1
|
Dietary fat quality impacts metabolic impairments of type 2 diabetes risk differently in male and female CD-1 ® mice. Br J Nutr 2022; 128:1013-1028. [PMID: 34605388 DOI: 10.1017/s0007114521004001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/24/2023]
Abstract
Metabolic impairments associated with type 2 diabetes, including insulin resistance and loss of glycaemic control, disproportionately impact the elderly. Lifestyle interventions, such as manipulation of dietary fat quality (i.e. fatty acid (FA) composition), have been shown to favourably modulate metabolic health. Yet, whether or not chronic consumption of beneficial FAs can protect against metabolic derangements and disease risk during ageing is not well defined. We sought to evaluate whether long-term dietary supplementation of fish-, dairy- or echium-derived FAs to the average FA profile in a U.S. American diet may offset metabolic impairments in males and females during ageing. One-month-old CD-1® mice were fed isoenergetic, high-fat (40 %) diets with the fat content composed of either 100 % control fat blend (CO) or 70 % CO with 30 % fish oil, dairy fat or echium oil for 13 months. Every 3 months, parameters of glucose homoeostasis were evaluated via glucose and insulin tolerance tests. Glucose tolerance improved in males consuming a diet supplemented with fish oil or echium oil as ageing progressed, but not in females. Yet, females were more metabolically protected than males regardless of age. Additionally, Spearman correlations were performed between indices of glucose homoeostasis and previously reported measurements of diet-derived FA content in tissues and colonic bacterial composition, which also revealed sex-specific associations. This study provides evidence that long-term dietary fat quality influences risk factors of metabolic diseases during ageing in a sex-dependent manner; thus, sex is a critical factor to be considered in future dietary strategies to mitigate type 2 diabetes risk.
Collapse
|
2
|
Di Sotto A, Vitalone A, Di Giacomo S. Plant-Derived Nutraceuticals and Immune System Modulation: An Evidence-Based Overview. Vaccines (Basel) 2020; 8:E468. [PMID: 32842641 PMCID: PMC7563161 DOI: 10.3390/vaccines8030468] [Citation(s) in RCA: 36] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2020] [Revised: 08/17/2020] [Accepted: 08/19/2020] [Indexed: 12/15/2022] Open
Abstract
Immunomodulators are agents able to affect the immune system, by boosting the immune defences to improve the body reaction against infectious or exogenous injuries, or suppressing the abnormal immune response occurring in immune disorders. Moreover, immunoadjuvants can support immune system acting on nonimmune targets, thus improving the immune response. The modulation of inflammatory pathways and microbiome can also contribute to control the immune function. Some plant-based nutraceuticals have been studied as possible immunomodulating agents due to their multiple and pleiotropic effects. Being usually more tolerable than pharmacological treatments, their adjuvant contribution is approached as a desirable nutraceutical strategy. In the present review, the up to date knowledge about the immunomodulating properties of polysaccharides, fatty acids and labdane diterpenes have been analyzed, in order to give scientific basic and clinical evidence to support their practical use. Since promising evidence in preclinical studies, limited and sometimes confusing results have been highlighted in clinical trials, likely due to low methodological quality and lacking standardization. More investigations of high quality and specificity are required to describe in depth the usefulness of these plant-derived nutraceuticals in the immune system modulation, for health promoting and disease preventing purposes.
Collapse
Affiliation(s)
- Antonella Di Sotto
- Department of Physiology and Pharmacology, Sapienza University of Rome, P.le Aldo Moro 5, 00185 Rome, Italy;
| | - Annabella Vitalone
- Department of Physiology and Pharmacology, Sapienza University of Rome, P.le Aldo Moro 5, 00185 Rome, Italy;
| | | |
Collapse
|
3
|
Aranaz P, Zabala M, Romo-Hualde A, Navarro-Herrera D, López-Yoldi M, Vizmanos JL, Martínez JA, Milagro FI, González-Navarro CJ. A combination of borage seed oil and quercetin reduces fat accumulation and improves insulin sensitivity in obese rats. Food Funct 2020; 11:4512-4524. [PMID: 32391533 DOI: 10.1039/d0fo00504e] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
The metabolic properties of omega-6 fatty acid consumption are being increasingly accepted. We had previously observed that supplementation with a borage seed oil (BSO), as a source of linoleic (18:2n-6; LA) and gamma-linolenic (18:3n-6; GLA) acids, reduces body weight and visceral adiposity and improves insulin sensitivity in a diet-induced obesity model of Wistar rats. Here, it was investigated whether the anti-obesogenic properties of BSO could be maintained in a pre-obese model of rats, and if these effects are enhanced by a combination with low doses of quercetin, together with its potential role in the regulation of the adipocyte biology. The combination of BSO and quercetin during 8 weeks was able to ameliorate glucose intolerance and insulin resistance, and to improve liver steatosis. Although no effects were observed on body weight, animals supplemented with this combination exhibited a lower proportion of visceral adiposity. In addition, in vitro differentiation of epididymal adipose-precursor cells of the BSO-treated animals exhibited a down-regulation of Fasn, Glut4, Pparg and Srebp1 genes, in comparison with the control group. Finally, in vitro evaluation of the components of BSO demonstrated that the anti-adipogenic activity of quercetin was significantly potentiated by the combination with both LA and GLA through the down-regulation of different adipogenesis-key genes in 3T3-L1 cells. All these data suggest that omega-6 fatty acids LA and GLA, and their natural sources such as BSO, could be combined with quercetin to potentiate their effects in the prevention of the excess of adiposity and the insulin resistance.
Collapse
Affiliation(s)
- Paula Aranaz
- Center for Nutrition Research, School of Pharmacy and Nutrition, University of Navarra, Spain.
| | | | | | | | | | | | | | | | | |
Collapse
|
4
|
Piper K, Garelnabi M. Eicosanoids: Atherosclerosis and cardiometabolic health. J Clin Transl Endocrinol 2020; 19:100216. [PMID: 32071878 PMCID: PMC7013337 DOI: 10.1016/j.jcte.2020.100216] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2019] [Revised: 01/22/2020] [Accepted: 02/01/2020] [Indexed: 02/08/2023] Open
Abstract
Cardiovascular diseases (CVD) have been the leading causes of death in the U.S. for nearly a century. Numerous studies have linked eicosanoids to cardiometabolic disease. Objectives and Methods: This review summaries recent advances and innovative research in eicosanoids and CVD. Numerous review articles and their original human or animal studies were assessed in the relevant and recent studies. OUTCOME We identified and discussed recent trends in eicosanoids known for their roles in CVD. Their subsequent relationships were assessed for any possible implications associated with consumption of different dietary lipids, essentially omega fatty acids. Eicosanoids have been heavily sought after over recent decades for their direct role in mediating the enhancement and resolution of acute immune responses. Given the short half-life of these oxidized lipid metabolites, studies on atherosclerosis have had to rely on the metabolites that are actively involved in eicosanoid production, signaling or redox reactions as markers for atherosclerosis-related molecular behaviors. CONCLUSION Further investigations expending current knowledge, should be applied to narrow the specific class and species of eicosanoids responsible for inciting inflammation especially in the context of recent clinical studies assessing the role of dietary lipid in cardiovascular diseases.
