1
|
Müller AK, Albrecht F, Rohrer C, Koeberle A, Werz O, Schlörmann W, Glei M, Lorkowski S, Wallert M. Olive Oil Extracts and Oleic Acid Attenuate the LPS-Induced Inflammatory Response in Murine RAW264.7 Macrophages but Induce the Release of Prostaglandin E2. Nutrients 2021; 13:4437. [PMID: 34959989 PMCID: PMC8703532 DOI: 10.3390/nu13124437] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2021] [Revised: 12/08/2021] [Accepted: 12/09/2021] [Indexed: 12/11/2022] Open
Abstract
Olive oil contains high amounts of oleic acid (OA). Although OA has been described to inhibit inflammatory processes, the effects of olive oil on cellular mechanisms remain poorly understood. Therefore, we compared the effects of major fatty acids (FA) from olive oil with those of olive oil extracts (OOE) on inflammatory mediators and alterations in the cellular phospholipid composition in murine macrophages. Upon treatment with different OOE, FA compositions of lipopolysaccharide (LPS)-stimulated murine RAW264.7 macrophages were analyzed using gas chromatography. Olive oil extracts and OA significantly reduced the LPS-induced expression of inducible nitric oxide synthase (iNos), cyclooxygenase (Cox2), and interleukin-6 mRNA. In addition, a significant decrease in Cox2 and iNos protein expression was observed. The formation of nitric oxide was significantly reduced, while the formation of prostaglandin (PG) E2 from arachidonic acid significantly increased after treatment with OOE or OA. The latter was associated with a shift in the phospholipid FA composition from arachidonic acid to OA, resulting in an elevated availability of arachidonic acid. Together, OOE and OA mediate anti-inflammatory effects in vitro but increase the release of arachidonic acid and hereinafter PGE2, likely due to elongation of OA and competitive incorporation of fatty acids into membrane phospholipids.
Collapse
Affiliation(s)
- Anke Katharina Müller
- Department of Nutritional Biochemistry and Physiology, Institute of Nutritional Science, Friedrich Schiller University Jena, 07743 Jena, Germany; (A.K.M.); (F.A.); (C.R.); (S.L.)
- Competence Cluster for Nutrition and Cardiovascular Health (nutriCARD), Halle-Jena-Leipzig, 07743 Jena, Germany; (W.S.); (M.G.)
| | - Franziska Albrecht
- Department of Nutritional Biochemistry and Physiology, Institute of Nutritional Science, Friedrich Schiller University Jena, 07743 Jena, Germany; (A.K.M.); (F.A.); (C.R.); (S.L.)
| | - Carsten Rohrer
- Department of Nutritional Biochemistry and Physiology, Institute of Nutritional Science, Friedrich Schiller University Jena, 07743 Jena, Germany; (A.K.M.); (F.A.); (C.R.); (S.L.)
- Competence Cluster for Nutrition and Cardiovascular Health (nutriCARD), Halle-Jena-Leipzig, 07743 Jena, Germany; (W.S.); (M.G.)
| | - Andreas Koeberle
- Center for Molecular Biosciences Innsbruck (CMBI), Michael Popp Institute, University of Innsbruck, 6020 Innsbruck, Austria;
- Department of Pharmaceutical/Medicinal Chemistry, Institute of Pharmacy, Friedrich Schiller University Jena, 07743 Jena, Germany;
| | - Oliver Werz
- Department of Pharmaceutical/Medicinal Chemistry, Institute of Pharmacy, Friedrich Schiller University Jena, 07743 Jena, Germany;
| | - Wiebke Schlörmann
- Competence Cluster for Nutrition and Cardiovascular Health (nutriCARD), Halle-Jena-Leipzig, 07743 Jena, Germany; (W.S.); (M.G.)
- Department of Applied Nutritional Toxicology, Institute of Nutritional Science, Friedrich Schiller University Jena, 07743 Jena, Germany
| | - Michael Glei
- Competence Cluster for Nutrition and Cardiovascular Health (nutriCARD), Halle-Jena-Leipzig, 07743 Jena, Germany; (W.S.); (M.G.)
- Department of Applied Nutritional Toxicology, Institute of Nutritional Science, Friedrich Schiller University Jena, 07743 Jena, Germany
| | - Stefan Lorkowski
- Department of Nutritional Biochemistry and Physiology, Institute of Nutritional Science, Friedrich Schiller University Jena, 07743 Jena, Germany; (A.K.M.); (F.A.); (C.R.); (S.L.)
- Competence Cluster for Nutrition and Cardiovascular Health (nutriCARD), Halle-Jena-Leipzig, 07743 Jena, Germany; (W.S.); (M.G.)
| | - Maria Wallert
- Department of Nutritional Biochemistry and Physiology, Institute of Nutritional Science, Friedrich Schiller University Jena, 07743 Jena, Germany; (A.K.M.); (F.A.); (C.R.); (S.L.)
- Competence Cluster for Nutrition and Cardiovascular Health (nutriCARD), Halle-Jena-Leipzig, 07743 Jena, Germany; (W.S.); (M.G.)
| |
Collapse
|
2
|
Son HK, Xiang H, Park S, Lee J, Lee JJ, Jung S, Ha JH. Partial Replacement of Dietary Fat with Polyunsaturated Fatty Acids Attenuates the Lipopolysaccharide-Induced Hepatic Inflammation in Sprague-Dawley Rats Fed a High-Fat Diet. INTERNATIONAL JOURNAL OF ENVIRONMENTAL RESEARCH AND PUBLIC HEALTH 2021; 18:ijerph182010986. [PMID: 34682732 PMCID: PMC8535618 DOI: 10.3390/ijerph182010986] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/21/2021] [Revised: 10/13/2021] [Accepted: 10/16/2021] [Indexed: 12/17/2022]
Abstract
In this study, we investigated whether the partial replacement of dietary fat with polyunsaturated fatty acids (PUFAs) ameliorated the lipopolysaccharide (LPS)-induced hepatic inflammation in rats fed a high-fat diet. Male Sprague-Dawley rats were divided into three groups and provided each of the following diets: (1) high-fat diet (HFD), (2) HFD with perilla oil (PO), and (3) HFD with corn oil (CO). After 12 weeks of dietary intervention, the rats were intraperitoneally injected with LPS (5 mg/kg) from Escherichia coli O55:B5 or phosphate-buffered saline (PBS). Following LPS stimulation, serum insulin levels were increased, while PO and CO lowered the serum levels of glucose and insulin. In the liver, LPS increased the triglyceride levels, while PO and CO alleviated the LPS-induced hepatic triglyceride accumulation. In the LPS injected rats, the mRNA expression of genes related to inflammation and endoplasmic reticulum (ER) stress was attenuated by PO and CO in the liver. Furthermore, hepatic levels of proteins involved in the nuclear factor kappa-light-chain-enhancer of activated B cells/mitogen-activated protein kinase pathways, antioxidant response, and ER stress were lowered by PO- and CO-replacement. Therefore, the partial replacement of dietary fat with PUFAs alleviates LPS-induced hepatic inflammation during HFD consumption, which may decrease metabolic abnormalities.
