1
|
Ren J, Ren A, Huang Z, Deng X, Jiang Z, Xue Y, Fu Z, Smith LE, Ke M, Gong Y. Metabolomic Profiling of Long-Chain Polyunsaturated Fatty Acid Oxidation in Adults with Retinal Vein Occlusion: A Case-Control Study. Am J Clin Nutr 2023; 118:579-590. [PMID: 37454758 DOI: 10.1016/j.ajcnut.2023.07.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2023] [Revised: 07/04/2023] [Accepted: 07/10/2023] [Indexed: 07/18/2023] Open
Abstract
BACKGROUND Long-chain polyunsaturated fatty acids (LCPUFAs) and their metabolites are closely related to neovascular eye diseases. However, the clinical significance of their oxylipins in retinal vein occlusion (RVO) remains inconclusive. OBJECTIVES This case-control study aimed to explore metabolomic profiles of LCPUFA oxidation in RVO and to identify potential indicators for diagnosis and pathologic progression. METHODS The plasma concentrations of ω-3 (n-3) and ω-6 (n-6) LCPUFA and their oxylipins in 44 adults with RVO and 36 normal controls were analyzed using ultraperformance liquid chromatography tandem mass spectrometry. Univariate analysis combined with principal component and orthogonal projections to latent structure discriminant analysis was used to screen differential metabolites. Aortic ring and choroidal explant sprouting assays were used to investigate the effects of 5-oxo-eicosatetraenoic acids (ETE) on angiogenesis ex vivo. Tubule formation and wound healing assays were performed to verify its effects on human retinal microvascular endothelial cell functions. RESULTS Higher ω-6 and lower ω-3 LCPUFA plasma concentrations were measured in the adults with RVO compared with control (odds ratio [OR]: 2.34; 95% confidence interval [CI]: 1.42, 3.86; P < 0.001; OR: 0.28; 95% CI: 0.15, 0.51; P < 0.001). Metabolomic analysis revealed 20 LCPUFA and their oxylipins dysregulated in RVO, including increased arachidonic acid (ω-6, OR: 1.85; 95% CI: 1.18, 2.90; P < 0.001) and its lipoxygenase product 5-oxo-ETE (OR: 11.76; 95% CI: 3.73, 37.11; P < 0.001), as well as decreased docosahexaenoic acid (ω-3, OR: 0.13; 95% CI: 0.05, 0.33; P < 0.001). Interestingly, 5-oxo-ETE was downregulated in ischemic compared with nonischemic central RVO. Exogenous 5-oxo-ETE attenuated aortic ring and choroidal explant sprouting and inhibited tubule formation and migration of human retinal microvascular endothelial cells in a dose-dependent manner, possibly via suppressing the vascular endothelial growth factor signaling pathway. CONCLUSIONS The plasma concentrations of ω-6 and ω-3 LCPUFA and their oxylipins were associated with RVO. The ω-6 LCPUFA-derived metabolite 5-oxo-ETE was a potential marker of RVO development and progression.
Collapse
Affiliation(s)
- Jiangbo Ren
- Department of Biological Repositories, Zhongnan Hospital of Wuhan University, Wuhan, China; Department of Ophthalmology, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Anli Ren
- Department of Biological Repositories, Zhongnan Hospital of Wuhan University, Wuhan, China; Department of Ophthalmology, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Zhengrong Huang
- Department of Biological Repositories, Zhongnan Hospital of Wuhan University, Wuhan, China; Tumor Precision Diagnosis and Treatment Technology and Translational Medicine, Hubei Engineering Research Center, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Xizhi Deng
- Department of Ophthalmology, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Ziyu Jiang
- Department of Biological Repositories, Zhongnan Hospital of Wuhan University, Wuhan, China; Tumor Precision Diagnosis and Treatment Technology and Translational Medicine, Hubei Engineering Research Center, Zhongnan Hospital of Wuhan University, Wuhan, China; Human Genetics Resource Preservation Center of Wuhan University, Wuhan University, Wuhan, China
| | - Yanni Xue
- Department of Ophthalmology, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Zhongjie Fu
- Department of Ophthalmology, Boston Children's Hospital, Harvard Medical School, Boston, MA, United States
| | - Lois Eh Smith
- Department of Ophthalmology, Boston Children's Hospital, Harvard Medical School, Boston, MA, United States
| | - Min Ke
- Department of Ophthalmology, Zhongnan Hospital of Wuhan University, Wuhan, China.
| | - Yan Gong
- Department of Biological Repositories, Zhongnan Hospital of Wuhan University, Wuhan, China; Tumor Precision Diagnosis and Treatment Technology and Translational Medicine, Hubei Engineering Research Center, Zhongnan Hospital of Wuhan University, Wuhan, China; Human Genetics Resource Preservation Center of Wuhan University, Wuhan University, Wuhan, China.
