1
|
Yoshimura T, Okamura T, Yuge H, Hosomi Y, Kimura T, Ushigome E, Nakanishi N, Sasano R, Ogata T, Hamaguchi M, Fukui M. Gut dysbiosis induced by a high-salt diet aggravates atherosclerosis by increasing the absorption of saturated fatty acids in ApoE-deficient mice. J Clin Biochem Nutr 2025; 76:210-220. [PMID: 40151404 PMCID: PMC11936735 DOI: 10.3164/jcbn.24-163] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2024] [Accepted: 09/27/2024] [Indexed: 03/29/2025] Open
Abstract
Excessive salt intake has been associated with gut dysbiosis and increased cardiovascular risk. This study investigates the role of gut dysbiosis induced by a high-salt diet in the progression of atherosclerosis in ApoE-deficient mice. Sixteen-week-old male ApoE-deficient mice were fed either a high-fat, high-sucrose diet or high-fat, high-sucrose diet supplemented with 4% NaCl for eight weeks. The group on the HFHSD with high salt showed significant progression of atherosclerosis compared to the high-fat, high-sucrose diet group. Analysis of the gut microbiota revealed reduced abundance of beneficial bacteria such as Allobaculum spp., Lachnospiraceae, and Alphaproteobacteria in the high-salt group. Additionally, this group exhibited increased expression of the Cd36 gene, a transporter of long-chain fatty acids, in the small intestine. Serum and aortic levels of saturated fatty acids, known contributors to atherosclerosis, were markedly elevated in the high-salt group. These findings suggest that a high-salt diet exacerbates atherosclerosis by altering gut microbiota and increasing the absorption of saturated fatty acids through upregulation of intestinal fatty acid transporters. This study provides new insights into how dietary salt can influence cardiovascular health through its effects on the gut microbiome and lipid metabolism.
Collapse
Affiliation(s)
- Takashi Yoshimura
- Department of Endocrinology and Metabolism, Kyoto Prefectural University of Medicine, Graduate School of Medical Science, 465 Kajii-cho, Kamigyo-ku, Kyoto 602-8566, Japan
| | - Takuro Okamura
- Department of Endocrinology and Metabolism, Kyoto Prefectural University of Medicine, Graduate School of Medical Science, 465 Kajii-cho, Kamigyo-ku, Kyoto 602-8566, Japan
| | - Hiroki Yuge
- Department of Endocrinology and Metabolism, Kyoto Prefectural University of Medicine, Graduate School of Medical Science, 465 Kajii-cho, Kamigyo-ku, Kyoto 602-8566, Japan
| | - Yukako Hosomi
- Department of Endocrinology and Metabolism, Kyoto Prefectural University of Medicine, Graduate School of Medical Science, 465 Kajii-cho, Kamigyo-ku, Kyoto 602-8566, Japan
| | - Tomonori Kimura
- Department of Endocrinology and Metabolism, Kyoto Prefectural University of Medicine, Graduate School of Medical Science, 465 Kajii-cho, Kamigyo-ku, Kyoto 602-8566, Japan
| | - Emi Ushigome
- Department of Endocrinology and Metabolism, Kyoto Prefectural University of Medicine, Graduate School of Medical Science, 465 Kajii-cho, Kamigyo-ku, Kyoto 602-8566, Japan
| | - Naoko Nakanishi
- Department of Endocrinology and Metabolism, Kyoto Prefectural University of Medicine, Graduate School of Medical Science, 465 Kajii-cho, Kamigyo-ku, Kyoto 602-8566, Japan
| | | | - Takehiro Ogata
- Department of Pathology and Cell Regulation, Kyoto Prefectural University of Medicine, Graduate School of Medical Science, 465 Kajii-cho, Kamigyo-ku, Kyoto 602-8566, Japan
| | - Masahide Hamaguchi
- Department of Endocrinology and Metabolism, Kyoto Prefectural University of Medicine, Graduate School of Medical Science, 465 Kajii-cho, Kamigyo-ku, Kyoto 602-8566, Japan
| | - Michiaki Fukui
- Department of Endocrinology and Metabolism, Kyoto Prefectural University of Medicine, Graduate School of Medical Science, 465 Kajii-cho, Kamigyo-ku, Kyoto 602-8566, Japan
| |
Collapse
|
2
|
Benaiges D, Goday A, Casajoana A, Flores-Le Roux JA, Fitó M, Pozo OJ, Serra C, Pera M, Llauradó G, Climent E, Villatoro M, Lazaro I, Castañer O, Pedro-Botet J. Short-term effects of gastric bypass versus sleeve gastrectomy on high LDL cholesterol: The BASALTO randomized clinical trial. Cardiovasc Diabetol 2024; 23:205. [PMID: 38879559 PMCID: PMC11180388 DOI: 10.1186/s12933-024-02296-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/26/2024] [Accepted: 06/04/2024] [Indexed: 06/19/2024] Open
Abstract
BACKGROUND There has been a substantial increase in the use of laparoscopic sleeve gastrectomy (SG) to treat morbid obesity despite observational evidence demonstrating the superiority of Roux-en-Y gastric bypass (RYGB) for reducing low-density lipoprotein (LDL) cholesterol. The main aim was to ascertain whether high LDL cholesterol levels should be considered when selecting the most appropriate surgical procedure for each patient (RYGB or SG). METHODS In this single-center, randomized clinical trial using intention-to-treat analysis, 38 patients with severe obesity and elevated levels of LDL cholesterol were randomly assigned to undergo RYGB or SG. The primary outcome was LDL cholesterol remission at 12 months, defined as LDL cholesterol < 3.36 nmol/l without lipid-lowering medications. Secondary outcomes included changes in weight, other comorbidities, qualitative lipoprotein traits, cholesterol esters, glycoproteins, cholesterol absorption and synthesis metabolites and complications. RESULTS Intention-to-treat analysis revealed that LDL cholesterol remission occurred in 66.6% of RYGB patients compared to 27.8% of SG patients (p = 0.019). Among patients completing follow-up, RYGB demonstrated superior remission (80.0% vs. 29.4%, p = 0.005). Exclusive benefits of RYGB included a reduction in large, medium, and small LDL particles. Cholesterol absorption markers showed differential behavior after both techniques: campesterol (Δ -15.2 µg/mg, 95% CI -30.2 to -0.1) decreased after RYGB, and sitosterol (Δ 21.1 µg/mg, 95% CI 0.9 to 41.2), cholestanol (Δ 30.6 µg/mg, 95% CI 14.8 to 57.9) and campesterol (Δ 18.4 µg/mg, 95% CI 4.4 to 32.3) increased after SG. No differences in weight loss, cholesterol esters, glycoproteins, cholesterol synthesis metabolites or postoperative complications were observed between techniques. CONCLUSION In conclusion, RYGB is superior to SG in terms of short-term of high LDL cholesterol remission. Furthermore, RYGB also led to a greater improvement in lipoprotein parameters that confer an atherogenic profile. Therefore, the presence of elevated levels of LDL cholesterol should be considered when determining the optimal bariatric surgery procedure for each patient. TRIAL REGISTRATION Clinicaltrials.gov number, NCT03975478).
Collapse
Affiliation(s)
- David Benaiges
- Department of Endocrinology and Nutrition, Hospital del Mar, Passeig Marítim, 25-29, Barcelona, 08003, Spain.
- Department of Medicine, Universitat Pompeu Fabra, Plaça de la Mercè, 10-12, Barcelona, 08002, Spain.
- Unit of Cardiovascular Risk and Nutrition, Institut Hospital del Mar d'Investigacions Mèdiques (IMIM), Dr. Aiguader, 80, Barcelona, 08003, Spain.
- Consorci Sanitari de l'Alt Penedès i Garraf, Vilafranca del Penedès, Spain.
- CiberOBN. Instituto de Salud Carlos III, Avenida Monforte de Lemos, 3-5. Pabellón 11. Planta 0, Madrid, 28029, Spain.
| | - Albert Goday
- Department of Endocrinology and Nutrition, Hospital del Mar, Passeig Marítim, 25-29, Barcelona, 08003, Spain
- Unit of Cardiovascular Risk and Nutrition, Institut Hospital del Mar d'Investigacions Mèdiques (IMIM), Dr. Aiguader, 80, Barcelona, 08003, Spain
- CiberOBN. Instituto de Salud Carlos III, Avenida Monforte de Lemos, 3-5. Pabellón 11. Planta 0, Madrid, 28029, Spain
- Department of Medicine, Universitat Autònoma de Barcelona. Plaça Cívica, Bellaterra, Barcelona, 08193, Spain
| | - Anna Casajoana
- Unit of Cardiovascular Risk and Nutrition, Institut Hospital del Mar d'Investigacions Mèdiques (IMIM), Dr. Aiguader, 80, Barcelona, 08003, Spain
- Esophago-Gastric and Bariatric Surgery Unit, Department of Surgery, Hospital del Mar, Passeig Marítim, 25-29, Barcelona, 08003, Spain
| | - Juana A Flores-Le Roux
- Department of Endocrinology and Nutrition, Hospital del Mar, Passeig Marítim, 25-29, Barcelona, 08003, Spain
- Department of Medicine, Universitat Pompeu Fabra, Plaça de la Mercè, 10-12, Barcelona, 08002, Spain
- Unit of Cardiovascular Risk and Nutrition, Institut Hospital del Mar d'Investigacions Mèdiques (IMIM), Dr. Aiguader, 80, Barcelona, 08003, Spain
| | - Montserrat Fitó
- Unit of Cardiovascular Risk and Nutrition, Institut Hospital del Mar d'Investigacions Mèdiques (IMIM), Dr. Aiguader, 80, Barcelona, 08003, Spain
- CiberOBN. Instituto de Salud Carlos III, Avenida Monforte de Lemos, 3-5. Pabellón 11. Planta 0, Madrid, 28029, Spain
| | - Oscar J Pozo
- Applied Metabolomics Research Group, Neurosciences Research Program, IMIM (Hospital del Mar Research Institute), Dr. Aiguader 88, 08003, Barcelona, Spain
| | - Carme Serra
- Department of Endocrinology and Nutrition, Hospital del Mar, Passeig Marítim, 25-29, Barcelona, 08003, Spain
| | - Manuel Pera
- Esophago-Gastric and Bariatric Surgery Unit, Department of Surgery, Hospital del Mar, Passeig Marítim, 25-29, Barcelona, 08003, Spain
| | - Gemma Llauradó
- Department of Endocrinology and Nutrition, Hospital del Mar, Passeig Marítim, 25-29, Barcelona, 08003, Spain
- Department of Medicine, Universitat Pompeu Fabra, Plaça de la Mercè, 10-12, Barcelona, 08002, Spain
- Unit of Cardiovascular Risk and Nutrition, Institut Hospital del Mar d'Investigacions Mèdiques (IMIM), Dr. Aiguader, 80, Barcelona, 08003, Spain
| | - Elisenda Climent
- Department of Endocrinology and Nutrition, Hospital del Mar, Passeig Marítim, 25-29, Barcelona, 08003, Spain
- Department of Medicine, Universitat Pompeu Fabra, Plaça de la Mercè, 10-12, Barcelona, 08002, Spain
- Unit of Cardiovascular Risk and Nutrition, Institut Hospital del Mar d'Investigacions Mèdiques (IMIM), Dr. Aiguader, 80, Barcelona, 08003, Spain
| | - Montserrat Villatoro
- Department of Endocrinology and Nutrition, Hospital del Mar, Passeig Marítim, 25-29, Barcelona, 08003, Spain
| | - Iolanda Lazaro
- Unit of Cardiovascular Risk and Nutrition, Institut Hospital del Mar d'Investigacions Mèdiques (IMIM), Dr. Aiguader, 80, Barcelona, 08003, Spain
- CiberOBN. Instituto de Salud Carlos III, Avenida Monforte de Lemos, 3-5. Pabellón 11. Planta 0, Madrid, 28029, Spain
| | - Olga Castañer
- Unit of Cardiovascular Risk and Nutrition, Institut Hospital del Mar d'Investigacions Mèdiques (IMIM), Dr. Aiguader, 80, Barcelona, 08003, Spain
- CiberOBN. Instituto de Salud Carlos III, Avenida Monforte de Lemos, 3-5. Pabellón 11. Planta 0, Madrid, 28029, Spain
- Ciber Epidemiología y Salud Pública (CiberESP), Instituto de Salud Carlos III, Avenida Monforte de Lemos, 3-5. Pabellón 11. Planta 0, Madrid, 28029, Spain
| | - Juan Pedro-Botet
- Department of Endocrinology and Nutrition, Hospital del Mar, Passeig Marítim, 25-29, Barcelona, 08003, Spain
- Unit of Cardiovascular Risk and Nutrition, Institut Hospital del Mar d'Investigacions Mèdiques (IMIM), Dr. Aiguader, 80, Barcelona, 08003, Spain
- Department of Medicine, Universitat Autònoma de Barcelona. Plaça Cívica, Bellaterra, Barcelona, 08193, Spain
| |
Collapse
|
3
|
Lomonosova A, Gognieva D, Suvorov A, Silantyev A, Abasheva A, Vasina Y, Abdullaev M, Nartova A, Eroshchenko N, Kazakova V, Komarov R, Dzyundzya A, Danilova E, Shchekochikhin D, Kopylov P. The Blood Plasma Lipidomic Profile in Atherosclerosis of the Brachiocephalic Arteries. Biomedicines 2024; 12:1279. [PMID: 38927486 PMCID: PMC11201046 DOI: 10.3390/biomedicines12061279] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2024] [Revised: 06/04/2024] [Accepted: 06/07/2024] [Indexed: 06/28/2024] Open
Abstract
According to the World Health Organization, ischemic stroke is the second leading cause of death in the world. Frequently, it is caused by brachiocephalic artery (BCA) atherosclerosis. Timely detection of atherosclerosis and its unstable course can allow for a timely response to potentially dangerous changes and reduce the risk of vascular complications. Omics technologies allow us to identify new biomarkers that we can use in diagnosing diseases. This research included 90 blood plasma samples. The study group comprised 52 patients with severe atherosclerotic lesions BCA, and the control group comprised 38 patients with no BCA atherosclerosis. Targeted and panoramic lipidomic profiling of their blood plasma was carried out. There was a statistically significant difference (p < 0.05) in the values of the indices saturated fatty acids (FAs), unsaturated FAs, monounsaturated FAs, omega-3, and omega-6. Based on the results on the blood plasma lipidome, we formed models that have a fairly good ability to determine atherosclerotic lesions of the brachiocephalic arteries, as well as a model for identifying unstable atherosclerotic plaques. According only to the panoramic lipidome data, divided into groups according to stable and unstable atherosclerotic plaques, a significant difference was taken into account: p value < 0.05 and abs (fold change) > 2. Unfortunately, we did not observe significant differences according to the established plasma panoramic lipidome criteria between patients with stable and unstable plaques. Omics technologies allow us to obtain data about any changes in the body. According to our data, statistically significant differences in lipidomic profiling were obtained when comparing groups with or without BCA atherosclerosis.
