51
|
Prokopiou E, Kolovos P, Kalogerou M, Neokleous A, Papagregoriou G, Deltas C, Malas S, Georgiou T. Therapeutic potential of omega-3 fatty acids supplementation in a mouse model of dry macular degeneration. BMJ Open Ophthalmol 2017; 1:e000056. [PMID: 29354704 PMCID: PMC5721630 DOI: 10.1136/bmjophth-2016-000056] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2016] [Revised: 04/13/2017] [Accepted: 05/07/2017] [Indexed: 01/27/2023] Open
Abstract
Purpose To evaluate the therapeutic effects of omega-3 (ω-3) and omega-6 (ω-6) fatty acids in the CCL2-/- model of dry age-related macular degeneration (AMD). The blood level of eicosapentaenoic acid (EPA) and arachidonic acid (AA) served to adjust the treatment dosage (AA/EPA=1-1.5). Methods Nine-month-old animals were allocated to different groups: (A) C57BL/6 untreated , (B) CCL2-/- untreated, (C) CCL2-/- treated with ω-3+ω-6, and (D) CCL2-/- treated with ω-3. Treatment was daily administered by gavage for 3 months. Fatty acids analysis was performed and retinas were histologically examined. Three-month-old wild type mice were used for comparison purposes. Real-time PCR and Western blot were performed for retinal inflammatory mediators. Results Increased EPA and decreased AA levels were observed in both blood and retinas in the treatment groups. The outer nuclear layer thickness was increased in groups C (90.0±7.8 μm) and D (125.6±9.8 μm) [corrected] compared with groups B (65.6±3.0 μm) and A (71.1±4.2 μm), and in young mice, it was 98.0±3.9 μm. A decrease in NF-κB expression was noted in the treatment groups. Interleukin (IL) 18 protein levels demonstrated a significant reduction in the ω-3-treated group only. Conclusion Supplementation with ω-3+ω-6 or ω-3 alone (AA/EPA=1-1.5) suggests a protective mechanism in the CCL2-/- animal model of dry AMD, with a more beneficial effect when ω-3 are used alone. Our findings indicated that inflammation is not the only determining factor; perhaps a regenerative process might be involved following administration of ω-3 fatty acids.
Collapse
Affiliation(s)
| | | | - Maria Kalogerou
- Ophthalmos Research and Educational Institute, Nicosia, Cyprus
| | | | - Gregory Papagregoriou
- Department of Biological Sciences, Molecular Medicine Research Centre and Laboratory of Molecular and Medical Genetics, University of Cyprus, Nicosia, Cyprus
| | - Constantinos Deltas
- Department of Biological Sciences, Molecular Medicine Research Centre and Laboratory of Molecular and Medical Genetics, University of Cyprus, Nicosia, Cyprus
| | - Stavros Malas
- Developmental and Functional Genetics Group, The Cyprus Institute of Neurology and Genetics, Nicosia, Cyprus
| | - Tassos Georgiou
- Ophthalmos Research and Educational Institute, Nicosia, Cyprus
| |
Collapse
|
52
|
Hammer SS, Busik JV. The role of dyslipidemia in diabetic retinopathy. Vision Res 2017; 139:228-236. [PMID: 28545981 DOI: 10.1016/j.visres.2017.04.010] [Citation(s) in RCA: 43] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2017] [Revised: 04/06/2017] [Accepted: 04/13/2017] [Indexed: 02/08/2023]
Abstract
Diabetic retinopathy (DR) affects over 93million people worldwide and is the number one cause of blindness among working age adults. These indicators coupled with the projected rise of patients diagnosed with diabetes, makes DR a serious and prevalent vision threating disease. Data from recent clinical trials demonstrate that in addition to the well accepted role of hyperglycemia, dyslipidemia is an important, but often overlooked factor in the development of DR. The central aim of this review article is to showcase the critical role of dyslipidemia in DR progression as well as highlight novel therapeutic solutions that take advantage of the vital roles lipid metabolism plays in DR progression.
Collapse
Affiliation(s)
- Sandra S Hammer
- Department of Physiology, Michigan State University, East Lansing, MI, United States
| | - Julia V Busik
- Department of Physiology, Michigan State University, East Lansing, MI, United States.