| |
Collapse
|
2
|
Bo Q, Xie Y, Lin Q, Fu L, Hu C, Zhang Z, Meng Q, Xu F, Wang G, Miao Z, Wang H, Xu D. Docosahexaenoic acid protects against lipopolysaccharide-induced fetal growth restriction via inducing the ubiquitination and degradation of NF-κB p65 in placental trophoblasts. J Nutr Biochem 2023; 118:109359. [PMID: 37085060 DOI: 10.1016/j.jnutbio.2023.109359] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2022] [Revised: 02/17/2023] [Accepted: 04/14/2023] [Indexed: 04/23/2023]
Abstract
Lipopolysaccharide (LPS) could induce adverse birth outcomes by evoking inflammation. We investigated the effect and mechanism of docosahexaenoic acid (DHA) on LPS-induced placental inflammation and fetal growth restriction (FGR). In vivo, pregnant CD-1 mice were divided into four groups: Ctrl, DHA, LPS and DHA+LPS group. We found that DHA pretreatment reduced the incidence of FGR induced by LPS and activated the expression of peroxisome proliferators-activated receptor gamma (PPARγ) in placental tissue. Moreover, the LPS-induced increase of mRNA levels of Tnf-α, Il-6, Il-1β, Mip-2 and Kc in placental tissue was significantly attenuated by DHA pretreatment. A similar effect of DHA was observed in serum of pregnant mice and amniotic fluid. In contrast, the levels of the IL-10 were significantly increased after DHA pretreatment. In vitro, we clarified that DHA antagonized the activation of the NF-κB signaling pathway induced by LPS, which was dependent on PPARγ. Subsequently, CHX (translation inhibitor) was used to indicated that PPARγ significantly increased the degradation rate of p65, an effect that was inhibited by MG132 (proteasome inhibitor) treatment. Finally, it was confirmed that the activation of PPARγ could significantly promote the ubiquitination and degradation of p65. Our results suggested that DHA alleviated LPS-induced inflammatory responses and FGR by activating PPARγ expression, leading to p65 ubiquitination and degradation.
Collapse
Affiliation(s)
- Qingli Bo
- Department of Nutrition and Food Hygiene, School of Public Health, Anhui Medical University, Hefei 230032, China; Key Laboratory of Environmental Toxicology of Anhui Higher Education Institutes, Hefei 230032, China
| | - Yali Xie
- Department of Nutrition and Food Hygiene, School of Public Health, Anhui Medical University, Hefei 230032, China
| | - Qiulin Lin
- Department of Nutrition and Food Hygiene, School of Public Health, Anhui Medical University, Hefei 230032, China
| | - Lin Fu
- Key Laboratory of Environmental Toxicology of Anhui Higher Education Institutes, Hefei 230032, China
| | - Chunqiu Hu
- Department of Nutrition and Food Hygiene, School of Public Health, Anhui Medical University, Hefei 230032, China
| | - Zhiqiang Zhang
- Department of Nutrition and Food Hygiene, School of Public Health, Anhui Medical University, Hefei 230032, China
| | - Qingchong Meng
- Department of Nutrition and Food Hygiene, School of Public Health, Anhui Medical University, Hefei 230032, China
| | - Feixiang Xu
- Department of Toxicology, School of Public Health, Anhui Medical University, Hefei 230032, China;; Key Laboratory of Environmental Toxicology of Anhui Higher Education Institutes, Hefei 230032, China
| | - Guoxiu Wang
- Department of Nutrition and Food Hygiene, School of Public Health, Anhui Medical University, Hefei 230032, China
| | - Ziyang Miao
- Department of Nutrition and Food Hygiene, School of Public Health, Anhui Medical University, Hefei 230032, China
| | - Hua Wang
- Department of Toxicology, School of Public Health, Anhui Medical University, Hefei 230032, China;; Key Laboratory of Environmental Toxicology of Anhui Higher Education Institutes, Hefei 230032, China
| | - Dexiang Xu
- Department of Toxicology, School of Public Health, Anhui Medical University, Hefei 230032, China;; Key Laboratory of Environmental Toxicology of Anhui Higher Education Institutes, Hefei 230032, China.
| |
Collapse
|
3
|
Ghasemi Darestani N, Bahrami A, Mozafarian MR, Esmalian Afyouni N, Akhavanfar R, Abouali R, Moradian A. Association of Polyunsaturated Fatty Acid Intake on Inflammatory Gene Expression and Multiple Sclerosis: A Systematic Review and Meta-Analysis. Nutrients 2022; 14:4627. [PMID: 36364885 PMCID: PMC9656750 DOI: 10.3390/nu14214627] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2022] [Revised: 10/24/2022] [Accepted: 10/31/2022] [Indexed: 11/06/2022] Open
Abstract
The health benefits of omega-3 fatty acid (FA) supplementation on inflammatory gene expression (IGE) and multiple sclerosis (MS) are becoming more evident. However, an overview of the results from randomized controlled trials is lacking. This study aimed to conduct a meta-analysis to evaluate the effect of omega-3 fatty acid intake on MS (based on the criteria of the Expanded Disability Status Scale (EDSS)) and inflammatory gene expression (IGE). A search was conducted of PubMed, EMBASE, and Web of Science for cohort studies published from the inception of the database up to May 2022 that assessed the associations of omega-3 polyunsaturated fatty acids (n-3 PUFAs), docosahexaenoic acid (DHA), α-linolenic acid (ALA), and eicosapentaenoic acid (EPA) with EDSS and inflammatory gene expression (peroxisome proliferator-activated receptor gamma (PPAR-γ), tumor necrosis factor-alpha (TNF-α), interleukin-1 (IL-1), interleukin-6 (IL-6), and interleukin-8 (IL-8)) outcomes. For the highest vs. lowest comparison, the relative risk (RR) estimates with a 95% confidence interval (CI) were pooled using the random-effect model. In total, 13 cohort studies with 1353 participants were included in the meta-analysis during periods of 3 to 144 weeks. A significant inverse relationship was found between DHA and EDSS scores (RR: 1.05; 95% CI: 0.62, 1.48; p < 0.00001). Our results also showed that omega-3 FAs significantly upregulated the gene expression of PPAR-γ (RR: 0.95; 95% CI: 0.52, 1.38; p < 0.03) and downregulated the expression of TNF-α (RR: −0.15; 95% CI: −0.99, 0.70; p < 0.00001) and IL-1 (RR: −0.60; 95% CI: −1.02, −0.18; p < 0.003). There was no clear evidence of publication bias with Egger’s tests for inflammatory gene expression (p = 0.266). Moreover, n-3 PUFAs and EPA were not significantly associated with EDSS scores (p > 0.05). In this meta-analysis of cohort studies, blood omega-3 FA concentrations were inversely related to inflammatory gene expression (IGE) and EDSS score, which indicates that they may hold great potential markers for the diagnosis, prognosis, and management of MS. However, further clinical trials are required to confirm the potential effects of the omega-3 FAs on MS disease management.