Collapse
Affiliation(s)
- Anastasiia Lomonosova
- World-Class Research Center «Digital Biodesign and Personalized Healthcare», I.M. Sechenov First Moscow State Medical University (Sechenov University), 119048 Moscow, Russia; (D.G.); (A.S.); (A.S.); (A.A.); (Y.V.); (M.A.); (A.N.); (P.K.)
| | - Daria Gognieva
- World-Class Research Center «Digital Biodesign and Personalized Healthcare», I.M. Sechenov First Moscow State Medical University (Sechenov University), 119048 Moscow, Russia; (D.G.); (A.S.); (A.S.); (A.A.); (Y.V.); (M.A.); (A.N.); (P.K.)
| | - Aleksandr Suvorov
- World-Class Research Center «Digital Biodesign and Personalized Healthcare», I.M. Sechenov First Moscow State Medical University (Sechenov University), 119048 Moscow, Russia; (D.G.); (A.S.); (A.S.); (A.A.); (Y.V.); (M.A.); (A.N.); (P.K.)
| | - Artemy Silantyev
- World-Class Research Center «Digital Biodesign and Personalized Healthcare», I.M. Sechenov First Moscow State Medical University (Sechenov University), 119048 Moscow, Russia; (D.G.); (A.S.); (A.S.); (A.A.); (Y.V.); (M.A.); (A.N.); (P.K.)
| | - Alina Abasheva
- World-Class Research Center «Digital Biodesign and Personalized Healthcare», I.M. Sechenov First Moscow State Medical University (Sechenov University), 119048 Moscow, Russia; (D.G.); (A.S.); (A.S.); (A.A.); (Y.V.); (M.A.); (A.N.); (P.K.)
| | - Yana Vasina
- World-Class Research Center «Digital Biodesign and Personalized Healthcare», I.M. Sechenov First Moscow State Medical University (Sechenov University), 119048 Moscow, Russia; (D.G.); (A.S.); (A.S.); (A.A.); (Y.V.); (M.A.); (A.N.); (P.K.)
| | - Magomed Abdullaev
- World-Class Research Center «Digital Biodesign and Personalized Healthcare», I.M. Sechenov First Moscow State Medical University (Sechenov University), 119048 Moscow, Russia; (D.G.); (A.S.); (A.S.); (A.A.); (Y.V.); (M.A.); (A.N.); (P.K.)
| | - Anna Nartova
- World-Class Research Center «Digital Biodesign and Personalized Healthcare», I.M. Sechenov First Moscow State Medical University (Sechenov University), 119048 Moscow, Russia; (D.G.); (A.S.); (A.S.); (A.A.); (Y.V.); (M.A.); (A.N.); (P.K.)
| | - Nikolay Eroshchenko
- Laboratory of Molecular Modeling and Chemistry of Natural Compounds, Institute of Molecular Theranostics, Scientific and Technological Park of Biomedicine, I.M. Sechenov First Moscow State Medical University (Sechenov University), 119048 Moscow, Russia; (N.E.); (E.D.)
| | - Viktoriia Kazakova
- Laboratory of Pathophysiology Federal Research and Clinical Center of Physical-Chemical Medicine of Federal Medical Biological Agency, 119435 Moscow, Russia
| | - Roman Komarov
- Department of Cardiovascular Surgery, Institute for Professional Education, I.M. Sechenov First Moscow State Medical University (Sechenov University), 119048 Moscow, Russia; (R.K.); (A.D.)
| | - Andrey Dzyundzya
- Department of Cardiovascular Surgery, Institute for Professional Education, I.M. Sechenov First Moscow State Medical University (Sechenov University), 119048 Moscow, Russia; (R.K.); (A.D.)
| | - Elena Danilova
- Laboratory of Molecular Modeling and Chemistry of Natural Compounds, Institute of Molecular Theranostics, Scientific and Technological Park of Biomedicine, I.M. Sechenov First Moscow State Medical University (Sechenov University), 119048 Moscow, Russia; (N.E.); (E.D.)
- Department of Analytic Chemistry, Faculty of Chemistry, Lomonosov Moscow State University, 119991 Moscow, Russia
| | - Dmitry Shchekochikhin
- Department of Cardiology, Functional and Ultrasound Diagnostics, N.V. Sklifosovsky Institute for Clinical Medicine, I.M. Sechenov First Moscow State Medical University (Sechenov University), 119048 Moscow, Russia;
- Moscow State Healthcare Institution, City Clinical Hospital №1, 8 Leninsky Ave., 119049 Moscow, Russia
| | - Philipp Kopylov
- World-Class Research Center «Digital Biodesign and Personalized Healthcare», I.M. Sechenov First Moscow State Medical University (Sechenov University), 119048 Moscow, Russia; (D.G.); (A.S.); (A.S.); (A.A.); (Y.V.); (M.A.); (A.N.); (P.K.)
| |
Collapse
|
4
|
Braun RJ, Swanson JMJ. Capturing the Liquid-Crystalline Phase Transformation: Implications for Protein Targeting to Sterol Ester-Rich Lipid Droplets. MEMBRANES 2022; 12:949. [PMID: 36295707 PMCID: PMC9607156 DOI: 10.3390/membranes12100949] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 09/02/2022] [Revised: 09/23/2022] [Accepted: 09/24/2022] [Indexed: 06/16/2023]
Abstract
Lipid droplets are essential organelles that store and traffic neutral lipids. The phospholipid monolayer surrounding their neutral lipid core engages with a highly dynamic proteome that changes according to cellular and metabolic conditions. Recent work has demonstrated that when the abundance of sterol esters increases above a critical concentration, such as under conditions of starvation or high LDL exposure, the lipid droplet core can undergo an amorphous to liquid-crystalline phase transformation. Herein, we study the consequences of this transformation on the physical properties of lipid droplets that are thought to regulate protein association. Using simulations of different sterol-ester concentrations, we have captured the liquid-crystalline phase transformation at the molecular level, highlighting the alignment of sterol esters in alternating orientations to form concentric layers. We demonstrate how ordering in the core permeates into the neutral lipid/phospholipid interface, changing the magnitude and nature of neutral lipid intercalation and inducing ordering in the phospholipid monolayer. Increased phospholipid packing is concomitant with altered surface properties, including smaller area per phospholipid and substantially reduced packing defects. Additionally, the ordering of sterol esters in the core causes less hydration in more ordered regions. We discuss these findings in the context of their expected consequences for preferential protein recruitment to lipid droplets under different metabolic conditions.
Collapse
|
5
|
Leikin-Frenkel A, Cohen H, Keshet R, Shnerb-GanOr R, Kandel-Kfir M, Harari A, Hollander KS, Shaish A, Harats D, Kamari Y. The effect of α-linolenic acid enrichment in perinatal diets in preventing high fat diet-induced SCD1 increased activity and lipid disarray in adult offspring of low density lipoprotein receptor knockout (LDLRKO) mice. Prostaglandins Leukot Essent Fatty Acids 2022; 184:102475. [PMID: 35940045 DOI: 10.1016/j.plefa.2022.102475] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/25/2022] [Revised: 07/20/2022] [Accepted: 07/21/2022] [Indexed: 12/29/2022]
Abstract
The present study examined the effects of maternal perinatal dietary ALA enrichment on the high fat diet (HFD)-induced lipid disarray in the adult offspring of low density lipoprotein receptor knock-out (LDLRKO) mice. Female LDLRKO mice received, during pregnancy and lactation, isocaloric diets with either corn oil, RD, or flax oil, ALA. The weaning offspring was given a regular chow diet for a washout period of eight weeks, which was followed by HFD for eight weeks. Plasma and liver lipids and SCD1 activity were then analyzed. The HFD-fed RD adult offspring had substantially higher plasma cholesterol levels than the HFD-fed ALA offspring (15.7 versus 9.7 mmole/l, p<0.00001) and non-alcoholic fatty liver disease (NAFLD) (65.0 versus 23.9 mg/g lipids, p<0.00001). Liver lipids oleic acid (OA) content and monounsaturated to saturated fatty acids (MUFA/SAT) ratio, were two times lower in RD compared to ALA (p<0.0001). The threefold HFD-induced SCD1 raised activity (p<0.00001), and OA produced from SA, observed in RD adult offspring were prevented by perinatal ALA. In conclusion, the resilience of SCD1 to HFD- induced increased activity may account for the beneficial effects of perinatal ALA dietary enrichment in preventing NAFLD and hypercholesterolemia from occurring in adult LDLRKO offspring mice.
Collapse
Affiliation(s)
- A Leikin-Frenkel
- The Bert W. Strassburger Metabolic Center, Sheba Medical Center, Tel-Hashomer, 5265601, Israel; Sackler School of Medicine, Tel-Aviv University, Tel-Aviv, 69978, Israel.