| |
Collapse
|
53
|
Hu J, Geyer A, Dziumbla S, Awwad K, Zeldin DC, Schunck WH, Popp R, Frömel T, Fleming I. Role of Müller cell cytochrome P450 2c44 in murine retinal angiogenesis. Prostaglandins Other Lipid Mediat 2017; 133:93-102. [PMID: 28442442 DOI: 10.1016/j.prostaglandins.2017.04.002] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2017] [Revised: 04/17/2017] [Accepted: 04/19/2017] [Indexed: 12/11/2022]
Abstract
Polyunsaturated fatty acids (PUFA) and their cytochrome P450 (CYP450) metabolites have been linked to angiogenesis and vessel homeostasis. However, the role of individual CYP isoforms and their endogenous metabolites in those processes are not clear. Here, we focused on the role of Cyp2c44 in postnatal retinal angiogenesis and report that Cyp2c44 is highly expressed in Müller glial cells in the retina. The constitutive as well as inducible postnatal genetic deletion of Cyp2c44 resulted in an increased vessel network density without affecting vessel radial expansion during the first postnatal week. This phenotype was associated with an increased endothelial cell proliferation and attenuated Notch signaling. LC-MS/MS analyses revealed that levels of hydroxydocosahexaenoic acids (HDHA), i.e., 10-, 17- and 20-HDHA were significantly elevated in retinas from 5day old Cyp2c44-/- mice compared to their wild-type littermates. Enzymatic activity assays revealed that HDHAs were potential substrates for Cyp2c44 which could account for the increased levels of HDHAs in retinas from Cyp2c44-/- mice. These data indicate that Cyp2c44 is expressed in the murine retina and, like the soluble epoxide hydrolase, is expressed in Müller glia cells. The enhanced endothelial cell proliferation and Notch inhibition seen in retinas from Cyp2c44-deficient mice indicate a role for Cyp2c44-derived lipid mediators in physiological angiogenesis.
Collapse
Affiliation(s)
- Jiong Hu
- Institute for Vascular Signalling, Centre for Molecular Medicine, Goethe University, Frankfurt am Main, D-60596, Germany; German Center of Cardiovascular Research (DZHK) Partner site Rhein-Main, Frankfurt am Main, Germany
| | - Alexandra Geyer
- Institute for Vascular Signalling, Centre for Molecular Medicine, Goethe University, Frankfurt am Main, D-60596, Germany
| | - Sarah Dziumbla
- Institute for Vascular Signalling, Centre for Molecular Medicine, Goethe University, Frankfurt am Main, D-60596, Germany; German Center of Cardiovascular Research (DZHK) Partner site Rhein-Main, Frankfurt am Main, Germany
| | - Khader Awwad
- Institute for Vascular Signalling, Centre for Molecular Medicine, Goethe University, Frankfurt am Main, D-60596, Germany
| | - Darryl C Zeldin
- Division of Intramural Research, National Institute of Environmental Health Sciences, National Institutes of Health, Research Triangle Park, NC, USA
| | - Wolf-Hagen Schunck
- Max-Delbrück Center for Molecular Medicine, Robert-Rössle-Str 10, 13092 Berlin, Germany
| | - Rüdiger Popp
- Institute for Vascular Signalling, Centre for Molecular Medicine, Goethe University, Frankfurt am Main, D-60596, Germany
| | - Timo Frömel
- Institute for Vascular Signalling, Centre for Molecular Medicine, Goethe University, Frankfurt am Main, D-60596, Germany; German Center of Cardiovascular Research (DZHK) Partner site Rhein-Main, Frankfurt am Main, Germany
| | - Ingrid Fleming
- Institute for Vascular Signalling, Centre for Molecular Medicine, Goethe University, Frankfurt am Main, D-60596, Germany; German Center of Cardiovascular Research (DZHK) Partner site Rhein-Main, Frankfurt am Main, Germany.
| |
Collapse
|
54
|
Takamura M, Kurokawa K, Ootsuji H, Inoue O, Okada H, Nomura A, Kaneko S, Usui S. Long-Term Administration of Eicosapentaenoic Acid Improves Post-Myocardial Infarction Cardiac Remodeling in Mice by Regulating Macrophage Polarization. J Am Heart Assoc 2017; 6:JAHA.116.004560. [PMID: 28223437 PMCID: PMC5523759 DOI: 10.1161/jaha.116.004560] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
Background Consumption of n‐3 fatty acids reduces the incidence of cardiovascular mortality in populations that consume diets rich in fish oil. Eicosapentaenoic acid (EPA) is an n‐3 fatty acid known to reduce the frequency of nonfatal coronary events; however, the frequency of mortality after myocardial infarction (MI) is not reduced. The aims of this study were to determine whether long‐term administration of EPA regulated cardiac remodeling after MI and to elucidate the underlying therapeutic mechanisms of EPA. Methods and Results C57BL/6J mice were divided into control (phosphate‐buffered saline–treated) and EPA‐treated groups. After 28 days of treatment, the mice were subjected to either sham surgery or MI by left anterior descending coronary artery ligation. Mortality due to MI or heart failure was significantly lower in the EPA‐treated mice than in the phosphate‐buffered saline–treated mice. However, the incidence of cardiac rupture was comparable between the EPA‐treated mice and the phosphate‐buffered saline–treated mice after MI. Echocardiographic tests indicated that EPA treatment attenuated post‐MI cardiac remodeling by preventing issues such as left ventricular systolic dysfunction and left ventricle dilatation 28 days after MI induction. Moreover, during the chronic remodeling phase, ie, 28 days after MI, flow cytometry demonstrated that EPA treatment significantly inhibited polarization toward proinflammatory M1 macrophages, but not anti‐inflammatory M2 macrophages, in the infarcted heart. Furthermore, EPA treatment attenuated fibrosis in the noninfarcted remote areas during the chronic phase. Conclusions Long‐term administration of EPA improved the prognosis of and attenuated chronic cardiac remodeling after MI by modulating the activation of proinflammatory M1 macrophages.