Collapse
Affiliation(s)
- Nadia Ghasemi Darestani
- School of Medicine, Isfahan University of Medical Sciences, Isfahan 8174673461, Iran; (N.G.D.); (R.A.)
| | - Abolfazl Bahrami
- Department of Animal Science, College of Agriculture and Natural Resources, University of Tehran, Tehran 1417643184, Iran
- Biomedical Center for Systems Biology Science Munich, Ludwig-Maximilians-University, 80333 Munich, Germany
| | - Mohammad Reza Mozafarian
- Department of Nutrition, School of Health and Nutrition, Bushehr University of Medical Sciences, Bushehr 75, Iran;
| | - Nazgol Esmalian Afyouni
- Isfahan Neurosciences Research Center, Alzahra Research Institute, Isfahan University of Medical Sciences, Isfahan 8174673461, Iran;
| | - Roozbeh Akhavanfar
- School of Medicine, Isfahan University of Medical Sciences, Isfahan 8174673461, Iran; (N.G.D.); (R.A.)
| | - Reza Abouali
- Department of Health Sciences, Interdisciplinary Research Center of Autoimmune Diseases—IRCAD, Università del Piemonte Orientale, 13100 Novara, Italy;
| | - Arsalan Moradian
- Department of Pharmacology and Toxicology, Faculty of Pharmacy and Pharmaceutical Sciences, Pharmacist, Isfahan University of Medical Sciences, Isfahan 8174673461, Iran
| |
Collapse
|
4
|
Badia-Soteras A, de Vries J, Dykstra W, Broersen LM, Verkuyl JM, Smit AB, Verheijen MHG. High-Throughput Analysis of Astrocyte Cultures Shows Prevention of Reactive Astrogliosis by the Multi-Nutrient Combination Fortasyn Connect. Cells 2022; 11:cells11091428. [PMID: 35563732 PMCID: PMC9099974 DOI: 10.3390/cells11091428] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2021] [Revised: 04/07/2022] [Accepted: 04/20/2022] [Indexed: 12/23/2022] Open
Abstract
Astrocytes are specialized glial cells that tile the central nervous system (CNS) and perform numerous essential functions. Astrocytes react to various forms of CNS insults by altering their morphology and molecular profile, through a process known as reactive astrogliosis. Accordingly, astrocyte reactivity is apparent in many neurodegenerative diseases, among which one is Alzheimer’s disease (AD). Recent clinical trials on early-stage AD have demonstrated that Fortasyn Connect (FC), a multi-nutrient combination providing specific precursors and cofactors for phospholipid synthesis, helps to maintain neuronal functional connectivity and cognitive performance of patients. Several studies have shown that FC may act through its effects on neuronal survival and synaptogenesis, leading to reduced astrocyte reactivity, but whether FC can directly counteract astrocyte reactivity remains to be elucidated. Hence, we developed an in vitro model of reactive astrogliosis using the pro-inflammatory cytokines TNF-α and IFN-γ together with an automated high-throughput assay (AstroScan) to quantify molecular and morphological changes that accompany reactive astrogliosis. Next, we showed that FC is potent in preventing cytokine-induced reactive astrogliosis, a finding that might be of high relevance to understand the beneficial effects of FC-based interventions in the context of neurodegenerative diseases.