| | - H Cohen
- The Bert W. Strassburger Metabolic Center, Sheba Medical Center, Tel-Hashomer, 5265601, Israel; Sackler School of Medicine, Tel-Aviv University, Tel-Aviv, 69978, Israel
| | - R Keshet
- The Bert W. Strassburger Metabolic Center, Sheba Medical Center, Tel-Hashomer, 5265601, Israel
| | - R Shnerb-GanOr
- The Bert W. Strassburger Metabolic Center, Sheba Medical Center, Tel-Hashomer, 5265601, Israel
| | - M Kandel-Kfir
- The Bert W. Strassburger Metabolic Center, Sheba Medical Center, Tel-Hashomer, 5265601, Israel
| | - A Harari
- The Bert W. Strassburger Metabolic Center, Sheba Medical Center, Tel-Hashomer, 5265601, Israel
| | - K S Hollander
- Sackler School of Medicine, Tel-Aviv University, Tel-Aviv, 69978, Israel
| | - A Shaish
- The Bert W. Strassburger Metabolic Center, Sheba Medical Center, Tel-Hashomer, 5265601, Israel; Achva Academic College, Israel
| | - D Harats
- The Bert W. Strassburger Metabolic Center, Sheba Medical Center, Tel-Hashomer, 5265601, Israel; Sackler School of Medicine, Tel-Aviv University, Tel-Aviv, 69978, Israel
| | - Y Kamari
- The Bert W. Strassburger Metabolic Center, Sheba Medical Center, Tel-Hashomer, 5265601, Israel; Sackler School of Medicine, Tel-Aviv University, Tel-Aviv, 69978, Israel
| |
Collapse
|
6
|
Linseed, Baru, and Coconut Oils: NMR-Based Metabolomics, Leukocyte Infiltration Potential In Vivo, and Their Oil Characterization. Are There Still Controversies? Nutrients 2022; 14:nu14061161. [PMID: 35334818 PMCID: PMC8950264 DOI: 10.3390/nu14061161] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2022] [Revised: 02/27/2022] [Accepted: 03/02/2022] [Indexed: 02/04/2023] Open
Abstract
Different fatty acid proportions produce potential inflammatory and metabolic changes in organisms. However, the evidence for how each fatty acid mediates the metabolic pathway, and its lipid stability remains controversial. To resolve this controversy, the present study investigated the metabolic effects of cold-pressed linseed (LG), coconut (CG), and baru (BG) oils in comparison to those of soybean oil (SG) in mice, in terms of their oil characterization and stability. The quality analysis showed less oxidative behavior among PUFA-rich oils (SO, BO, and LO, with induction periods lower than 2 h compared to 39.8 h for CG), besides the high contents of tocopherols and carotenoids in SG and LG. In the experimental study, CG presented higher triglyceride (257.93 ± 72.30) and VLDL-cholesterol levels (51.59 ± 14.46, p < 0.05), while LG reduced LDL levels (59.29 ± 7.56, p < 0.05) when compared to SG (183.14 ± 22.06, 36.63 ± 4.41 and 131.63 ± 29.0, respectively). For visceral fats, the adiposity index was lower for BG (7.32 ± 3.13) and CG (9.58 ± 1.02, p < 0.05) in relation to SG (12.53 ± 2.80), and for leukocyte recruitment, CG presented lower polymorphonuclear (PMN) (p < 0.0001) and mononuclear (MN) (p < 0.05) cell infiltration, demonstrating anti-inflammatory potential. In NMR-based metabolomics, although CG presented higher values for the glucose, lactate, and LDL/VLDL ratio, this group also evidenced high levels of choline, a lipotropic metabolite. Our study emphasized the controversies of saturated fatty acids, which impair serum lipids, while alfa-linolenic acid presented cardioprotective effects. However, coconut oil also has a positive immunomodulatory pathway and was found to reduce visceral bodyfat in mice. Therefore, for future applications, we suggest a combination of lauric and al-fa-linolenic acid sources, which are present in coconut and linseed oil, respectively. This combination could be less obesogenic and inflammatory and exert cardioprotective action.
Collapse
|
7
|
McGranaghan P, Kirwan JA, Garcia-Rivera MA, Pieske B, Edelmann F, Blaschke F, Appunni S, Saxena A, Rubens M, Veledar E, Trippel TD. Lipid Metabolite Biomarkers in Cardiovascular Disease: Discovery and Biomechanism Translation from Human Studies. Metabolites 2021; 11:621. [PMID: 34564437 PMCID: PMC8470800 DOI: 10.3390/metabo11090621] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2021] [Revised: 08/30/2021] [Accepted: 09/06/2021] [Indexed: 12/12/2022] Open
Abstract
Lipids represent a valuable target for metabolomic studies since altered lipid metabolism is known to drive the pathological changes in cardiovascular disease (CVD). Metabolomic technologies give us the ability to measure thousands of metabolites providing us with a metabolic fingerprint of individual patients. Metabolomic studies in humans have supported previous findings into the pathomechanisms of CVD, namely atherosclerosis, apoptosis, inflammation, oxidative stress, and insulin resistance. The most widely studied classes of lipid metabolite biomarkers in CVD are phospholipids, sphingolipids/ceramides, glycolipids, cholesterol esters, fatty acids, and acylcarnitines. Technological advancements have enabled novel strategies to discover individual biomarkers or panels that may aid in the diagnosis and prognosis of CVD, with sphingolipids/ceramides as the most promising class of biomarkers thus far. In this review, application of metabolomic profiling for biomarker discovery to aid in the diagnosis and prognosis of CVD as well as metabolic abnormalities in CVD will be discussed with particular emphasis on lipid metabolites.
Collapse
Affiliation(s)
- Peter McGranaghan
- Department of Internal Medicine and Cardiology, Charité Campus Virchow-Klinikum, 13353 Berlin, Germany; (P.M.); (B.P.); (F.E.); (F.B.)
- Baptist Health South Florida, Miami, FL 33143, USA; (A.S.); (M.R.); (E.V.)
| | - Jennifer A. Kirwan
- Metabolomics Platform, Berlin Institute of Health at Charité Universitätsmedizin Berlin, 13353 Berlin, Germany; (J.A.K.); (M.A.G.-R.)
- Max Delbrück Center for Molecular Research, 13125 Berlin, Germany
- School of Veterinary Medicine and Science, University of Nottingham, Leicestershire LE12 5RD, UK
| | - Mariel A. Garcia-Rivera
- Metabolomics Platform, Berlin Institute of Health at Charité Universitätsmedizin Berlin, 13353 Berlin, Germany; (J.A.K.); (M.A.G.-R.)
- Max Delbrück Center for Molecular Research, 13125 Berlin, Germany
| | - Burkert Pieske
- Department of Internal Medicine and Cardiology, Charité Campus Virchow-Klinikum, 13353 Berlin, Germany; (P.M.); (B.P.); (F.E.); (F.B.)
- DZHK (German Centre for Cardiovascular Research), 13353 Berlin, Germany
- Berlin Institute of Health, 13353 Berlin, Germany
- German Heart Center Berlin, Department of Cardiology, 13353 Berlin, Germany
| | - Frank Edelmann
- Department of Internal Medicine and Cardiology, Charité Campus Virchow-Klinikum, 13353 Berlin, Germany; (P.M.); (B.P.); (F.E.); (F.B.)
- DZHK (German Centre for Cardiovascular Research), 13353 Berlin, Germany
- German Heart Center Berlin, Department of Cardiology, 13353 Berlin, Germany
| | - Florian Blaschke
- Department of Internal Medicine and Cardiology, Charité Campus Virchow-Klinikum, 13353 Berlin, Germany; (P.M.); (B.P.); (F.E.); (F.B.)
- DZHK (German Centre for Cardiovascular Research), 13353 Berlin, Germany
| | - Sandeep Appunni
- Department of Biochemistry, Government Medical College, Kozhikode, Kerala 673008, India;
| | - Anshul Saxena
- Baptist Health South Florida, Miami, FL 33143, USA; (A.S.); (M.R.); (E.V.)
| | - Muni Rubens
- Baptist Health South Florida, Miami, FL 33143, USA; (A.S.); (M.R.); (E.V.)
| | - Emir Veledar
- Baptist Health South Florida, Miami, FL 33143, USA; (A.S.); (M.R.); (E.V.)
- Department of Biostatistics, Florida International University, Miami, FL 33199, USA
- Division of Cardiology, Emory University School of Medicine, Atlanta, GA 30322, USA
| | - Tobias Daniel Trippel
- Department of Internal Medicine and Cardiology, Charité Campus Virchow-Klinikum, 13353 Berlin, Germany; (P.M.); (B.P.); (F.E.); (F.B.)
- DZHK (German Centre for Cardiovascular Research), 13353 Berlin, Germany
| |
Collapse
|
8
|
You J, Qi S, Du Y, Wang C, Su G. Multiple Bioinformatics Analyses of Integrated Gene Expression Profiling Data and Verification of Hub Genes Associated with Diabetic Retinopathy. Med Sci Monit 2020; 26:e923146. [PMID: 32294661 PMCID: PMC7177039 DOI: 10.12659/msm.923146] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
Background Diabetic retinopathy (DR) is a serious complication of diabetes that can lead to blindness. This study aimed to identify the core genes and molecular functions involved in DR through multiple bioinformatics analyses. Material/Methods The mRNA gene profiles of human DR tissues from the GSE60436 and GSE53257 datasets were assessed with R software and integrated to identify the co-expressed differentially expressed genes (DEGs). Multiple bioinformatics analyses were used: Gene Ontology (GO) analysis, signaling pathway analysis, and hub gene prediction. Quantitative reverse transcription-PCR (qRT-PCR) was used to verify the hub genes. Results The Database for Annotation, Visualization and Integrated Discovery (DAVID) online tool suggested that the biological processes of the DEGs focused on mitochondrial transport, the cellular components focused on mitochondria, and molecular functions focused on catalytic activity. The results provided by DAVID were consistent with those provided by STRING and the GeneMANIA online database. All the DEGs function in metabolic pathways, consistent with the g: Profiler online analysis results. The protein-protein interaction (PPI) networks forecasted by STRING and GeneMANIA were entered into Cytoscape for cytoHubba degree analysis. The hub genes predicted by cytoHubba suggested that fumarate hydratase (FH) might be relevant to DR. qRT-PCR suggested that the expression of FH was higher in DR retinal tissues than in normal control tissues. Conclusions Multiple bioinformatics analyses verified that FH could be used as a potential diagnostic marker and new therapeutic target of DR.
Collapse
Affiliation(s)
- Jiaxin You
- Department of Ophthalmology, The Second Hospital of Jilin University, Changchun, Jilin, China (mainland)
| | - Shounan Qi
- Department of Ophthalmology, The Second Hospital of Jilin University, Changchun, Jilin, China (mainland)
| | - Yang Du
- Department of Ophthalmology, The Second Hospital of Jilin University, Changchun, Jilin, China (mainland)
| | - Chenguang Wang
- Department of Ophthalmology, The Second Hospital of Jilin University, Changchun, Jilin, China (mainland)
| | - Guanfang Su
- Department of Ophthalmology, The Second Hospital of Jilin University, Changchun, Jilin, China (mainland)
| |
Collapse
|
9
|
Bowman E, Funderburg NT. Lipidome Abnormalities and Cardiovascular Disease Risk in HIV Infection. Curr HIV/AIDS Rep 2020; 16:214-223. [PMID: 30993515 DOI: 10.1007/s11904-019-00442-9] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
PURPOSE OF REVIEW Human immunodeficiency virus (HIV) infection and its treatment with antiretroviral therapy (ART) are associated with lipid abnormalities that may enhance cardiovascular disease risk (CVD). RECENT FINDINGS Chronic inflammation persists in HIV+ individuals, and complex relationships exist among lipids and inflammation, as immune activation may be both a cause and a consequence of lipid abnormalities in HIV infection. Advances in mass spectrometry-based techniques now allow for detailed measurements of individual lipid species; improved lipid measurement might better evaluate CVD risk compared with the prognostic value of traditional assessments. Lipidomic analyses have begun to characterize dynamic changes in lipid composition during HIV infection and following treatment with ART, and further investigation may identify novel lipid biomarkers predictive of adverse outcomes. Developing strategies to improve management of comorbidities in the HIV+ population is important, and statin therapy and lifestyle modifications, including diet and exercise, may help to improve lipid levels and mitigate CVD risk.
Collapse
Affiliation(s)
- Emily Bowman
- School of Health and Rehabilitation Sciences, Division of Medical Laboratory Science, Ohio State University College of Medicine, 453 W. 10th Ave. 535A Atwell Hall, Columbus, OH, 43210, USA
| | - Nicholas T Funderburg
- School of Health and Rehabilitation Sciences, Division of Medical Laboratory Science, Ohio State University College of Medicine, 453 W. 10th Ave. 535A Atwell Hall, Columbus, OH, 43210, USA.
| |
Collapse
|
10
|
Abstract
The effect of dietary fats on cardiometabolic diseases, including cardiovascular diseases and type 2 diabetes mellitus, has generated tremendous interest. Many earlier investigations focused on total fat and conventional fat classes (such as saturated and unsaturated fats) and their influence on a limited number of risk factors. However, dietary fats comprise heterogeneous molecules with diverse structures, and growing research in the past two decades supports correspondingly complex health effects of individual dietary fats. Moreover, health effects of dietary fats might be modified by additional factors, such as accompanying nutrients and food-processing methods, emphasizing the importance of the food sources. Accordingly, the rapidly increasing scientific findings on dietary fats and cardiometabolic diseases have generated debate among scientists, caused confusion for the general public and present challenges for translation into dietary advice and policies. This Review summarizes the evidence on the effects of different dietary fats and their food sources on cell function and on risk factors and clinical events of cardiometabolic diseases. The aim is not to provide an exhaustive review but rather to focus on the most important evidence from randomized controlled trials and prospective cohort studies and to highlight current areas of controversy and the most relevant future research directions for understanding how to improve the prevention and management of cardiometabolic diseases through optimization of dietary fat intake.
