi Zhu
- Tianjin Key Laboratory of Metabolic Diseases and Department of Physiology and Pathophysiology, Tianjin Medical University, Tianjin, China.,Department of Physiology and Pathophysiology, Peking University Health Science Center, Beijing, China
| |
Collapse
|
15
|
Wüstner D, Lund FW, Röhrl C, Stangl H. Potential of BODIPY-cholesterol for analysis of cholesterol transport and diffusion in living cells. Chem Phys Lipids 2016; 194:12-28. [DOI: 10.1016/j.chemphyslip.2015.08.007] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2015] [Revised: 08/07/2015] [Accepted: 08/12/2015] [Indexed: 01/04/2023]
|
16
|
Zanetti M, Grillo A, Losurdo P, Panizon E, Mearelli F, Cattin L, Barazzoni R, Carretta R. Omega-3 Polyunsaturated Fatty Acids: Structural and Functional Effects on the Vascular Wall. BIOMED RESEARCH INTERNATIONAL 2015; 2015:791978. [PMID: 26301252 PMCID: PMC4537737 DOI: 10.1155/2015/791978] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/25/2014] [Accepted: 01/26/2015] [Indexed: 01/24/2023]
Abstract
Omega-3 polyunsaturated fatty acids (n-3 PUFA) consumption is associated with reduced cardiovascular disease risk. Increasing evidence demonstrating a beneficial effect of n-3 PUFA on arterial wall properties is progressively emerging. We reviewed the recent available evidence for the cardiovascular effects of n-3 PUFA focusing on structural and functional properties of the vascular wall. In experimental studies and clinical trials n-3 PUFA have shown the ability to improve arterial hemodynamics by reducing arterial stiffness, thus explaining some of its cardioprotective properties. Recent studies suggest beneficial effects of n-3 PUFA on endothelial activation, which are likely to improve vascular function. Several molecular, cellular, and physiological pathways influenced by n-3 PUFA can affect arterial wall properties and therefore interfere with the atherosclerotic process. Although the relative weight of different physiological and molecular mechanisms and the dose-response on arterial wall properties have yet to be determined, n-3 PUFA have the potential to beneficially impact arterial wall remodeling and cardiovascular outcomes by targeting arterial wall stiffening and endothelial dysfunction.
Collapse
Affiliation(s)
- Michela Zanetti
- Department of Medicine, Surgery and Health Sciences, University of Trieste, Strada di Fiume 447, 34149 Trieste, Italy
| | - Andrea Grillo
- Department of Medicine, Surgery and Health Sciences, University of Trieste, Strada di Fiume 447, 34149 Trieste, Italy
| | - Pasquale Losurdo
- Department of Medicine, Surgery and Health Sciences, University of Trieste, Strada di Fiume 447, 34149 Trieste, Italy
| | - Emiliano Panizon
- Department of Medicine, Surgery and Health Sciences, University of Trieste, Strada di Fiume 447, 34149 Trieste, Italy
| | - Filippo Mearelli
- Department of Medicine, Surgery and Health Sciences, University of Trieste, Strada di Fiume 447, 34149 Trieste, Italy
| | - Luigi Cattin
- Department of Medicine, Surgery and Health Sciences, University of Trieste, Strada di Fiume 447, 34149 Trieste, Italy
| | - Rocco Barazzoni
- Department of Medicine, Surgery and Health Sciences, University of Trieste, Strada di Fiume 447, 34149 Trieste, Italy
| | - Renzo Carretta
- Department of Medicine, Surgery and Health Sciences, University of Trieste, Strada di Fiume 447, 34149 Trieste, Italy
| |
Collapse
|
17
|
Li Y, He PP, Zhang DW, Zheng XL, Cayabyab FS, Yin WD, Tang CK. Lipoprotein lipase: from gene to atherosclerosis. Atherosclerosis 2014; 237:597-608. [PMID: 25463094 DOI: 10.1016/j.atherosclerosis.2014.10.016] [Citation(s) in RCA: 79] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/09/2014] [Revised: 10/13/2014] [Accepted: 10/13/2014] [Indexed: 01/21/2023]
Abstract
Lipoprotein lipase (LPL) is a key enzyme in lipid metabolism and responsible for catalyzing lipolysis of triglycerides in lipoproteins. LPL is produced mainly in adipose tissue, skeletal and heart muscle, as well as in macrophage and other tissues. After synthesized, it is secreted and translocated to the vascular lumen. LPL expression and activity are regulated by a variety of factors, such as transcription factors, interactive proteins and nutritional state through complicated mechanisms. LPL with different distributions may exert distinct functions and have diverse roles in human health and disease with close association with atherosclerosis. It may pose a pro-atherogenic or an anti-atherogenic effect depending on its locations. In this review, we will discuss its gene, protein, synthesis, transportation and biological functions, and then focus on its regulation and relationship with atherosclerosis and potential underlying mechanisms. The goal of this review is to provide basic information and novel insight for further studies and therapeutic targets.
