251
|
Scott MA, Woolums AR, Swiderski CE, Perkins AD, Nanduri B, Smith DR, Karisch BB, Epperson WB, Blanton JR. Whole blood transcriptomic analysis of beef cattle at arrival identifies potential predictive molecules and mechanisms that indicate animals that naturally resist bovine respiratory disease. PLoS One 2020; 15:e0227507. [PMID: 31929561 PMCID: PMC6957175 DOI: 10.1371/journal.pone.0227507] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2019] [Accepted: 12/19/2019] [Indexed: 12/12/2022] Open
Abstract
Bovine respiratory disease (BRD) is a multifactorial disease complex and the leading infectious disease in post-weaned beef cattle. Clinical manifestations of BRD are recognized in beef calves within a high-risk setting, commonly associated with weaning, shipping, and novel feeding and housing environments. However, the understanding of complex host immune interactions and genomic mechanisms involved in BRD susceptibility remain elusive. Utilizing high-throughput RNA-sequencing, we contrasted the at-arrival blood transcriptomes of 6 beef cattle that ultimately developed BRD against 5 beef cattle that remained healthy within the same herd, differentiating BRD diagnosis from production metadata and treatment records. We identified 135 differentially expressed genes (DEGs) using the differential gene expression tools edgeR and DESeq2. Thirty-six of the DEGs shared between these two analysis platforms were prioritized for investigation of their relevance to infectious disease resistance using WebGestalt, STRING, and Reactome. Biological processes related to inflammatory response, immunological defense, lipoxin metabolism, and macrophage function were identified. Production of specialized pro-resolvin mediators (SPMs) and endogenous metabolism of angiotensinogen were increased in animals that resisted BRD. Protein-protein interaction modeling of gene products with significantly higher expression in cattle that naturally acquire BRD identified molecular processes involving microbial killing. Accordingly, identification of DEGs in whole blood at arrival revealed a clear distinction between calves that went on to develop BRD and those that resisted BRD. These results provide novel insight into host immune factors that are present at the time of arrival that confer protection from BRD.
Collapse
Affiliation(s)
- Matthew A. Scott
- Department of Pathobiology and Population Medicine, Mississippi State University, Mississippi State, MS, United States of America
- * E-mail:
| | - Amelia R. Woolums
- Department of Pathobiology and Population Medicine, Mississippi State University, Mississippi State, MS, United States of America
| | - Cyprianna E. Swiderski
- Department of Clinical Sciences, Mississippi State University, Mississippi State, MS, United States of America
| | - Andy D. Perkins
- Department of Computer Science and Engineering, Mississippi State University, Mississippi State, MS, United States of America
| | - Bindu Nanduri
- Department of Basic Sciences, Mississippi State University College of Veterinary Medicine, Mississippi State University, Mississippi State, MS, United States of America
| | - David R. Smith
- Department of Pathobiology and Population Medicine, Mississippi State University, Mississippi State, MS, United States of America
| | - Brandi B. Karisch
- Department of Animal and Dairy Sciences, Mississippi State University, Mississippi State, MS, United States of America
| | - William B. Epperson
- Department of Pathobiology and Population Medicine, Mississippi State University, Mississippi State, MS, United States of America
| | - John R. Blanton
- Department of Animal and Dairy Sciences, Mississippi State University, Mississippi State, MS, United States of America
| |
Collapse
|
252
|
Edwards JM, McCarthy CG, Wenceslau CF. The Obligatory Role of the Acetylcholine-Induced Endothelium-Dependent Contraction in Hypertension: Can Arachidonic Acid Resolve this Inflammation? Curr Pharm Des 2020; 26:3723-3732. [PMID: 32303165 PMCID: PMC7542659 DOI: 10.2174/1381612826666200417150121] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2020] [Accepted: 04/10/2020] [Indexed: 02/07/2023]
Abstract
The endothelium produces many substances that can regulate vascular tone. Acetylcholine is a widely used pharmacological tool to assess endothelial function. In general, acetylcholine binds to G-protein coupled muscarinic receptors that mediate a transient elevation in intracellular, free calcium. This intracellular rise in calcium is responsible for triggering several cellular responses, including the synthesis of nitric oxide, endothelium- derived hyperpolarizing factor, and eicosanoids derived from arachidonic acid. Endothelial arachidonic acid metabolism is also an important signaling pathway for mediating inflammation. Therefore, in conditions with sustained and excessive inflammation such as hypertension, arachidonic acid serves as a substrate for the synthesis of several vasoconstrictive metabolites, predominantly via the cyclooxygenase and lipoxygenase enzymes. Cyclooxygenase and lipoxygenase products can then activate G-protein coupled receptors expressed on vascular smooth muscle cells to causes contractile responses. As a result, acetylcholine-induced contraction due to arachidonic acid is a commonly observed feature of endothelial dysfunction and vascular inflammation in hypertension. In this review, we will critically analyze the literature supporting this concept, as well as address the potential underlying mechanisms, including the possibility that arachidonic acid signaling is diverted away from the synthesis of pro-resolving metabolites in conditions such as hypertension.