Collapse
Affiliation(s)
- Aina Badia-Soteras
- Department of Molecular and Cellular Neurobiology, Center for Neurogenomics and Cognitive Research, Faculty of Earth and Life Sciences, Vrije Universiteit, 1081 HV Amsterdam, The Netherlands; (A.B.-S.); (J.d.V.); (W.D.); (A.B.S.)
| | - Janneke de Vries
- Department of Molecular and Cellular Neurobiology, Center for Neurogenomics and Cognitive Research, Faculty of Earth and Life Sciences, Vrije Universiteit, 1081 HV Amsterdam, The Netherlands; (A.B.-S.); (J.d.V.); (W.D.); (A.B.S.)
| | - Werner Dykstra
- Department of Molecular and Cellular Neurobiology, Center for Neurogenomics and Cognitive Research, Faculty of Earth and Life Sciences, Vrije Universiteit, 1081 HV Amsterdam, The Netherlands; (A.B.-S.); (J.d.V.); (W.D.); (A.B.S.)
| | - Laus M. Broersen
- Danone Nutricia Research, 3584 CT Utrecht, The Netherlands; (L.M.B.); (J.M.V.)
| | - Jan Martin Verkuyl
- Danone Nutricia Research, 3584 CT Utrecht, The Netherlands; (L.M.B.); (J.M.V.)
| | - August B. Smit
- Department of Molecular and Cellular Neurobiology, Center for Neurogenomics and Cognitive Research, Faculty of Earth and Life Sciences, Vrije Universiteit, 1081 HV Amsterdam, The Netherlands; (A.B.-S.); (J.d.V.); (W.D.); (A.B.S.)
| | - Mark H. G. Verheijen
- Department of Molecular and Cellular Neurobiology, Center for Neurogenomics and Cognitive Research, Faculty of Earth and Life Sciences, Vrije Universiteit, 1081 HV Amsterdam, The Netherlands; (A.B.-S.); (J.d.V.); (W.D.); (A.B.S.)
- Correspondence:
| |
Collapse
|
5
|
Effect of omega-3 fatty acid supplementation on gene expression of inflammation, oxidative stress and cardiometabolic parameters: Systematic review and meta-analysis. J Funct Foods 2021. [DOI: 10.1016/j.jff.2021.104619] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
|
6
|
Huang X, Yi S, Hu J, Du Z, Wang Q, Ye Z, Su G, Kijlstra A, Yang P. Linoleic acid inhibits in vitro function of human and murine dendritic cells, CD4 +T cells and retinal pigment epithelial cells. Graefes Arch Clin Exp Ophthalmol 2020; 259:987-998. [PMID: 33079282 DOI: 10.1007/s00417-020-04972-6] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2020] [Revised: 09/29/2020] [Accepted: 10/07/2020] [Indexed: 02/07/2023] Open
Abstract
PURPOSE Increased linoleic acid (LA) was observed in acute anterior uveitis (AAU) patient feces in our previous study. To investigate the immunoregulatory effect of LA, we studied the effect of LA on human and murine dendritic cells (DCs), CD4+T cells, and retinal pigment epithelial (RPE) cells in vitro. METHODS The level of LA in feces from AAU patients and healthy individuals was measured by gas chromatography coupled with a mass spectrometer (GC-MS). The immunoregulatory effect of LA on human and murine DCs, CD4+ T cells, and RPE cells was evaluated by enzyme linked immunosorbent assay (ELISA) and flow cytometry (FCM). The effect of LA on DCs was evaluated by Tandem mass tag (TMT)-based proteomics analysis. RESULTS Increased LA was observed in feces from AAU patients (1018.35 ± 900.01 mg/kg) as compared with healthy individuals (472.55 ± 365.49 mg/kg, p = 0.0136). LA attenuated the antigen-presenting function of human and murine DCs by decreasing the expression of CD40, the secretion of IL-6 and IL-12p70, and the ability to shift naïve T cells towards T helper type 1 (Th1) and Th17 cells. LA also inhibited the secretion of MCP-1 and IL-8 from RPE cells. Proteomics analysis showed differential expression of 28 proteins, including squalene epoxidase (SQLE), farnesyl-diphosphate farnesyltransferase 1 (FDFT1), and cytochrome P450 family 51 subfamily A member 1 (CYP51A1), in LA-treated DCs compared with controls. LA also accelerated the apoptosis of DCs from healthy individuals. CONCLUSION LA inhibited the function of human and murine DCs, CD4+T cells, and RPE cells, regulated the expression of proteins, and promoted the apoptosis of human DCs. These results collectively suggest that LA might decrease the function of immune cells in vitro, and further studies are needed to investigate its role in the pathogenesis of AAU.
Collapse
Affiliation(s)
- Xinyue Huang
- The First Affiliated Hospital of Chongqing Medical University, Chongqing Key Laboratory of Ophthalmology, and Chongqing Eye Institute, Chongqing, People's Republic of China
| | - Shenglan Yi
- The First Affiliated Hospital of Chongqing Medical University, Chongqing Key Laboratory of Ophthalmology, and Chongqing Eye Institute, Chongqing, People's Republic of China
| | - Jianping Hu
- The First Affiliated Hospital of Chongqing Medical University, Chongqing Key Laboratory of Ophthalmology, and Chongqing Eye Institute, Chongqing, People's Republic of China
| | - Ziyu Du
- The First Affiliated Hospital of Chongqing Medical University, Chongqing Key Laboratory of Ophthalmology, and Chongqing Eye Institute, Chongqing, People's Republic of China
| | - Qingfeng Wang
- The First Affiliated Hospital of Chongqing Medical University, Chongqing Key Laboratory of Ophthalmology, and Chongqing Eye Institute, Chongqing, People's Republic of China
| | - Zi Ye
- The First Affiliated Hospital of Chongqing Medical University, Chongqing Key Laboratory of Ophthalmology, and Chongqing Eye Institute, Chongqing, People's Republic of China
| | - Guannan Su
- The First Affiliated Hospital of Chongqing Medical University, Chongqing Key Laboratory of Ophthalmology, and Chongqing Eye Institute, Chongqing, People's Republic of China
| | - Aize Kijlstra
- University Eye Clinic Maastricht, Maastricht, Netherlands
| | - Peizeng Yang
- The First Affiliated Hospital of Chongqing Medical University, Chongqing Key Laboratory of Ophthalmology, and Chongqing Eye Institute, Chongqing, People's Republic of China.