Collapse
Affiliation(s)
- Masayuki Takamura
- Department of Disease Control and Homeostasis, Graduate School of Medical Science, Kanazawa University, Kanazawa, Japan
| | - Keisuke Kurokawa
- Department of Disease Control and Homeostasis, Graduate School of Medical Science, Kanazawa University, Kanazawa, Japan
| | - Hiroshi Ootsuji
- Department of Disease Control and Homeostasis, Graduate School of Medical Science, Kanazawa University, Kanazawa, Japan
| | - Oto Inoue
- Department of Disease Control and Homeostasis, Graduate School of Medical Science, Kanazawa University, Kanazawa, Japan
| | - Hikari Okada
- Department of Disease Control and Homeostasis, Graduate School of Medical Science, Kanazawa University, Kanazawa, Japan
| | - Ayano Nomura
- Department of Disease Control and Homeostasis, Graduate School of Medical Science, Kanazawa University, Kanazawa, Japan
| | - Shuichi Kaneko
- Department of Disease Control and Homeostasis, Graduate School of Medical Science, Kanazawa University, Kanazawa, Japan
| | - Soichiro Usui
- Department of Disease Control and Homeostasis, Graduate School of Medical Science, Kanazawa University, Kanazawa, Japan
| |
Collapse
|
55
|
Omega-3 fatty acids and cytochrome P450-derived eicosanoids in cardiovascular diseases: Which actions and interactions modulate hemodynamics? Prostaglandins Other Lipid Mediat 2017; 128-129:34-42. [DOI: 10.1016/j.prostaglandins.2017.01.004] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2016] [Revised: 01/05/2017] [Accepted: 01/18/2017] [Indexed: 12/24/2022]
|
56
|
Gong Y, Shao Z, Fu Z, Edin ML, Sun Y, Liegl RG, Wang Z, Liu CH, Burnim SB, Meng SS, Lih FB, SanGiovanni JP, Zeldin DC, Hellström A, Smith LEH. Fenofibrate Inhibits Cytochrome P450 Epoxygenase 2C Activity to Suppress Pathological Ocular Angiogenesis. EBioMedicine 2016; 13:201-211. [PMID: 27720395 PMCID: PMC5264653 DOI: 10.1016/j.ebiom.2016.09.025] [Citation(s) in RCA: 35] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2016] [Revised: 09/23/2016] [Accepted: 09/28/2016] [Indexed: 12/19/2022] Open
Abstract
Neovascular eye diseases including retinopathy of prematurity, diabetic retinopathy and age-related-macular-degeneration are major causes of blindness. Fenofibrate treatment in type 2 diabetes patients reduces progression of diabetic retinopathy independent of its peroxisome proliferator-activated receptor (PPAR)α agonist lipid lowering effect. The mechanism is unknown. Fenofibrate binds to and inhibits cytochrome P450 epoxygenase (CYP)2C with higher affinity than to PPARα. CYP2C metabolizes ω-3 long-chain polyunsaturated fatty acids (LCPUFAs). While ω-3 LCPUFA products from other metabolizing pathways decrease retinal and choroidal neovascularization, CYP2C products of both ω-3 and ω-6 LCPUFAs promote angiogenesis. We hypothesized that fenofibrate inhibits retinopathy by reducing CYP2C ω-3 LCPUFA (and ω-6 LCPUFA) pro-angiogenic metabolites. Fenofibrate reduced retinal and choroidal neovascularization in PPARα-/-mice and augmented ω-3 LCPUFA protection via CYP2C inhibition. Fenofibrate suppressed retinal and choroidal neovascularization in mice overexpressing human CYP2C8 in endothelial cells and reduced plasma levels of the pro-angiogenic ω-3 LCPUFA CYP2C8 product, 19,20-epoxydocosapentaenoic acid. 19,20-epoxydocosapentaenoic acid reversed fenofibrate-induced suppression of angiogenesis ex vivo and suppression of endothelial cell functions in vitro. In summary fenofibrate suppressed retinal and choroidal neovascularization via CYP2C inhibition as well as by acting as an agonist of PPARα. Fenofibrate augmented the overall protective effects of ω-3 LCPUFAs on neovascular eye diseases. Fenofibrate inhibits retinal and choroidal neovascularization by inhibiting CYP2C activity as well as by activating PPARα. Fenofibrate augments the protective effects of ω-3 LCPUFAs on pathological ocular angiogenesis. Inhibition of CYP2C is a potential therapeutic approach for treatment of proliferative retinopathy and neovascular AMD.