Collapse
Affiliation(s)
- Jonnelle M. Edwards
- Center for Hypertension and Precision Medicine, Department of Physiology and Pharmacology, University of Toledo College of Medicine & Life Sciences, Toledo, OH, USA
| | - Cameron G. McCarthy
- Center for Hypertension and Precision Medicine, Department of Physiology and Pharmacology, University of Toledo College of Medicine & Life Sciences, Toledo, OH, USA
| | - Camilla F. Wenceslau
- Center for Hypertension and Precision Medicine, Department of Physiology and Pharmacology, University of Toledo College of Medicine & Life Sciences, Toledo, OH, USA
| |
Collapse
|
253
|
Stockert K. Synopsis. ALLERGIEPRÄVENTION 2020. [PMCID: PMC7121829 DOI: 10.1007/978-3-662-58140-7_7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Akute entzündliche Reaktionen bzw. der akute Infekt mit Restitutio ad integrum laufen in einer perfekt modulierten Kaskade ab, bei dem eine akute inflammatorische Einleitungsphase von einer antiinflammatorischen Phase und einer Entzündungsauflösungsphase abgelöst werden.
Collapse
|
254
|
Poorani R, Bhatt AN, Das UN. Modulation of pro-inflammatory and pro-resolution mediators by γ-linolenic acid: an important element in radioprotection against ionizing radiation. Arch Med Sci 2020; 16:1448-1456. [PMID: 33224346 PMCID: PMC7667419 DOI: 10.5114/aoms.2020.92469] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/15/2018] [Accepted: 01/16/2019] [Indexed: 01/25/2023] Open
Abstract
INTRODUCTION The current study explored the radio-protective property of γ-linolenic acid (GLA) in C57BL/6J mice against low linear energy transfer ionizing radiation (IR; X-rays) and its modulatory effect on the production of lipid mediators such as prostaglandin E2 (PGE2), leukotriene E4 and lipoxin A4 (LXA4) in mice plasma. METHODS The effect of GLA pre-treatment on radiation induced inflammation was assessed by estimating plasma levels of high mobility group box 1 protein (HMGB1), TMOP/NO and various anti-oxidant enzymes. RESULTS γ-linolenic acid pre-treated mice exposed to lethal IR dose (7.5 Gy) showed a decrease in plasma levels of HMGB1, PGE2 and LXA4 and a fall in TMOP/NO ratio and improvement in anti-oxidant enzymes: catalase, glutathione transferase and glutathione peroxidase compared to IR mice, suggesting that GLA suppresses IR-induced inflammation and restores the pro- vs. anti-oxidant ratio to near normal, which could explain its radioprotective action. CONCLUSIONS GLA showed radioprotective action.