| |
Collapse
|
7
|
Millan-Linares MC, Toscano R, Lemus-Conejo A, Martin ME, Pedroche J, Millan F, Montserrat-de la Paz S. GPETAFLR, a biopeptide from Lupinus angustifolius L., protects against oxidative and inflammatory damage in retinal pigment epithelium cells. J Food Biochem 2019; 43:e12995. [PMID: 31659814 DOI: 10.1111/jfbc.12995] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2019] [Revised: 06/22/2019] [Accepted: 06/26/2019] [Indexed: 01/23/2023]
Abstract
GPETAFLR, an octapeptide released from the enzymatic hydrolysis of lupine (Lupinus angustifolius L.) protein, has demonstrated anti-inflammatory effect in myeloid lineage. This work aims to evaluate in retinal pigment epithelium (RPE) cells the protective role of GPETAFLR on both oxidative and inflammatory markers known to be involved in age-related macular degeneration (AMD). In comparison with stimulated control cells, GPETAFLR increased glutathione production and diminished the secretion and gene expression of VEFG, IL-1β, IL-6, IFNγ, and TNF-α, as well as reactive oxygen species, and nitrite output. Our findings reveal that GPETAFLR, a novel plant peptide, is able to protect against RPE oxidative stress and inflammation. Taken together, these results strongly support innovative nutritional strategies considering Lupinus angustifolius L. as source of proteins to prevent the onset and progression of AMD. PRACTICAL APPLICATIONS: We reveal a novel nutraceutical impact of GPETAFLR peptide in human RPE cells to prevent oxidative and inflammatory mediators. Our results support that the intake of Lupine angustifolius L., proposed to be a reservoir of GPETAFLR, could lessen the functional decay of RPE cells, leading therefore to a slowdown of the progress of AMD during age. Not only this work, but also future simple clinical studies should raise new nutritional strategies focused on understanding the etiological role of the foods, nutrition, and metabolism in the pathogenesis of ocular disorders.
Collapse
Affiliation(s)
| | - Rocio Toscano
- Department of Food & Health, Instituto de la Grasa, CSIC, Seville, Spain.,Department of Medical Biochemistry, Molecular Biology, and Immunology, School of Medicine, Universidad de Sevilla, Seville, Spain
| | - Ana Lemus-Conejo
- Department of Food & Health, Instituto de la Grasa, CSIC, Seville, Spain.,Department of Medical Biochemistry, Molecular Biology, and Immunology, School of Medicine, Universidad de Sevilla, Seville, Spain
| | - Maria E Martin
- Department of Cell Biology, Faculty of Biology, Universidad de Sevilla, Seville, Spain
| | - Justo Pedroche
- Department of Food & Health, Instituto de la Grasa, CSIC, Seville, Spain
| | - Francisco Millan
- Department of Food & Health, Instituto de la Grasa, CSIC, Seville, Spain
| | - Sergio Montserrat-de la Paz
- Department of Medical Biochemistry, Molecular Biology, and Immunology, School of Medicine, Universidad de Sevilla, Seville, Spain
| |
Collapse
|
8
|
Huang X, Zhen J, Dong S, Zhang H, Van Halm-Lutterodt N, Yuan L. DHA and vitamin E antagonized the Aβ25–35-mediated neuron oxidative damage through activation of Nrf2 signaling pathways and regulation of CD36, SRB1 and FABP5 expression in PC12 cells. Food Funct 2019; 10:1049-1061. [DOI: 10.1039/c8fo01713a] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
Abstract
The present study was designed to explore the neuroprotective effects of docosahexaenoic acid (DHA) and/or vitamin E (VE) in vitro.