Collapse
|
5
|
Yu F, Tie Y, Zhang Y, Wang Z, Yu L, Zhong L, Zhang C. Circular RNA expression profiles and bioinformatic analysis in coronary heart disease. Epigenomics 2020; 12:439-454. [PMID: 32043895 DOI: 10.2217/epi-2019-0369] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
Aim: We aimed to identify the expression profile and role of circular RNAs (circRNAs) in coronary heart disease (CHD). Materials & methods: We performed sequence analysis of circRNAs in peripheral blood mononuclear cells of 70 CHD patients and 30 controls. Eight selected circRNAs were validated using quantitative real-time polymerase chain reaction (qRT-PCR) in human atherosclerotic coronary arteries. Results: In total, 2283 downregulated and 85 upregulated circRNAs were identified in CHD. Parental genes of top 100 dysregulated-circRNAs are related to metabolism and protein modification, and 12 circRNAs might upregulate their CHD-related parental genes through miRNA sponges. Of the eight circRNAs validated in atherosclerotic coronary arteries by qRT-PCR, six were consistent with sequencing results of peripheral blood mononuclear cells. Conclusion: As potential ceRNAs, dysregulated circRNAs may be involved in CHD pathophysiology.
Collapse
Affiliation(s)
- Fangpu Yu
- The Key Laboratory of Cardiovascular Remodeling & Function Research, Chinese Ministry of Education, Chinese Ministry of Health & Chinese Academy of Medical Sciences, & The State & Shandong Province Joint Key Laboratory of Translational Cardiovascular Medicine, Qilu Hospital of Shandong University, Jinan 250012, Shandong, PR China
| | - Yuanyuan Tie
- The Key Laboratory of Cardiovascular Remodeling & Function Research, Chinese Ministry of Education, Chinese Ministry of Health & Chinese Academy of Medical Sciences, & The State & Shandong Province Joint Key Laboratory of Translational Cardiovascular Medicine, Qilu Hospital of Shandong University, Jinan 250012, Shandong, PR China
| | - Ya Zhang
- The Key Laboratory of Cardiovascular Remodeling & Function Research, Chinese Ministry of Education, Chinese Ministry of Health & Chinese Academy of Medical Sciences, & The State & Shandong Province Joint Key Laboratory of Translational Cardiovascular Medicine, Qilu Hospital of Shandong University, Jinan 250012, Shandong, PR China
| | - Zunzhe Wang
- The Key Laboratory of Cardiovascular Remodeling & Function Research, Chinese Ministry of Education, Chinese Ministry of Health & Chinese Academy of Medical Sciences, & The State & Shandong Province Joint Key Laboratory of Translational Cardiovascular Medicine, Qilu Hospital of Shandong University, Jinan 250012, Shandong, PR China
| | - Liwen Yu
- The Key Laboratory of Cardiovascular Remodeling & Function Research, Chinese Ministry of Education, Chinese Ministry of Health & Chinese Academy of Medical Sciences, & The State & Shandong Province Joint Key Laboratory of Translational Cardiovascular Medicine, Qilu Hospital of Shandong University, Jinan 250012, Shandong, PR China
| | - Lin Zhong
- The Cardiology Department of Yuhuangding Hospital, Qingdao University School of Medicine, Yantai 264000, Shandong, PR China
| | - Cheng Zhang
- The Key Laboratory of Cardiovascular Remodeling & Function Research, Chinese Ministry of Education, Chinese Ministry of Health & Chinese Academy of Medical Sciences, & The State & Shandong Province Joint Key Laboratory of Translational Cardiovascular Medicine, Qilu Hospital of Shandong University, Jinan 250012, Shandong, PR China
| |
Collapse
|
6
|
Fan R, Kim J, You M, Giraud D, Toney AM, Shin SH, Kim SY, Borkowski K, Newman JW, Chung S. α-Linolenic acid-enriched butter attenuated high fat diet-induced insulin resistance and inflammation by promoting bioconversion of n-3 PUFA and subsequent oxylipin formation. J Nutr Biochem 2020; 76:108285. [PMID: 31760228 PMCID: PMC6995772 DOI: 10.1016/j.jnutbio.2019.108285] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2019] [Revised: 09/01/2019] [Accepted: 11/04/2019] [Indexed: 12/18/2022]
Abstract
α-Linolenic acid (ALA) is an essential fatty acid and the precursor for long-chain n-3 PUFA. However, biosynthesis of n-3 PUFA is limited in a Western diet likely due to an overabundance of n-6 PUFA. We hypothesized that dietary reduction of n-6/n-3 PUFA ratio is sufficient to promote the biosynthesis of long-chain n-3 PUFA, leading to an attenuation of high fat (HF) diet-induced obesity and inflammation. C57BL/6 J mice were fed a HF diet from ALA-enriched butter (n3Bu, n-6/n-3=1) in comparison with isocaloric HF diets from either conventional butter lacking both ALA and LA (Bu, n-6/n-3=6), or margarine containing a similar amount of ALA and abundant LA (Ma, n-6/n-3=6). Targeted lipidomic analyses revealed that n3Bu feeding promoted the bioconversion of long-chain n-3 PUFA and their oxygenated metabolites (oxylipins) derived from ALA and EPA. The n3Bu supplementation attenuated hepatic TG accumulation and adipose tissue inflammation, resulting in improved insulin sensitivity. Decreased inflammation by n3Bu feeding was attributed to the suppression of NF-κB activation and M1 macrophage polarization. Collectively, our work suggests that dietary reduction of the n-6/n-3 PUFA ratio, as well as total n-3 PUFA consumed, is a crucial determinant that facilitates n-3 PUFA biosynthesis and subsequent lipidomic modifications, thereby conferring metabolic benefits against obesity-induced inflammation and insulin resistance.