Collapse
Affiliation(s)
- Rangachar Poorani
- Institute of Nuclear Medicine and Allied Sciences, DRDO, Delhi, India
- BioScience Research Centre and Department of Medicine, GVP Hospital and Medical College, Visakhapatnam, India
| | - Anant N. Bhatt
- Institute of Nuclear Medicine and Allied Sciences, DRDO, Delhi, India
| | - Undurti N. Das
- BioScience Research Centre and Department of Medicine, GVP Hospital and Medical College, Visakhapatnam, India
- UND Life Sciences, Battle Ground, WA, USA
| |
Collapse
|
255
|
Fussbroich D, Colas RA, Eickmeier O, Trischler J, Jerkic SP, Zimmermann K, Göpel A, Schwenger T, Schaible A, Henrich D, Baer P, Zielen S, Dalli J, Beermann C, Schubert R. A combination of LCPUFA ameliorates airway inflammation in asthmatic mice by promoting pro-resolving effects and reducing adverse effects of EPA. Mucosal Immunol 2020; 13:481-492. [PMID: 31907365 PMCID: PMC7181394 DOI: 10.1038/s41385-019-0245-2] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2018] [Revised: 11/24/2019] [Accepted: 12/02/2019] [Indexed: 02/04/2023]
Abstract
Lipid mediators derived from omega (n)-3 and n-6 long-chain polyunsaturated fatty acids (LCPUFA) play key roles in bronchoconstriction, airway inflammation, and resolution processes in asthma. This study compared the effects of dietary supplementation with either a combination of LCPUFAs or eicosapentaenoic acid (EPA) alone to investigate whether the combination has superior beneficial effects on the outcome of asthmatic mice. Mice were sensitized with house dust mite (HDM) extract, and subsequently supplemented with either a combination of LCPUFAs or EPA alone in a recall asthma model. After the final HDM and LCPUFA administration, airway hyperresponsiveness (AHR), bronchoalveolar lavages, and lung histochemistry were examined. Lipid mediator profiles were determined by liquid chromatography coupled with tandem mass spectrometry (LC-MS-MS). The LCPUFA combination reduced AHR, eosinophilic inflammation, and inflammatory cytokines (IL-5, IFN-γ, and IL-6) in asthmatic mice, whereas EPA enhanced inflammation. The combination of LCPUFAs was more potent in downregulating EPA-derived LTB5 and LTC5 and in supporting DHA-derived RvD1 and RvD4 (2.22-fold and 2.58-fold higher levels) than EPA alone. Ex vivo experiments showed that LTB5 contributes to granulocytes' migration and M1-polarization in monocytes. Consequently, the LCPUFA combination ameliorated airway inflammation by inhibiting adverse effects of EPA and promoting pro-resolving effects supporting the lipid mediator-dependent resolution program.
Collapse
Affiliation(s)
- D. Fussbroich
- grid.430588.2Department of Food Technology, University of Applied Sciences Fulda, Fulda, Germany ,0000 0004 1936 9721grid.7839.5Division for Allergy, Pneumology and Cystic Fibrosis, Department for Children and Adolescence, Goethe-University, Frankfurt/Main, Germany ,0000 0004 1936 9721grid.7839.5Faculty of Biological Sciences, Goethe University Frankfurt/Main, Frankfurt/Main, Germany
| | - R. A. Colas
- 0000 0001 2171 1133grid.4868.2Lipid Mediator Unit, William Harvey Research Institute, Bart’s and the London School of Medicine, Queen Mary University of London, London, UK
| | - O. Eickmeier
- 0000 0004 1936 9721grid.7839.5Division for Allergy, Pneumology and Cystic Fibrosis, Department for Children and Adolescence, Goethe-University, Frankfurt/Main, Germany
| | - J. Trischler
- 0000 0004 1936 9721grid.7839.5Division for Allergy, Pneumology and Cystic Fibrosis, Department for Children and Adolescence, Goethe-University, Frankfurt/Main, Germany
| | - S. P. Jerkic
- 0000 0004 1936 9721grid.7839.5Division for Allergy, Pneumology and Cystic Fibrosis, Department for Children and Adolescence, Goethe-University, Frankfurt/Main, Germany
| | - K. Zimmermann
- grid.430588.2Department of Food Technology, University of Applied Sciences Fulda, Fulda, Germany
| | - A. Göpel
- grid.430588.2Department of Food Technology, University of Applied Sciences Fulda, Fulda, Germany
| | - T. Schwenger
- grid.430588.2Department of Food Technology, University of Applied Sciences Fulda, Fulda, Germany
| | - A. Schaible
- 0000 0004 1936 9721grid.7839.5Department of Trauma, Hand & Reconstructive Surgery, Goethe-University, Frankfurt/Main, Germany
| | - D. Henrich
- 0000 0004 1936 9721grid.7839.5Department of Trauma, Hand & Reconstructive Surgery, Goethe-University, Frankfurt/Main, Germany
| | - P. Baer
- 0000 0004 1936 9721grid.7839.5Division of Nephrology, Department of Internal Medicine III, Goethe-University, Frankfurt/Main, Germany
| | - S. Zielen
- 0000 0004 1936 9721grid.7839.5Division for Allergy, Pneumology and Cystic Fibrosis, Department for Children and Adolescence, Goethe-University, Frankfurt/Main, Germany
| | - J. Dalli
- 0000 0001 2171 1133grid.4868.2Lipid Mediator Unit, William Harvey Research Institute, Bart’s and the London School of Medicine, Queen Mary University of London, London, UK ,0000 0001 2171 1133grid.4868.2Centre for inflammation and Therapeutic Innovation, Queen Mary University of London, London, UK
| | - C. Beermann
- grid.430588.2Department of Food Technology, University of Applied Sciences Fulda, Fulda, Germany
| | - R. Schubert
- 0000 0004 1936 9721grid.7839.5Division for Allergy, Pneumology and Cystic Fibrosis, Department for Children and Adolescence, Goethe-University, Frankfurt/Main, Germany
| |
Collapse
|
256
|
Lordan R, Redfern S, Tsoupras A, Zabetakis I. Inflammation and cardiovascular disease: are marine phospholipids the answer? Food Funct 2020; 11:2861-2885. [DOI: 10.1039/c9fo01742a] [Citation(s) in RCA: 35] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
This review presents the latest research on the cardioprotective effects of n-3 fatty acids (FA) and n-3 FA bound to polar lipids (PL). Overall, n-3 PL may have enhanced bioavailability and potentially bioactivityversusfree FA and ester forms of n-3 FA.