Collapse
Affiliation(s)
- Xiaochen Huang
- School of Public Health
- Capital Medical University
- Beijing 100069
- P.R. China
| | - Jie Zhen
- School of Public Health
- Capital Medical University
- Beijing 100069
- P.R. China
| | - Shengqi Dong
- School of Public Health
- Capital Medical University
- Beijing 100069
- P.R. China
| | - Huiqiang Zhang
- School of Public Health
- Capital Medical University
- Beijing 100069
- P.R. China
| | | | - Linhong Yuan
- School of Public Health
- Capital Medical University
- Beijing 100069
- P.R. China
| |
Collapse
|
9
|
Das M, Das S. Docosahexaenoic Acid (DHA) Induced Morphological Differentiation of Astrocytes Is Associated with Transcriptional Upregulation and Endocytosis of β 2-AR. Mol Neurobiol 2018; 56:2685-2702. [PMID: 30054857 DOI: 10.1007/s12035-018-1260-0] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2018] [Accepted: 07/17/2018] [Indexed: 12/24/2022]
Abstract
Docosahexaenoic acid (DHA), an important ω-3 fatty acid, is abundantly present in the central nervous system and is important in every step of brain development. Much of this knowledge has been based on studies of the role of DHA in the function of the neurons, and reports on its effect on the glial cells are few and far between. We have previously reported that DHA facilitates astrocyte differentiation in primary culture. We have further explored the signaling mechanism associated with this event. It was observed that a sustained activation of the extracellular signal-regulated kinase (ERK) appeared to be critical for DHA-induced differentiation of the cultured astrocytes. Prior exposure to different endocytic inhibitors blocked both ERK activation and differentiation of the astrocytes during DHA treatment suggesting that the observed induction of ERK-2 was purely endosomal. Unlike the β1-adrenergic receptor (β1-AR) antagonist, atenolol, pre-treatment of the cells with the β2-adrenergic receptor (β2-AR) antagonist, ICI-118,551 inhibited the DHA-induced differentiation process, indicating a downstream involvement of β2-AR in the differentiation process. qRT-PCR and western blot analysis demonstrated a significant induction in the mRNA and protein expression of β2-AR at 18-24 h of DHA treatment, suggesting that the induction of β2-AR may be due to transcriptional upregulation. Moreover, DHA caused activation of PKA at 6 h, followed by activation of downstream cAMP response element-binding protein, a known transcription factor for β2-AR. Altogether, the observations suggest that DHA upregulates β2-AR in astrocytes, which undergo endocytosis and signals for sustained endosomal ERK activation to drive the differentiation process.
Collapse
Affiliation(s)
- Moitreyi Das
- Neurobiology Division, Cell Biology & Physiology Department, CSIR-Indian Institute of Chemical Biology, 4 Raja S. C. Mullick Road, Jadavpur, Kolkata, 700032, India
| | - Sumantra Das
- Neurobiology Division, Cell Biology & Physiology Department, CSIR-Indian Institute of Chemical Biology, 4 Raja S. C. Mullick Road, Jadavpur, Kolkata, 700032, India.
| |
Collapse
|
10
|
Deng Q, Wang Y, Wang C, Ji B, Cong R, Zhao L, Chen P, Zang X, Lu F, Han F, Huang F. Dietary supplementation with omega-3 polyunsaturated fatty acid-rich oils protects against visible-light-induced retinal damage in vivo. Food Funct 2018; 9:2469-2479. [DOI: 10.1039/c7fo01168g] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Abstract
Dietary ω-3 PUFA-rich oils protect against visible-light-induced retinal damage.
Collapse
|
11
|
Jamieson KL, Endo T, Darwesh AM, Samokhvalov V, Seubert JM. Cytochrome P450-derived eicosanoids and heart function. Pharmacol Ther 2017; 179:47-83. [DOI: 10.1016/j.pharmthera.2017.05.005] [Citation(s) in RCA: 82] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
|
12
|
Qiu S, Wei Y, Zhou X, Jiang Z, Zhang T, Jiang X, Zhang S. Intravitreal injection of docosahexaenoic acid attenuated photoreceptor cell injury in a NaIO 3 -induced age-related macular degeneration rat model. Neurosci Lett 2017; 657:53-61. [DOI: 10.1016/j.neulet.2017.07.041] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2017] [Revised: 07/07/2017] [Accepted: 07/24/2017] [Indexed: 12/11/2022]
|
13
|
Song EA, Lim JW, Kim H. Docosahexaenoic acid inhibits IL-6 expression via PPARγ-mediated expression of catalase in cerulein-stimulated pancreatic acinar cells. Int J Biochem Cell Biol 2017; 88:60-68. [PMID: 28483666 DOI: 10.1016/j.biocel.2017.05.011] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2016] [Revised: 04/28/2017] [Accepted: 05/04/2017] [Indexed: 02/07/2023]
Abstract
Cerulein pancreatitis mirrors human acute pancreatitis. In pancreatic acinar cells exposed to cerulein, reactive oxygen species (ROS) mediate inflammatory signaling by Janus kinase (JAK) 2/signal transducer and activator of transcription (STAT) 3, and cytokine induction. Docosahexaenoic acid (DHA) acts as an agonist of peroxisome proliferator activated receptor γ (PPARγ), which mediates the expression of some antioxidant enzymes. We hypothesized that DHA may induce PPARγ-target catalase expression and reduce ROS levels, leading to the inhibition of JAK2/STAT3 activation and IL-6 expression in cerulein-stimulated acinar cells. Pancreatic acinar AR42J cells were treated with DHA in the presence or absence of the PPARγ antagonist GW9662, or treated with the PPARγ agonist troglitazone, and then stimulated with cerulein. Expression of IL-6 and catalase, ROS levels, JAK2/STAT3 activation, and nuclear translocation of PPARγ were assessed. DHA suppressed the increase in ROS, JAK2/STAT3 activation, and IL-6 expression induced nuclear translocation of PPARγ and catalase expression in cerulein-stimulated AR42J cells. Troglitazone inhibited the cerulein-induced increase in ROS and IL-6 expression, but induced catalase expression similar to DHA in AR42J cells. GW9662 abolished the inhibitory effect of DHA on cerulein-induced increase in ROS and IL-6 expression in AR42J cells. DHA-induced expression of catalase was suppressed by GW9662 in cerulein-stimulated AR42J cells. Thus, DHA induces PPARγ activation and catalase expression, which inhibits ROS-mediated activation of JAK2/STAT3 and IL-6 expression in cerulein-stimulated pancreatic acinar cells.