Collapse
Affiliation(s)
- Yuan Li
- Institute of Cardiovascular Research, Key Laboratory for Atherosclerology of Hunan Province, Hunan Province Cooperative Innovation Center for Molecular Target New Drug Discovery, Life Science Research Center, University of South China, Hengyang, Hunan 421001, China
| | - Ping-Ping He
- Institute of Cardiovascular Research, Key Laboratory for Atherosclerology of Hunan Province, Hunan Province Cooperative Innovation Center for Molecular Target New Drug Discovery, Life Science Research Center, University of South China, Hengyang, Hunan 421001, China; School of Nursing, University of South China, Hengyang, Hunan 421001, China
| | - Da-Wei Zhang
- Department of Pediatrics and Group on the Molecular and Cell Biology of Lipids, University of Alberta, Edmonton, Alberta T6G 2S2, Canada
| | - Xi-Long Zheng
- Department of Biochemistry and Molecular Biology, The Libin Cardiovascular Institute of Alberta, The Cumming School of Medicine, The University of Calgary, Health Sciences Center, 3330 Hospital Dr NW, Calgary, Alberta T2N 4N1, Canada
| | - Fracisco S Cayabyab
- Department of Surgery, College of Medicine, University of Saskatchewan, Saskatoon, Saskatchewan, Canada
| | - Wei-Dong Yin
- Institute of Cardiovascular Research, Key Laboratory for Atherosclerology of Hunan Province, Hunan Province Cooperative Innovation Center for Molecular Target New Drug Discovery, Life Science Research Center, University of South China, Hengyang, Hunan 421001, China.
| | - Chao-Ke Tang
- Institute of Cardiovascular Research, Key Laboratory for Atherosclerology of Hunan Province, Hunan Province Cooperative Innovation Center for Molecular Target New Drug Discovery, Life Science Research Center, University of South China, Hengyang, Hunan 421001, China.
| |
Collapse
|
18
|
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
|
19
|
Chang CL, Torrejon C, Jung UJ, Graf K, Deckelbaum RJ. Incremental replacement of saturated fats by n-3 fatty acids in high-fat, high-cholesterol diets reduces elevated plasma lipid levels and arterial lipoprotein lipase, macrophages and atherosclerosis in LDLR-/- mice. Atherosclerosis 2014; 234:401-9. [PMID: 24747115 DOI: 10.1016/j.atherosclerosis.2014.03.022] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/06/2014] [Revised: 02/28/2014] [Accepted: 03/18/2014] [Indexed: 01/18/2023]
Abstract
OBJECTIVE Effects of progressive substitution of dietary n-3 fatty acids (FA) for saturated FA (SAT) on modulating risk factors for atherosclerosis have not been fully defined. Our previous reports demonstrate that SAT increased, but n-3 FA decreased, arterial lipoprotein lipase (LpL) levels and arterial LDL-cholesterol deposition early in atherogenesis. We now questioned whether incremental increases in dietary n-3 FA can counteract SAT-induced pro-atherogenic effects in atherosclerosis-prone LDL-receptor knockout (LDLR-/-) mice and have identified contributing mechanisms. METHODS AND RESULTS Mice were fed chow or high-fat diets enriched in SAT, n-3, or a combination of both SAT and n-3 in ratios of 3:1 (S:n-3 3:1) or 1:1 (S:n-3 1:1). Each diet resulted in the expected changes in fatty acid composition in blood and aorta for each feeding group. SAT-fed mice became hyperlipidemic. By contrast, n-3 inclusion decreased plasma lipid levels, especially cholesterol. Arterial LpL and macrophage levels were increased over 2-fold in SAT-fed mice but these were decreased with incremental replacement with n-3 FA. n-3 FA partial inclusion markedly decreased expression of pro-inflammatory markers (CD68, IL-6, and VCAM-1) in aorta. SAT diets accelerated advanced atherosclerotic lesion development, whereas all n-3 FA-containing diets markedly slowed atherosclerotic progression. CONCLUSION Mechanisms whereby dietary n-3 FA may improve adverse cardiovascular effects of high-SAT, high-fat diets include improving plasma lipid profiles, increasing amounts of n-3 FA in plasma and the arterial wall. Even low levels of replacement of SAT by n-3 FA effectively reduce arterial lipid deposition by decreasing aortic LpL, macrophages and pro-inflammatory markers.