Collapse
Affiliation(s)
- Rong Fan
- Department of Nutrition and Health Sciences, University of Nebraska, Lincoln, NE
| | - Judy Kim
- Department of Nutrition and Health Sciences, University of Nebraska, Lincoln, NE
| | - Mikyoung You
- Department of Nutrition and Health Sciences, University of Nebraska, Lincoln, NE
| | - David Giraud
- Department of Nutrition and Health Sciences, University of Nebraska, Lincoln, NE
| | - Ashley M Toney
- Department of Nutrition and Health Sciences, University of Nebraska, Lincoln, NE
| | - Seung-Ho Shin
- Sunseo Omega Inc, University of Nebraska Innovation Campus, Lincoln, NE
| | - So-Youn Kim
- Olson Center for Women's Health, Department of Obstetrics and Gynecology, and Fred & Pamela Buffett Cancer Center, University of Nebraska Medical Center, Omaha, NE
| | - Kamil Borkowski
- West Coast Metabolomics Center, Genome Center, University of California Davis, Davis, CA
| | - John W Newman
- West Coast Metabolomics Center, Genome Center, University of California Davis, Davis, CA; Obesity and Metabolism Research Unit, USDA-ARS-WHNRC, Davis, CA
| | - Soonkyu Chung
- Department of Nutrition and Health Sciences, University of Nebraska, Lincoln, NE.
| |
Collapse
|
7
|
Wang Z, Sequeira RC, Zabalawi M, Madenspacher J, Boudyguina E, Ou T, Nelson JM, Nie Y, Zhao Q, Fessler MB, Zhu X. Myeloid atg5 deletion impairs n-3 PUFA-mediated atheroprotection. Atherosclerosis 2020; 295:8-17. [PMID: 31978760 DOI: 10.1016/j.atherosclerosis.2020.01.004] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/10/2019] [Revised: 11/13/2019] [Accepted: 01/10/2020] [Indexed: 12/13/2022]
Abstract
BACKGROUND AND AIMS Dietary long-chain (≥20 carbons) n-3 polyunsaturated fatty acids (PUFAs) reduce atherosclerosis and enhance macrophage autophagy activation. How macrophage autophagy impacts atherosclerotic progression, particularly when comparing dietary n-3 PUFA supplementation vs. saturated fat feeding, is unknown. METHODS We generated myeloid-specific autophagy-deficient and control mice in the Ldlr-/- background by transplanting bone marrow from myeloid-specific autophagy-related (atg) 5 knockout mice and wild type controls into irradiated Ldlr-/- recipients. After 7 weeks for recovery from radiation, mice were fed an atherogenic diet containing 0.2% cholesterol and 20% calories as palm oil (PO diet), or 10% calories as PO plus 10% calories as fish oil (FO diet) for 16 weeks. RESULTS Compared to PO, FO significantly reduced plasma cholesterol, triglyceride, hepatic neutral lipid, and aortic caspase-1 cleavage, but increased aortic efferocytosis, leading to attenuated atherosclerosis in Ldlr-/- mice receiving wild type bone marrow. Myeloid atg5 deletion had little impact on plasma lipid concentrations and hepatic neutral lipid content, regardless of diet. Myeloid atg5 deletion increased aortic caspase-1 cleavage, decreased aortic efferocytosis and worsened atherosclerosis only in the FO-fed Ldlr-/- mice. CONCLUSIONS Deficient myeloid autophagy significantly attenuated FO-induced atheroprotection, suggesting that dietary n-3 PUFAs reduce atherosclerosis, in part, by activation of macrophage autophagy.
Collapse
Affiliation(s)
- Zhan Wang
- Department of Internal Medicine, Section on Molecular Medicine, Wake Forest School of Medicine, Winston-Salem, NC, 27157, USA
| | - Russel C Sequeira
- Department of Internal Medicine, Section on Molecular Medicine, Wake Forest School of Medicine, Winston-Salem, NC, 27157, USA
| | - Manal Zabalawi
- Department of Internal Medicine, Section on Molecular Medicine, Wake Forest School of Medicine, Winston-Salem, NC, 27157, USA
| | - Jennifer Madenspacher
- Immunity, Inflammation and Disease Laboratory, National Institute of Environmental Health Sciences, NIH, Research Triangle Park, NC, 27709, USA
| | - Elena Boudyguina
- Department of Internal Medicine, Section on Molecular Medicine, Wake Forest School of Medicine, Winston-Salem, NC, 27157, USA
| | - Tiantong Ou
- Department of Internal Medicine, Section on Molecular Medicine, Wake Forest School of Medicine, Winston-Salem, NC, 27157, USA
| | - Jonathan M Nelson
- Department of Internal Medicine, Section on Molecular Medicine, Wake Forest School of Medicine, Winston-Salem, NC, 27157, USA
| | - Yan Nie
- Department of Internal Medicine, Section on Molecular Medicine, Wake Forest School of Medicine, Winston-Salem, NC, 27157, USA
| | - Qingxia Zhao
- Department of Internal Medicine, Section on Molecular Medicine, Wake Forest School of Medicine, Winston-Salem, NC, 27157, USA
| | - Michael B Fessler
- Immunity, Inflammation and Disease Laboratory, National Institute of Environmental Health Sciences, NIH, Research Triangle Park, NC, 27709, USA
| | - Xuewei Zhu
- Department of Internal Medicine, Section on Molecular Medicine, Wake Forest School of Medicine, Winston-Salem, NC, 27157, USA; Department of Microbiology and Immunology, Wake Forest School of Medicine, Winston-Salem, NC, 27157, USA.
| |
Collapse
|
8
|
Navarro-Herrera D, Aranaz P, Eder-Azanza L, Zabala M, Romo-Hualde A, Hurtado C, Calavia D, López-Yoldi M, Martínez JA, González-Navarro CJ, Vizmanos JL. Borago officinalis seed oil (BSO), a natural source of omega-6 fatty acids, attenuates fat accumulation by activating peroxisomal beta-oxidation both in C. elegans and in diet-induced obese rats. Food Funct 2018; 9:4340-4351. [PMID: 30043014 DOI: 10.1039/c8fo00423d] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2023]
Abstract
Obesity is a medical condition with increasing prevalence, characterized by an accumulation of excess fat that could be improved using some bioactive compounds. However, many of these compounds with in vitro activity fail to respond in vivo, probably due to the sophistication of the physiological energy regulatory networks. In this context, C. elegans has emerged as a plausible model for the identification and characterization of the effect of such compounds on fat storage in a complete organism. However, the results obtained in such a simple model are not easily extrapolated to more complex organisms such as mammals, which hinders its application in the short term. Therefore, it is necessary to obtain new experimental data about the evolutionary conservation of the mechanisms of fat loss between worms and mammals. Previously, we found that some omega-6 fatty acids promote fat loss in C. elegans by up-regulation of peroxisomal fatty acid β-oxidation in an omega-3 independent manner. In this work, we prove that the omega-6 fatty acids' effects on worms are also seen when they are supplemented with a natural omega-6 source (borage seed oil, BSO). Additionally, we explore the anti-obesity effects of two doses of BSO in a diet-induced obesity rat model, validating the up-regulation of peroxisomal fatty acid β-oxidation. The supplementation with BSO significantly reduces body weight gain and energy efficiency and prevents white adipose tissue accumulation without affecting food intake. Moreover, BSO also increases serum HDL-cholesterol levels, improves insulin resistance and promotes the down-regulation of Cebpa, an adipogenesis-related gene. Therefore, we conclude that the effects of omega-6 fatty acids are highly conserved between worms and obesity-induced mammals, so these compounds could be considered to treat or prevent obesity-related disorders.