Collapse
Affiliation(s)
- Eun Ah Song
- Department of Food and Nutrition, Brain Korea 21 PLUS Project, College of Human Ecology, Yonsei University, Seoul 03722, Republic of Korea
| | - Joo Weon Lim
- Department of Food and Nutrition, Brain Korea 21 PLUS Project, College of Human Ecology, Yonsei University, Seoul 03722, Republic of Korea
| | - Hyeyoung Kim
- Department of Food and Nutrition, Brain Korea 21 PLUS Project, College of Human Ecology, Yonsei University, Seoul 03722, Republic of Korea.
| |
Collapse
|
14
|
Rodrigues HG, Vinolo MAR, Sato FT, Magdalon J, Kuhl CMC, Yamagata AS, Pessoa AFM, Malheiros G, dos Santos MF, Lima C, Farsky SH, Camara NOS, Williner MR, Bernal CA, Calder PC, Curi R. Oral Administration of Linoleic Acid Induces New Vessel Formation and Improves Skin Wound Healing in Diabetic Rats. PLoS One 2016; 11:e0165115. [PMID: 27764229 PMCID: PMC5072690 DOI: 10.1371/journal.pone.0165115] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2016] [Accepted: 10/06/2016] [Indexed: 12/28/2022] Open
Abstract
Introduction Impaired wound healing has been widely reported in diabetes. Linoleic acid (LA) accelerates the skin wound healing process in non-diabetic rats. However, LA has not been tested in diabetic animals. Objectives We investigated whether oral administration of pure LA improves wound healing in streptozotocin-induced diabetic rats. Methods Dorsal wounds were induced in streptozotocin-induced type-1 diabetic rats treated or not with LA (0.22 g/kg b.w.) for 10 days. Wound closure was daily assessed for two weeks. Wound tissues were collected at specific time-points and used to measure fatty acid composition, and contents of cytokines, growth factors and eicosanoids. Histological and qPCR analyses were employed to examine the dynamics of cell migration during the healing process. Results LA reduced the wound area 14 days after wound induction. LA also increased the concentrations of cytokine-induced neutrophil chemotaxis (CINC-2αβ), tumor necrosis factor-α (TNF-α) and leukotriene B4 (LTB4), and reduced the expression of macrophage chemoattractant protein-1 (MCP-1) and macrophage inflammatory protein-1 (MIP-1). These results together with the histological analysis, which showed accumulation of leukocytes in the wound early in the healing process, indicate that LA brought forward the inflammatory phase and improved wound healing in diabetic rats. Angiogenesis was induced by LA through elevation in tissue content of key mediators of this process: vascular-endothelial growth factor (VEGF) and angiopoietin-2 (ANGPT-2). Conclusions Oral administration of LA hastened wound closure in diabetic rats by improving the inflammatory phase and angiogenesis.
Collapse
Affiliation(s)
- Hosana G. Rodrigues
- School of Applied Sciences, University of Campinas, Limeira, Brazil
- Department of Physiology and Biophysics, Institute of Biomedical Sciences, Sao Paulo University, Sao Paulo, Brazil
- * E-mail:
| | - Marco A. R. Vinolo
- Department of Genetics, Evolution and Bioagents, Institute of Biology, University of Campinas, Campinas, Brazil
| | - Fabio T. Sato
- Department of Genetics, Evolution and Bioagents, Institute of Biology, University of Campinas, Campinas, Brazil
| | - Juliana Magdalon
- Department of Physiology and Biophysics, Institute of Biomedical Sciences, Sao Paulo University, Sao Paulo, Brazil
| | | | - Ana S. Yamagata
- Department of Physiology and Biophysics, Institute of Biomedical Sciences, Sao Paulo University, Sao Paulo, Brazil
| | - Ana Flávia M. Pessoa
- Cell and Developmental Biology Department, Institute of Biomedical Sciences, Sao Paulo University, Sao Paulo, Brazil
| | - Gabriella Malheiros
- Cell and Developmental Biology Department, Institute of Biomedical Sciences, Sao Paulo University, Sao Paulo, Brazil
| | - Marinilce F. dos Santos
- Cell and Developmental Biology Department, Institute of Biomedical Sciences, Sao Paulo University, Sao Paulo, Brazil
| | - Camila Lima
- Department of Clinical and Toxicology Analyses, School of Pharmaceutical Sciences, Sao Paulo University, Sao Paulo, Brazil
| | - Sandra H. Farsky
- Department of Clinical and Toxicology Analyses, School of Pharmaceutical Sciences, Sao Paulo University, Sao Paulo, Brazil
| | - Niels O. S. Camara
- Department of Immunology, Institute of Biomedical Sciences, Sao Paulo University, Sao Paulo, Brazil
| | - Maria R. Williner
- Food Sciences and Nutrition, School of Biochemistry and Biological Sciences, National University of Litoral, Santa Fé, Argentina