Collapse
Affiliation(s)
- Chuchun L Chang
- Institute of Human Nutrition, College of Physicians and Surgeons, Columbia University, New York, NY, USA
| | - Claudia Torrejon
- Institute of Human Nutrition, College of Physicians and Surgeons, Columbia University, New York, NY, USA; Campus Norte Hospital Roberto del Río, University of Chile, Santiago, Chile
| | - Un Ju Jung
- Institute of Human Nutrition, College of Physicians and Surgeons, Columbia University, New York, NY, USA
| | - Kristin Graf
- Campus Norte Hospital Roberto del Río, University of Chile, Santiago, Chile
| | - Richard J Deckelbaum
- Institute of Human Nutrition, College of Physicians and Surgeons, Columbia University, New York, NY, USA; Department of Pediatrics, College of Physicians and Surgeons, Columbia University, New York, NY, USA.
| |
Collapse
|
20
|
Afonso MDS, Castilho G, Lavrador MSF, Passarelli M, Nakandakare ER, Lottenberg SA, Lottenberg AM. The impact of dietary fatty acids on macrophage cholesterol homeostasis. J Nutr Biochem 2014; 25:95-103. [DOI: 10.1016/j.jnutbio.2013.10.001] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2013] [Revised: 09/11/2013] [Accepted: 10/03/2013] [Indexed: 11/16/2022]
|
21
|
Brodeur MR, Bouvet C, Barrette M, Moreau P. Palmitic acid increases medial calcification by inducing oxidative stress. J Vasc Res 2013; 50:430-41. [PMID: 24080574 DOI: 10.1159/000354235] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2012] [Accepted: 07/05/2013] [Indexed: 12/12/2022] Open
Abstract
BACKGROUND Aortic medial calcification is a cellular-regulated process leading to arterial stiffness. Although epidemiological studies have suggested an association between the saturation of fatty acids (FA) and arterial stiffness, there is no evidence that saturated FA can induce arterial calcification. This study investigated the capacity of palmitic acid (PA) to induce medial calcification and the signaling pathway(s) implicated in this process. METHODS Rat aortic segments and vascular smooth muscle cells (VSMC) were exposed to calcification medium supplemented with PA. In vivo, rats were treated with warfarin to induce calcification and fed a PA-enriched diet. RESULTS In vitro and ex vivo, palmitate increases calcification and ROS production. Palmitate increases extracellular-signal-regulated kinase (ERK1/2) phosphorylation and osteogenic gene expression. Inhibition of NADPH oxidase with apocynin or an siRNA prevents these effects. ERK1/2 inhibition attenuates the amplification of osteogenic gene expression and calcification induced by palmitate. In vivo, a PA-enriched diet amplified medial calcification and pulse wave velocity (PWV). These effects are mediated by ROS production as indicated by the inhibition of calcification and PWV normalization in rats concomitantly treated with apocynin. CONCLUSION ROS induction by palmitate leads to ERK1/2 phosphorylation and subsequently induces the osteogenic differentiation of VSMC. © 2013 S. Karger AG, Basel.
Collapse
Affiliation(s)
- Mathieu R Brodeur
- Laboratoire de Pharmacologie Vasculaire, Faculté de Pharmacie, Université de Montréal, Montréal, Qué., Canada
| | | | | | | |
Collapse
|
22
|
Abstract
PURPOSE OF REVIEW This article provides an updated review on mechanistic and molecular studies relating to the effects of n-3 fatty acids (FA) on inhibiting atherogenesis. RECENT FINDINGS The effects of n-3 FA on modulating arterial lipoprotein lipase levels link to changes in lipid deposition in the arterial wall. Lipoprotein lipase expression in the arterial wall also relates to local macrophage-mediated inflammatory processes. Increasing evidence suggests that n-3 FA ameliorate inflammation, another key component in the development of atherosclerosis, including decreases in proinflammatory cytokine production. n-3 FA inhibit atherogenic signaling pathways and modulate the phenotypes of inflammatory leukocytes and their recruitment in the arterial wall. SUMMARY New mechanistic insights into the antiatherogenic action of n-3 FA have emerged. These studies may contribute to future therapeutic advances in preventing mortality and morbidity associated with atherosclerosis.
Collapse
Affiliation(s)
- Chuchun L Chang
- Institute of Human Nutrition and Department of Pediatrics, College of Physicians and Surgeons, Columbia University, New York, New York 10032, USA
| | | |
Collapse
|
23
|
Araujo M, Santos T, Lourenço M, Takahira R, Machado L, Carvalho L. Avaliação de colesterol e triglicerídeos séricos em cães saudáveis suplementados com ômega n-3. ARQ BRAS MED VET ZOO 2012. [DOI: 10.1590/s0102-09352012000600013] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
A análise da concentração sérica de colesterol e triglicerídeos foi realizada em 20 cães, sem raça definida, saudáveis, 10 machos e 10 fêmeas, previamente e após a suplementação por 30 dias com ácidos graxos poli-insaturados de cadeia longa derivados do ômega n-3 (497mg ácido docosa-hexaenoico e 780mg ácido eicosapentanoico). A concentração sérica de colesterol apresentou redução significativa após a suplementação em ambos os sexos (271,6±79,8mg/dL; 236,2±67,6mg/dL, antes e após suplementação, respectivamente). Em relação à concentração sérica de triglicerídeos, houve redução apenas nas fêmeas (57,8±12,1mg/dL; 45,2±7,8mg/dL, antes e após suplementação, respectivamente), não havendo efeito da suplementação nos machos.