Collapse
Affiliation(s)
- Ronan Lordan
- Department of Biological Sciences
- University of Limerick
- Limerick
- Ireland
- Health Research Institute (HRI)
| | - Shane Redfern
- Department of Biological Sciences
- University of Limerick
- Limerick
- Ireland
| | - Alexandros Tsoupras
- Department of Biological Sciences
- University of Limerick
- Limerick
- Ireland
- Health Research Institute (HRI)
| | - Ioannis Zabetakis
- Department of Biological Sciences
- University of Limerick
- Limerick
- Ireland
- Health Research Institute (HRI)
| |
Collapse
|
257
|
Zirnheld KH, Warner DR, Warner JB, Hardesty JE, McClain CJ, Kirpich IA. Dietary fatty acids and bioactive fatty acid metabolites in alcoholic liver disease. LIVER RESEARCH 2019. [DOI: 10.1016/j.livres.2019.10.001] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
|
258
|
Windisch HS, Fink P. Transcriptome sequencing of a keystone aquatic herbivore yields insights on the temperature-dependent metabolism of essential lipids. BMC Genomics 2019; 20:894. [PMID: 31752680 PMCID: PMC6873670 DOI: 10.1186/s12864-019-6268-y] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2019] [Accepted: 11/06/2019] [Indexed: 11/10/2022] Open
Abstract
Background Nutritional quality of phytoplankton is a major determinant of the trophic transfer efficiency at the plant-herbivore interface in freshwater food webs. In particular, the phytoplankton’s content of the essential polyunsaturated omega-3 fatty acid eicosapentaenoic acid (EPA) has been repeatedly shown to limit secondary production in the major zooplankton herbivore genus Daphnia. Despite extensive research efforts on the biological model organism Daphnia, and the availability of several Daphnia genomes, little is known regarding the molecular mechanisms underlying the limitations in Daphnia related to dietary EPA availability. Results We used RNA-seq to analyse the transcriptomic response of Daphnia magna which were fed with two different diets — each with or without supplementation of EPA — at two different temperature levels (15 and 20 °C). The transcripts were mapped to the D. magna genome assembly version 2.4, containing 26,646 translations. When D. magna fed on green alga, changing the temperature provoked a differential expression of 2001 transcripts, and in cyanobacteria-fed daphnia, 3385 transcripts were affected. The supplementation of EPA affected 1635 (on the green algal diet), or 175 transcripts (on the cyanobacterial diet), respectively. Combined effects for diet and temperature were also observed (669 for the green algal and 128 transcripts for the cyanobacterial diet). Searching for orthologous genes (COG-analysis) yielded a functional overview of the altered transcriptomes. Cross-matched transcript sets from both feed types were compiled to illuminate core responses to the factors temperature and EPA-supplementation. Conclusions Our highly controlled eco-physiological experiments revealed an orchestrated response of genes involved in the transformation and signalling of essential fatty acids, including eicosanoid-signalling pathways with potential immune functions. We provide an overview of downstream-regulated genes, which contribute to enhance growth and reproductive output. We also identified numerous EPA-responsive candidate genes of yet unknown function, which constitute new targets for future studies on the molecular basis of EPA-dependent effects at the freshwater plant-herbivore interface.