Collapse
|
11
|
Visscher M, Moerman AM, Burgers PC, Van Beusekom HMM, Luider TM, Verhagen HJM, Van der Steen AFW, Van der Heiden K, Van Soest G. Data Processing Pipeline for Lipid Profiling of Carotid Atherosclerotic Plaque with Mass Spectrometry Imaging. JOURNAL OF THE AMERICAN SOCIETY FOR MASS SPECTROMETRY 2019; 30:1790-1800. [PMID: 31250318 PMCID: PMC6695360 DOI: 10.1007/s13361-019-02254-y] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/24/2018] [Revised: 04/25/2019] [Accepted: 05/20/2019] [Indexed: 05/09/2023]
Abstract
Atherosclerosis is a lipid and inflammation-driven disease of the arteries that is characterized by gradual buildup of plaques in the vascular wall. A so-called vulnerable plaque, consisting of a lipid-rich necrotic core contained by a thin fibrous cap, may rupture and trigger thrombus formation, which can lead to ischemia in the heart (heart attack) or in the brain (stroke). In this study, we present a protocol to investigate the lipid composition of advanced human carotid plaques using matrix-assisted laser desorption ionization (MALDI) mass spectrometry imaging (MSI), providing a framework that should enable the discrimination of vulnerable from stable plaques based on lipid composition. We optimized the tissue preparation and imaging methods by systematically analyzing data from three specimens: two human carotid endarterectomy samples (advanced plaque) and one autopsy sample (early stage plaque). We show a robust data reduction method and evaluate the variability of the endarterectomy samples. We found diacylglycerols to be more abundant in a thrombotic area compared to other plaque areas and could distinguish advanced plaque from early stage plaque based on cholesteryl ester composition. We plan to use this systematic approach to analyze a larger dataset of carotid atherosclerotic plaques.
Collapse
Affiliation(s)
- Mirjam Visscher
- Department of Biomedical Engineering, Thorax Center, Erasmus MC, Rotterdam, The Netherlands.
| | - Astrid M Moerman
- Department of Biomedical Engineering, Thorax Center, Erasmus MC, Rotterdam, The Netherlands
| | - Peter C Burgers
- Department of Neurology, Laboratory of Neuro-Oncology, Erasmus MC, Rotterdam, The Netherlands
| | - Heleen M M Van Beusekom
- Department of Experimental Cardiology, Thorax Center, Erasmus MC, Rotterdam, The Netherlands
| | - Theo M Luider
- Department of Neurology, Laboratory of Neuro-Oncology, Erasmus MC, Rotterdam, The Netherlands
| | - Hence J M Verhagen
- Department of Vascular and Endovascular Surgery, Erasmus Medical Center, Rotterdam, The Netherlands
| | - Antonius F W Van der Steen
- Department of Biomedical Engineering, Thorax Center, Erasmus MC, Rotterdam, The Netherlands
- Medical Delta, Delft, Rotterdam, The Netherlands
- Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China
| | - Kim Van der Heiden
- Department of Biomedical Engineering, Thorax Center, Erasmus MC, Rotterdam, The Netherlands
| | - Gijs Van Soest
- Department of Biomedical Engineering, Thorax Center, Erasmus MC, Rotterdam, The Netherlands
| |
Collapse
|
12
|
Connecting the Dots Between Fatty Acids, Mitochondrial Function, and DNA Methylation in Atherosclerosis. Curr Atheroscler Rep 2017; 19:36. [PMID: 28735349 DOI: 10.1007/s11883-017-0673-y] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
PURPOSE OF REVIEW The quest for factors and mechanisms responsible for aberrant DNA methylation in human disease-including atherosclerosis-is a promising area of research. This review focuses on the role of fatty acids (FAs) as modulators of DNA methylation-in particular the role of mitochondrial beta-oxidation in FA-induced changes in DNA methylation during the progression of atherosclerosis. RECENT FINDINGS Recent publications have advanced the knowledge in all areas touched by this review: the causal role of lipids in shaping the DNA methylome, the associations between chronic degenerative disease and mitochondrial function, the lipid composition of the atheroma, and the relevance of DNA hypermethylation in atherosclerosis. Evidence is beginning to emerge, linking the dynamics of FA type abundance, mitochondrial function, and DNA methylation in the atheroma and systemically. In particular, this review highlights mitochondrial beta-oxidation as an important regulator of DNA methylation in metabolic disease. Despite the many questions still unanswered, this area of research promises to identify mechanisms and molecular factors that establish a pathological gene expression pattern in atherosclerosis.
Collapse
|
13
|
Shen L, Yang Y, Ou T, Key CCC, Tong SH, Sequeira RC, Nelson JM, Nie Y, Wang Z, Boudyguina E, Shewale SV, Zhu X. Dietary PUFAs attenuate NLRP3 inflammasome activation via enhancing macrophage autophagy. J Lipid Res 2017; 58:1808-1821. [PMID: 28729463 PMCID: PMC5580895 DOI: 10.1194/jlr.m075879] [Citation(s) in RCA: 67] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2017] [Revised: 07/18/2017] [Indexed: 12/20/2022] Open
Abstract
Dietary PUFAs reduce atherosclerosis and macrophage inflammation, but how nucleotide-binding oligomerization domain leucine-rich repeat-containing receptor protein (NLRP3) inflammasome activation and autophagy influence PUFA-mediated atheroprotection is poorly understood. We fed Ldlr-/- mice diets containing 10% (calories) palm oil (PO) and 0.2% cholesterol, supplemented with an additional 10% of calories as PO, fish oil (FO), echium oil (EO, containing 18:4 n-3), or borage oil (BO, containing 18:3 n-6). Inflammasome activation, autophagic flux, and mitochondrial function were measured in peritoneal macrophages, blood monocytes, or liver from diet-fed mice. Compared with PO, dietary PUFAs (FO, EO, or BO) markedly inhibited inflammasome activation, shown by 1) less macrophage IL-1β secretion and caspase-1 cleavage in response to NLRP3 inflammasome activators, 2) less IL-1β secretion and caspase-1 cleavage from liver or hepatocytes in response to lipopolysaccharide (LPS), and 3) attenuated caspase-1 activity in blood monocytes. Furthermore, PUFA-enriched diets increased LC3-II expression in macrophage, aorta, and liver samples and reduced numbers of dysfunctional mitochondria in macrophages in response to LPS and palmitate, suggesting enhanced autophagic activation. Dietary PUFAs did not attenuate NLRP3 inflammasome activation in atg5-deficient macrophages, indicating that autophagic activation is critical for the PUFA-mediated inflammasome inactivation. In conclusion, dietary PUFAs reduce atherosclerosis, in part, by activation of macrophage autophagy and attenuation of NLRP3 inflammasome activation.
Collapse
Affiliation(s)
- Lulu Shen
- Department of Internal Medicine, Section on Molecular Medicine, Wake Forest School of Medicine, Winston-Salem, NC
| | - Yan Yang
- Department of Internal Medicine, Section on Molecular Medicine, Wake Forest School of Medicine, Winston-Salem, NC
| | - Tiantong Ou
- Department of Internal Medicine, Section on Molecular Medicine, Wake Forest School of Medicine, Winston-Salem, NC
| | - Chia-Chi C Key
- Department of Internal Medicine, Section on Molecular Medicine, Wake Forest School of Medicine, Winston-Salem, NC
| | - Sarah H Tong
- Prestige Department of Poultry Science, North Carolina State University, Raleigh, NC
| | - Russel C Sequeira
- Department of Internal Medicine, Section on Molecular Medicine, Wake Forest School of Medicine, Winston-Salem, NC
| | - Jonathan M Nelson
- Department of Internal Medicine, Section on Molecular Medicine, Wake Forest School of Medicine, Winston-Salem, NC
| | - Yan Nie
- Department of Internal Medicine, Section on Molecular Medicine, Wake Forest School of Medicine, Winston-Salem, NC
| | - Zhan Wang
- Department of Internal Medicine, Section on Molecular Medicine, Wake Forest School of Medicine, Winston-Salem, NC
| | - Elena Boudyguina
- Department of Internal Medicine, Section on Molecular Medicine, Wake Forest School of Medicine, Winston-Salem, NC
| | - Swapnil V Shewale
- Department of Internal Medicine, Section on Molecular Medicine, Wake Forest School of Medicine, Winston-Salem, NC
| | - Xuewei Zhu
- Department of Internal Medicine, Section on Molecular Medicine, Wake Forest School of Medicine, Winston-Salem, NC
| |
Collapse
|
14
|
Ferguson JJ, Stojanovski E, MacDonald-Wicks L, Garg ML. Fat type in phytosterol products influence their cholesterol-lowering potential: A systematic review and meta-analysis of RCTs. Prog Lipid Res 2016; 64:16-29. [DOI: 10.1016/j.plipres.2016.08.002] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2016] [Revised: 08/02/2016] [Accepted: 08/02/2016] [Indexed: 12/29/2022]
|
15
|
Abstract
Suboptimal nutrition is a leading cause of poor health. Nutrition and policy science have advanced rapidly, creating confusion yet also providing powerful opportunities to reduce the adverse health and economic impacts of poor diets. This review considers the history, new evidence, controversies, and corresponding lessons for modern dietary and policy priorities for cardiovascular diseases, obesity, and diabetes mellitus. Major identified themes include the importance of evaluating the full diversity of diet-related risk pathways, not only blood lipids or obesity; focusing on foods and overall diet patterns, rather than single isolated nutrients; recognizing the complex influences of different foods on long-term weight regulation, rather than simply counting calories; and characterizing and implementing evidence-based strategies, including policy approaches, for lifestyle change. Evidence-informed dietary priorities include increased fruits, nonstarchy vegetables, nuts, legumes, fish, vegetable oils, yogurt, and minimally processed whole grains; and fewer red meats, processed (eg, sodium-preserved) meats, and foods rich in refined grains, starch, added sugars, salt, and trans fat. More investigation is needed on the cardiometabolic effects of phenolics, dairy fat, probiotics, fermentation, coffee, tea, cocoa, eggs, specific vegetable and tropical oils, vitamin D, individual fatty acids, and diet-microbiome interactions. Little evidence to date supports the cardiometabolic relevance of other popular priorities: eg, local, organic, grass-fed, farmed/wild, or non-genetically modified. Evidence-based personalized nutrition appears to depend more on nongenetic characteristics (eg, physical activity, abdominal adiposity, gender, socioeconomic status, culture) than genetic factors. Food choices must be strongly supported by clinical behavior change efforts, health systems reforms, novel technologies, and robust policy strategies targeting economic incentives, schools and workplaces, neighborhood environments, and the food system. Scientific advances provide crucial new insights on optimal targets and best practices to reduce the burdens of diet-related cardiometabolic diseases.
Collapse
Affiliation(s)
- Dariush Mozaffarian
- From Friedman School of Nutrition Science & Policy, Tufts University, Boston, MA.
| |
Collapse
|
16
|
Abudoukelimu M, Fu ZY, Maimaiti A, Ma YT, Abudu M, Zhu Q, Adi D, Yang YN, Li XM, Xie X, Liu F, Chen BD. The association of cholesterol absorption gene Numb polymorphism with Coronary Artery Disease among Han Chinese and Uighur Chinese in Xinjiang, China. Lipids Health Dis 2015; 14:120. [PMID: 26415596 PMCID: PMC4587863 DOI: 10.1186/s12944-015-0102-6] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2015] [Accepted: 08/23/2015] [Indexed: 12/25/2022] Open
Abstract
BACKGROUND Hypercholesterolemia is a major risk factor for coronary artery disease (CAD). As Numb is an important regulating factor for intestinal cholesterol absorption and plasma cholesterol level, the aim of the present study is to assess the association between human Numb gene polymorphism and CAD among Han and Uighur Chinese. METHODS We have conducted two independent case-control studies in Han Chinese (384 CAD patients and 433 controls) and Uighur Chinese (506 CAD patients and 351 controls) subjects. All subjects were genotyped for four kinds of SNPs (rs12435797, rs2108552, rs1019075 and rs17781919) and SNP is used as a genetic marker for human Numb gene. Genotyping was undertaken using TaqMan SNP genotyping assay, and the subjects' ethnicity and gender were considered in the analysis. RESULTS We found that rs2108552 was associated with CAD in the dominant model (CC vs CG + GG) for the total Han Chinese population (n = 200) and Han Chinese males (n = 115) (P = 0.004 and P = 0.001, respectively). The difference remained statistically significant after multivariate adjustment (total: OR = 1.687, P = 0.004; male: OR = 1.498, P = 0.006). Further, for the total (n = 817) and male (n = 490) Han Chinese, the frequency of the haplotype (T-C-T-C) was significantly higher in the CAD patients than in the controls (P = 0.004 and P = 0.002), and the frequency of the haplotype (G-G-T-C) was significantly lower in the CAD patients than in the control subjects (P = 0.013, P = 0.007). In addition, for the total (n = 857) and male (n = 582) Uighur Chinese, we observed that rs12435797 was associated with CAD in an additive and recessive model (P = 0.021 and P = 0.009; P = 0.048 and P = 0.034). However, the difference did not remain statistically significant after multivariate adjustment. The overall distribution of rs2108552, rs1019075 and rs17781919 genotypes, alleles and the frequency of the haplotype established by four SNPs showed no significant difference between CAD patients and control subjects in the total, male and female Uighur Chinese. CONCLUSIONS The results of this study indicate that CC genotype of rs2108552 and T-C-T-C haplotypes in Numb gene is a possible risk genetic marker and G allele and G-G-T-C haplotypes is a possible protective genetic marker for CAD in male Han Chinese.