Collapse
|
24
|
Nyrén R, Chang CL, Lindström P, Barmina A, Vorrsjö E, Ali Y, Juntti-Berggren L, Bensadoun A, Young SG, Olivecrona T, Olivecrona G. Localization of lipoprotein lipase and GPIHBP1 in mouse pancreas: effects of diet and leptin deficiency. BMC PHYSIOLOGY 2012. [PMID: 23186339 PMCID: PMC3537605 DOI: 10.1186/1472-6793-12-14] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
BACKGROUND Lipoprotein lipase (LPL) hydrolyzes triglycerides in plasma lipoproteins and enables uptake of lipolysis products for energy production or storage in tissues. Our aim was to study the localization of LPL and its endothelial anchoring protein glycosylphosphatidylinositol-anchored high density lipoprotein-binding protein 1 (GPIHBP1) in mouse pancreas, and effects of diet and leptin deficiency on their expression patterns. For this, immunofluorescence microscopy was used on pancreatic tissue from C57BL/6 mouse embryos (E18), adult mice on normal or high-fat diet, and adult ob/ob-mice treated or not with leptin. The distribution of LPL and GPIHBP1 was compared to insulin, glucagon and CD31. Heparin injections were used to discriminate between intracellular and extracellular LPL. RESULTS In the exocrine pancreas LPL was found in capillaries, and was mostly co-localized with GPIHBP1. LPL was releasable by heparin, indicating localization on cell surfaces. Within the islets, most of the LPL was associated with beta cells and could not be released by heparin, indicating that the enzyme remained mostly within cells. Staining for LPL was found also in the glucagon-producing alpha cells, both in embryos (E18) and in adult mice. Only small amounts of LPL were found together with GPIHBP1 within the capillaries of islets. Neither a high fat diet nor fasting/re-feeding markedly altered the distribution pattern of LPL or GPIHBP1 in mouse pancreas. Islets from ob/ob mice appeared completely deficient of LPL in the beta cells, while LPL-staining was normal in alpha cells and in the exocrine pancreas. Leptin treatment of ob/ob mice for 12 days reversed this pattern, so that most of the islets expressed LPL in beta cells. CONCLUSIONS We conclude that both LPL and GPIHBP1 are present in mouse pancreas, and that LPL expression in beta cells is dependent on leptin.
Collapse
Affiliation(s)
- Rakel Nyrén
- Department of Medical Biosciences/Physiological Chemistry, Umeå University, Umeå, Sweden
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
25
|
Jung UJ, Torrejon C, Chang CL, Hamai H, Worgall TS, Deckelbaum RJ. Fatty acids regulate endothelial lipase and inflammatory markers in macrophages and in mouse aorta: a role for PPARγ. Arterioscler Thromb Vasc Biol 2012; 32:2929-37. [PMID: 23042820 DOI: 10.1161/atvbaha.112.300188] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
OBJECTIVE Macrophage endothelial lipase (EL) is associated with increased atherosclerosis and inflammation. Because of their anti-inflammatory properties we hypothesized that n-3 fatty acids, in contrast to saturated fatty acids, would lower macrophages and arterial EL and inflammatory markers. METHODS AND RESULTS Murine J774 and peritoneal macrophages were incubated with eicosapentaenoic acid or palmitic acid in the presence or absence of lipopolysaccaride (LPS). LPS increased EL mRNA and protein. Palmitic acid alone or with LPS dose-dependently increased EL mRNA and protein. In contrast, eicosapentaenoic acid dose-dependently abrogated effects of LPS or palmitic acid on increasing EL expression. EL expression closely linked to peroxisome proliferator activated receptor (PPAR)γ expression. Eicosapentaenoic acid blocked rosiglitazone (a PPARγ agonist)-mediated EL activation and GW9662 (a PPARγ antagonist)-blocked palmitic acid-mediated EL stimulation. Eicosapentaenoic acid alone or with LPS blunted LPS-mediated stimulation of macrophage proinflammatory interleukin-6, interleukin-12p40, and toll-like receptor-4 mRNA and increased anti-inflammatory interleukin-10 and mannose receptor mRNA. In vivo studies in low density lipoprotein receptor knockout mice showed that high saturated fat rich diets, but not n-3 diets, increased arterial EL, PPARγ, and proinflammatory cytokine mRNA. CONCLUSIONS n-3 fatty acids, in contrast to saturated fatty acids, decrease EL in parallel with modulating pro- and anti-inflammatory markers, and these effects on EL link to PPARγ.