Collapse
Affiliation(s)
- David Navarro-Herrera
- University of Navarra, School of Sciences, Department of Biochemistry and Genetics, Pamplona, Spain.
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
9
|
Lowering the n-6/n-3 PUFAs ratio inhibits the formation of THP-1 macrophage-derived foam cell. Lipids Health Dis 2018; 17:125. [PMID: 29801502 PMCID: PMC5970467 DOI: 10.1186/s12944-018-0772-y] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2018] [Accepted: 05/10/2018] [Indexed: 12/22/2022] Open
Abstract
Background The balance between n-6 and n-3 PUFAs is an important determinant in the risk for cardiovascular disease. The study was to investigate the influence of the n-6 and n-3 PUFAs ratio on the formation of THP-1 monocyte-derived foam cells and explore the probable mechanism of anti-atherosclerosis. Methods THP-1 monocyte cells were cultured with PMA and ox-LDL to establish a foam-cell model, while treated with different ratios of n-6 to n-3 PUFAs for 48 h. The cholesterol of foam cells was measured by a cholesterol assay kit. The levels of IL-6 and TNFα in supernatant were detected with ELISA methods. The expressions of CD36, ABCA1, ACAT1, PPARγ and LXRα mRNA were detected with real-time PCR. Results Compared with the foam cell model group, the low and middle ratio of n-6 to n-3 PUFAs groups decreased the intracellular concentration of cholesterol (P < 0.01), but the high n-6/n-3 PUFAs ratio did not. Fatty acids decreased the level of IL-6 and TNFα in supernatant in a ratio-dependent manner. Fatty acids treatment also decreased the expressions of CD36、ACTA1、PPARγ、LXRα mRNA in a ratio-dependent manner. Conclusions Lowering the ratios of n-6 to n-3 PUFAs can decrease the secretion of inflammatory cytokines then reduce the expressions of CD36 and ACAT1 mRNA. As well, it can decrease the expressions of CD36 mRNA through the PPARγ pathway. This leads to less cholesterol ingestion into the cells and decreased synthesis of cholesteryl ester, which inhibits the formation of the foam cells, further preventing the occurrence and development of atherosclerosis.
Collapse
|
10
|
Gromovsky AD, Schugar RC, Brown AL, Helsley RN, Burrows AC, Ferguson D, Zhang R, Sansbury BE, Lee RG, Morton RE, Allende DS, Parks JS, Spite M, Brown JM. Δ-5 Fatty Acid Desaturase FADS1 Impacts Metabolic Disease by Balancing Proinflammatory and Proresolving Lipid Mediators. Arterioscler Thromb Vasc Biol 2018; 38:218-231. [PMID: 29074585 PMCID: PMC5746431 DOI: 10.1161/atvbaha.117.309660] [Citation(s) in RCA: 67] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2017] [Accepted: 10/08/2017] [Indexed: 02/07/2023]
Abstract
OBJECTIVE Human genetic variants near the FADS (fatty acid desaturase) gene cluster (FADS1-2-3) are strongly associated with cardiometabolic traits including dyslipidemia, fatty liver, type 2 diabetes mellitus, and coronary artery disease. However, mechanisms underlying these genetic associations are unclear. APPROACH AND RESULTS Here, we specifically investigated the physiological role of the Δ-5 desaturase FADS1 in regulating diet-induced cardiometabolic phenotypes by treating hyperlipidemic LDLR (low-density lipoprotein receptor)-null mice with antisense oligonucleotides targeting the selective knockdown of Fads1. Fads1 knockdown resulted in striking reorganization of both ω-6 and ω-3 polyunsaturated fatty acid levels and their associated proinflammatory and proresolving lipid mediators in a highly diet-specific manner. Loss of Fads1 activity promoted hepatic inflammation and atherosclerosis, yet was associated with suppression of hepatic lipogenesis. Fads1 knockdown in isolated macrophages promoted classic M1 activation, whereas suppressing alternative M2 activation programs, and also altered systemic and tissue inflammatory responses in vivo. Finally, the ability of Fads1 to reciprocally regulate lipogenesis and inflammation may rely in part on its role as an effector of liver X receptor signaling. CONCLUSIONS These results position Fads1 as an underappreciated regulator of inflammation initiation and resolution, and suggest that endogenously synthesized arachidonic acid and eicosapentaenoic acid are key determinates of inflammatory disease progression and liver X receptor signaling.
Collapse
MESH Headings
- Animals
- Aorta/enzymology
- Aorta/pathology
- Aortic Diseases/enzymology
- Aortic Diseases/genetics
- Aortic Diseases/pathology
- Arachidonic Acid/metabolism
- Atherosclerosis/enzymology
- Atherosclerosis/genetics
- Atherosclerosis/pathology
- Cells, Cultured
- Delta-5 Fatty Acid Desaturase
- Disease Models, Animal
- Dyslipidemias/enzymology
- Dyslipidemias/genetics
- Dyslipidemias/pathology
- Eicosapentaenoic Acid/metabolism
- Fatty Acid Desaturases/genetics
- Fatty Acid Desaturases/metabolism
- Inflammation/enzymology
- Inflammation/genetics
- Inflammation/pathology
- Inflammation Mediators/metabolism
- Lipogenesis
- Liver/metabolism
- Liver X Receptors/metabolism
- Macrophage Activation
- Macrophages, Peritoneal/enzymology
- Macrophages, Peritoneal/pathology
- Mice, Inbred C57BL
- Mice, Knockout
- Oligonucleotides, Antisense/genetics
- Oligonucleotides, Antisense/metabolism
- Plaque, Atherosclerotic
- Receptors, LDL/deficiency
- Receptors, LDL/genetics
Collapse
Affiliation(s)
- Anthony D Gromovsky
- From the Department of Cellular and Molecular Medicine, Lerner Research Institute (A.D.G., R.C.S., A.L.B., R.N.H., A.C.B., D.F., R.Z., R.E.M., J.M.B.) and Department of Anatomical Pathology (D.S.A.), Cleveland Clinic, OH; Center for Experimental Therapeutics and Reperfusion Injury, Department of Anesthesiology, Perioperative and Pain Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA (B.E.S., M.S.); Cardiovascular Group, Antisense Drug Discovery, Ionis Pharmaceuticals, Inc, Carlsbad, CA (R.G.L.); and Department of Internal Medicine-Section on Molecular Medicine, Wake Forest School of Medicine, Winston-Salem, NC (J.S.P.)