Collapse
Affiliation(s)
- Mayila Abudoukelimu
- Department of Cardiology, The First Affiliated Hospital of Xinjiang Medical University, Urumqi, 830001, People's Republic of China.
- Xinjiang Key Laboratory of Cardiovascular Disease Research, Urumqi, 830001, People's Republic of China.
- Present address: The First Affiliated Hospital of Xinjiang Medical University, Li Yu Shan South Road 137, Urumqi, 830001, People's Republic of China.
- Present address: Xinjiang Key Laboratory of Cardiovascular Disease Research, Li Yu Shan South Road 137, Urumqi, 830001, People's Republic of China.
| | - Zhen-Yan Fu
- Department of Cardiology, The First Affiliated Hospital of Xinjiang Medical University, Urumqi, 830001, People's Republic of China.
- Xinjiang Key Laboratory of Cardiovascular Disease Research, Urumqi, 830001, People's Republic of China.
- Present address: The First Affiliated Hospital of Xinjiang Medical University, Li Yu Shan South Road 137, Urumqi, 830001, People's Republic of China.
- Present address: Xinjiang Key Laboratory of Cardiovascular Disease Research, Li Yu Shan South Road 137, Urumqi, 830001, People's Republic of China.
| | - Ailifeire Maimaiti
- Department of Cardiology, The First Affiliated Hospital of Xinjiang Medical University, Urumqi, 830001, People's Republic of China.
- Xinjiang Key Laboratory of Cardiovascular Disease Research, Urumqi, 830001, People's Republic of China.
- Present address: The First Affiliated Hospital of Xinjiang Medical University, Li Yu Shan South Road 137, Urumqi, 830001, People's Republic of China.
- Present address: Xinjiang Key Laboratory of Cardiovascular Disease Research, Li Yu Shan South Road 137, Urumqi, 830001, People's Republic of China.
| | - Yi-Tong Ma
- Department of Cardiology, The First Affiliated Hospital of Xinjiang Medical University, Urumqi, 830001, People's Republic of China.
- Xinjiang Key Laboratory of Cardiovascular Disease Research, Urumqi, 830001, People's Republic of China.
- Present address: The First Affiliated Hospital of Xinjiang Medical University, Li Yu Shan South Road 137, Urumqi, 830001, People's Republic of China.
- Present address: Xinjiang Key Laboratory of Cardiovascular Disease Research, Li Yu Shan South Road 137, Urumqi, 830001, People's Republic of China.
| | - Minawaer Abudu
- The First Affiliated Hospital of Xinjiang Medical University, Urumqi, 830001, People's Republic of China.
- Present address: The First Affiliated Hospital of Xinjiang Medical University, Li Yu Shan South Road 137, Urumqi, 830001, People's Republic of China.
| | - Qing Zhu
- Department of Cardiology, The First Affiliated Hospital of Xinjiang Medical University, Urumqi, 830001, People's Republic of China.
- Xinjiang Key Laboratory of Cardiovascular Disease Research, Urumqi, 830001, People's Republic of China.
- Present address: The First Affiliated Hospital of Xinjiang Medical University, Li Yu Shan South Road 137, Urumqi, 830001, People's Republic of China.
- Present address: Xinjiang Key Laboratory of Cardiovascular Disease Research, Li Yu Shan South Road 137, Urumqi, 830001, People's Republic of China.
| | - Dilare Adi
- Department of Cardiology, The First Affiliated Hospital of Xinjiang Medical University, Urumqi, 830001, People's Republic of China.
- Xinjiang Key Laboratory of Cardiovascular Disease Research, Urumqi, 830001, People's Republic of China.
- Present address: The First Affiliated Hospital of Xinjiang Medical University, Li Yu Shan South Road 137, Urumqi, 830001, People's Republic of China.
- Present address: Xinjiang Key Laboratory of Cardiovascular Disease Research, Li Yu Shan South Road 137, Urumqi, 830001, People's Republic of China.
| | - Yi-Ning Yang
- Department of Cardiology, The First Affiliated Hospital of Xinjiang Medical University, Urumqi, 830001, People's Republic of China.
- Xinjiang Key Laboratory of Cardiovascular Disease Research, Urumqi, 830001, People's Republic of China.
- Present address: The First Affiliated Hospital of Xinjiang Medical University, Li Yu Shan South Road 137, Urumqi, 830001, People's Republic of China.
- Present address: Xinjiang Key Laboratory of Cardiovascular Disease Research, Li Yu Shan South Road 137, Urumqi, 830001, People's Republic of China.
| | - Xiao-Mei Li
- Department of Cardiology, The First Affiliated Hospital of Xinjiang Medical University, Urumqi, 830001, People's Republic of China.
- Xinjiang Key Laboratory of Cardiovascular Disease Research, Urumqi, 830001, People's Republic of China.
- Present address: The First Affiliated Hospital of Xinjiang Medical University, Li Yu Shan South Road 137, Urumqi, 830001, People's Republic of China.
- Present address: Xinjiang Key Laboratory of Cardiovascular Disease Research, Li Yu Shan South Road 137, Urumqi, 830001, People's Republic of China.
| | - Xiang Xie
- Department of Cardiology, The First Affiliated Hospital of Xinjiang Medical University, Urumqi, 830001, People's Republic of China.
- Xinjiang Key Laboratory of Cardiovascular Disease Research, Urumqi, 830001, People's Republic of China.
- Present address: The First Affiliated Hospital of Xinjiang Medical University, Li Yu Shan South Road 137, Urumqi, 830001, People's Republic of China.
- Present address: Xinjiang Key Laboratory of Cardiovascular Disease Research, Li Yu Shan South Road 137, Urumqi, 830001, People's Republic of China.
| | - Fen Liu
- Xinjiang Key Laboratory of Cardiovascular Disease Research, Urumqi, 830001, People's Republic of China.
- Present address: Xinjiang Key Laboratory of Cardiovascular Disease Research, Li Yu Shan South Road 137, Urumqi, 830001, People's Republic of China.
| | - Bang-Dang Chen
- Xinjiang Key Laboratory of Cardiovascular Disease Research, Urumqi, 830001, People's Republic of China.
- Present address: Xinjiang Key Laboratory of Cardiovascular Disease Research, Li Yu Shan South Road 137, Urumqi, 830001, People's Republic of China.
| |
Collapse
|
17
|
Abstract
Identifying the mechanisms that convert a healthy vascular wall to an atherosclerotic wall is of major importance since the consequences may lead to a shortened lifespan. Classical risk factors (age, smoking, obesity, diabetes mellitus, hypertension, and dyslipidemia) may result in the progression of atherosclerotic lesions by processes including inflammation and lipid accumulation. Thus, the evaluation of blood lipids and the full lipid complement produced by cells, organisms, or tissues (lipidomics) is an issue of importance. In this review, we shall describe the recent progress in vascular health research using lipidomic advances. We will begin with an overview of vascular wall biology and lipids, followed by a short analysis of lipidomics. Finally, we shall focus on the clinical implications of lipidomics and studies that have examined lipidomic approaches and vascular health.
Collapse
Affiliation(s)
- Genovefa Kolovou
- Cardiology Department, Onassis Cardiac Surgery Center, Athens, Greece
| | - Vana Kolovou
- Cardiology Department, Onassis Cardiac Surgery Center, Athens, Greece ; Molecular Immunology Laboratory, Onassis Cardiac Surgery Center, Athens, Greece
| | - Sophie Mavrogeni
- Cardiology Department, Onassis Cardiac Surgery Center, Athens, Greece
| |
Collapse
|
18
|
Shewale SV, Boudyguina E, Zhu X, Shen L, Hutchins PM, Barkley RM, Murphy RC, Parks JS. Botanical oils enriched in n-6 and n-3 FADS2 products are equally effective in preventing atherosclerosis and fatty liver. J Lipid Res 2015; 56:1191-205. [PMID: 25921305 DOI: 10.1194/jlr.m059170] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2015] [Indexed: 01/02/2023] Open
Abstract
Echium oil (EO), which is enriched in 18:4 n-3, the immediate product of fatty acid desaturase 2 (FADS2) desaturation of 18:3 n-3, is as atheroprotective as fish oil (FO). The objective of this study was to determine whether botanical oils enriched in the FADS2 products 18:3 n-6 versus 18:4 n-3 are equally atheroprotective. LDL receptor KO mice were fed one of four atherogenic diets containing 0.2% cholesterol and 10% calories as palm oil (PO) plus 10% calories as: 1) PO; 2) borage oil (BO; 18:3 n-6 enriched); 3) EO (18:4 n-3 enriched); or 4) FO for 16 weeks. Mice fed BO, EO, and FO versus PO had significantly lower plasma total and VLDL cholesterol concentrations; hepatic neutral lipid content and inflammation, aortic CE content, aortic root intimal area and macrophage content; and peritoneal macrophage inflammation, CE content, and ex vivo chemotaxis. Atheromas lacked oxidized CEs despite abundant generation of macrophage 12/15 lipooxygenase-derived metabolites. We conclude that botanical oils enriched in 18:3 n-6 and 18:4 n-3 PUFAs beyond the rate-limiting FADS2 enzyme are equally effective in preventing atherosclerosis and hepatosteatosis compared with saturated/monounsaturated fat due to cellular enrichment of ≥20 PUFAs, reduced plasma VLDL, and attenuated macrophage inflammation.
Collapse
Affiliation(s)
- Swapnil V Shewale
- Departments of Internal Medicine-Section on Molecular Medicine, Wake Forest School of Medicine, Winston-Salem, NC 27157 Physiology/Pharmacology, Wake Forest School of Medicine, Winston-Salem, NC 27157
| | - Elena Boudyguina
- Departments of Internal Medicine-Section on Molecular Medicine, Wake Forest School of Medicine, Winston-Salem, NC 27157
| | - Xuewei Zhu
- Departments of Internal Medicine-Section on Molecular Medicine, Wake Forest School of Medicine, Winston-Salem, NC 27157
| | - Lulu Shen
- Departments of Internal Medicine-Section on Molecular Medicine, Wake Forest School of Medicine, Winston-Salem, NC 27157
| | - Patrick M Hutchins
- Department of Pharmacology, University of Colorado Denver, Aurora, CO 80045
| | - Robert M Barkley
- Department of Pharmacology, University of Colorado Denver, Aurora, CO 80045
| | - Robert C Murphy
- Department of Pharmacology, University of Colorado Denver, Aurora, CO 80045
| | - John S Parks
- Departments of Internal Medicine-Section on Molecular Medicine, Wake Forest School of Medicine, Winston-Salem, NC 27157 Biochemistry, Wake Forest School of Medicine, Winston-Salem, NC 27157
| |
Collapse
|
19
|
Macri EV, Lifshitz F, Alsina E, Juiz N, Zago V, Lezón C, Rodriguez PN, Schreier L, Boyer PM, Friedman SM. Monounsaturated fatty acids-rich diets in hypercholesterolemic-growing rats. Int J Food Sci Nutr 2015; 66:400-8. [PMID: 25830945 DOI: 10.3109/09637486.2015.1025719] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
The effects of replacing dietary saturated fat by different monounsaturated fatty acid (ω-9MUFA) sources on serum lipids, body fat and bone in growing hypercholesterolemic rats were studied. Rats received one of the six different diets: AIN-93G (control, C); extra virgin olive oil (OO) + C; high-oleic sunflower oil (HOSO) + C or atherogenic diet (AT) for 8 weeks; the remaining two groups received AT for 3 weeks and then, the saturated fat was replaced by an oil mixture of soybean oil added with OO or HOSO for 5 weeks. Rats consuming MUFA-rich diets showed the highest body fat, hepatic index and epididymal, intestinal and perirenal fat, and triglycerides. T-chol and non-HDL-chol were increased in HOSO rats but decreased in OO rats. Bone mineral content and density were higher in both OO and HOSO groups than in AT rats. This study casts caution to the generalization of the benefits of MUFA for the treatment of hypercholesterolemia.