Findings from clinical trials indicate that fenofibrate reduces the progression of proliferative diabetic retinopathy, but the mechanism of this effect is currently unknown. Dietary intake of ω-3 long-chain polyunsaturated fatty acids (LCPUFAs) is generally associated with a suppression of proliferative retinopathy and age-related macular degeneration acting through LCPUFA cyclooxygenase and lipoxygenase metabolites. However, cytochrome P450 epoxygenase (CYP)2C ω-3 and ω-6 LCPUFA metabolites promote retinopathy. Fenofibrate is a potent inhibitor of CYP2C. Our findings suggested that fenofibrate suppressed retinal and choroidal neovascularization via CYP2C inhibition. Combination therapy of dietary ω-3 LCPUFA supplementation with fenofibrate may be a promising approach to prevent incidence or progression of neovascular eye diseases.
Collapse
Affiliation(s)
- Yan Gong
- Department of Ophthalmology, Boston Children's Hospital, Harvard Medical School, Boston, MA 01248, United States
| | - Zhuo Shao
- Department of Ophthalmology, Boston Children's Hospital, Harvard Medical School, Boston, MA 01248, United States
| | - Zhongjie Fu
- Department of Ophthalmology, Boston Children's Hospital, Harvard Medical School, Boston, MA 01248, United States
| | - Matthew L Edin
- Division of Intramural Research, National Institute of Environmental Health Sciences, National Institutes of Health, Research Triangle Park, NC 27709, United States
| | - Ye Sun
- Department of Ophthalmology, Boston Children's Hospital, Harvard Medical School, Boston, MA 01248, United States
| | - Raffael G Liegl
- Department of Ophthalmology, Boston Children's Hospital, Harvard Medical School, Boston, MA 01248, United States
| | - Zhongxiao Wang
- Department of Ophthalmology, Boston Children's Hospital, Harvard Medical School, Boston, MA 01248, United States
| | - Chi-Hsiu Liu
- Department of Ophthalmology, Boston Children's Hospital, Harvard Medical School, Boston, MA 01248, United States
| | - Samuel B Burnim
- Department of Ophthalmology, Boston Children's Hospital, Harvard Medical School, Boston, MA 01248, United States
| | - Steven S Meng
- Department of Ophthalmology, Boston Children's Hospital, Harvard Medical School, Boston, MA 01248, United States
| | - Fred B Lih
- Division of Intramural Research, National Institute of Environmental Health Sciences, National Institutes of Health, Research Triangle Park, NC 27709, United States
| | - John Paul SanGiovanni
- Section on Nutritional Neurosciences, Laboratory of Membrane Biophysics and Biochemistry, National Institute on Alcohol Abuse and Alcoholism, National Institutes of Health, Bethesda, MD 20892, United States
| | - Darryl C Zeldin
- Division of Intramural Research, National Institute of Environmental Health Sciences, National Institutes of Health, Research Triangle Park, NC 27709, United States
| | - Ann Hellström
- Department of Ophthalmology, Sahlgrenska Academy at University of Gothenburg, Gothenburg 40530, Sweden
| | - Lois E H Smith
- Department of Ophthalmology, Boston Children's Hospital, Harvard Medical School, Boston, MA 01248, United States.