| | - Claudio A. Bernal
- Food Sciences and Nutrition, School of Biochemistry and Biological Sciences, National University of Litoral, Santa Fé, Argentina
| | - Philip C. Calder
- Human Development and Health Academic Unit, Faculty of Medicine, University of Southampton, Southampton, United Kingdom
- NIHR Southampton Biomedical Research Centre, University Hospital Southampton NHS Foundation Trust and University of Southampton, Southampton, United Kingdom
| | - Rui Curi
- Department of Physiology and Biophysics, Institute of Biomedical Sciences, Sao Paulo University, Sao Paulo, Brazil
| |
Collapse
|
15
|
Lian S, Xia Y, Nguyen TT, Ung TT, Yoon HJ, Kim NH, Kim KK, Jung YD. Docosahexaenoic Acid Inhibits Tumor Promoter-Induced Urokinase-Type Plasminogen Activator Receptor by Suppressing PKCδ- and MAPKs-Mediated Pathways in ECV304 Human Endothelial Cells. PLoS One 2016; 11:e0163395. [PMID: 27654969 PMCID: PMC5031411 DOI: 10.1371/journal.pone.0163395] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2016] [Accepted: 09/06/2016] [Indexed: 11/18/2022] Open
Abstract
The overexpression of urokinase-type plasminogen activator receptor (uPAR) is associated with inflammation and virtually all human cancers. Despite the fact that docosahexaenoic acid (DHA) has been reported to possess anti-inflammatory and anti-tumor properties, the negative regulation of uPAR by DHA is still undefined. Here, we investigated the effect of DHA on 12-O-tetradecanoylphorbol-13-acetate (TPA)-induced uPAR expression and the underlying molecular mechanisms in ECV304 human endothelial cells. DHA concentration-dependently inhibited TPA-induced uPAR. Specific inhibitors and mutagenesis studies showed that PKCδ, JNK1/2, Erk1/2, NF-κB, and AP-1 were critical for TPA-induced uPAR expression. Application of DHA suppressed TPA-induced translocation of PKCδ, activation of the JNK1/2 and Erk1/2 signaling pathways, and subsequent AP-1 and NF-κB transactivation. In conclusion, these observations suggest a novel role for DHA in reducing uPAR expression and cell invasion by inhibition of PKCδ, JNK1/2, and Erk1/2, and the reduction of AP-1 and NF-κB activation in ECV304 human endothelial cells.
Collapse
Affiliation(s)
- Sen Lian
- Research Institute of Medical Sciences, Chonnam National University Medical School, Gwangju 501-190, Republic of Korea
| | - Yong Xia
- Research Institute of Medical Sciences, Chonnam National University Medical School, Gwangju 501-190, Republic of Korea
| | - Thi Thinh Nguyen
- Research Institute of Medical Sciences, Chonnam National University Medical School, Gwangju 501-190, Republic of Korea
| | - Trong Thuan Ung
- Research Institute of Medical Sciences, Chonnam National University Medical School, Gwangju 501-190, Republic of Korea
| | - Hyun Joong Yoon
- Research Institute of Medical Sciences, Chonnam National University Medical School, Gwangju 501-190, Republic of Korea
| | - Nam Ho Kim
- Research Institute of Medical Sciences, Chonnam National University Medical School, Gwangju 501-190, Republic of Korea
| | - Kyung Keun Kim
- Research Institute of Medical Sciences, Chonnam National University Medical School, Gwangju 501-190, Republic of Korea
| | - Young Do Jung
- Research Institute of Medical Sciences, Chonnam National University Medical School, Gwangju 501-190, Republic of Korea
- * E-mail:
| |
Collapse
|
16
|
Montserrat-de la Paz S, Naranjo MC, Bermudez B, Lopez S, Moreda W, Abia R, Muriana FJG. Postprandial dietary fatty acids exert divergent inflammatory responses in retinal-pigmented epithelium cells. Food Funct 2016; 7:1345-53. [DOI: 10.1039/c6fo00136j] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Postprandial triglyceride-rich lipoproteins (TRLs) lead to a complex series of events that are potentially oxidative and inflammatory.
Collapse
Affiliation(s)
| | - M. Carmen Naranjo
- Laboratory of Cellular and Molecular Nutrition
- Instituto de la Grasa
- CSIC
- 41013 Seville
- Spain
| | - Beatriz Bermudez
- Department of Pharmacology
- School of Pharmacy
- University of Seville
- 41012 Seville
- Spain
| | - Sergio Lopez
- Laboratory of Cellular and Molecular Nutrition
- Instituto de la Grasa
- CSIC
- 41013 Seville
- Spain
| | - Wenceslao Moreda
- Laboratory of Olive Oil Quality
- Purity and Technology
- Instituto de la Grasa
- CSIC
- 41013 Seville
| | - Rocio Abia
- Laboratory of Cellular and Molecular Nutrition
- Instituto de la Grasa
- CSIC
- 41013 Seville
- Spain
| | | |
Collapse
|
17
|
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
|