Collapse
Affiliation(s)
- Un Ju Jung
- Institute of Human Nutrition, Columbia University, New York, NY 10032, USA
| | | | | | | | | | | |
Collapse
|
26
|
Abstract
Severe hypertriglyceridemia is associated with acute pancreatitis and can be a manifestation of lipoprotein lipase (LPL) deficiency. It is associated with a spectrum of disorders, ranging from heterozygous LPL deficiency allied with environmental factors to rare severe cases of homozygous LPL deficiency. The genes associated with reduced LPL activity include LPL, its cofactor apoC-2, a controlling protein apoA-5 and the LPL receptor GPI-HBP1. The effects of mutations are exacerbated by environmental factors such as diet, pregnancy and insulin resistance. Treatment of clinical LPL deficiency is by ultra-low-fat diet along with the use of fibrates, omega-3 fatty acids, niacin, statins and insulin-sensitizing therapies, depending on the extent of residual LPL activity. Novel therapies that target lipoprotein particle assembly through the antisense oligonucleotides or by interference with triglyceride-loading microsomal transport protein inhibitors offer new potential options for treating hypertriglyceridemia.
Collapse
Affiliation(s)
- Adie Viljoen
- Lister Hospital, Stevenage, Hertfordshire, SG1 4AB, UK
| | | |
Collapse
|
27
|
Annuzzi G, Rivellese AA, Wang H, Patti L, Vaccaro O, Riccardi G, Ebbesson SO, Comuzzie AG, Umans JG, Howard BV. Lipoprotein subfractions and dietary intake of n-3 fatty acid: the Genetics of Coronary Artery Disease in Alaska Natives study. Am J Clin Nutr 2012; 95:1315-22. [PMID: 22572646 PMCID: PMC3349453 DOI: 10.3945/ajcn.111.023887] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/14/2022] Open
Abstract
BACKGROUND Few studies have compared lipoprotein composition with dietary intake. OBJECTIVE The lipoprotein subfraction profile was evaluated in relation to diet in Alaska Eskimos at high cardiovascular risk but with a low frequency of hyperlipidemia and high intake of n-3 (omega-3) fatty acids. DESIGN A population-based sample (n = 1214) from the Norton Sound Region of Alaska underwent a physical examination and blood sampling. Analyses were from 977 individuals who did not have diabetes or use lipid-lowering medications and had complete dietary information (food-frequency questionnaire) and a lipoprotein subfraction profile (nuclear magnetic resonance spectroscopy). RESULTS After adjustment for age, BMI, total energy intake, and percentage of energy from fat, the intake of n-3 fatty acids was significantly associated with fewer large VLDLs (P = 0.022 in women, P = 0.064 in men), a smaller VLDL size (P = 0.018 and P = 0.036), more large HDLs (P = 0.179 and P = 0.021), and a larger HDL size (P = 0.004 and P = 0.001). After adjustment for carbohydrate and sugar intakes, large VLDLs (P = 0.042 and 0.018) and VLDL size (P = 0.011 and 0.025) remained negatively associated with n-3 fatty acid intake in women and men, and large HDLs (P = 0.067 and 0.005) and HDL size (P = 0.001 in both) remained positively associated with n-3 fatty acid intake in women and men. In addition, large LDLs (P = 0.040 and P = 0.025) were positively associated in both sexes, and LDL size (P = 0.006) showed a positive association in women. There were no significant relations with total LDL particles in either model. CONCLUSIONS Dietary n-3 fatty acids, independent of the reciprocal changes in carbohydrate and sugar intakes, are associated with an overall favorable lipoprotein profile in terms of cardiovascular risk. Because there are no relations with total LDL particles, the benefit may be related to cardiovascular processes other than atherosclerosis.
Collapse
Affiliation(s)
- Giovanni Annuzzi
- Department of Clinical and Experimental Medicine, Federico II University, Naples, Italy
| | | | | | | | | | | | | | | | | | | |
Collapse
|
28
|
Deckelbaum RJ, Torrejon C. The omega-3 fatty acid nutritional landscape: health benefits and sources. J Nutr 2012; 142:587S-591S. [PMID: 22323763 PMCID: PMC3278270 DOI: 10.3945/jn.111.148080] [Citation(s) in RCA: 111] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023] Open
Abstract
Dietary fatty acids (FA) are increasingly recognized as major biologic regulators and have properties that relate to health outcomes and disease. The longer chain, more bioactive (n-6) (or omega-6) FA and (n-3) (or omega-3) FA share similar elongation and desaturation enzymes in their conversion from the essential (n-6) FA, linoleic acid, and (n-3) FA, α-linolenic acid (ALA). Conversion from these essential FA is very inefficient. However, now for the (n-3) FA series, soy oil can be enriched with (n-3) stearidonic acid (SDA) to allow for much more efficient conversion to longer chain EPA. EPA and the longer chain DHA possess distinct physical and biological properties that generally impart properties to cells and tissue, which underlie their ability to promote health and prevent disease. Although active in a number of areas of human biology, mechanisms of action of EPA and DHA are perhaps best defined in cardiovascular disease. There is concern that to reach the intake recommendations of EPA and DHA, their supply from cold water fish will be insufficient. Gaps in understanding mechanisms of action of (n-3) FA in a number of health and disease areas as well as optimal sources and intake levels for each need to be defined by further research. Because of the inefficient conversion of ALA, the appearance of SDA in enriched soy oil offers a biologically effective and cost effective approach to providing a sustainable plant source for (n-3) FA in the future.