| |
Collapse
|
57
|
Sardell RJ, Bailey JNC, Courtenay MD, Whitehead P, Laux RA, Adams LD, Fortun JA, Brantley MA, Kovach JL, Schwartz SG, Agarwal A, Scott WK, Haines JL, Pericak-Vance MA. Whole exome sequencing of extreme age-related macular degeneration phenotypes. Mol Vis 2016; 22:1062-76. [PMID: 27625572 PMCID: PMC5007100] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2016] [Accepted: 08/27/2016] [Indexed: 11/29/2022] Open
Abstract
PURPOSE Demographic, environmental, and genetic risk factors for age-related macular degeneration (AMD) have been identified; however, a substantial portion of the variance in AMD disease risk and heritability remains unexplained. To identify AMD risk variants and generate hypotheses for future studies, we performed whole exome sequencing for 75 individuals whose phenotype was not well predicted by their genotype at known risk loci. We hypothesized that these phenotypically extreme individuals were more likely to carry rare risk or protective variants with large effect sizes. METHODS A genetic risk score was calculated in a case-control set of 864 individuals (467 AMD cases, 397 controls) based on 19 common (≥1% minor allele frequency, MAF) single nucleotide variants previously associated with the risk of advanced AMD in a large meta-analysis of advanced cases and controls. We then selected for sequencing 39 cases with bilateral choroidal neovascularization with the lowest genetic risk scores to detect risk variants and 36 unaffected controls with the highest genetic risk score to detect protective variants. After minimizing the influence of 19 common genetic risk loci on case-control status, we targeted single variants of large effect and the aggregate effect of weaker variants within genes and pathways. Single variant tests were conducted on all variants, while gene-based and pathway analyses were conducted on three subsets of data: 1) rare (≤1% MAF in the European population) stop, splice, or damaging missense variants, 2) all rare variants, and 3) all variants. All analyses controlled for the effects of age and sex. RESULTS No variant, gene, or pathway outside regions known to be associated with risk for advanced AMD reached genome-wide significance. However, we identified several variants with substantial differences in allele frequency between cases and controls with strong additive effects on affection status after controlling for age and sex. Protective effects trending toward significance were detected at two loci identified in single-variant analyses: an intronic variant in FBLN7 (the gene encoding fibulin 7) and at three variants near pyridoxal (pyridoxine, vitamin B6) kinase (PDXK). Aggregate rare-variant analyses suggested evidence for association at ASRGL1, a gene previously linked to photoreceptor cell death, and at BSDC1. In known AMD loci we also identified 29 novel or rare damaging missense or stop/splice variants in our sample of cases and controls. CONCLUSIONS Identified variants and genes may highlight regions important in the pathogenesis of AMD and are key targets for replication.
Collapse
Affiliation(s)
- Rebecca J. Sardell
- John P. Hussman Institute for Human Genomics, University of Miami Miller School of Medicine, Miami, FL
| | - Jessica N Cooke Bailey
- Department of Epidemiology and Biostatistics, Case Western Reserve University, Cleveland, OH
| | - Monique D. Courtenay
- John P. Hussman Institute for Human Genomics, University of Miami Miller School of Medicine, Miami, FL
| | - Patrice Whitehead
- John P. Hussman Institute for Human Genomics, University of Miami Miller School of Medicine, Miami, FL
| | - Reneé A. Laux
- Department of Epidemiology and Biostatistics, Case Western Reserve University, Cleveland, OH
| | - Larry D. Adams
- John P. Hussman Institute for Human Genomics, University of Miami Miller School of Medicine, Miami, FL
| | - Jorge A. Fortun
- Bascom Palmer Eye Institute, University of Miami Miller School of Medicine, Miami, FL
| | - Milam A. Brantley
- Department of Ophthalmology and Visual Sciences, Vanderbilt University School of Medicine, Nashville, TN
| | - Jaclyn L. Kovach
- Bascom Palmer Eye Institute, University of Miami Miller School of Medicine, Miami, FL
| | - Stephen G. Schwartz
- Bascom Palmer Eye Institute, University of Miami Miller School of Medicine, Miami, FL
| | - Anita Agarwal
- Department of Ophthalmology and Visual Sciences, Vanderbilt University School of Medicine, Nashville, TN
| | - William K. Scott
- John P. Hussman Institute for Human Genomics, University of Miami Miller School of Medicine, Miami, FL
| | - Jonathan L. Haines
- Department of Epidemiology and Biostatistics, Case Western Reserve University, Cleveland, OH
| | - Margaret A. Pericak-Vance
- John P. Hussman Institute for Human Genomics, University of Miami Miller School of Medicine, Miami, FL
| |
Collapse
|
58
|
Markworth JF, Kaur G, Miller EG, Larsen AE, Sinclair AJ, Maddipati KR, Cameron-Smith D. Divergent shifts in lipid mediator profile following supplementation with n-3 docosapentaenoic acid and eicosapentaenoic acid. FASEB J 2016; 30:3714-3725. [PMID: 27461565 DOI: 10.1096/fj.201600360r] [Citation(s) in RCA: 67] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2016] [Accepted: 07/11/2016] [Indexed: 12/31/2022]
Abstract
In contrast to the well-characterized effects of specialized proresolving lipid mediators (SPMs) derived from eicosapentaenoic acid (EPA) and docosahexaenoic acid (DHA), little is known about the metabolic fate of the intermediary long-chain (LC) n-3 polyunsaturated fatty acid (PUFA) docosapentaenoic acid (DPA). In this double blind crossover study, shifts in circulating levels of n-3 and n-6 PUFA-derived bioactive lipid mediators were quantified by an unbiased liquid chromatography-tandem mass spectrometry lipidomic approach. Plasma was obtained from human subjects before and after 7 d of supplementation with pure n-3 DPA, n-3 EPA or placebo (olive oil). DPA supplementation increased the SPM resolvin D5n-3DPA (RvD5n-3DPA) and maresin (MaR)-1, the DHA vicinal diol 19,20-dihydroxy-DPA and n-6 PUFA derived 15-keto-PG E2 (15-keto-PGE2). EPA supplementation had no effect on any plasma DPA or DHA derived mediators, but markedly elevated monohydroxy-eicosapentaenoic acids (HEPEs), including the e-series resolvin (RvE) precursor 18-HEPE; effects not observed with DPA supplementation. These data show that dietary n-3 DPA and EPA have highly divergent effects on human lipid mediator profile, with no overlap in PUFA metabolites formed. The recently uncovered biologic activity of n-3 DPA docosanoids and their marked modulation by dietary DPA intake reveals a unique and specific role of n-3 DPA in human physiology.-Markworth, J. F., Kaur, G., Miller, E. G., Larsen, A. E., Sinclair, A. J., Maddipati, K. R., Cameron-Smith, D. Divergent shifts in lipid mediator profile following supplementation with n-3 docosapentaenoic acid and eicosapentaenoic acid.