Collapse
Affiliation(s)
- Hee-Kyoung Son
- Research Center for Industrialization of Natural Neutralization, Dankook University, Cheonan 31116, Korea; (H.-K.S.); (H.X.); (S.P.); (J.L.)
| | - Huo Xiang
- Research Center for Industrialization of Natural Neutralization, Dankook University, Cheonan 31116, Korea; (H.-K.S.); (H.X.); (S.P.); (J.L.)
- Department of Food Science and Nutrition, Dankook University, Cheonan 31116, Korea
| | - Seohyun Park
- Research Center for Industrialization of Natural Neutralization, Dankook University, Cheonan 31116, Korea; (H.-K.S.); (H.X.); (S.P.); (J.L.)
- Department of Food Science and Nutrition, Dankook University, Cheonan 31116, Korea
| | - Jisu Lee
- Research Center for Industrialization of Natural Neutralization, Dankook University, Cheonan 31116, Korea; (H.-K.S.); (H.X.); (S.P.); (J.L.)
- Department of Food Science and Nutrition, Dankook University, Cheonan 31116, Korea
| | - Jae-Joon Lee
- Department of Food and Nutrition, Chosun University, Gwangju 61452, Korea;
| | - Sunyoon Jung
- Research Center for Industrialization of Natural Neutralization, Dankook University, Cheonan 31116, Korea; (H.-K.S.); (H.X.); (S.P.); (J.L.)
- Department of Food Science and Nutrition, Dankook University, Cheonan 31116, Korea
- Correspondence: (S.J.); (J.-H.H.); Tel.: +82-31-8005-3186 (S.J.); +82-41-550-3479 (J.-H.H.)
| | - Jung-Heun Ha
- Research Center for Industrialization of Natural Neutralization, Dankook University, Cheonan 31116, Korea; (H.-K.S.); (H.X.); (S.P.); (J.L.)
- Department of Food Science and Nutrition, Dankook University, Cheonan 31116, Korea
- Correspondence: (S.J.); (J.-H.H.); Tel.: +82-31-8005-3186 (S.J.); +82-41-550-3479 (J.-H.H.)
| |
Collapse
|
3
|
Zhao Y, Chen Y, Wang J, Zhu X, Wang K, Li Y, Zhou F. Ginkgolide J protects human synovial cells SW982 via suppression of p38‑dependent production of pro‑inflammatory mediators. Mol Med Rep 2021; 24:555. [PMID: 34080024 PMCID: PMC8188640 DOI: 10.3892/mmr.2021.12194] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2020] [Accepted: 02/08/2021] [Indexed: 11/21/2022] Open
Abstract
Fibroblast-like synoviocytes (FLS) in the synovial lining play a key role in the pathological process of rheumatoid arthritis (RA), which produce pro-inflammatory mediators to perpetuate inflammation and proteases to contribute to cartilage destruction. Ginkgolide J (GJ) is a subclass of ginkgolides (GGs) that exhibits anti-inflammatory activity. In the present study, the protective effect of GJ on lipopolysaccharide (LPS)-treated human synovial cells SW982 and its related mechanisms were investigated using various methods, including ELISA, Griess assay, western blotting, immunofluorescence analysis and p38 kinase activity assay. The results revealed that GJ pretreatment significantly attenuated LPS-induced excess production of pro-inflammatory mediators in SW982 cells via suppression of tumor necrosis factor-α/interleukin (IL)-1β/IL-18/NF-κB/NLR family pyrin domain containing 3, prostaglandin E2/cyclooxygenase-2 and inducible nitric oxide synthase/nitric oxide signaling. Mechanistic studies revealed that p38 activation contributed to the LPS-induced inflammatory response, and GJ pretreatment dose-dependently attenuated p38 activation, indicating that the suppressive effect of GJ was achieved by targeting p38 signaling. These findings may contribute to the prevention and treatment of RA.
Collapse
Affiliation(s)
- Yujie Zhao
- NHC Key Laboratory of Nuclear Medicine, Jiangsu Key Laboratory of Molecular Nuclear Medicine, Jiangsu Institute of Nuclear Medicine, Wuxi, Jiangsu 214063, P.R. China
| | - Yuan Chen
- NHC Key Laboratory of Nuclear Medicine, Jiangsu Key Laboratory of Molecular Nuclear Medicine, Jiangsu Institute of Nuclear Medicine, Wuxi, Jiangsu 214063, P.R. China
| | - Jiayi Wang
- Department of Rheumatology, Shanghai East Hospital, Tongji University School of Medicine, Shanghai 200120, P.R. China
| | - Xue Zhu
- NHC Key Laboratory of Nuclear Medicine, Jiangsu Key Laboratory of Molecular Nuclear Medicine, Jiangsu Institute of Nuclear Medicine, Wuxi, Jiangsu 214063, P.R. China
| | - Ke Wang
- NHC Key Laboratory of Nuclear Medicine, Jiangsu Key Laboratory of Molecular Nuclear Medicine, Jiangsu Institute of Nuclear Medicine, Wuxi, Jiangsu 214063, P.R. China
| | - Yue Li
- Faculty of Pharmacy, The University of Sydney, Sydney, NSW 2006, Australia
| | - Fanfan Zhou
- Faculty of Pharmacy, The University of Sydney, Sydney, NSW 2006, Australia
| |
Collapse
|
4
|
Fattahi N, Shahbazi MA, Maleki A, Hamidi M, Ramazani A, Santos HA. Emerging insights on drug delivery by fatty acid mediated synthesis of lipophilic prodrugs as novel nanomedicines. J Control Release 2020; 326:556-598. [PMID: 32726650 DOI: 10.1016/j.jconrel.2020.07.012] [Citation(s) in RCA: 42] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2020] [Revised: 07/09/2020] [Accepted: 07/10/2020] [Indexed: 12/25/2022]
Abstract
Many drug molecules that are currently in the market suffer from short half-life, poor absorption, low specificity, rapid degradation, and resistance development. The design and development of lipophilic prodrugs can provide numerous benefits to overcome these challenges. Fatty acids (FAs), which are lipophilic biomolecules constituted of essential components of the living cells, carry out many necessary functions required for the development of efficient prodrugs. Chemical conjugation of FAs to drug molecules may change their pharmacodynamics/pharmacokinetics in vivo and even their toxicity profile. Well-designed FA-based prodrugs can also present other benefits, such as improved oral bioavailability, promoted tumor targeting efficiency, controlled drug release, and enhanced cellular penetration, leading to improved therapeutic efficacy. In this review, we discuss diverse drug molecules conjugated to various unsaturated FAs. Furthermore, various drug-FA conjugates loaded into various nanostructure delivery systems, including liposomes, solid lipid nanoparticles, emulsions, nano-assemblies, micelles, and polymeric nanoparticles, are reviewed. The present review aims to inspire readers to explore new avenues in prodrug design based on the various FAs with or without nanostructured delivery systems.