Collapse
Affiliation(s)
- Heidrun S Windisch
- Heinrich-Heine-University, Institute for Cell Biology and Zoology, Universitätsstrasse 1, 40225, Düsseldorf, Germany. .,Fraunhofer IME, Institute for Molecular Ecology, Am Aberg 1, 57392, Schmallenberg, Germany.
| | - Patrick Fink
- Institute for Zoology, University of Cologne, Zülpicher Strasse 47b, 50674, Köln, Germany.,Department River Ecology, Helmholtz Centre for Environmental Science, Brückstrasse 3a, 39114, Magdeburg, Germany.,Department Aquatic Ecosystem Analysis and Management, Helmholtz Centre for Environmental Science, Brückstrasse 3a, 39114, Magdeburg, Germany
| |
Collapse
|
259
|
The Role of Maresins in Inflammatory Pain: Function of Macrophages in Wound Regeneration. Int J Mol Sci 2019; 20:ijms20235849. [PMID: 31766461 PMCID: PMC6928948 DOI: 10.3390/ijms20235849] [Citation(s) in RCA: 37] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2019] [Revised: 11/12/2019] [Accepted: 11/19/2019] [Indexed: 02/06/2023] Open
Abstract
Although acute inflammatory responses are host-protective and generally self-limited, unresolved and delayed resolution of acute inflammation can lead to further tissue damage and chronic inflammation. The mechanism of pain induction under inflammatory conditions has been studied extensively; however, the mechanism of pain resolution is not fully understood. The resolution of inflammation is a biosynthetically active process, involving specialized pro-resolving mediators (SPMs). In particular, maresins (MaRs) are synthesized from docosahexaenoic acid (DHA) by macrophages and have anti-inflammatory and pro-resolving capacities as well as tissue regenerating and pain-relieving properties. A new class of macrophage-derived molecules—MaR conjugates in tissue regeneration (MCTRs)—has been reported to regulate phagocytosis and the repair and regeneration of damaged tissue. Macrophages not only participate in the biosynthesis of SPMs, but also play an important role in phagocytosis. They exhibit different phenotypes categorized as proinflammatory M1-like phenotypes and anti-inflammatory M2 phenotypes that mediate both harmful and protective functions, respectively. However, the signaling mechanisms underlying macrophage functions and phenotypic changes have not yet been fully established. Recent studies report that MaRs help resolve inflammatory pain by enhancing macrophage phagocytosis and shifting cytokine release to the anti-inflammatory M2 phenotypes. Consequently, this review elucidated the characteristics of MaRs and macrophages, focusing on the potent action of MaRs to enhance the M2 macrophage phenotype profiles that possess the ability to alleviate inflammatory pain.
Collapse
|
260
|
Oliveira Perucci L, Pereira Santos TA, Campi Santos P, Ribeiro Teixeira LC, Nessralla Alpoim P, Braga Gomes K, Pires Sousa L, Sant'Ana Dusse LM, Talvani A. Pre-eclampsia is associated with reduced resolvin D1 and maresin 1 to leukotriene B4 ratios in the plasma. Am J Reprod Immunol 2019; 83:e13206. [PMID: 31679164 DOI: 10.1111/aji.13206] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2019] [Revised: 10/16/2019] [Accepted: 10/30/2019] [Indexed: 12/11/2022] Open
Abstract
PROBLEM Omega-3 and omega-6 fatty acids can be endogenously converted into mediators with pro-inflammatory (eg, leukotriene B4/LTB4) or anti-inflammatory/pro-resolving activities (eg, resolvin D1/RvD1 and maresin 1/MaR1). Recent data indicate an imbalance of LTB4 and MaR1 levels in pre-eclampsia (PE), but the relative production of these mediators, including RvD1, and the role of these mediators in the disease pathogenesis remain unclear. Therefore, this study aimed to investigate the plasma levels of LTB4, RvD1, and MaR1 in pregnant women with or without PE and non-pregnant controls and their association with clinical/laboratory parameters of PE women. METHOD OF STUDY LTB4, RvD1, and MaR1 plasma levels were measured by competitive enzyme immunoassay in 19 non-pregnant, 20 normotensive pregnant, and 21 PE women. RESULTS Plasma concentrations of LTB4 were higher and RvD1 were lower in PE women than in normotensive pregnant women, who presented higher levels of LTB4 and similar levels of RvD1 to non-pregnant women. MaR1 levels did not differ among the groups. Pre-eclampsia women had decreased RvD1/LTB4 and MaR1/LTB4 ratios. Considering only the PE group, positive correlations were observed among all the mediators tested, between LTB4 and white blood cell count and between RvD1 and creatinine levels. However, all lipid mediators correlated negatively with body mass index before pregnancy. LTB4 also correlated negatively with maternal age. CONCLUSION Our findings suggest that the PE state results in systemic overproduction of LTB4 in relation to RvD1 and MaR1, and that these lipid mediators may be involved with the disease pathogenesis.