Collapse
Affiliation(s)
| | - Gunveen Kaur
- Centre for Physical Activity and Nutrition Research, School of Exercise and Nutrition Sciences, Deakin University, Melbourne, Victoria, Australia
| | - Eliza G Miller
- Centre for Physical Activity and Nutrition Research, School of Exercise and Nutrition Sciences, Deakin University, Melbourne, Victoria, Australia
| | - Amy E Larsen
- Department of Physiology, Anatomy and Microbiology, La Trobe University Melbourne, Victoria, Australia
| | | | - Krishna Rao Maddipati
- Department of Pathology, Lipidomics Core Facility, Wayne State University, Detroit, Michigan, USA; and.,Karmanos Cancer Institute, Wayne State University, Detroit, Michigan, USA
| | | |
Collapse
|
59
|
Schunck WH. EPA and/or DHA? A test question on the principles and opportunities in utilizing the therapeutic potential of omega-3 fatty acids. J Lipid Res 2016; 57:1608-11. [PMID: 27436589 DOI: 10.1194/jlr.c071084] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
|
60
|
Marion-Letellier R, Savoye G, Ghosh S. Fatty acids, eicosanoids and PPAR gamma. Eur J Pharmacol 2015; 785:44-49. [PMID: 26632493 DOI: 10.1016/j.ejphar.2015.11.004] [Citation(s) in RCA: 187] [Impact Index Per Article: 20.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2015] [Revised: 11/02/2015] [Accepted: 11/02/2015] [Indexed: 12/25/2022]
Abstract
Peroxisome proliferator-activated receptor γ (PPARγ) belongs to the family of nuclear nuclear receptors and is mainly expressed in adipose tissue, hematopoietic cells and the large intestine. Contrary to other nuclear receptors that mainly bind a single specific ligand, there are numerous natural PPARγ ligands, in particular fatty acids or their derivatives called eicosanoids. PPARγ have pleiotropic functions: (i) glucose and lipid metabolism regulation, (ii) anti-inflammatory properties, (iii) oxidative stress inhibition, (iv) improvement of endothelial function. Its role has been mainly studied by the use synthetic agonists. In this review, we will focus on the effects of PPARγ mediated through fatty acids and how these have beneficial health properties.
Collapse
Affiliation(s)
- Rachel Marion-Letellier
- INSERM Unit UMR1073, Rouen University and Rouen University hospital, 22, boulevard Gambetta, 76183 Rouen cedex, France.
| | - Guillaume Savoye
- INSERM Unit UMR1073, Rouen University and Rouen University hospital, 22, boulevard Gambetta, 76183 Rouen cedex, France; Department of Gastroenterology, Rouen University Hospital, 1 rue de Germont, 76031 Rouen cedex, France
| | - Subrata Ghosh
- Division of Gastroenterology, University of Calgary, Alberta, Canada
| |
Collapse
|
61
|
Shoda H, Yanai R, Yoshimura T, Nagai T, Kimura K, Sobrin L, Connor KM, Sakoda Y, Tamada K, Ikeda T, Sonoda KH. Dietary Omega-3 Fatty Acids Suppress Experimental Autoimmune Uveitis in Association with Inhibition of Th1 and Th17 Cell Function. PLoS One 2015; 10:e0138241. [PMID: 26393358 PMCID: PMC4578775 DOI: 10.1371/journal.pone.0138241] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2015] [Accepted: 08/27/2015] [Indexed: 12/21/2022] Open
Abstract
Omega (ω)–3 long-chain polyunsaturated fatty acids (LCPUFAs) inhibit the production of inflammatory mediators and thereby contribute to the regulation of inflammation. Experimental autoimmune uveitis (EAU) is a well-established animal model of autoimmune retinal inflammation. To investigate the potential effects of dietary intake of ω-3 LCPUFAs on uveitis, we examined the anti-inflammatory properties of these molecules in comparison with ω-6 LCPUFAs in a mouse EAU model. C57BL/6 mice were fed a diet containing ω-3 LCPUFAs or ω-6 LCPUFAs for 2 weeks before as well as after the induction of EAU by subcutaneous injection of a fragment of human interphotoreceptor retinoid-binding protein emulsified with complete Freund’s adjuvant. Both clinical and histological scores for uveitis were smaller for mice fed ω-3 LCPUFAs than for those fed ω-6 LCPUFAs. The concentrations of the T helper 1 (Th1) cytokine interferon-γ and the Th17 cytokine interleukin-17 in intraocular fluid as well as the production of these cytokines by lymph node cells were reduced for mice fed ω-3 LCPUFAs. Furthermore, the amounts of mRNAs for the Th1- and Th17-related transcription factors T-bet and RORγt, respectively, were reduced both in the retina and in lymph node cells of mice fed ω-3 LCPUFAs. Our results thus show that a diet enriched in ω-3 LCPUFAs suppressed uveitis in mice in association with inhibition of Th1 and Th17 cell function.