Collapse
Affiliation(s)
- Elisa V Macri
- Department of Biochemistry, School of Dentistry, University of Buenos Aires , Buenos Aires , Argentina
| | | | | | | | | | | | | | | | | | | |
Collapse
|
20
|
|
21
|
Babu AS, Veluswamy SK, Arena R, Guazzi M, Lavie CJ. Virgin Coconut Oil and Its Potential Cardioprotective Effects. Postgrad Med 2015; 126:76-83. [DOI: 10.3810/pgm.2014.11.2835] [Citation(s) in RCA: 40] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023]
|
22
|
Siri-Tarino PW, Chiu S, Bergeron N, Krauss RM. Saturated Fats Versus Polyunsaturated Fats Versus Carbohydrates for Cardiovascular Disease Prevention and Treatment. Annu Rev Nutr 2015; 35:517-43. [PMID: 26185980 PMCID: PMC4744652 DOI: 10.1146/annurev-nutr-071714-034449] [Citation(s) in RCA: 160] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
The effects of saturated fatty acids (SFAs) on cardiovascular disease (CVD) risk are modulated by the nutrients that replace them and their food matrices. Replacement of SFAs with polyunsaturated fatty acids has been associated with reduced CVD risk, although there is heterogeneity in both fatty acid categories. In contrast, replacement of SFAs with carbohydrates, particularly sugar, has been associated with no improvement or even a worsening of CVD risk, at least in part through effects on atherogenic dyslipidemia, a cluster of traits including small, dense low-density lipoprotein particles. The effects of dietary SFAs on insulin sensitivity, inflammation, vascular function, and thrombosis are less clear. There is growing evidence that SFAs in the context of dairy foods, particularly fermented dairy products, have neutral or inverse associations with CVD. Overall dietary patterns emphasizing vegetables, fish, nuts, and whole versus processed grains form the basis of heart-healthy eating and should supersede a focus on macronutrient composition.
Collapse
Affiliation(s)
- Patty W. Siri-Tarino
- Atherosclerosis Research Program, Children’s Hospital Oakland Research Institute, Oakland, California 94609
| | - Sally Chiu
- Atherosclerosis Research Program, Children’s Hospital Oakland Research Institute, Oakland, California 94609
| | - Nathalie Bergeron
- Atherosclerosis Research Program, Children’s Hospital Oakland Research Institute, Oakland, California 94609
- College of Pharmacy, Touro University California, Vallejo, California 94594
| | - Ronald M. Krauss
- Atherosclerosis Research Program, Children’s Hospital Oakland Research Institute, Oakland, California 94609
| |
Collapse
|
23
|
Jones PJH, MacKay DS, Senanayake VK, Pu S, Jenkins DJA, Connelly PW, Lamarche B, Couture P, Kris-Etherton PM, West SG, Liu X, Fleming JA, Hantgan RR, Rudel LL. High-oleic canola oil consumption enriches LDL particle cholesteryl oleate content and reduces LDL proteoglycan binding in humans. Atherosclerosis 2014; 238:231-8. [PMID: 25528432 DOI: 10.1016/j.atherosclerosis.2014.12.010] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/15/2014] [Revised: 11/27/2014] [Accepted: 12/02/2014] [Indexed: 11/25/2022]
Abstract
Oleic acid consumption is considered cardio-protective according to studies conducted examining effects of the Mediterranean diet. However, animal models have shown that oleic acid consumption increases LDL particle cholesteryl oleate content which is associated with increased LDL-proteoglycan binding and atherosclerosis. The objective was to examine effects of varying oleic, linoleic and docosahexaenoic acid consumption on human LDL-proteoglycan binding in a non-random subset of the Canola Oil Multi-center Intervention Trial (COMIT) participants. COMIT employed a randomized, double-blind, five-period, cross-over trial design. Three of the treatment oil diets: 1) a blend of corn/safflower oil (25:75); 2) high oleic canola oil; and 3) DHA-enriched high oleic canola oil were selected for analysis of LDL-proteoglycan binding in 50 participants exhibiting good compliance. LDL particles were isolated from frozen plasma by gel filtration chromatography and LDL cholesteryl esters quantified by mass-spectrometry. LDL-proteoglycan binding was assessed using surface plasmon resonance. LDL particle cholesterol ester fatty acid composition was sensitive to the treatment fatty acid compositions, with the main fatty acids in the treatments increasing in the LDL cholesterol esters. The corn/safflower oil and high-oleic canola oil diets lowered LDL-proteoglycan binding relative to their baseline values (p = 0.0005 and p = 0.0012, respectively). At endpoint, high-oleic canola oil feeding resulted in lower LDL-proteoglycan binding than corn/safflower oil (p = 0.0243) and DHA-enriched high oleic canola oil (p = 0.0249), although high-oleic canola oil had the lowest binding at baseline (p = 0.0344). Our findings suggest that high-oleic canola oil consumption in humans increases cholesteryl oleate percentage in LDL, but in a manner not associated with a rise in LDL-proteoglycan binding.
Collapse
Affiliation(s)
- Peter J H Jones
- Richardson Centre for Functional Foods and Nutraceuticals, University of Manitoba, Winnipeg, MB, Canada.
| | - Dylan S MacKay
- Richardson Centre for Functional Foods and Nutraceuticals, University of Manitoba, Winnipeg, MB, Canada
| | - Vijitha K Senanayake
- Richardson Centre for Functional Foods and Nutraceuticals, University of Manitoba, Winnipeg, MB, Canada
| | - Shuaihua Pu
- Richardson Centre for Functional Foods and Nutraceuticals, University of Manitoba, Winnipeg, MB, Canada
| | | | | | - Benoît Lamarche
- Institute of Nutrition and Functional Foods, Laval University, Quebec City, QC, Canada
| | - Patrick Couture
- Institute of Nutrition and Functional Foods, Laval University, Quebec City, QC, Canada
| | | | - Sheila G West
- Department of Nutritional Sciences, Pennsylvania State University, PA, USA; Department of Biobehavioral Health, Pennsylvania State University, PA, USA
| | - Xiaoran Liu
- Department of Nutritional Sciences, Pennsylvania State University, PA, USA
| | - Jennifer A Fleming
- Department of Nutritional Sciences, Pennsylvania State University, PA, USA
| | - Roy R Hantgan
- Department of Biochemistry Wake Forest University School of Medicine, Winston-Salem, NC, USA
| | - Lawrence L Rudel
- Department of Biochemistry Wake Forest University School of Medicine, Winston-Salem, NC, USA; Department of Pathology, Section on Lipid Sciences, Wake Forest University School of Medicine, Winston-Salem, NC, USA
| |
Collapse
|
24
|
Zhang J, Sawyer JK, Marshall SM, Kelley KL, Davis MA, Wilson MD, Brown JM, Rudel LL. Cholesterol esters (CE) derived from hepatic sterol O-acyltransferase 2 (SOAT2) are associated with more atherosclerosis than CE from intestinal SOAT2. Circ Res 2014; 115:826-33. [PMID: 25239141 PMCID: PMC4209196 DOI: 10.1161/circresaha.115.304378] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
RATIONALE Cholesterol esters (CE), especially cholesterol oleate, generated by hepatic and intestinal sterol O-acyltransferase 2 (SOAT2) play a critical role in cholesterol homeostasis. However, it is unknown whether the contribution of intestine-derived CE from SOAT2 would have similar effects in promoting atherosclerosis progression as for liver-derived CE. OBJECTIVE To test whether, in low-density lipoprotein receptor null (LDLr(-/-)) mice, the conditional knockout of intestinal SOAT2 (SOAT2(SI-/SI-)) or hepatic SOAT2 (SOAT2(L-/L-)) would equally limit atherosclerosis development compared with the global deletion of SOAT2 (SOAT2(-/-)). METHODS AND RESULTS SOAT2 conditional knockout mice were bred with LDLr(-/-) mice creating LDLr(-/-) mice with each of the specific SOAT2 gene deletions. All mice then were fed an atherogenic diet for 16 weeks. SOAT2(SI-/SI-)LDLr(-/-) and SOAT2(-/-)LDLr(-/-) mice had significantly lower levels of intestinal cholesterol absorption, more fecal sterol excretion, and lower biliary cholesterol levels. Analysis of plasma LDL showed that all mice with SOAT2 gene deletions had LDL CE with reduced percentages of cholesterol palmitate and cholesterol oleate. Each of the LDLr(-/-) mice with SOAT2 gene deletions had lower accumulations of total cholesterol and CE in the liver compared with control mice. Finally, aortic atherosclerosis development was significantly lower in all mice with global or tissue-restricted SOAT2 gene deletions. Nevertheless, SOAT2(-/-)LDLr(-/-) and SOAT2(L-/L-)LDLr(-/-) mice had less aortic CE accumulation and smaller aortic lesions than SOAT2(SI-/SI-)LDLr(-/-) mice. CONCLUSIONS SOAT2-derived CE from both the intestine and liver significantly contribute to the development of atherosclerosis, although the CE from the hepatic enzyme appeared to promote more atherosclerosis development.
Collapse
Affiliation(s)
- Jun Zhang
- From the Section on Molecular Medicine, Department of Internal Medicine, Wake Forest University School of Medicine, Winston-Salem, NC (J.Z., J.K.S., S.M.M., K.L.K., M.A.D., M.D.W., L.L.R.); and Department of Cellular and Molecular Medicine, Cleveland Clinic Lerner Research Institute, OH (S.M.M., J.M.B.)
| | - Janet K Sawyer
- From the Section on Molecular Medicine, Department of Internal Medicine, Wake Forest University School of Medicine, Winston-Salem, NC (J.Z., J.K.S., S.M.M., K.L.K., M.A.D., M.D.W., L.L.R.); and Department of Cellular and Molecular Medicine, Cleveland Clinic Lerner Research Institute, OH (S.M.M., J.M.B.)
| | - Stephanie M Marshall
- From the Section on Molecular Medicine, Department of Internal Medicine, Wake Forest University School of Medicine, Winston-Salem, NC (J.Z., J.K.S., S.M.M., K.L.K., M.A.D., M.D.W., L.L.R.); and Department of Cellular and Molecular Medicine, Cleveland Clinic Lerner Research Institute, OH (S.M.M., J.M.B.)
| | - Kathryn L Kelley
- From the Section on Molecular Medicine, Department of Internal Medicine, Wake Forest University School of Medicine, Winston-Salem, NC (J.Z., J.K.S., S.M.M., K.L.K., M.A.D., M.D.W., L.L.R.); and Department of Cellular and Molecular Medicine, Cleveland Clinic Lerner Research Institute, OH (S.M.M., J.M.B.)
| | - Matthew A Davis
- From the Section on Molecular Medicine, Department of Internal Medicine, Wake Forest University School of Medicine, Winston-Salem, NC (J.Z., J.K.S., S.M.M., K.L.K., M.A.D., M.D.W., L.L.R.); and Department of Cellular and Molecular Medicine, Cleveland Clinic Lerner Research Institute, OH (S.M.M., J.M.B.)
| | - Martha D Wilson
- From the Section on Molecular Medicine, Department of Internal Medicine, Wake Forest University School of Medicine, Winston-Salem, NC (J.Z., J.K.S., S.M.M., K.L.K., M.A.D., M.D.W., L.L.R.); and Department of Cellular and Molecular Medicine, Cleveland Clinic Lerner Research Institute, OH (S.M.M., J.M.B.)
| | - J Mark Brown
- From the Section on Molecular Medicine, Department of Internal Medicine, Wake Forest University School of Medicine, Winston-Salem, NC (J.Z., J.K.S., S.M.M., K.L.K., M.A.D., M.D.W., L.L.R.); and Department of Cellular and Molecular Medicine, Cleveland Clinic Lerner Research Institute, OH (S.M.M., J.M.B.)
| | - Lawrence L Rudel
- From the Section on Molecular Medicine, Department of Internal Medicine, Wake Forest University School of Medicine, Winston-Salem, NC (J.Z., J.K.S., S.M.M., K.L.K., M.A.D., M.D.W., L.L.R.); and Department of Cellular and Molecular Medicine, Cleveland Clinic Lerner Research Institute, OH (S.M.M., J.M.B.).
| |
Collapse
|
25
|
Papadopoulou E, Stanner S. Questioning current recommendations on fatty acids and their role in heart health. NUTR BULL 2014. [DOI: 10.1111/nbu.12100] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023]
|
26
|
Wang P, Li J, Wang P, Hu CR, Zhang D, Sturek M, Cheng JX. Label-free quantitative imaging of cholesterol in intact tissues by hyperspectral stimulated Raman scattering microscopy. Angew Chem Int Ed Engl 2013; 52:13042-6. [PMID: 24127161 PMCID: PMC3932421 DOI: 10.1002/anie.201306234] [Citation(s) in RCA: 68] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2013] [Indexed: 01/28/2023]
Abstract
A finger on the pulse: Current molecular analysis of cells and tissues routinely relies on separation, enrichment, and subsequent measurements by various assays. Now, a platform of hyperspectral stimulated Raman scattering microscopy has been developed for the fast, quantitative, and label-free imaging of biomolecules in intact tissues using spectroscopic fingerprints as the contrast mechanism.