| | - Rebecca C Schugar
- From the Department of Cellular and Molecular Medicine, Lerner Research Institute (A.D.G., R.C.S., A.L.B., R.N.H., A.C.B., D.F., R.Z., R.E.M., J.M.B.) and Department of Anatomical Pathology (D.S.A.), Cleveland Clinic, OH; Center for Experimental Therapeutics and Reperfusion Injury, Department of Anesthesiology, Perioperative and Pain Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA (B.E.S., M.S.); Cardiovascular Group, Antisense Drug Discovery, Ionis Pharmaceuticals, Inc, Carlsbad, CA (R.G.L.); and Department of Internal Medicine-Section on Molecular Medicine, Wake Forest School of Medicine, Winston-Salem, NC (J.S.P.)
| | - Amanda L Brown
- From the Department of Cellular and Molecular Medicine, Lerner Research Institute (A.D.G., R.C.S., A.L.B., R.N.H., A.C.B., D.F., R.Z., R.E.M., J.M.B.) and Department of Anatomical Pathology (D.S.A.), Cleveland Clinic, OH; Center for Experimental Therapeutics and Reperfusion Injury, Department of Anesthesiology, Perioperative and Pain Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA (B.E.S., M.S.); Cardiovascular Group, Antisense Drug Discovery, Ionis Pharmaceuticals, Inc, Carlsbad, CA (R.G.L.); and Department of Internal Medicine-Section on Molecular Medicine, Wake Forest School of Medicine, Winston-Salem, NC (J.S.P.)
| | - Robert N Helsley
- From the Department of Cellular and Molecular Medicine, Lerner Research Institute (A.D.G., R.C.S., A.L.B., R.N.H., A.C.B., D.F., R.Z., R.E.M., J.M.B.) and Department of Anatomical Pathology (D.S.A.), Cleveland Clinic, OH; Center for Experimental Therapeutics and Reperfusion Injury, Department of Anesthesiology, Perioperative and Pain Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA (B.E.S., M.S.); Cardiovascular Group, Antisense Drug Discovery, Ionis Pharmaceuticals, Inc, Carlsbad, CA (R.G.L.); and Department of Internal Medicine-Section on Molecular Medicine, Wake Forest School of Medicine, Winston-Salem, NC (J.S.P.)
| | - Amy C Burrows
- From the Department of Cellular and Molecular Medicine, Lerner Research Institute (A.D.G., R.C.S., A.L.B., R.N.H., A.C.B., D.F., R.Z., R.E.M., J.M.B.) and Department of Anatomical Pathology (D.S.A.), Cleveland Clinic, OH; Center for Experimental Therapeutics and Reperfusion Injury, Department of Anesthesiology, Perioperative and Pain Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA (B.E.S., M.S.); Cardiovascular Group, Antisense Drug Discovery, Ionis Pharmaceuticals, Inc, Carlsbad, CA (R.G.L.); and Department of Internal Medicine-Section on Molecular Medicine, Wake Forest School of Medicine, Winston-Salem, NC (J.S.P.)
| | - Daniel Ferguson
- From the Department of Cellular and Molecular Medicine, Lerner Research Institute (A.D.G., R.C.S., A.L.B., R.N.H., A.C.B., D.F., R.Z., R.E.M., J.M.B.) and Department of Anatomical Pathology (D.S.A.), Cleveland Clinic, OH; Center for Experimental Therapeutics and Reperfusion Injury, Department of Anesthesiology, Perioperative and Pain Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA (B.E.S., M.S.); Cardiovascular Group, Antisense Drug Discovery, Ionis Pharmaceuticals, Inc, Carlsbad, CA (R.G.L.); and Department of Internal Medicine-Section on Molecular Medicine, Wake Forest School of Medicine, Winston-Salem, NC (J.S.P.)
| | - Renliang Zhang
- From the Department of Cellular and Molecular Medicine, Lerner Research Institute (A.D.G., R.C.S., A.L.B., R.N.H., A.C.B., D.F., R.Z., R.E.M., J.M.B.) and Department of Anatomical Pathology (D.S.A.), Cleveland Clinic, OH; Center for Experimental Therapeutics and Reperfusion Injury, Department of Anesthesiology, Perioperative and Pain Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA (B.E.S., M.S.); Cardiovascular Group, Antisense Drug Discovery, Ionis Pharmaceuticals, Inc, Carlsbad, CA (R.G.L.); and Department of Internal Medicine-Section on Molecular Medicine, Wake Forest School of Medicine, Winston-Salem, NC (J.S.P.)
| | - Brian E Sansbury
- From the Department of Cellular and Molecular Medicine, Lerner Research Institute (A.D.G., R.C.S., A.L.B., R.N.H., A.C.B., D.F., R.Z., R.E.M., J.M.B.) and Department of Anatomical Pathology (D.S.A.), Cleveland Clinic, OH; Center for Experimental Therapeutics and Reperfusion Injury, Department of Anesthesiology, Perioperative and Pain Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA (B.E.S., M.S.); Cardiovascular Group, Antisense Drug Discovery, Ionis Pharmaceuticals, Inc, Carlsbad, CA (R.G.L.); and Department of Internal Medicine-Section on Molecular Medicine, Wake Forest School of Medicine, Winston-Salem, NC (J.S.P.)
| | - Richard G Lee
- From the Department of Cellular and Molecular Medicine, Lerner Research Institute (A.D.G., R.C.S., A.L.B., R.N.H., A.C.B., D.F., R.Z., R.E.M., J.M.B.) and Department of Anatomical Pathology (D.S.A.), Cleveland Clinic, OH; Center for Experimental Therapeutics and Reperfusion Injury, Department of Anesthesiology, Perioperative and Pain Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA (B.E.S., M.S.); Cardiovascular Group, Antisense Drug Discovery, Ionis Pharmaceuticals, Inc, Carlsbad, CA (R.G.L.); and Department of Internal Medicine-Section on Molecular Medicine, Wake Forest School of Medicine, Winston-Salem, NC (J.S.P.)
| | - Richard E Morton
- From the Department of Cellular and Molecular Medicine, Lerner Research Institute (A.D.G., R.C.S., A.L.B., R.N.H., A.C.B., D.F., R.Z., R.E.M., J.M.B.) and Department of Anatomical Pathology (D.S.A.), Cleveland Clinic, OH; Center for Experimental Therapeutics and Reperfusion Injury, Department of Anesthesiology, Perioperative and Pain Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA (B.E.S., M.S.); Cardiovascular Group, Antisense Drug Discovery, Ionis Pharmaceuticals, Inc, Carlsbad, CA (R.G.L.); and Department of Internal Medicine-Section on Molecular Medicine, Wake Forest School of Medicine, Winston-Salem, NC (J.S.P.)