Collapse
Affiliation(s)
- Nadia Fattahi
- Department of Chemistry, Faculty of Science, University of Zanjan, P.O. Box 45195-313, Zanjan, Iran; Trita Nanomedicine Research Center (TNRC), Trita Third Millennium Pharmaceuticals, 45331-55681 Zanjan, Iran
| | - Mohammad-Ali Shahbazi
- Drug Research Program, Division of Pharmaceutical Chemistry and Technology, Faculty of Pharmacy, University of Helsinki, Helsinki FI-00014, Finland; Department of Pharmaceutical Nanotechnology, School of Pharmacy, Zanjan University of Medical Sciences, Zanjan, Iran; Zanjan Pharmaceutical Nanotechnology Research Center (ZPNRC), Zanjan University of Medical Sciences, Zanjan, Iran.
| | - Aziz Maleki
- Department of Pharmaceutical Nanotechnology, School of Pharmacy, Zanjan University of Medical Sciences, Zanjan, Iran; Zanjan Pharmaceutical Nanotechnology Research Center (ZPNRC), Zanjan University of Medical Sciences, Zanjan, Iran
| | - Mehrdad Hamidi
- Trita Nanomedicine Research Center (TNRC), Trita Third Millennium Pharmaceuticals, 45331-55681 Zanjan, Iran; Department of Pharmaceutical Nanotechnology, School of Pharmacy, Zanjan University of Medical Sciences, Zanjan, Iran; Zanjan Pharmaceutical Nanotechnology Research Center (ZPNRC), Zanjan University of Medical Sciences, Zanjan, Iran.
| | - Ali Ramazani
- Department of Chemistry, Faculty of Science, University of Zanjan, P.O. Box 45195-313, Zanjan, Iran; Research Institute of Modern Biological Techniques (RIMBT), University of Zanjan, P.O. Box 45195-313, Zanjan, Iran
| | - Hélder A Santos
- Drug Research Program, Division of Pharmaceutical Chemistry and Technology, Faculty of Pharmacy, University of Helsinki, Helsinki FI-00014, Finland; Helsinki Institute of Life Science (HiLIFE), Faculty of Pharmacy, University of Helsinki, Helsinki FI-00014, Finland.
| |
Collapse
|
5
|
Zhang J, Tang X, Huang C, Liu Z, Ye Y. Oleic Acid Copolymer as A Novel Upconversion Nanomaterial to Make Doxorubicin-Loaded Nanomicelles with Dual Responsiveness to pH and NIR. Pharmaceutics 2020; 12:pharmaceutics12070680. [PMID: 32698309 PMCID: PMC7408047 DOI: 10.3390/pharmaceutics12070680] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2020] [Revised: 07/16/2020] [Accepted: 07/17/2020] [Indexed: 12/29/2022] Open
Abstract
Oleic acid (OA) as main component of plant oil is an important solvent but seldom used in the nanocarrier of anticancer drugs because of strong hydrophobicity and little drug release. In order to develop a new type of OA nanomaterial with dual responses to pH and near infrared light (NIR) to achieve the intelligent delivery of anticancer drugs. The novel OA copolymer (mPEG-PEI-(NBS, OA)) was synthesized by grafting OA and o-nitrobenzyl succinate (NBS) onto mPEGylated polyethyleneimine (mPEG-PEI) by amidation reaction. It was further conjugated with NaYF4:Yb3+/Er3+ nanoparticles, and encapsulated doxorubicin (DOX) through self-assembly to make upconversion nanomicelles with dual response to pH and NIR. Drug release behavior of DOX, physicochemical characteristics of the nanomicelles were evaluated, along with its cytotoxic profile, as well as the degree of cellular uptake in A549 cells. The encapsulation efficiency and drug loading capacity of DOX in the nanomicelles were 73.84% ± 0.58% and 4.62% ± 0.28%, respectively, and the encapsulated DOX was quickly released in an acidic environment exposed to irradiation at 980 nm. The blank nanomicelles exhibited low cytotoxicity and excellent biocompatibility by MTT assay against A549 cells. The DOX-loaded nanomicelles showed remarkable cytotoxicity to A549 cells under NIR, and promoted the cellular uptake of DOX into the cytoplasm and nucleus of cancer cells. OA copolymer can effectively deliver DOX to cancer cells and achieve tumor targeting through a dual response to pH and NIR.
Collapse
Affiliation(s)
| | | | | | | | - Yong Ye
- Correspondence: ; Tel.: +86-20-87110234
| |
Collapse
|
6
|
Adam GO, Kim GB, Lee SJ, Lee H, Kang HS, Kim SJ. Red Ginseng Reduces Inflammatory Response via Suppression MAPK/P38 Signaling and p65 Nuclear Proteins Translocation in Rats and Raw 264.7 Macrophage. THE AMERICAN JOURNAL OF CHINESE MEDICINE 2019; 47:1589-1609. [PMID: 31645122 DOI: 10.1142/s0192415x19500812] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
Abstract
Lipopolysaccharides (LPS) cause systemic inflammatory responses, which are characterized by high mortality and multiple signs, including metabolic disturbances, respiratory acidosis, hypotension, and vital organs disorder. Cytokines secretion and oxidative stress are the main features of the disease. Diagnosis and treatment of systemic inflammation (SI) remain a challenge. Korean Red Ginseng (RG) is one of medicinal herbs that showed a potent anti-oxidant effect. We aimed to study the protective effects of RG on systemic inflammatory response in rats and RAW 264.7 macrophage cells induced by LPS. The rats were treated with water and alcohol extracts of RG for four weeks to prevent the inflammatory response. The result showed that LPS toxin increased morbidity and mortality, and induced liver, kidney, and lung injuries manifested by deteriorated biomarkers. Hypotension, hypomagnesemia, acidosis, and oxidative stress were observed in septic rats. However, RG extracts attenuated liver, kidney, and lung enzymes and metabolites in treated groups via its anti-inflammatory and anti-oxidant properties. Furthermore, RG improved magnesium and blood pressure in the treated groups. RAW 264.7 macrophage cells exposed to LPS disturbance in translocation of p65 and MAPK/p38. Nevertheless, RG-pretreated cells did not significantly alter. In conclusion, RG reduced the rates of mortality and morbidity of treated rats - liver, kidney, and lung injuries were protected in the treated groups through the potentiation of anti-oxidant defense. RG was able to conserve mitochondrial function, inhibiting the activation of MAPK/p38 signaling and suppressing NF-κB p65 cytoplasm-nucleus transport. Further studies are needed to examine the effects on chronic conditions in animal models and human.