Collapse
Affiliation(s)
- Ping Wang
- Weldon School of Biomedical Engineering, Purdue University, West
Lafayette, IN 47906 (USA)
| | - Junjie Li
- Department of Biological Sciences, Purdue University, West
Lafayette, IN 47906 (USA)
| | - Pu Wang
- Weldon School of Biomedical Engineering, Purdue University, West
Lafayette, IN 47906 (USA)
| | - Chun-Rui Hu
- Weldon School of Biomedical Engineering, Purdue University, West
Lafayette, IN 47906 (USA)
| | - Delong Zhang
- Department of Chemistry, Purdue University, West Lafayette, IN
47906 (USA)
| | - Michael Sturek
- Department of Cellular and Integrative Physiology, Indiana
University Medical School, Indianapolis, IN 46202 (USA)
| | - Ji-Xin Cheng
- Weldon School of Biomedical Engineering, Purdue University, West
Lafayette, IN 47906 (USA)
| |
Collapse
|
27
|
Wang P, Li J, Wang P, Hu CR, Zhang D, Sturek M, Cheng JX. Label-Free Quantitative Imaging of Cholesterol in Intact Tissues by Hyperspectral Stimulated Raman Scattering Microscopy. Angew Chem Int Ed Engl 2013. [DOI: 10.1002/ange.201306234] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2023]
|
28
|
Bonamassa B, Moschetta A. Atherosclerosis: lessons from LXR and the intestine. Trends Endocrinol Metab 2013; 24:120-8. [PMID: 23158108 DOI: 10.1016/j.tem.2012.10.004] [Citation(s) in RCA: 52] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/27/2012] [Revised: 10/12/2012] [Accepted: 10/18/2012] [Indexed: 12/17/2022]
Abstract
Modulation of the cholesterol-sensing liver X receptors (LXRs) and their downstream targets has emerged as promising therapeutic avenues in atherosclerosis. The intestine is important for its unique capabilities to act as a gatekeeper for cholesterol absorption and to participate in the process of cholesterol elimination in the feces and reverse cholesterol transport (RCT). Pharmacological and genetic intestine-specific LXR activation have been shown to protect against atherosclerosis. In this review we discuss the LXR-targeted molecular players in the enterocytes as well as the intestine-driven pathways contributing to cholesterol homeostasis with therapeutic potential as targets in the prevention and treatment of atherosclerosis..
Collapse
Affiliation(s)
- Barbara Bonamassa
- Laboratory of Lipid Metabolism and Cancer, Department of Translational Pharmacology, Consorzio Mario Negri Sud, Via Nazionale 8/A, 66030 Santa Maria Imbaro (CH), Italy
| | | |
Collapse
|
29
|
Rong JX, Blachford C, Feig JE, Bander I, Mayne J, Kusunoki J, Miller C, Davis M, Wilson M, Dehn S, Thorp E, Tabas I, Taubman MB, Rudel LL, Fisher EA. ACAT inhibition reduces the progression of preexisting, advanced atherosclerotic mouse lesions without plaque or systemic toxicity. Arterioscler Thromb Vasc Biol 2012; 33:4-12. [PMID: 23139293 DOI: 10.1161/atvbaha.112.252056] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
OBJECTIVE Acyl-CoA:cholesterol acyltransferase (ACAT) converts cholesterol to cholesteryl esters in plaque foam cells. Complete deficiency of macrophage ACAT has been shown to increase atherosclerosis in hypercholesterolemic mice because of cytotoxicity from free cholesterol accumulation, whereas we previously showed that partial ACAT inhibition by Fujirebio compound F1394 decreased early atherosclerosis development. In this report, we tested F1394 effects on preestablished, advanced lesions of apolipoprotein-E-deficient mice. METHODS AND RESULTS Apolipoprotein-E-deficient mice on Western diet for 14 weeks developed advanced plaques, and were either euthanized (Baseline), or continued on Western diet with or without F1394 and euthanized after 14 more weeks. F1394 was not associated with systemic toxicity. Compared with the baseline group, lesion size progressed in both groups; however, F1394 significantly retarded plaque progression and reduced plaque macrophage, free and esterified cholesterol, and tissue factor contents compared with the untreated group. Apoptosis of plaque cells was not increased, consistent with the decrease in lesional free cholesterol. There was no increase in plaque necrosis and unimpaired efferocytosis (phagocytic clearance of apoptotic cells). The effects of F1394 were independent of changes in plasma cholesterol levels. CONCLUSIONS Partial ACAT inhibition by F1394 lowered plaque cholesterol content and had other antiatherogenic effects in advanced lesions in apolipoprotein-E-deficient mice without overt systemic or plaque toxicity, suggesting the continued potential of ACAT inhibition for the clinical treatment of atherosclerosis, in spite of recent trial data.
Collapse
Affiliation(s)
- James X Rong
- Marc and Ruti Bell Vascular Biology and Disease Research Program of the Leon H. Charney Division of Cardiology and the Department of Medicine (Cardiology), New York University School of Medicine, Smilow 7, 522 First Ave, New York, NY 10029, USA
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
30
|
Effects of body weight and season on serum lipid concentrations in sloth bears (Melursus ursinus ursinus). J Zoo Wildl Med 2012; 42:373-81. [PMID: 22950308 DOI: 10.1638/2008-0213.1] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
Serum lipid levels were measured in 66 healthy sloth bears (Melursus ursinus ursinus) living under semicaptive conditions with access to natural food resources in the Bannerghatta Biological Park (Karnataka, India), a portion of their native habitat range in the Indian peninsula. Total cholesterol, triglycerides, high-density lipoprotein (HDL) cholesterol, and low-density lipoprotein cholesterol levels were analyzed. The effects of age, body weight, and season on these lipid parameters were statistically evaluated. There were no correlations between age and any of the serum lipid parameters analyzed. Positive correlations of body weight to both triglyceride and HDL cholesterol levels in these bears were identified. In addition, seasonal trends in physiological serum lipid values, potentially due to variations in the sloth bear diet, were identified. Serum triglyceride levels were higher during postmonsoon season and cholesterol levels were higher during winter compared to other seasons. Serum lipid values obtained from sloth bears in this study were also compared to previously published data on other members of the family Ursidae. This is the first report of serum lipid values as a reference for sloth bears. These values can be used as sensitive predictors of overall health and nutritional status to aid in the captive management and feeding of these bears.
Collapse
|
31
|
Zhang J, Kelley KL, Marshall SM, Davis MA, Wilson MD, Sawyer JK, Farese RV, Brown JM, Rudel LL. Tissue-specific knockouts of ACAT2 reveal that intestinal depletion is sufficient to prevent diet-induced cholesterol accumulation in the liver and blood. J Lipid Res 2012; 53:1144-52. [PMID: 22460046 PMCID: PMC3351821 DOI: 10.1194/jlr.m024356] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2012] [Revised: 03/20/2012] [Indexed: 11/20/2022] Open
Abstract
Acyl-CoA:cholesterol acyltransferase 2 (ACAT2) generates cholesterol esters (CE) for packaging into newly synthesized lipoproteins and thus is a major determinant of blood cholesterol levels. ACAT2 is expressed exclusively in the small intestine and liver, but the relative contributions of ACAT2 expression in these tissues to systemic cholesterol metabolism is unknown. We investigated whether CE derived from the intestine or liver would differentially affect hepatic and plasma cholesterol homeostasis. We generated liver-specific (ACAT2(L-/L-)) and intestine-specific (ACAT2(SI-/SI-)) ACAT2 knockout mice and studied dietary cholesterol-induced hepatic lipid accumulation and hypercholesterolemia. ACAT2(SI-/SI-) mice, in contrast to ACAT2(L-/L-) mice, had blunted cholesterol absorption. However, specific deletion of ACAT2 in the intestine generated essentially a phenocopy of the conditional knockout of ACAT2 in the liver, with reduced levels of plasma very low-density lipoprotein and hepatic CE, yet hepatic-free cholesterol does not build up after high cholesterol intake. ACAT2(L-/L-) and ACAT2(SI-/SI-) mice were equally protected from diet-induced hepatic CE accumulation and hypercholesterolemia. These results suggest that inhibition of intestinal or hepatic ACAT2 improves atherogenic hyperlipidemia and limits hepatic CE accumulation in mice and that depletion of intestinal ACAT2 is sufficient for most of the beneficial effects on cholesterol metabolism. Inhibitors of ACAT2 targeting either tissue likely would be beneficial for atheroprotection.
Collapse
Affiliation(s)
- Jun Zhang
- Section on Lipid Sciences, Department of Pathology, Wake Forest University School of Medicine, Winston-Salem, NC 27157
| | - Kathryn L. Kelley
- Section on Lipid Sciences, Department of Pathology, Wake Forest University School of Medicine, Winston-Salem, NC 27157
| | - Stephanie M. Marshall
- Section on Lipid Sciences, Department of Pathology, Wake Forest University School of Medicine, Winston-Salem, NC 27157
| | - Matthew A. Davis
- Section on Lipid Sciences, Department of Pathology, Wake Forest University School of Medicine, Winston-Salem, NC 27157
| | - Martha D. Wilson
- Section on Lipid Sciences, Department of Pathology, Wake Forest University School of Medicine, Winston-Salem, NC 27157
| | - Janet K. Sawyer
- Section on Lipid Sciences, Department of Pathology, Wake Forest University School of Medicine, Winston-Salem, NC 27157
| | - Robert V. Farese
- Departments of Medicine, Biochemistry & Biophysics, Gladstone Institute of Cardiovascular Disease, University of California, San Francisco, CA 94158
| | - J. Mark Brown
- Section on Lipid Sciences, Department of Pathology, Wake Forest University School of Medicine, Winston-Salem, NC 27157
| | - Lawrence L. Rudel
- Section on Lipid Sciences, Department of Pathology, Wake Forest University School of Medicine, Winston-Salem, NC 27157
| |
Collapse
|
32
|
Suhalim J, Chung CY, Lilledahl M, Lim R, Levi M, Tromberg B, Potma E. Characterization of cholesterol crystals in atherosclerotic plaques using stimulated Raman scattering and second-harmonic generation microscopy. Biophys J 2012; 102:1988-95. [PMID: 22768956 PMCID: PMC3328706 DOI: 10.1016/j.bpj.2012.03.016] [Citation(s) in RCA: 95] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2012] [Revised: 03/05/2012] [Accepted: 03/12/2012] [Indexed: 02/04/2023] Open
Abstract
Cholesterol crystals (ChCs) have been identified as a major factor of plaque vulnerability and as a potential biomarker for atherosclerosis. Yet, due to the technical challenge of selectively detecting cholesterol in its native tissue environment, the physiochemical role of ChCs in atherosclerotic progression remains largely unknown. In this work, we demonstrate the utility of hyperspectral stimulated Raman scattering (SRS) microscopy combined with second-harmonic generation (SHG) microscopy to selectively detect ChC. We show that despite the polarization sensitivity of the ChC Raman spectrum, cholesterol monohydrate crystals can be reliably discriminated from aliphatic lipids, from structural proteins of the tissue matrix and from other condensed structures, including cholesteryl esters. We also show that ChCs exhibit a nonvanishing SHG signal, corroborating the noncentrosymmetry of the crystal lattice composed of chiral cholesterol molecules. However, combined hyperspectral SRS and SHG imaging reveals that not all SHG-active structures with solidlike morphologies can be assigned to ChCs. This study exemplifies the merit of combining SRS and SHG microscopy for an enhanced label-free chemical analysis of crystallized structures in diseased tissue.