Collapse
Affiliation(s)
- Richard J. Deckelbaum
- Institute of Human Nutrition, Department of Pediatrics, Columbia University Medical Center, New York, NY 10032; and Institute of Nutrition and Food Technology, University of Chile, Santiago, Chile,To whom correspondence should be addressed. E-mail:
| | - Claudia Torrejon
- Institute of Human Nutrition, Department of Pediatrics, Columbia University Medical Center, New York, NY 10032; and Institute of Nutrition and Food Technology, University of Chile, Santiago, Chile
| |
Collapse
|
29
|
Ahn J, Lee H, Chung CH, Ha T. High fat diet induced downregulation of microRNA-467b increased lipoprotein lipase in hepatic steatosis. Biochem Biophys Res Commun 2011; 414:664-9. [PMID: 21986524 DOI: 10.1016/j.bbrc.2011.09.120] [Citation(s) in RCA: 51] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2011] [Accepted: 09/22/2011] [Indexed: 02/08/2023]
Abstract
Non-alcoholic fatty liver disease (NAFLD) is characterized by hepatic fat accumulation and is presently the most common chronic liver disease. However, the mechanisms underlying the development of steatosis remain unclear. MicroRNAs (miRNAs) are small non-coding RNAs that modulate a variety of biological functions. We have investigated the role of miRNA in the development of steatosis. We found that miR-467b expression is significantly downregulated in liver tissues of high-fat diet fed mice and in steatosis-induced hepatocytes. The downregulation of miR-467b resulted in the upregulation of hepatic lipoprotein lipase (LPL), the direct target of miR-467b. Moreover, the interaction between miR-467b and LPL was associated with insulin resistance, a major cause of NAFLD. These results suggest that downregulation of miR-467b is involved in the development of hepatic steatosis by modulating the expression of its target, LPL.
Collapse
Affiliation(s)
- Jiyun Ahn
- Functional Food Research Center, Research Division for Emerging Innovative Technology, Korea Food Research Institute, Seoungnam-si, Gyeonggi-do 463-746, South Korea
| | | | | | | |
Collapse
|
30
|
n-3 fatty acids ameliorate hepatic steatosis and dysfunction after LXR agonist ingestion in mice. Biochim Biophys Acta Mol Cell Biol Lipids 2011; 1811:491-7. [PMID: 21704188 DOI: 10.1016/j.bbalip.2011.06.003] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2011] [Revised: 05/16/2011] [Accepted: 06/01/2011] [Indexed: 01/10/2023]
Abstract
UNLABELLED Liver X receptor (LXR) agonists slow atherogenesis, but cause hepatic steatosis and dysfunction in part by increasing expression of sterol regulatory element binding protein 1-c (SREBP1-c), a transcription factor that upregulates fatty acid (FA) synthesis. n-3 FAs decrease hepatic FA synthesis by down-regulating SREBP1-c. To test the hypothesis that n-3 FAs decrease hepatic steatosis in mice given LXR agonist, C57BL/6 mice received daily gavage of an LXR agonist T0901317 (LXR(T)) or vehicle for 4weeks with concomitant intakes chow or high-fat diets enriched in saturated fat (SAT) or n-3 fat (n-3). Mice on LXR(T) and SAT developed hepatomegaly with a large increase in size and number of hepatic lipid droplets; an n-3 diet reduced liver weight/body weight with decreased hepatic steatosis and triglyceride levels. Effects of n-3 diet on hepatic lipogenesis were linked to a blunting of LXR(T) upregulation of hepatic SREBP1-c and FA synthase mRNA. n-3 diets also normalized LXR(T)-mediated increases of plasma ALT and AST levels, whereas SAT diet increased these markers. CONCLUSION These studies suggest that n-3 FAs when given together with LXR agonists have the potential to improve both hepatic steatosis and hepatotoxicity in humans that might receive LXR agonists to decrease risk of atherosclerosis.