Collapse
Affiliation(s)
- Gareeballah Osman Adam
- Department of Pharmacology and Toxicology, College of Veterinary Medicine, Chonbuk National University, Iksan Campus, 79 Gobong-ro, Iksan-si, Jeollabuk-do 54596, Republic of Korea.,Department of Veterinary Medicine and Surgery, College of Veterinary Medicine, Sudan University of Science and Technology, P.O. Box No. 204, Hilat Kuku, Khartoum, Sudan
| | - Gi-Beum Kim
- Department of Pharmacology and Toxicology, College of Veterinary Medicine, Chonbuk National University, Iksan Campus, 79 Gobong-ro, Iksan-si, Jeollabuk-do 54596, Republic of Korea
| | - Sei-Jin Lee
- Korea Basic Science Institute Jeonju Center, Deokjin-gu, Jeonju-si, Jeollabuk-do 54896, Republic of Korea
| | - Heeryung Lee
- Department of Pharmacology and Toxicology, College of Veterinary Medicine, Chonbuk National University, Iksan Campus, 79 Gobong-ro, Iksan-si, Jeollabuk-do 54596, Republic of Korea.,Hansarang Animal Clinic, 27 Seongbuk-ro, Seoul 02880, Republic of Korea
| | - Hyung-Sub Kang
- Department of Pharmacology and Toxicology, College of Veterinary Medicine, Chonbuk National University, Iksan Campus, 79 Gobong-ro, Iksan-si, Jeollabuk-do 54596, Republic of Korea
| | - Shang-Jin Kim
- Department of Pharmacology and Toxicology, College of Veterinary Medicine, Chonbuk National University, Iksan Campus, 79 Gobong-ro, Iksan-si, Jeollabuk-do 54596, Republic of Korea
| |
Collapse
|
7
|
Kunz M, Simon JC, Saalbach A. Psoriasis: Obesity and Fatty Acids. Front Immunol 2019; 10:1807. [PMID: 31417571 PMCID: PMC6684944 DOI: 10.3389/fimmu.2019.01807] [Citation(s) in RCA: 50] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2019] [Accepted: 07/17/2019] [Indexed: 01/10/2023] Open
Abstract
Psoriasis is chronic inflammatory skin disease affecting skin, joints, cardiovascular system, brain, and metabolism. The pathogenesis of psoriasis is mediated by a complex interplay between the immune system, inflammatory mediators of different pathways, e.g., TNF-alpha and the IL-23/IL-17 pathways, psoriasis-associated susceptibility loci, autoantigens, and multiple environmental factors. Psoriasis is triggered by the combination of genetic and environmental factors. A novel environmental risk factor with rising importance is obesity. Several studies proved that obesity is an independent risk factor for the onset and severity of psoriasis. Due to the dramatic increase of obesity worldwide this minireview focuses on obesity as a major environmental risk factor for psoriasis and the mechanisms of obesity-mediated exacerbation of psoriasis.
Collapse
Affiliation(s)
- Manfred Kunz
- Department of Dermatology, Venereology and Allergology, University of Leipzig, Leipzig, Germany
| | - Jan C Simon
- Department of Dermatology, Venereology and Allergology, University of Leipzig, Leipzig, Germany
| | - Anja Saalbach
- Department of Dermatology, Venereology and Allergology, University of Leipzig, Leipzig, Germany
| |
Collapse
|
8
|
In Vitro Digested Nut Oils Attenuate the Lipopolysaccharide-Induced Inflammatory Response in Macrophages. Nutrients 2019; 11:nu11030503. [PMID: 30818812 PMCID: PMC6471109 DOI: 10.3390/nu11030503] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2019] [Revised: 02/15/2019] [Accepted: 02/24/2019] [Indexed: 12/22/2022] Open
Abstract
Nut consumption is known for its health benefits, in particular in inflammatory diseases. A possible mechanism for these effects could be their beneficial fatty acid composition. Nuts mainly contain mono- and polyunsaturated fatty acids, which have anti-inflammatory properties. However, studies investigating the effects of nut extracts on inflammatory processes on the molecular level are rare. We therefore prepared oily nut extracts after in vitro digestion and saponification of the fat-soluble constituents. Besides chromatographic analysis, cell culture experiments were performed using murine macrophages (RAW264.7) to study the capacity of different nut extracts (hazelnut, almond, walnut, macadamia, and pistachio) to modulate inflammatory processes. Oleic acid was the main fatty acid in hazelnut, almond, macadamia, and pistachio extracts. Both oily nut extracts and pure oleic acid significantly reduced the LPS-induced expression of iNos, Cox2, Tnfα, Il1β, and Il6 mRNAs. iNos protein expression was down-regulated followed by reduced nitric oxide formation. Thus, nut extracts at concentrations achievable in the digestive tract inhibit the expression and formation of inflammatory mediators in macrophages. Hence, a beneficial contribution of nut consumption to inflammatory diseases can be assumed. We are convinced that these results provide new insights on the molecular mechanisms involved in the health-beneficial effects of nuts.
Collapse
|
9
|
Herbert D, Franz S, Popkova Y, Anderegg U, Schiller J, Schwede K, Lorz A, Simon JC, Saalbach A. High-Fat Diet Exacerbates Early Psoriatic Skin Inflammation Independent of Obesity: Saturated Fatty Acids as Key Players. J Invest Dermatol 2018; 138:1999-2009. [DOI: 10.1016/j.jid.2018.03.1522] [Citation(s) in RCA: 55] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2018] [Revised: 03/12/2018] [Accepted: 03/18/2018] [Indexed: 12/13/2022]
|
10
|
Sun B, Luo C, Cui W, Sun J, He Z. Chemotherapy agent-unsaturated fatty acid prodrugs and prodrug-nanoplatforms for cancer chemotherapy. J Control Release 2017; 264:145-159. [DOI: 10.1016/j.jconrel.2017.08.034] [Citation(s) in RCA: 76] [Impact Index Per Article: 10.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2017] [Revised: 08/23/2017] [Accepted: 08/23/2017] [Indexed: 12/22/2022]
|
11
|
Giacoppo S, Rajan TS, Iori R, Rollin P, Bramanti P, Mazzon E. The α-cyclodextrin complex of the Moringa isothiocyanate suppresses lipopolysaccharide-induced inflammation in RAW 264.7 macrophage cells through Akt and p38 inhibition. Inflamm Res 2017; 66:487-503. [PMID: 28289752 DOI: 10.1007/s00011-017-1033-7] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2016] [Revised: 02/17/2017] [Accepted: 02/27/2017] [Indexed: 12/14/2022] Open
Abstract
In the last decades, a growing need to discover new compounds for the prevention and treatment of inflammatory diseases has led researchers to consider drugs derived from natural products as a valid option in the treatment of inflammation-associated disorders. The purpose of the present study was to investigate the anti-inflammatory effects of a new formulation of Moringa oleifera-derived 4-(α-L-rhamnopyranosyloxy)benzyl isothiocyanate as a complex with alpha-cyclodextrin (moringin + α-CD) on lipopolysaccharide (LPS)-stimulated RAW 264.7 macrophage cells, a common model used for inflammation studies. In buffered/aqueous solution, the moringin + α-CD complex has enhanced the water solubility and stability of this isothiocyanate by forming a stable inclusion system. Our results showed that moringin + α-CD inhibits the production of inflammatory mediators in LPS-stimulated macrophages by down-regulation of pro-inflammatory cytokines (TNF-α and IL-1β), by preventing IκB-α phosphorylation, translocation of the nuclear factor-κB (NF-κB), and also via the suppression of Akt and p38 phosphorylation. In addition, as a consequence of upstream inhibition of the inflammatory pathway following treatment with moringin + α-CD, the modulation of the oxidative stress (results focused on the expression of iNOS and nitrotyrosine) and apoptotic pathway (Bax and Bcl-2) was demonstrated. Therefore, moringin + α-CD appears to be a new relevant helpful tool to use in clinical practice for inflammation-associated disorders.