| | - Daniela S Allende
- From the Department of Cellular and Molecular Medicine, Lerner Research Institute (A.D.G., R.C.S., A.L.B., R.N.H., A.C.B., D.F., R.Z., R.E.M., J.M.B.) and Department of Anatomical Pathology (D.S.A.), Cleveland Clinic, OH; Center for Experimental Therapeutics and Reperfusion Injury, Department of Anesthesiology, Perioperative and Pain Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA (B.E.S., M.S.); Cardiovascular Group, Antisense Drug Discovery, Ionis Pharmaceuticals, Inc, Carlsbad, CA (R.G.L.); and Department of Internal Medicine-Section on Molecular Medicine, Wake Forest School of Medicine, Winston-Salem, NC (J.S.P.)
| | - John S Parks
- From the Department of Cellular and Molecular Medicine, Lerner Research Institute (A.D.G., R.C.S., A.L.B., R.N.H., A.C.B., D.F., R.Z., R.E.M., J.M.B.) and Department of Anatomical Pathology (D.S.A.), Cleveland Clinic, OH; Center for Experimental Therapeutics and Reperfusion Injury, Department of Anesthesiology, Perioperative and Pain Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA (B.E.S., M.S.); Cardiovascular Group, Antisense Drug Discovery, Ionis Pharmaceuticals, Inc, Carlsbad, CA (R.G.L.); and Department of Internal Medicine-Section on Molecular Medicine, Wake Forest School of Medicine, Winston-Salem, NC (J.S.P.)
| | - Matthew Spite
- From the Department of Cellular and Molecular Medicine, Lerner Research Institute (A.D.G., R.C.S., A.L.B., R.N.H., A.C.B., D.F., R.Z., R.E.M., J.M.B.) and Department of Anatomical Pathology (D.S.A.), Cleveland Clinic, OH; Center for Experimental Therapeutics and Reperfusion Injury, Department of Anesthesiology, Perioperative and Pain Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA (B.E.S., M.S.); Cardiovascular Group, Antisense Drug Discovery, Ionis Pharmaceuticals, Inc, Carlsbad, CA (R.G.L.); and Department of Internal Medicine-Section on Molecular Medicine, Wake Forest School of Medicine, Winston-Salem, NC (J.S.P.)
| | - J Mark Brown
- From the Department of Cellular and Molecular Medicine, Lerner Research Institute (A.D.G., R.C.S., A.L.B., R.N.H., A.C.B., D.F., R.Z., R.E.M., J.M.B.) and Department of Anatomical Pathology (D.S.A.), Cleveland Clinic, OH; Center for Experimental Therapeutics and Reperfusion Injury, Department of Anesthesiology, Perioperative and Pain Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA (B.E.S., M.S.); Cardiovascular Group, Antisense Drug Discovery, Ionis Pharmaceuticals, Inc, Carlsbad, CA (R.G.L.); and Department of Internal Medicine-Section on Molecular Medicine, Wake Forest School of Medicine, Winston-Salem, NC (J.S.P.).
| |
Collapse
|
11
|
Morrow NM, Huff MW. Knockdown of Δ-5 Fatty Acid Desaturase Is More Than Just a Fad. Arterioscler Thromb Vasc Biol 2017; 38:6-8. [PMID: 29282245 DOI: 10.1161/atvbaha.117.310382] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Affiliation(s)
- Nadya M Morrow
- From the Robarts Research Institute, London, Ontario, Canada; and Department of Medicine and Department of Biochemistry, Schulich School of Medicine and Dentistry, The University of Western Ontario, London, Canada
| | - Murray W Huff
- From the Robarts Research Institute, London, Ontario, Canada; and Department of Medicine and Department of Biochemistry, Schulich School of Medicine and Dentistry, The University of Western Ontario, London, Canada.
| |
Collapse
|
12
|
Quinn WJ, Wan M, Shewale SV, Gelfer R, Rader DJ, Birnbaum MJ, Titchenell PM. mTORC1 stimulates phosphatidylcholine synthesis to promote triglyceride secretion. J Clin Invest 2017; 127:4207-4215. [PMID: 29035283 DOI: 10.1172/jci96036] [Citation(s) in RCA: 64] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2017] [Accepted: 09/05/2017] [Indexed: 12/13/2022] Open
Abstract
Liver triacylglycerol (TAG) synthesis and secretion are closely linked to nutrient availability. After a meal, hepatic TAG formation from fatty acids is decreased, largely due to a reduction in circulating free fatty acids (FFA). Despite the postprandial decrease in FFA-driven esterification and oxidation, VLDL-TAG secretion is maintained to support peripheral lipid delivery and metabolism. The regulatory mechanisms underlying the postprandial control of VLDL-TAG secretion remain unclear. Here, we demonstrated that the mTOR complex 1 (mTORC1) is essential for this sustained VLDL-TAG secretion and lipid homeostasis. In murine models, the absence of hepatic mTORC1 reduced circulating TAG, despite hepatosteatosis, while activation of mTORC1 depleted liver TAG stores. Additionally, mTORC1 promoted TAG secretion by regulating phosphocholine cytidylyltransferase α (CCTα), the rate-limiting enzyme involved in the synthesis of phosphatidylcholine (PC). Increasing PC synthesis in mice lacking mTORC1 rescued hepatosteatosis and restored TAG secretion. These data identify mTORC1 as a major regulator of phospholipid biosynthesis and subsequent VLDL-TAG secretion, leading to increased postprandial TAG secretion.