Collapse
Affiliation(s)
- Jeffrey L. Suhalim
- Beckman Laser Institute, University of California, Irvine, California
- Department of Biomedical Engineering, University of California, Irvine, California
| | - Chao-Yu Chung
- Department of Chemistry, University of California, Irvine, California
| | - Magnus B. Lilledahl
- Department of Physics, Norwegian University of Science and Technology, Trondheim, Norway
| | - Ryan S. Lim
- Beckman Laser Institute, University of California, Irvine, California
| | - Moshe Levi
- Division of Renal Diseases and Hypertension, Department of Medicine, University of Colorado Denver, Aurora, Colorado
| | - Bruce J. Tromberg
- Beckman Laser Institute, University of California, Irvine, California
- Department of Biomedical Engineering, University of California, Irvine, California
| | - Eric O. Potma
- Beckman Laser Institute, University of California, Irvine, California
- Department of Chemistry, University of California, Irvine, California
| |
Collapse
|
33
|
Leonarduzzi G, Gamba P, Gargiulo S, Biasi F, Poli G. Inflammation-related gene expression by lipid oxidation-derived products in the progression of atherosclerosis. Free Radic Biol Med 2012; 52:19-34. [PMID: 22037514 DOI: 10.1016/j.freeradbiomed.2011.09.031] [Citation(s) in RCA: 70] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/03/2011] [Revised: 09/16/2011] [Accepted: 09/24/2011] [Indexed: 12/31/2022]
Abstract
Vascular areas of atherosclerotic development persist in a state of inflammation, and any further inflammatory stimulus in the subintimal area elicits a proatherogenic response; this alters the behavior of the artery wall cells and recruits further inflammatory cells. In association with the inflammatory response, oxidative events are also involved in the development of atherosclerotic plaques. It is now unanimously recognized that lipid oxidation-derived products are key players in the initiation and progression of atherosclerotic lesions. Oxidized lipids, derived from oxidatively modified low-density lipoproteins (LDLs), which accumulate in the intima, strongly modulate inflammation-related gene expression, through involvement of various signaling pathways. In addition, considerable evidence supports a proatherogenic role of a large group of potent bioactive lipids called eicosanoids, which derive from oxidation of arachidonic acid, a component of membrane phospholipids. Of note, LDL lipid oxidation products might regulate eicosanoid production, modulating the enzymatic degradation of arachidonic acid by cyclooxygenases and lipoxygenases; these enzymes might also directly contribute to LDL oxidation. This review provides a comprehensive overview of current knowledge on signal transduction pathways and inflammatory gene expression, modulated by lipid oxidation-derived products, in the progression of atherosclerosis.
Collapse
|
34
|
Isoform-specific inhibitors of ACATs: recent advances and promising developments. Future Med Chem 2011; 3:2039-61. [DOI: 10.4155/fmc.11.158] [Citation(s) in RCA: 37] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Acyl-CoA:cholesterol acyltransferase (ACAT) is a promising therapeutic target for cardiovascular diseases. Although a number of synthetic ACAT inhibitors have been developed, they have failed to show efficacy in clinical trials. Now, the presence of two ACAT isoforms with distinct functions, ACAT1 and ACAT2, has been discovered. Thus, the selectivity of ACAT inhibitors toward the two isoforms is important for their development as novel anti-atherosclerotic agents. The selectivity study indicated that fungal pyripyropene A (PPPA) is only an ACAT2-specific inhibitor. Furthermore, PPPA proved orally active in atherogenic mouse models, indicating it possessed cholesterol-lowering and atheroprotective activities. Certain PPPA derivatives, semi-synthetically prepared, possessed more potent and selective in vitro activity than PPPA against ACAT2. This review covers these studies and describes the future prospects of ACAT2-specific inhibitors.
Collapse
|
35
|
Affiliation(s)
- Dariush Mozaffarian
- Division of Cardiovascular Medicine and Channing Laboratory, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA.
| | | | | |
Collapse
|
36
|
Stegemann C, Drozdov I, Shalhoub J, Humphries J, Ladroue C, Didangelos A, Baumert M, Allen M, Davies AH, Monaco C, Smith A, Xu Q, Mayr M. Comparative lipidomics profiling of human atherosclerotic plaques. ACTA ACUST UNITED AC 2011; 4:232-42. [PMID: 21511877 DOI: 10.1161/circgenetics.110.959098] [Citation(s) in RCA: 158] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
BACKGROUND We sought to perform a systematic lipid analysis of atherosclerotic plaques using emerging mass spectrometry techniques. METHODS AND RESULTS A chip-based robotic nanoelectrospray platform interfaced to a triple quadrupole mass spectrometer was adapted to analyze lipids in tissue sections and extracts from human endarterectomy specimens by shotgun lipidomics. Eighteen scans for different lipid classes plus additional scans for fatty acids resulted in the detection of 150 lipid species from 9 different classes of which 24 were detected in endarterectomies only. Further analyses focused on plaques from symptomatic and asymptomatic patients and stable versus unstable regions within the same lesion. Polyunsaturated cholesteryl esters with long-chain fatty acids and certain sphingomyelin species showed the greatest relative enrichment in plaques compared to plasma and formed part of a lipid signature for vulnerable and stable plaque areas in a systems-wide network analysis. In principal component analyses, the combination of lipid species across different classes provided a better separation of stable and unstable areas than individual lipid classes. CONCLUSIONS This comprehensive analysis of plaque lipids demonstrates the potential of lipidomics for unraveling the lipid heterogeneity within atherosclerotic lesions.
Collapse
|
37
|
Iggman D, Risérus U. Role of different dietary saturated fatty acids for cardiometabolic risk. ACTA ACUST UNITED AC 2011. [DOI: 10.2217/clp.11.7] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
|
38
|
Siri-Tarino PW, Sun Q, Hu FB, Krauss RM. Saturated fatty acids and risk of coronary heart disease: modulation by replacement nutrients. Curr Atheroscler Rep 2010; 12:384-90. [PMID: 20711693 PMCID: PMC2943062 DOI: 10.1007/s11883-010-0131-6] [Citation(s) in RCA: 237] [Impact Index Per Article: 15.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Despite the well-established observation that substitution of saturated fats for carbohydrates or unsaturated fats increases low-density lipoprotein (LDL) cholesterol in humans and animal models, the relationship of saturated fat intake to risk for atherosclerotic cardiovascular disease in humans remains controversial. A critical question is what macronutrient should be used to replace saturated fat. Substituting polyunsaturated fat for saturated fat reduces LDL cholesterol and the total cholesterol to high-density lipoprotein cholesterol ratio. However, replacement of saturated fat by carbohydrates, particularly refined carbohydrates and added sugars, increases levels of triglyceride and small LDL particles and reduces high-density lipoprotein cholesterol, effects that are of particular concern in the context of the increased prevalence of obesity and insulin resistance. Epidemiologic studies and randomized clinical trials have provided consistent evidence that replacing saturated fat with polyunsaturated fat, but not carbohydrates, is beneficial for coronary heart disease. Therefore, dietary recommendations should emphasize substitution of polyunsaturated fat and minimally processed grains for saturated fat.
Collapse
Affiliation(s)
- Patty W. Siri-Tarino
- Atherosclerosis Research, Children’s Hospital Oakland Research Institute, 5700 Martin Luther King Junior Way, Oakland, CA 94609 USA
| | - Qi Sun
- Channing Laboratory, Department of Medicine, Brigham and Women’s Hospital and Harvard Medical School, 181 Longwood Avenue, Boston, MA 02115 USA
| | - Frank B. Hu
- Departments of Nutrition and Epidemiology, Harvard School of Public Health, 665 Huntington Avenue, Boston, MA USA
| | - Ronald M. Krauss
- Atherosclerosis Research, Children’s Hospital Oakland Research Institute, 5700 Martin Luther King Junior Way, Oakland, CA 94609 USA
| |
Collapse
|
39
|
Abstract
Dietary interventions have been consistently proposed as a part of a comprehensive strategy to lower the incidence and severity of coronary heart disease (CHD), in the process providing long-term cardioprotection. Replacement of dietary saturated fatty acids (SFA) with higher intakes of monounsaturated fatty acids (MUFA) and polyunsaturated fatty acids (PUFA) has been reported to be inversely associated with risk of CHD. The observed lower incidence of CHD among populations consuming a Mediterranean-type diet, mainly enriched in MUFA from olive oil, has long supported the belief that MUFA are an optimal substitution for SFA. However, both epidemiologic and interventional studies suggest that although substituting MUFA-rich foods for SFA-rich foods in the diet can potentially lower total plasma cholesterol concentrations, this substitution does not lower the extent of coronary artery atherosclerosis. In addition, although recent evidence suggests that the source of MUFA (animal fat vs vegetable oils) may differentially influence the correlation between MUFA intake and CHD mortality, animal studies suggest that neither source is cardioprotective.
Collapse
Affiliation(s)
- Chiara Degirolamo
- Department of Translational Pharmacology, Consorzio Mario Negri Sud, via Nazionale 8/A, 66030, S. Maria Imbaro, CH, Italy,
| | - Lawrence L. Rudel
- Department of Pathology, Section on Lipid Sciences, Wake Forest University School of Medicine, Medical Center Boulevard, Winston-Salem, NC 27157-1040, USA,
| |
Collapse
|
40
|
Degirolamo C, Kelley KL, Wilson MD, Rudel LL. Dietary n-3 LCPUFA from fish oil but not alpha-linolenic acid-derived LCPUFA confers atheroprotection in mice. J Lipid Res 2010; 51:1897-905. [PMID: 20154006 DOI: 10.1194/jlr.m005058] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023] Open
Abstract
The atheroprotective potential of n-3 alpha-linolenic acid (ALA) has not yet been fully determined, even in murine models of atherosclerosis. We tested whether ALA-derived, n-3 long chain polyunsaturated fatty acids (LCPUFA) could offer atheroprotection in a dose-dependent manner. Apolipoprotein B (ApoB)100/100LDLr-/- mice were fed with diets containing two levels of ALA from flaxseed oil for 16 weeks. Fish oil- and cis-monounsaturated-fat-enriched diets were used as positive and negative controls, respectively. The mice fed cis-monounsaturated fat and ALA-enriched diets exhibited equivalent plasma total cholesterol (TPC) and LDL-cholesterol (LDL-c) levels; only mice fed the fish-oil diet had lower TPC and LDL-c concentrations. Plasma LDL-CE fatty acid composition analysis showed that ALA-enriched diets lowered the percentage of atherogenic cholesteryl oleate compared with cis-monounsaturated-fat diet (44% versus 55.6%) but not as efficiently as the fish-oil diet (32.4%). Although both ALA and fish-oil diets equally enriched hepatic phospholipids with eicosapentaenoic acid (EPA) and ALA-enriched diets lowered hepatic cholesteryl ester (CE) levels compared with cis-monounsaturated-fat diet, only fish oil strongly protected from atherosclerosis. These outcomes indicate that dietary n-3 LCPUFA from fish oil and n-3 LCPUFA (mostly EPA) synthesized endogenously from ALA were not equally atheroprotective in these mice.
Collapse
Affiliation(s)
- Chiara Degirolamo
- Department of Pathology, Wake Forest University School of Medicine, Winston-Salem, NC, USA
| | | | | | | |
Collapse
|
41
|
Boyanovsky BB, Li X, Shridas P, Sunkara M, Morris AJ, Webb NR. Bioactive products generated by group V sPLA(2) hydrolysis of LDL activate macrophages to secrete pro-inflammatory cytokines. Cytokine 2010; 50:50-7. [PMID: 20138782 DOI: 10.1016/j.cyto.2009.12.009] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2009] [Revised: 12/16/2009] [Accepted: 12/18/2009] [Indexed: 01/02/2023]
Abstract
OBJECTIVE Previous studies have established that hydrolysis of LDL by Group V secretory phospholipase A(2) (GV sPLA(2)) generates a modified particle capable of inducing macrophage foam cell formation. The aim of the present study was to determine whether GV sPLA(2)-hydrolyzed LDL (GV-LDL) produces pro-atherogenic effects in macrophages independent of cholesterol accumulation. METHODS AND RESULTS J-774 cells incubated with GV-LDL produced more TNF-alpha and IL-6 compared to cells incubated with control-LDL. Indirect immunofluorescence showed that GV-LDL but not control-LDL induced nuclear translocation of NFkappaB. Inhibitors of NFkappaB activation, effectively blocked cytokine production induced by GV-LDL. Control-LDL and GV-LDL were separated from albumin present in reaction mixtures by ultracentrifugation. The albumin fraction derived from GV-LDL contained 80% of the FFA generated and was more potent than the re-isolated GV-LDL in inducing pro-inflammatory cytokine secretion. Linoleic acid (18:2) and oleic acid (18:1) were the most abundant FFAs generated, whereas newly formed lyso-PCs contained 14:0 (myristic), 16:1 (palmitic), and 18:2 fatty acyl groups. Experiments with synthetic FFA showed that 18:1 induced J-774 cells to secrete TNF-alpha and IL-6. CONCLUSIONS These results indicate that in addition to promoting atherosclerotic lipid accumulation in macrophages, GV sPLA(2) hydrolysis of LDL leads to activation of NFkappaB, a key regulator of inflammation.
Collapse
Affiliation(s)
- Boris B Boyanovsky
- Department of Internal Medicine Endocrinology Division, University of Kentucky, Lexington, 40536, USA.
| | | | | | | | | | | |
Collapse
|
42
|
|
43
|
Cocoa Butter and Safflower Oil Elicit Different Effects on Hepatic Gene Expression and Lipid Metabolism in Rats. Lipids 2009; 44:1011-27. [DOI: 10.1007/s11745-009-3352-0] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2009] [Accepted: 09/18/2009] [Indexed: 01/06/2023]
|