Collapse
Affiliation(s)
- Sabrina Giacoppo
- IRCCS Centro Neurolesi "Bonino-Pulejo", Via Provinciale Palermo, Contrada Casazza, 98124, Messina, Italy
| | - Thangavelu Soundara Rajan
- IRCCS Centro Neurolesi "Bonino-Pulejo", Via Provinciale Palermo, Contrada Casazza, 98124, Messina, Italy
| | - Renato Iori
- Consiglio per la ricerca in agricoltura e l'analisi dell'economia agraria, Centro di ricerca Agricoltura e Ambiente (CREA-AA), Via di Corticella 133, 40128, Bologna, Italy
| | - Patrick Rollin
- Université d'Orléans et CNRS, ICOA, UMR 7311, BP 6759, 45067, Orléans, France
| | - Placido Bramanti
- IRCCS Centro Neurolesi "Bonino-Pulejo", Via Provinciale Palermo, Contrada Casazza, 98124, Messina, Italy
| | - Emanuela Mazzon
- IRCCS Centro Neurolesi "Bonino-Pulejo", Via Provinciale Palermo, Contrada Casazza, 98124, Messina, Italy.
| |
Collapse
|
12
|
Chang CF, Liao KC, Chen CH. 2-Phenylnaphthalene Derivatives Inhibit Lipopolysaccharide-Induced Pro-Inflammatory Mediators by Downregulating of MAPK/NF-κB Pathways in RAW 264.7 Macrophage Cells. PLoS One 2017; 12:e0168945. [PMID: 28060845 PMCID: PMC5218479 DOI: 10.1371/journal.pone.0168945] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2016] [Accepted: 12/08/2016] [Indexed: 11/24/2022] Open
Abstract
The anti-inflammatory pharmacological effect of eight 2-phenylnaphthalenes (PNAP-1−PNAP-8) on lipopolysaccharide (LPS)-induced RAW 264.7 (a mouse cell line) was investigated. Among them, 6,7-dihydroxy-2-(4′-hydroxyphenyl)naphthalene (PNAP-6) and 2-(4′-aminophenyl)-6,7-dimethoxynaphthalene (PNAP-8) exhibited the best anti-inflammatory activity in this study. PNAP-6 and PNAP-8 not only significantly decreased the expression of inducible nitric oxide synthase and cyclooxygenase-II, but also inhibited the production of nitric oxide, interleukin-6, and tumor necrosis factor-α in LPS stimulated cells. Moreover, PNAP-6 and PNAP-8 inhibited nuclear factor (NF)-κB activation by decreasing the degradation of IκB and nuclear translocation of NF-κB subunit (p65). In addition, PNAP-6 and PNAP-8 also attenuated the phosphorylation of ERK, p38, and JNK. These results suggest that PNAP-6 and PNAP-8 exert anti-inflammatory activities by down regulating NF-κB activation and the mitogen-activated protein kinase signaling pathway in LPS-stimulated Raw 264.7 cells. This is the first study demonstrating that PNAPs can inhibit LPS-induced pro-inflammatory mediators in macrophages cells.
Collapse
Affiliation(s)
- Chi-Fen Chang
- Department of Anatomy, School of Medicine, China Medical University, Taichung, Taiwan
- * E-mail:
| | - Kang-Chun Liao
- Department of Biomedical Imaging and Radiological Science, China Medical University, Taichung, Taiwan
| | - Chung-Hwan Chen
- Department of Orthopedics, Kaohsiung Municipal Ta-Tung Hospital, Kaohsiung Medical University, Kaohsiung, Taiwan
- Department of Orthopedics, College of Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan
- Orthopaedic Research Center, Kaohsiung Medical University, Kaohsiung, Taiwan
| |
Collapse
|
13
|
Lin S, Zhang Y, Long Y, Wan H, Che L, Lin Y, Xu S, Feng B, Li J, Wu D, Fang Z. Mammary inflammatory gene expression was associated with reproductive stage and regulated by docosahexenoic acid: in vitro and in vivo studies. Lipids Health Dis 2016; 15:215. [PMID: 27938408 PMCID: PMC5148867 DOI: 10.1186/s12944-016-0386-1] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2016] [Accepted: 12/02/2016] [Indexed: 01/15/2023] Open
Abstract
Background Periparturient mastitis is the most prevalent disease affecting lactating animals. However, it has long been relied on antibiotics to deal with mastitis, leading to a potential threat to food safety. This study was aimed to investigate the expression of pro-inflammatory cytokines in mammary glands of sows around parturition when mastitis and oxidative stress usually occur, and evaluate the anti-inflammatory effect of docosahexenoic acid (DHA) in porcine mammary epithelial cells (PMEC) challenged by lipopolysaccharide (LPS). Methods Mammary tissues and blood samples were collected from seven pregnant sows at different reproductive stages. Primarily cultured PMEC at passage 4 were assigned to four treatments: basal medium (control), basal medium with LPS (10 μg/mL) (LPS treatment), basal medium with LPS (10 μg/mL) and DHA (100 or 200 μM) (LPS + DHA treatments), and cell samples were harvested after 24 h incubation. The measurements included oxidative stress markers in blood samples and gene expression in mammary tissues and PMEC samples. Results Serum α-tocopherol concentration was lower at parturition than at day 90 of gestation and day 28 post parturition, while serum malondialdehyde concentration was higher at day 28 post parturition than at day 90 of gestation. Higher interleukin (IL)-1β mRNA abundance while lower LPS binding protein mRNA abundance in mammary tissues were observed at day 90 of gestation compared with that at parturition and at day 28 and 35 post parturition. Mammary tumor necrosis factor (TNF)-α mRNA abundance were lower at parturition than at day 90 of gestation and day 28 and 35 post parturition, whereas mammary IL-8 mRNA abundance were lower at parturition than at day 35 post parturition. In the PMEC experiment, compared with the control, increased mRNA abundances of Toll-like receptor (TLR)-4 downstream target, myeloid differentiation factor 88 (MyD88), IL-6 and IL-8 were observed in LPS treatment, whereas DHA appeared to decrease mRNA abundances of MyD88, IL-6 and IL-8 induced by LPS. Conclusions The down-regulated expression of pro-inflammatory cytokines in mammary tissues and aggravated systemic oxidative stress at parturition suggest that sows are in a vulnerable status during periparturient period. DHA appears to attenuate inflammatory responses in LPS-challenged PMEC through modulation of TLR4 signalling pathway.