Collapse
Affiliation(s)
| | - Min Wan
- Institute for Diabetes, Obesity, and Metabolism, and
| | - Swapnil V Shewale
- Cardiovascular Institute, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | | | - Daniel J Rader
- Cardiovascular Institute, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Morris J Birnbaum
- Institute for Diabetes, Obesity, and Metabolism, and.,Internal Medicine, Pfizer Inc., Cambridge, Massachusetts, USA
| | - Paul M Titchenell
- Institute for Diabetes, Obesity, and Metabolism, and.,Department of Physiology Perelman School of Medicine at the University of Pennsylvania, Philadelphia, Pennsylvania, USA
| |
Collapse
|
13
|
Csonka C, Baranyai T, Tiszlavicz L, Fébel H, Szűcs G, Varga ZV, Sárközy M, Puskás LG, Antal O, Siska A, Földesi I, Ferdinandy P, Czakó L, Csont T. Isolated hypercholesterolemia leads to steatosis in the liver without affecting the pancreas. Lipids Health Dis 2017; 16:144. [PMID: 28750643 PMCID: PMC5532767 DOI: 10.1186/s12944-017-0537-z] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2017] [Accepted: 07/13/2017] [Indexed: 02/06/2023] Open
Abstract
Background Lipid accumulation in the liver and pancreas is primarily caused by combined hyperlipidemia. However, the effect of isolated hypercholesterolemia without hypertriglyceridemia is not fully described. Therefore, our aim was to investigate whether hypercholesterolemia alone leads to alterations both in hepatic and pancreatic lipid panel and histology in rats. Methods Male Wistar rats were fed with 2% cholesterol +0.25% cholate-supplemented diet or standard chow for 12 weeks. Blood was collected at weeks 0, 4, 8 and 12 to measure serum cholesterol and triglyceride levels. At week 12, both the pancreas and the liver were isolated for further histological and biochemical analysis. Hepatic and plasma fatty acid composition was assessed by gas chromatography. Expression of mRNA of major enzymes involved in saturated/unsaturated fatty acid synthesis was analyzed by qPCR. In separate experiments serum enzyme activities and insulin levels were measured at week 9. Results At week 12, rats fed with 2% cholesterol +0.25% cholate-supplemented diet were characterized by elevated serum cholesterol (4.09 ± 0.20 vs. 2.89 ± 0.22 mmol/L, *p < 0.05) while triglyceride (2.27 ± 0.05 vs. 2.03 ± 0.03 mmol/L) and glucose levels (5.32 ± 0.14 vs. 5.23 ± 0.10 mmol/L) remained unchanged. Isolated hypercholesterolemia increased hepatic lipid accumulation, hepatic cholesterol (5.86 ± 0.22 vs. 1.60 ± 0.15 ng/g tissue, *p < 0.05) and triglyceride contents (19.28 ± 1.42 vs. 6.78 ± 0.71 ng/g tissue, *p < 0.05), and hepatic nitrotyrosine level (4.07 ± 0.52 vs. 2.59 ± 0.31 ng/mg protein, *p < 0.05). The histology and tissue lipid content of the pancreas was not affected. Serum total protein level, alanine aminotransferase (ALT) and aspartate aminotransferase (AST) activities remained unchanged in response to isolated hypercholesterolemia while serum alkaline phosphatase activity (ALP) significantly increased. Plasma insulin levels did not change in response to isolated hypercholesterolemia suggesting an intact endocrine function of the pancreas. Isolated hypercholesterolemia caused a significantly increased hepatic and serum fatty acid level associated with a marked alteration of fatty acid composition. Hepatic expression of Δ9-desaturase (SCD1) was increased 4.92×, while expression of Δ5-desaturase and Δ6-desaturase were decreased (0.447× and 0.577×, respectively) due to isolated hypercholesterolemia. Conclusions Isolated hypercholesterolemia leads to hepatic steatosis and marked alterations in the hepatic lipid profile without affecting the pancreas. Altered fatty acid profile might mediate harmful effects of cholesterol in the liver.
Collapse
Affiliation(s)
- Csaba Csonka
- Metabolic Diseases and Cell Signaling Research Group, Department of Biochemistry, University of Szeged, Dóm tér 9, Szeged, H-6720, Hungary
| | - Tamás Baranyai
- Metabolic Diseases and Cell Signaling Research Group, Department of Biochemistry, University of Szeged, Dóm tér 9, Szeged, H-6720, Hungary.,1st Department of Internal Medicine, University of Szeged, Szeged, Hungary.,Department of Pharmacology and Pharmacotherapy, Semmelweis University, Budapest, Hungary
| | | | - Hedvig Fébel
- Research Institute for Animal Breeding, Nutrition and Meat Science, Herceghalom, Szeged, Hungary
| | - Gergő Szűcs
- Metabolic Diseases and Cell Signaling Research Group, Department of Biochemistry, University of Szeged, Dóm tér 9, Szeged, H-6720, Hungary.,Department of Physiology, Anatomy and Neuroscience, Faculty of Science and Informatics, University of Szeged, Szeged, Hungary
| | - Zoltán V Varga
- Metabolic Diseases and Cell Signaling Research Group, Department of Biochemistry, University of Szeged, Dóm tér 9, Szeged, H-6720, Hungary.,Department of Pharmacology and Pharmacotherapy, Semmelweis University, Budapest, Hungary
| | - Márta Sárközy
- Metabolic Diseases and Cell Signaling Research Group, Department of Biochemistry, University of Szeged, Dóm tér 9, Szeged, H-6720, Hungary
| | - László G Puskás
- Institute of Genetics, Biological Research Center, Hungarian Academy of Sciences, Szeged, Hungary
| | - Otilia Antal
- Institute of Genetics, Biological Research Center, Hungarian Academy of Sciences, Szeged, Hungary
| | - Andrea Siska
- Department of Laboratory Medicine, Faculty of Medicine, University of Szeged, Szeged, Hungary
| | - Imre Földesi
- Department of Laboratory Medicine, Faculty of Medicine, University of Szeged, Szeged, Hungary
| | - Péter Ferdinandy
- Department of Pharmacology and Pharmacotherapy, Semmelweis University, Budapest, Hungary.,Pharmahungary Group, Szeged, Hungary
| | - László Czakó
- 1st Department of Internal Medicine, University of Szeged, Szeged, Hungary
| | - Tamás Csont
- Metabolic Diseases and Cell Signaling Research Group, Department of Biochemistry, University of Szeged, Dóm tér 9, Szeged, H-6720, Hungary.
| |
Collapse
|
14
|
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
|
15
|
Shewale SV, Brown AL, Bi X, Boudyguina E, Sawyer JK, Alexander-Miller MA, Parks JS. In vivo activation of leukocyte GPR120/FFAR4 by PUFAs has minimal impact on atherosclerosis in LDL receptor knockout mice. J Lipid Res 2016; 58:236-246. [PMID: 27811230 DOI: 10.1194/jlr.m072769] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2016] [Indexed: 01/15/2023] Open
Abstract
G protein-coupled receptor (GPR)120/FFA receptor (FFAR)4 (GPR120/FFAR4) activation by n-3 PUFAs attenuates inflammation, but its impact on atherosclerosis is unknown. We determined whether in vivo activation of leukocyte GPR120/FFAR4 by n-3 versus n-6 PUFAs is atheroprotective. Leukocyte GPR120/FFAR4 WT or KO mice in the LDL receptor KO background were generated by bone marrow transplantation. Mice were fed one of the four atherogenic diets containing 0.2% cholesterol and 10% calories as palm oil (PO) + 10% calories as: 1) PO, 2) fish oil (FO; 20:5 n-3 and 22:6 n-3 enriched), 3) echium oil (EO; 18:4 n-3 enriched), or 4) borage oil (BO; 18:3 n-6 enriched) for 16 weeks. Compared with PO, mice fed BO, EO, and FO had significantly reduced plasma cholesterol, TG, VLDL cholesterol, hepatic neutral lipid, and atherosclerosis that were equivalent for WT and KO mice. In BO-, EO-, and FO-fed mice, but not PO-fed mice, lack of leukocyte GPR120/FFAR4 resulted in neutrophilia, pro-inflammatory Ly6Chi monocytosis, increased aortic root monocyte recruitment, and increased hepatic inflammatory gene expression. In conclusion, leukocyte GPR120 expression has minimal effects on dietary PUFA-induced plasma lipid/lipoprotein reduction and atheroprotection, and there is no distinction between n-3 versus n-6 PUFAs in activating anti-inflammatory effects of leukocyte GPR120/FFAR4 in vivo.