Collapse
Affiliation(s)
- Hiromi Shoda
- Department of Ophthalmology, Yamaguchi University Graduate School of Medicine, Ube, Yamaguchi, Japan
- Department of Ophthalmology, Osaka Medical Collage, Osaka, Japan
| | - Ryoji Yanai
- Department of Ophthalmology, Yamaguchi University Graduate School of Medicine, Ube, Yamaguchi, Japan
- * E-mail:
| | - Takeru Yoshimura
- Angiogenesis Laboratory, Department of Ophthalmology, Massachusetts Eye and Ear Infirmary, Harvard Medical School, Cambridge, Massachusetts, United States of America
| | - Tomohiko Nagai
- Department of Ophthalmology, Yamaguchi University Graduate School of Medicine, Ube, Yamaguchi, Japan
| | - Kazuhiro Kimura
- Department of Ophthalmology, Yamaguchi University Graduate School of Medicine, Ube, Yamaguchi, Japan
| | - Lucia Sobrin
- Angiogenesis Laboratory, Department of Ophthalmology, Massachusetts Eye and Ear Infirmary, Harvard Medical School, Cambridge, Massachusetts, United States of America
| | - Kip M. Connor
- Angiogenesis Laboratory, Department of Ophthalmology, Massachusetts Eye and Ear Infirmary, Harvard Medical School, Cambridge, Massachusetts, United States of America
| | - Yukimi Sakoda
- Department of Immunology, Yamaguchi University Graduate School of Medicine, Ube, Yamaguchi, Japan
| | - Koji Tamada
- Department of Immunology, Yamaguchi University Graduate School of Medicine, Ube, Yamaguchi, Japan
| | - Tsunehiko Ikeda
- Department of Ophthalmology, Osaka Medical Collage, Osaka, Japan
| | - Koh-Hei Sonoda
- Department of Ophthalmology, Yamaguchi University Graduate School of Medicine, Ube, Yamaguchi, Japan
| |
Collapse
|
62
|
Nakanishi A, Tsukamoto I. n-3 polyunsaturated fatty acids stimulate osteoclastogenesis through PPARγ-mediated enhancement of c-Fos expression, and suppress osteoclastogenesis through PPARγ-dependent inhibition of NFkB activation. J Nutr Biochem 2015; 26:1317-27. [PMID: 26303404 DOI: 10.1016/j.jnutbio.2015.06.007] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2015] [Revised: 06/17/2015] [Accepted: 06/17/2015] [Indexed: 11/17/2022]
Abstract
n-3 polyunsaturated fatty acids (PUFAs), eicosapentaenoic acid (EPA) and docosahexaenoic acid (DHA) have been reported to suppress osteoclastogenesis in vivo. In this study, the effect of PUFAs on receptor for activation of nuclear factor-κB ligand (RANKL)-induced osteoclastogenesis was examined using bone marrow-derived monocytes/macrophage precursor cells (BMMs) or bone marrow cells (BMCs) in vitro. EPA and DHA stimulated the osteoclastic differentiation of BMMs, but n-6 PUFAs, linoleic acid and arachidonic acid had no effect. The stimulation of osteoclastogenesis of BMMs by EPA and DHA was associated with enhancement of the gene expressions of c-Fos, tartrate-resistant acid phosphatase, cathepsin K and peroxisome proliferator-activated receptor-γ (PPARγ) and the protein levels of c-Fos, PPARγ and nuclear factor of activated T cells, cytoplasmic, calcineurin-dependent-1 (NFATc1). The PPARγ agonists, rosiglitazone and GW1929, also stimulated the osteoclastogenesis of BMMs. The PPARγ antagonists, T0070907 and GW9662, inhibited the stimulations of osteoclastogenesis and c-Fos expression by EPA or DHA. However, EPA and DHA inhibited the osteoclastogenesis in BMCs including BMMs and mesenchymal stem cells (MSCs). This inhibition was associated with suppression of the expression of RANKL and nuclear factor-κB (NFκB)-regulating genes, cyclooxygenase 2, TNFα and IL-6 in BMCs and MSCs. The agonists and antagonists of PPARγ showed that the inhibitions of NFκB transcriptional activity and osteoclastogenesis by EPA and DHA were PPARγ-dependent. These results suggest that EPA and DHA directly act on BMMs and stimulate osteoclastogenesis through enhancing c-Fos expression mediated by PPARγ but suppress osteoclastogenesis through the PPARγ-dependent inhibition of NFκB activation of MSCs in BMCs.