Collapse
Affiliation(s)
- Sen Lin
- Key Laboratory for Animal Disease Resistance Nutrition of the Ministry of Education, Animal Nutrition Institute, Sichuan Agricultural University, Chengdu, 611130, People's Republic of China
| | - Yalin Zhang
- Key Laboratory for Animal Disease Resistance Nutrition of the Ministry of Education, Animal Nutrition Institute, Sichuan Agricultural University, Chengdu, 611130, People's Republic of China
| | - Yanrong Long
- Key Laboratory for Animal Disease Resistance Nutrition of the Ministry of Education, Animal Nutrition Institute, Sichuan Agricultural University, Chengdu, 611130, People's Republic of China
| | - Haifeng Wan
- Key Laboratory for Animal Disease Resistance Nutrition of the Ministry of Education, Animal Nutrition Institute, Sichuan Agricultural University, Chengdu, 611130, People's Republic of China
| | - Lianqiang Che
- Key Laboratory for Animal Disease Resistance Nutrition of the Ministry of Education, Animal Nutrition Institute, Sichuan Agricultural University, Chengdu, 611130, People's Republic of China
| | - Yan Lin
- Key Laboratory for Animal Disease Resistance Nutrition of the Ministry of Education, Animal Nutrition Institute, Sichuan Agricultural University, Chengdu, 611130, People's Republic of China
| | - Shengyu Xu
- Key Laboratory for Animal Disease Resistance Nutrition of the Ministry of Education, Animal Nutrition Institute, Sichuan Agricultural University, Chengdu, 611130, People's Republic of China
| | - Bin Feng
- Key Laboratory for Animal Disease Resistance Nutrition of the Ministry of Education, Animal Nutrition Institute, Sichuan Agricultural University, Chengdu, 611130, People's Republic of China
| | - Jian Li
- Key Laboratory for Animal Disease Resistance Nutrition of the Ministry of Education, Animal Nutrition Institute, Sichuan Agricultural University, Chengdu, 611130, People's Republic of China
| | - De Wu
- Key Laboratory for Animal Disease Resistance Nutrition of the Ministry of Education, Animal Nutrition Institute, Sichuan Agricultural University, Chengdu, 611130, People's Republic of China
| | - Zhengfeng Fang
- Key Laboratory for Animal Disease Resistance Nutrition of the Ministry of Education, Animal Nutrition Institute, Sichuan Agricultural University, Chengdu, 611130, People's Republic of China.
| |
Collapse
|
14
|
Stelzner K, Herbert D, Popkova Y, Lorz A, Schiller J, Gericke M, Klöting N, Blüher M, Franz S, Simon JC, Saalbach A. Free fatty acids sensitize dendritic cells to amplify TH1/TH17-immune responses. Eur J Immunol 2016; 46:2043-53. [PMID: 27214608 DOI: 10.1002/eji.201546263] [Citation(s) in RCA: 59] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2015] [Revised: 04/25/2016] [Accepted: 05/18/2016] [Indexed: 12/20/2022]
Abstract
Obesity is associated with body fat gain and impaired glucose metabolism. Here, we identified both body fat gain in obesity and impaired glucose metabolism as two independent risk factors for increased serum levels of free fatty acids (FFAs). Since obesity is associated with increased and/or delayed resolution of inflammation observed in various chronic inflammatory diseases such as psoriasis, we investigated the impact of FFAs on human monocyte-derived and mouse bone marrow-derived dendritic cell (DCs) functions relevant for the pathogenesis of chronic inflammation. FFAs such as palmitic acid (PA) and oleic acid (OA) did not affect the pro-inflammatory immune response of DCs. In contrast, PA and OA sensitize DCs resulting in augmented secretion of TH1/TH17-instructive cytokines upon pro-inflammatory stimulation. Interestingly, obesity in mice worsened a TH1/TH17-driven psoriasis-like skin inflammation. Strong correlation of the amount of total FFA, PA, and OA in serum with the severity of skin inflammation points to a critical role of FFA in obesity-mediated exacerbation of skin inflammation. Our data suggest that increased levels of FFAs might be a predisposing factor promoting a TH1/TH17-mediated inflammation such as psoriasis in response to an inflammatory danger signal.
Collapse
Affiliation(s)
- Kristin Stelzner
- Department of Dermatology, Venerology and Allergology, Medical Faculty of University of Leipzig, Germany
| | - Diana Herbert
- Department of Dermatology, Venerology and Allergology, Medical Faculty of University of Leipzig, Germany
| | - Yulia Popkova
- Institute of Medical Physics and Biophysics, Medical Faculty of University of Leipzig, Germany
| | - Axel Lorz
- Department of Dermatology, Venerology and Allergology, Medical Faculty of University of Leipzig, Germany
| | - Jürgen Schiller
- Institute of Medical Physics and Biophysics, Medical Faculty of University of Leipzig, Germany
| | - Martin Gericke
- Institute of Anatomy, Medical Faculty of University of Leipzig, Germany
| | - Nora Klöting
- IFB Adiposity Diseases, University of Leipzig, Germany
| | - Matthias Blüher
- Department of Medicine, Medical Faculty of University of Leipzig, Germany
| | - Sandra Franz
- Department of Dermatology, Venerology and Allergology, Medical Faculty of University of Leipzig, Germany
| | - Jan C Simon
- Department of Dermatology, Venerology and Allergology, Medical Faculty of University of Leipzig, Germany
| | - Anja Saalbach
- Department of Dermatology, Venerology and Allergology, Medical Faculty of University of Leipzig, Germany
| |
Collapse
|
15
|
Caspar-Bauguil S, Kolditz CI, Lefort C, Vila I, Mouisel E, Beuzelin D, Tavernier G, Marques MA, Zakaroff-Girard A, Pecher C, Houssier M, Mir L, Nicolas S, Moro C, Langin D. Fatty acids from fat cell lipolysis do not activate an inflammatory response but are stored as triacylglycerols in adipose tissue macrophages. Diabetologia 2015; 58:2627-36. [PMID: 26245186 DOI: 10.1007/s00125-015-3719-0] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/25/2015] [Accepted: 07/09/2015] [Indexed: 01/28/2023]
Abstract
AIMS/HYPOTHESIS Activation of macrophages by fatty acids (FAs) is a potential mechanism linking obesity to adipose tissue (AT) inflammation and insulin resistance. Here, we investigated the effects of FAs released during adipocyte lipolysis on AT macrophages (ATMs). METHODS Human THP-1 macrophages were treated with media from human multipotent adipose-derived stem (hMADS) adipocytes stimulated with lipolytic drugs. Macrophages were also treated with mixtures of FAs and an inhibitor of Toll-like receptor 4, since this receptor is activated by saturated FAs. Levels of mRNA and the secretion of inflammation-related molecules were measured in macrophages. FA composition was determined in adipocytes, conditioned media and macrophages. The effect of chronic inhibition or acute activation of fat cell lipolysis on ATM response was investigated in vivo in mice. RESULTS Whereas palmitic acid alone activates THP-1, conditioned media from hMADS adipocyte lipolysis had no effect on IL, chemokine and cytokine gene expression, and secretion by macrophages. Mixtures of FAs representing de novo lipogenesis or habitual dietary conditions also had no effect. FAs derived from adipocyte lipolysis were taken up by macrophages and stored as triacylglycerol droplets. In vivo, chronic treatment with an antilipolytic drug did not modify gene expression and number of ATMs in mice with intact or defective Tlr4. Stimulation of adipocyte lipolysis increased storage of neutral lipids by macrophages without change in number and phenotype. CONCLUSIONS/INTERPRETATION Our data suggest that adipocyte lipolysis does not activate inflammatory pathways in ATMs, which instead may act as scavengers of FAs.