Collapse
|
31
|
Vijay-Kumar M, Vanegas SM, Patel N, Aitken JD, Ziegler TR, Ganji V. Fish oil rich diet in comparison to saturated fat rich diet offered protection against lipopolysaccharide-induced inflammation and insulin resistance in mice. Nutr Metab (Lond) 2011; 8:16. [PMID: 21388548 PMCID: PMC3066121 DOI: 10.1186/1743-7075-8-16] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2010] [Accepted: 03/09/2011] [Indexed: 01/22/2023] Open
Abstract
Background and Objective Systemic chronic inflammation is linked to metabolic syndrome, type-2 diabetes, and heart disease. Lipopolysaccharide (LPS), a Gram negative microbial product, triggers inflammation through toll-like-receptor-4 (TLR-4) signaling. It has been reported that dietary fatty acids also modulate inflammation through TLR-4. We investigated whether fish oil (FO) rich diet in comparison to saturated fat (SF) rich diet would confer protection from pathologies induced by LPS. Methods Twenty C57BL/6 mice were divided into two groups. One group received FO-diet and other received SF-diet ad libitum for 60 days. Diets were isocaloric containing 45% energy from fat. After 60-days of feeding, blood was collected after overnight fast. Mice were allowed to recover for 4-days, fasted for 5-hours, challenged with 100 ng/mL of LPS intraperitonially, and bled after 2-hours. After 7-days of recuperation, mice were challenged with 500 ng/mL of LPS intraperitonially and observed for physical health. Results Food intake was similar in FO- and SF-fed mice. FO-fed mice compared to SF-fed mice had significantly less body weight gain (P = 0.005), epididymal fat weight (P = 0.005), fasting blood glucose (70.8 vs 83.3 ng/dL; P < 0.05), HOMA-IR (5.0 vs 13.6; P < 0.019), and serum cholesterol (167 vs 94 mg/dL; P < 0.05). When challenged with LPS, FO-fed mice had significantly lower serum IL-1β compared to SF-fed mice (2.0 vs 30.0 pg/mL; P < 0.001). After LPS-challenge, SF-fed mice had higher mortality, lost more body weight, and had greater decrease in blood glucose compared to FO-fed mice. Conclusion Overall, FO-diet compared to SF-diet offered protection against deleterious effects of LPS in mice.
Collapse
Affiliation(s)
- Matam Vijay-Kumar
- Division of Nutrition, School of Health Professions, College of Health and Human Sciences, Georgia State University, Atlanta, GA 30302, USA.
| | | | | | | | | | | |
Collapse
|
32
|
Ooi LG, Ahmad R, Yuen KH, Liong MT. Lactobacillus gasseri [corrected] CHO-220 and inulin reduced plasma total cholesterol and low-density lipoprotein cholesterol via alteration of lipid transporters. J Dairy Sci 2011; 93:5048-58. [PMID: 20965319 DOI: 10.3168/jds.2010-3311] [Citation(s) in RCA: 60] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2010] [Accepted: 07/18/2010] [Indexed: 12/13/2022]
Abstract
This randomized, double-blind, placebo-controlled, and parallel-designed study was conducted to investigate the effect of a synbiotic product containing Lactobacillus gasseri [corrected] CHO-220 and inulin on lipid profiles of hypercholesterolemic men and women. Thirty-two hypercholesterolemic men and women with initial mean plasma cholesterol levels of 5.7±0.32 mmol/L were recruited for the 12-wk study. The subjects were randomly allocated to 2 groups; namely the treatment group (synbiotic product) and the control group (placebo), and each received 4 capsules of synbiotic or placebo daily. Our results showed that the mean body weight, energy, and nutrient intake of the subjects did not differ between the 2 groups over the study period. The supplementation of synbiotic reduced plasma total cholesterol and low-density lipoprotein (LDL)-cholesterol by 7.84 and 9.27%, respectively, compared with the control over 12 wk. Lipoproteins were subsequently subfractionated and characterized. The synbiotic supplementation resulted in a lower concentration of triglycerides in the very low, intermediate, low, and high-density lipoprotein particles compared with the control over 12 wk. The concentration of triglycerides in lipoproteins is positively correlated with an increased risk of atherosclerosis. Our results showed that the synbiotic might exhibit an atheropreventive characteristic. Cholesteryl ester (CE) in the high-density lipoprotein particles of the synbiotic group was also higher compared with the control, indicating greater transport of cholesterol in the form of CE to the liver for hydrolysis. This may have led to the reduced plasma total cholesterol level of the synbiotic group. The supplementation of synbiotic also reduced the concentration of CE in the LDL particles compared with the control, leading to the formation of smaller and denser particles that are more easily removed from blood. This supported the reduced LDL-cholesterol level of the synbiotic group compared with the control. Our present study showed that the synbiotic product improved plasma total- and LDL-cholesterol levels by modifying the interconnected pathways of lipid transporters. In addition, although Lactobacillus gasseri [corrected] CHO-220 could deconjugate bile, our results showed a statistically insignificant difference in the levels of conjugated, deconjugated, primary, and secondary bile acids between the synbiotic and control groups over 12 wk, indicating safety from bile-related toxicity.