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
| | - Amanda L Brown
- Departments of Internal Medicine/Section on Molecular Medicine, Wake Forest School of Medicine, Winston-Salem, NC 27157
| | - Xin Bi
- Departments of Internal Medicine/Section on Molecular Medicine, 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
| | - Janet K Sawyer
- Departments of Internal Medicine/Section on Molecular Medicine, Wake Forest School of Medicine, Winston-Salem, NC 27157
| | | | - 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
|
16
|
Cao Q, Cui X, Wu R, Zha L, Wang X, Parks JS, Yu L, Shi H, Xue B. Myeloid Deletion of α1AMPK Exacerbates Atherosclerosis in LDL Receptor Knockout (LDLRKO) Mice. Diabetes 2016; 65:1565-76. [PMID: 26822081 PMCID: PMC4878417 DOI: 10.2337/db15-0917] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/03/2015] [Accepted: 12/31/2015] [Indexed: 12/28/2022]
Abstract
Macrophage inflammation marks all stages of atherogenesis, and AMPK is a regulator of macrophage inflammation. We therefore generated myeloid α1AMPK knockout (MAKO) mice on the LDL receptor knockout (LDLRKO) background to investigate whether myeloid deletion of α1AMPK exacerbates atherosclerosis. When fed an atherogenic diet, MAKO/LDLRKO mice displayed exacerbated atherosclerosis compared with LDLRKO mice. To determine the underlying pathophysiological pathways, we characterized macrophage inflammation/chemotaxis and lipid/cholesterol metabolism in MAKO/LDLRKO mice. Myeloid deletion of α1AMPK increased macrophage inflammatory gene expression and enhanced macrophage migration and adhesion to endothelial cells. Remarkably, MAKO/LDLRKO mice also displayed higher composition of circulating chemotaxically active Ly-6C(high) monocytes, enhanced atherosclerotic plaque chemokine expression, and monocyte recruitment into plaques, leading to increased atherosclerotic plaque macrophage content and inflammation. MAKO/LDLRKO mice also exhibited higher plasma LDL and VLDL cholesterol content, increased circulating apolipoprotein B (apoB) levels, and higher liver apoB expression. We conclude that macrophage α1AMPK deficiency promotes atherogenesis in LDLRKO mice and is associated with enhanced macrophage inflammation and hypercholesterolemia and that macrophage α1AMPK may serve as a therapeutic target for prevention and treatment of atherosclerosis.
Collapse
Affiliation(s)
- Qiang Cao
- Department of Biology and Center for Obesity Reversal, Georgia State University, Atlanta, GA
| | - Xin Cui
- Department of Biology and Center for Obesity Reversal, Georgia State University, Atlanta, GA
| | - Rui Wu
- Department of Biology and Center for Obesity Reversal, Georgia State University, Atlanta, GA
| | - Lin Zha
- Department of Biology and Center for Obesity Reversal, Georgia State University, Atlanta, GA
| | - Xianfeng Wang
- Department of Internal Medicine, Wake Forest School of Medicine, Winston-Salem, NC
| | - John S Parks
- Section on Molecular Medicine, Department of Internal Medicine, Wake Forest School of Medicine, Winston-Salem, NC
| | - Liqing Yu
- Department of Animal and Avian Sciences, University of Maryland, College Park, MD
| | - Hang Shi
- Department of Biology and Center for Obesity Reversal, Georgia State University, Atlanta, GA
| | - Bingzhong Xue
- Department of Biology and Center for Obesity Reversal, Georgia State University, Atlanta, GA
| |
Collapse
|
17
|
Liu L, Hu Q, Wu H, Xue Y, Cai L, Fang M, Liu Z, Yao P, Wu Y, Gong Z. Protective role of n6/n3 PUFA supplementation with varying DHA/EPA ratios against atherosclerosis in mice. J Nutr Biochem 2016; 32:171-80. [DOI: 10.1016/j.jnutbio.2016.02.010] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2015] [Revised: 01/16/2016] [Accepted: 02/08/2016] [Indexed: 10/22/2022]
|
18
|
Abeywardena MY, Adams M, Dallimore J, Kitessa SM. Rise in DPA Following SDA-Rich Dietary Echium Oil Less Effective in Affording Anti-Arrhythmic Actions Compared to High DHA Levels Achieved with Fish Oil in Sprague-Dawley Rats. Nutrients 2016; 8:nu8010014. [PMID: 26742064 PMCID: PMC4728628 DOI: 10.3390/nu8010014] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2015] [Revised: 12/17/2015] [Accepted: 12/18/2015] [Indexed: 11/16/2022] Open
Abstract
Stearidonic acid (SDA; C18:4n-3) has been suggested as an alternative to fish oil (FO) for delivering health benefits of C ≥ 20 long-chain n-3 polyunsaturated fatty acids (LC n-3 PUFA). Echium oil (EO) represents a non-genetically-modified source of SDA available commercially. This study compared EO and FO in relation to alterations in plasma and tissue fatty acids, and for their ability to afford protection against ischemia-induced cardiac arrhythmia and ventricular fibrillation (VF). Rats were fed (12 weeks) diets supplemented with either EO or FO at three dose levels (1, 3 and 5% w/w; n = 18 per group). EO failed to influence C22:6n-3 (DHA) but increased C22:5n-3 (DPA) in tissues dose-dependently, especially in heart tissue. Conversely, DHA in hearts of FO rats showed dose-related elevation; 14.8%-24.1% of total fatty acids. Kidney showed resistance for incorporation of LC n-3 PUFA. Overall, FO provided greater cardioprotection than EO. At the highest dose level, FO rats displayed lower (p < 0.05) episodes of VF% (29% vs. 73%) and duration (22.7 ± 12.0 vs. 75.8 ± 17.1 s) than the EO group but at 3% EO was comparable to FO. We conclude that there is no endogenous conversion of SDA to DHA, and that DPA may be associated with limited cardiac benefit.
Collapse
Affiliation(s)
- Mahinda Y Abeywardena
- Commonwealth Scientific and Industrial Research Organisation (CSIRO) Food & Nutrition, Kintore Ave, Adelaide SA 5000, Australia.
| | - Michael Adams
- Commonwealth Scientific and Industrial Research Organisation (CSIRO) Food & Nutrition, Kintore Ave, Adelaide SA 5000, Australia.
| | - Julie Dallimore
- Commonwealth Scientific and Industrial Research Organisation (CSIRO) Food & Nutrition, Kintore Ave, Adelaide SA 5000, Australia.
| | - Soressa M Kitessa
- Commonwealth Scientific and Industrial Research Organisation (CSIRO) Food & Nutrition, Kintore Ave, Adelaide SA 5000, Australia.
| |
Collapse
|