Collapse
Affiliation(s)
- Sylvie Caspar-Bauguil
- Inserm, UMR1048, Institute of Metabolic and Cardiovascular Diseases, I2MC, Obesity Research Laboratory, Team 4, CHU Rangueil, 1 avenue Jean Poulhès, BP 84225, 31432, Toulouse Cedex 4, France
- University of Toulouse, UMR1048, Paul Sabatier University, Toulouse, France
- Toulouse University Hospitals, Department of Clinical Biochemistry, Toulouse, France
| | - Catherine-Ines Kolditz
- Inserm, UMR1048, Institute of Metabolic and Cardiovascular Diseases, I2MC, Obesity Research Laboratory, Team 4, CHU Rangueil, 1 avenue Jean Poulhès, BP 84225, 31432, Toulouse Cedex 4, France
- University of Toulouse, UMR1048, Paul Sabatier University, Toulouse, France
| | - Corinne Lefort
- Inserm, UMR1048, Institute of Metabolic and Cardiovascular Diseases, I2MC, Obesity Research Laboratory, Team 4, CHU Rangueil, 1 avenue Jean Poulhès, BP 84225, 31432, Toulouse Cedex 4, France
- University of Toulouse, UMR1048, Paul Sabatier University, Toulouse, France
| | - Isabelle Vila
- Inserm, UMR1048, Institute of Metabolic and Cardiovascular Diseases, I2MC, Obesity Research Laboratory, Team 4, CHU Rangueil, 1 avenue Jean Poulhès, BP 84225, 31432, Toulouse Cedex 4, France
- University of Toulouse, UMR1048, Paul Sabatier University, Toulouse, France
| | - Etienne Mouisel
- Inserm, UMR1048, Institute of Metabolic and Cardiovascular Diseases, I2MC, Obesity Research Laboratory, Team 4, CHU Rangueil, 1 avenue Jean Poulhès, BP 84225, 31432, Toulouse Cedex 4, France
- University of Toulouse, UMR1048, Paul Sabatier University, Toulouse, France
| | - Diane Beuzelin
- Inserm, UMR1048, Institute of Metabolic and Cardiovascular Diseases, I2MC, Obesity Research Laboratory, Team 4, CHU Rangueil, 1 avenue Jean Poulhès, BP 84225, 31432, Toulouse Cedex 4, France
- University of Toulouse, UMR1048, Paul Sabatier University, Toulouse, France
| | - Geneviève Tavernier
- Inserm, UMR1048, Institute of Metabolic and Cardiovascular Diseases, I2MC, Obesity Research Laboratory, Team 4, CHU Rangueil, 1 avenue Jean Poulhès, BP 84225, 31432, Toulouse Cedex 4, France
- University of Toulouse, UMR1048, Paul Sabatier University, Toulouse, France
| | - Marie-Adeline Marques
- Inserm, UMR1048, Institute of Metabolic and Cardiovascular Diseases, I2MC, Obesity Research Laboratory, Team 4, CHU Rangueil, 1 avenue Jean Poulhès, BP 84225, 31432, Toulouse Cedex 4, France
- University of Toulouse, UMR1048, Paul Sabatier University, Toulouse, France
| | - Alexia Zakaroff-Girard
- University of Toulouse, UMR1048, Paul Sabatier University, Toulouse, France
- Inserm, UMR1048, Cytometry Facility, Institute of Metabolic and Cardiovascular Diseases, Toulouse, France
- Inserm, UMR1048, Team 1, Institute of Metabolic and Cardiovascular Diseases, Toulouse, France
| | - Christiane Pecher
- University of Toulouse, UMR1048, Paul Sabatier University, Toulouse, France
- Inserm, UMR1048, Cytometry Facility, Institute of Metabolic and Cardiovascular Diseases, Toulouse, France
| | - Marianne Houssier
- Inserm, UMR1048, Institute of Metabolic and Cardiovascular Diseases, I2MC, Obesity Research Laboratory, Team 4, CHU Rangueil, 1 avenue Jean Poulhès, BP 84225, 31432, Toulouse Cedex 4, France
- University of Toulouse, UMR1048, Paul Sabatier University, Toulouse, France
| | - Lucile Mir
- Inserm, UMR1048, Institute of Metabolic and Cardiovascular Diseases, I2MC, Obesity Research Laboratory, Team 4, CHU Rangueil, 1 avenue Jean Poulhès, BP 84225, 31432, Toulouse Cedex 4, France
- University of Toulouse, UMR1048, Paul Sabatier University, Toulouse, France
| | - Sarah Nicolas
- Inserm, UMR1048, Institute of Metabolic and Cardiovascular Diseases, I2MC, Obesity Research Laboratory, Team 4, CHU Rangueil, 1 avenue Jean Poulhès, BP 84225, 31432, Toulouse Cedex 4, France
- University of Toulouse, UMR1048, Paul Sabatier University, Toulouse, France
| | - Cédric Moro
- Inserm, UMR1048, Institute of Metabolic and Cardiovascular Diseases, I2MC, Obesity Research Laboratory, Team 4, CHU Rangueil, 1 avenue Jean Poulhès, BP 84225, 31432, Toulouse Cedex 4, France
- University of Toulouse, UMR1048, Paul Sabatier University, Toulouse, France
| | - Dominique Langin
- Inserm, UMR1048, Institute of Metabolic and Cardiovascular Diseases, I2MC, Obesity Research Laboratory, Team 4, CHU Rangueil, 1 avenue Jean Poulhès, BP 84225, 31432, Toulouse Cedex 4, France.
- University of Toulouse, UMR1048, Paul Sabatier University, Toulouse, France.
- Toulouse University Hospitals, Department of Clinical Biochemistry, Toulouse, France.
| |
Collapse
|
16
|
Liu XB, Yang BX, Zhang L, Lu YZ, Gong MH, Tian JK. An in vivo and in vitro assessment of the anti-inflammatory, antinociceptive, and immunomodulatory activities of Clematis terniflora DC. extract, participation of aurantiamide acetate. JOURNAL OF ETHNOPHARMACOLOGY 2015; 169:287-294. [PMID: 25910534 DOI: 10.1016/j.jep.2015.04.009] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/05/2014] [Revised: 03/13/2015] [Accepted: 04/10/2015] [Indexed: 06/04/2023]
Abstract
AIM Clematis terniflora DC. has been widely used as a traditional Chinese medicine for the treatment of tonsillitis, rheumatoid arthritis, and prostatitis. Despite its widespread use in China, there are currently no studies systematically examined its therapeutic effects and mechanism of action. As such, the present study was conducted to evaluate the anti-inflammatory, antinociceptive, and immunomodulatory effects of C. terniflora DC. using rodent and cellular models. METHODS The anti-inflammatory properties of the 70% ethanol eluted fraction of the 70% ethanol extract of C. terniflora DC. (EECTD) were evaluated using the xylene-induced ear swelling test, the carrageenan-induced edema model, and the cotton pellet granuloma method. Its antinociceptive activities were determined using both the acetic acid-induced writhing test and the hot plate assay. In parallel, we conducted an in vitro assay in LPS-induced RAW264.7 cells to examine the anti-inflammatory effects of EECTD and its purified form, aurantiamide acetate (AA) on inhibition of nitric oxide (NO) and prostaglandin E2 (PGE2) release. RESULTS EECTD (300mg/kg) significantly reduced the number of writhing, extended the pain response latency, and suppressed xylene-induced ear swelling. Each EECTD treatment group also had significant inhibition of cotton granulation formation in addition to reduced carrageenan-induced paw edema. EECTD was also shown to alleviate signs of inflammation in histopathological paw sections. However, it had a less noticeable effect on mouse ear swelling in the delayed type hypersensitivity test. A purified compound was isolated from EECTD and its structure was identified as AA. In vitro experimental results showed that both EECTD and AA were able to significantly inhibit the release of pro-inflammatory cytokines NO and PGE2 on LPS-induced RAW264.7 cells. CONCLUSION These results suggest that EECTD has significant anti-inflammatory and antinociceptive activities, partially related to one of the active substances identified as AA. We hypothesize that these effects are related to its ability to inhibit the production of cytokines NO and PGE2. However, further work will be needed to determine its exact mechanism of action.