Collapse
Affiliation(s)
- L-G Ooi
- School of Industrial Technology, Universiti Sains Malaysia, Penang, Malaysia
| | | | | | | |
Collapse
|
33
|
Deckelbaum RJ. n-6 and n-3 Fatty acids and atherosclerosis: ratios or amounts? Arterioscler Thromb Vasc Biol 2010; 30:2325-6. [PMID: 21084701 DOI: 10.1161/atvbaha.110.214353] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
|
34
|
Chang CL, Seo T, Du CB, Accili D, Deckelbaum RJ. n-3 Fatty acids decrease arterial low-density lipoprotein cholesterol delivery and lipoprotein lipase levels in insulin-resistant mice. Arterioscler Thromb Vasc Biol 2010; 30:2510-7. [PMID: 20930167 DOI: 10.1161/atvbaha.110.215848] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/29/2023]
Abstract
OBJECTIVE To determine whether n-3 fatty acids (n-3) influence arterial cholesterol delivery and lipoprotein lipase (LpL) levels in insulin-resistant mice. METHODS AND RESULTS Insulin resistance contributes to risk of cardiovascular disease. It was previously reported that saturated fat (SAT) diets increased, but n-3 diets decreased, arterial low-density lipoprotein (LDL) cholesterol deposition from LDL total and selective uptake; this was associated with increased or decreased arterial LpL, respectively. Insulin receptor transgenic knockout mice (L1) were fed a chow, SAT, or n-3 diet for 12 weeks. Double-fluorescent boron dipyrromethene (BODIPY)-cholesteryl ester (CE) and Alexa dye-labeled human LDL were injected to separately trace LDL-CE and LDL-apolipoprotein B whole particle uptake. In contrast to SAT, n-3 diets markedly reduced all plasma lipids, ameliorating progression of insulin resistance. As opposed to SAT, n-3 reduced arterial LDL uptake, CE deposition, and selective uptake. Disparate patterns of CE deposition between diets were comparable with arterial LpL distribution; SAT induced high LpL levels throughout aortic media; LpL was limited only to intima in n-3-fed mice. CONCLUSIONS n-3 diets diminish arterial LDL-cholesterol deposition in mice with insulin resistance, and this is associated with changes in arterial LpL levels and distribution.
Collapse
Affiliation(s)
- Chuchun L Chang
- Institute of Human Nutrition, College of Physicians and Surgeons, Columbia University, New York, NY 10032, USA
| | | | | | | | | |
Collapse
|
35
|
Abstract
PURPOSE OF REVIEW There are strong epidemiologic connections between plasma triglycerides and atherosclerosis. We will consider to what extent this goes back to derangements of the lipoprotein lipase (LPL) system. The roles of hepatic lipase and endothelial lipase will also be touched upon. RECENT FINDINGS Understanding of LPL action has taken major steps with the discovery of lipase maturation factor 1 as a specific endoplasmic reticulum chaperon needed for proper folding of the lipases, glycosylphosphatidylinositol-anchored HDL-binding protein 1 as an endothelial cell protein needed for transport and binding of LPL and some angiopoietin-like proteins that can modulate LPL activity. Studies of genetic variants continue to support the important roles of the lipases in lipoprotein metabolism and in atherosclerosis. CONCLUSION There are several ways by which derangement of the lipases may contribute to atherogenesis. Lipase actions are major determinants of plasma lipoprotein patterns. LPL activity must be modulated in relation to the physiological situation (feeding, fasting, exercise, etc.). Fatty acids and monoglycerides generated must be efficiently removed so that they do not endanger the integrity of the endothelium, cause lipotoxic reactions or both. In addition, the lipases may cause binding and endocytosis of lipoprotein particles in the artery wall.
Collapse
Affiliation(s)
- Gunilla Olivecrona
- Department of Medical Biosciences, Section on Physiological Chemistry, Umeå University, Umeå, Sweden.
| | | |
Collapse
|
36
|
Sudheendran S, Chang CC, Deckelbaum RJ. N-3 vs. saturated fatty acids: effects on the arterial wall. Prostaglandins Leukot Essent Fatty Acids 2010; 82:205-9. [PMID: 20207121 PMCID: PMC2878127 DOI: 10.1016/j.plefa.2010.02.020] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Cardiovascular disease is a leading cause of death worldwide. Atherosclerosis and unstable plaques are underlying causes for cardiovascular diseases. Cardiovascular disease is associated with consumption of diets high in saturated fats. In contrast there is increasing evidence that higher intakes of dietary n-3 fatty acids decrease risk for cardiovascular disease. Recent studies are beginning to clarify how n-3 compared with saturated fatty acids influence cardiovascular disease risk via pathways in the arterial wall. In this paper we will review studies that report on mechanisms whereby dietary fatty acids affect atherosclerosis through modulation of arterial wall lipid deposition, inflammation, cell proliferation, and plaque vulnerability.
Collapse
Affiliation(s)
- S Sudheendran
- Department of Pediatrics, College of Physicians and Surgeons, Columbia University, New York, NY 10032, USA
| | | | | |
Collapse
|