Collapse
Affiliation(s)
- Atsuko Nakanishi
- Department of Food Science and Nutrition, Nara Women's University, Kita-uoya Nishi-machi, Nara, 630-8506, Japan
| | - Ikuyo Tsukamoto
- Department of Food Science and Nutrition, Nara Women's University, Kita-uoya Nishi-machi, Nara, 630-8506, Japan; Faculty of Clinical Nutrition, Hiroshima International University, 5-1-1, Hirokoshingai, Kure, Hiroshima, 737-0112, Japan.
| |
Collapse
|
63
|
Yonekawa Y, Miller JW, Kim IK. Age-Related Macular Degeneration: Advances in Management and Diagnosis. J Clin Med 2015; 4:343-59. [PMID: 26239130 PMCID: PMC4470128 DOI: 10.3390/jcm4020343] [Citation(s) in RCA: 90] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2014] [Revised: 12/24/2014] [Accepted: 01/20/2015] [Indexed: 12/27/2022] Open
Abstract
Age-related macular degeneration (AMD) is the most common cause of irreversible visual impairment in older populations in industrialized nations. AMD is a late-onset deterioration of photoreceptors and retinal pigment epithelium in the central retina caused by various environmental and genetic factors. Great strides in our understanding of AMD pathogenesis have been made in the past several decades, which have translated into revolutionary therapeutic agents in recent years. In this review, we describe the clinical and pathologic features of AMD and present an overview of current diagnosis and treatment strategies.
Collapse
Affiliation(s)
- Yoshihiro Yonekawa
- Retina Service, Department of Ophthalmology, Massachusetts Eye and Ear Infirmary, Harvard Medical School, Boston, MA 02114, USA.
| | - Joan W Miller
- Retina Service, Department of Ophthalmology, Massachusetts Eye and Ear Infirmary, Harvard Medical School, Boston, MA 02114, USA.
| | - Ivana K Kim
- Retina Service, Department of Ophthalmology, Massachusetts Eye and Ear Infirmary, Harvard Medical School, Boston, MA 02114, USA.
| |
Collapse
|
64
|
Abstract
The question whether dietary habits and lifestyle have influence on the course of multiple sclerosis (MS) is still a matter of debate, and at present, MS therapy is not associated with any information on diet and lifestyle. Here we show that dietary factors and lifestyle may exacerbate or ameliorate MS symptoms by modulating the inflammatory status of the disease both in relapsing-remitting MS and in primary-progressive MS. This is achieved by controlling both the metabolic and inflammatory pathways in the human cell and the composition of commensal gut microbiota. What increases inflammation are hypercaloric Western-style diets, characterized by high salt, animal fat, red meat, sugar-sweetened drinks, fried food, low fiber, and lack of physical exercise. The persistence of this type of diet upregulates the metabolism of human cells toward biosynthetic pathways including those of proinflammatory molecules and also leads to a dysbiotic gut microbiota, alteration of intestinal immunity, and low-grade systemic inflammation. Conversely, exercise and low-calorie diets based on the assumption of vegetables, fruit, legumes, fish, prebiotics, and probiotics act on nuclear receptors and enzymes that upregulate oxidative metabolism, downregulate the synthesis of proinflammatory molecules, and restore or maintain a healthy symbiotic gut microbiota. Now that we know the molecular mechanisms by which dietary factors and exercise affect the inflammatory status in MS, we can expect that a nutritional intervention with anti-inflammatory food and dietary supplements can alleviate possible side effects of immune-modulatory drugs and the symptoms of chronic fatigue syndrome and thus favor patient wellness.
Collapse
Affiliation(s)
- Paolo Riccio
- Department of Sciences, University of Basilicata, Potenza, Italy
| | - Rocco Rossano
- Department of Sciences, University of Basilicata, Potenza, Italy
| |
Collapse
|