Collapse
Affiliation(s)
- X B Liu
- College of Biomedical Engineering and Instrument Science, Zhejiang University, Hangzhou 310027, Zhejiang, China; Clinical Pharmaceutical Research Institute, Hunan Cancer Hospital/Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha 410013, Hunan, China.
| | - B X Yang
- College of Biomedical Engineering and Instrument Science, Zhejiang University, Hangzhou 310027, Zhejiang, China
| | - L Zhang
- College of Biomedical Engineering and Instrument Science, Zhejiang University, Hangzhou 310027, Zhejiang, China
| | - Y Z Lu
- College of Biomedical Engineering and Instrument Science, Zhejiang University, Hangzhou 310027, Zhejiang, China
| | - M H Gong
- Changshu Qiushi Technology Co. Ltd., Changshu 215500, Jiangsu, China
| | - J K Tian
- College of Biomedical Engineering and Instrument Science, Zhejiang University, Hangzhou 310027, Zhejiang, China.
| |
Collapse
|
17
|
Honda KL, Lamon-Fava S, Matthan NR, Wu D, Lichtenstein AH. EPA and DHA exposure alters the inflammatory response but not the surface expression of Toll-like receptor 4 in macrophages. Lipids 2014; 50:121-9. [PMID: 25408476 DOI: 10.1007/s11745-014-3971-y] [Citation(s) in RCA: 45] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2014] [Accepted: 11/03/2014] [Indexed: 12/31/2022]
Abstract
Dietary intake of eicosapentaenoic acid (EPA) and docosahexaenoic acid (DHA) and their respective enrichment in cell membranes have been negatively associated with atherosclerotic lesion development. This effect may be mediated, in part, by dampened inflammatory response of macrophages triggered by toll-like receptor 4 (TLR4) activation. This study investigated the influence of membrane fatty acid profile on TLR4-mediated inflammation in RAW 264.7 macrophages. Cells pretreated with myristic acid (MA), EPA, DHA or vehicle control for 24 h were stimulated with ultra-pure LPS, a specific TLR4 agonist, for 6 or 24 h, corresponding to early and late stages of TNFα and IL-6 protein induction. Treatment significantly increased cell membrane MA, EPA, and DHA by 4.5-, 20.6-, and 8.9-fold, respectively. MA significantly increased IL-6 secretion 6 h post-exposure to the fatty acid, but did not change TNFα secretion in response to any other treatment condition. EPA and DHA significantly reduced TNFα secretion by 36 and 41 %, respectively, in cells stimulated for 24 h but not 6 h. In contrast, EPA and DHA significantly reduced IL-6 secretion at both 6 h (67 and 72%, respectively) and 24 h (69 and 72%, respectively). MA or DHA treatment had no significant effect compared to vehicle on factors influencing cellular LPS recognition, including LPS-cell association, and cell surface expression of TLR4, TLR4-MD2 complex, and CD14. These data suggest that membrane fatty acid profiles influence the TLR4-mediated inflammatory response in macrophages, via mechanisms that occur downstream of TLR4 receptor activation.
Collapse
Affiliation(s)
- Kaori L Honda
- Cardiovascular Nutrition Laboratory, Jean Mayer USDA Human Nutrition Research Center on Aging at Tufts University, 711 Washington Street, Boston, MA, 02111, USA,
| | | | | | | | | |
Collapse
|
18
|
Chapman JR, Rangan GK. Are nonesterified fatty acids protective in chronic allograft nephropathy? Transplantation 2013; 95:1313-4. [PMID: 23736282 DOI: 10.1097/tp.0b013e31828b8fc6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Affiliation(s)
- Jeremy R Chapman
- Centre for Transplant and Renal Research, Westmead Millennium Institute, University of Sydney at Westmead Hospital, Westmead, New South Wales, Australia.
| | | |
Collapse
|
19
|
Murumalla RK, Gunasekaran MK, Padhan JK, Bencharif K, Gence L, Festy F, Césari M, Roche R, Hoareau L. Fatty acids do not pay the toll: effect of SFA and PUFA on human adipose tissue and mature adipocytes inflammation. Lipids Health Dis 2012; 11:175. [PMID: 23259689 PMCID: PMC3551671 DOI: 10.1186/1476-511x-11-175] [Citation(s) in RCA: 61] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2012] [Accepted: 12/18/2012] [Indexed: 12/27/2022] Open
Abstract
Background On the basis that high fat diet induces inflammation in adipose tissue, we wanted to test the effect of dietary saturated and polysunsaturated fatty acids on human adipose tissue and adipocytes inflammation. Moreover we wanted to determine if TLR2 and TLR4 are involved in this pathway. Methods Human adipose tissue and adipocytes primary cultures were treated with endotoxin-free BSA conjugated with SFA (lauric acid and palmitic acid - LA and PA) and PUFA (eicosapentaeneic acid, docosahexaenoic acid and oleic acid - EPA, DHA and OA) with or without LPS. Cytokines were then assayed by ELISA (TNF-alpha, IL-6 and MCP-1). In order to determine if TLR2 and TLR4 are activated by fatty acid (FA), we used HEK-Blue cells transfected by genes from TLR2 or TLR4 pathways associated with secreted alkaline phosphatase reporter gene. Results None of the FA tested in HEK-Blue cells were able to activate TLR2 or TLR4, which is concordant with the fact that after FA treatment, adipose tissue and adipocytes cytokines levels remain the same as controls. However, all the PUFA tested: DHA, EPA and to a lesser extent OA down-regulated TNF-alpha, IL-6 and MCP-1 secretion in human adipose tissue and adipocytes cultures. Conclusions This study first confirms that FA do not activate TLR2 and TLR4. Moreover by using endotoxin-free BSA, both SFA and PUFA tested were not proinflammatory in human adipose tissue and adipocytes model. More interestingly we showed that some PUFA exert an anti-inflammatory action in human adipose tissue and adipocytes model. These results are important since they clarify the relationship between dietary fatty acids and inflammation linked to obesity.
Collapse
Affiliation(s)
- Ravi Kumar Murumalla
- GEICO-Study Group on Chronic Inflammation and Obesity, Platform 'Cyclotron Reunion Ocean Indien' CYROI, 2 Rue Maxime Rivière, Sainte-Clotilde, Reunion Island 97490, France
| | | | | | | | | | | | | | | | | |
Collapse
|