Collapse
Affiliation(s)
- Luiza Oliveira Perucci
- Nucleus of Research on Biological Sciences, Federal University of Ouro Preto, Ouro Preto, Brazil
| | - Talita Adriana Pereira Santos
- Department of Clinical and Toxicological Analysis, Faculty of Pharmacy, Federal University of Minas Gerais, Belo Horizonte, Brazil
| | - Patrícia Campi Santos
- Departament of Microbiology, Institute of Biological Sciences, Federal University of Minas Gerais, Belo Horizonte, Brazil
| | - Lívia Cristina Ribeiro Teixeira
- Department of Clinical and Toxicological Analysis, Faculty of Pharmacy, Federal University of Minas Gerais, Belo Horizonte, Brazil
| | - Patrícia Nessralla Alpoim
- Department of Clinical and Toxicological Analysis, Faculty of Pharmacy, Federal University of Minas Gerais, Belo Horizonte, Brazil
| | - Karina Braga Gomes
- Department of Clinical and Toxicological Analysis, Faculty of Pharmacy, Federal University of Minas Gerais, Belo Horizonte, Brazil
| | - Lirlândia Pires Sousa
- Department of Clinical and Toxicological Analysis, Faculty of Pharmacy, Federal University of Minas Gerais, Belo Horizonte, Brazil
| | - Luci Maria Sant'Ana Dusse
- Department of Clinical and Toxicological Analysis, Faculty of Pharmacy, Federal University of Minas Gerais, Belo Horizonte, Brazil
| | - André Talvani
- Nucleus of Research on Biological Sciences, Federal University of Ouro Preto, Ouro Preto, Brazil.,Departament of Biological Sciences, Institute of Exact and Biological Sciences, Federal University of Ouro Preto, Ouro Preto, Brazil
| |
Collapse
|
261
|
Fattori V, Zaninelli TH, Rasquel-Oliveira FS, Casagrande R, Verri WA. Specialized pro-resolving lipid mediators: A new class of non-immunosuppressive and non-opioid analgesic drugs. Pharmacol Res 2019; 151:104549. [PMID: 31743775 DOI: 10.1016/j.phrs.2019.104549] [Citation(s) in RCA: 44] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/16/2019] [Revised: 11/04/2019] [Accepted: 11/14/2019] [Indexed: 12/19/2022]
Abstract
We now appreciate that the mechanism of resolution depends on an active and time-dependent biosynthetic shift from pro-inflammatory to pro-resolution mediators, the so-called specialized pro-resolving lipid mediators (SPMs). These SPMs are biosynthesized from the omega-3 fatty acids arachidonic acid (AA), eicosapentaenoic acid (EPA), docosapentaenoic acid (DPA), or docosahexaenoic acid (DHA). Despite effective for a fraction of patients with rheumatic diseases and neuropathic pain, current analgesic therapies such as biological agents, opioids, corticoids, and gabapentinoids cause unwanted side effects, such as immunosuppression, addiction, or induce analgesic tolerance. A growing body of evidence demonstrates that isolated SPMs show efficacy at very low doses and have been successively used as therapeutic drugs to treat pain and infection in experimental models showing no side effects. Moreover, SPMs work as immunoresolvents and some of them present long-lasting analgesic and anti-inflammatory effects (i.e. block pain without immunosuppressive effects). In this review, we focus on how SPMs block pain, infection and neuro-immune interactions and, therefore, emerge as a new class of non-immunosuppressive and non-opioid analgesic drugs.
Collapse
Affiliation(s)
- Victor Fattori
- Laboratory of Pain, Inflammation, Neuropathy, and Cancer, Department of Pathology, Center of Biological Sciences, Londrina State University, Londrina, Paraná, Brazil.
| | - Tiago H Zaninelli
- Laboratory of Pain, Inflammation, Neuropathy, and Cancer, Department of Pathology, Center of Biological Sciences, Londrina State University, Londrina, Paraná, Brazil
| | - Fernanda S Rasquel-Oliveira
- Laboratory of Pain, Inflammation, Neuropathy, and Cancer, Department of Pathology, Center of Biological Sciences, Londrina State University, Londrina, Paraná, Brazil
| | - Rubia Casagrande
- Laboratory of Antioxidants and Inflammation, Department of Pharmaceutical Sciences, Center of Health Sciences, Londrina State University, Londrina, Paraná, Brazil
| | - Waldiceu A Verri
- Laboratory of Pain, Inflammation, Neuropathy, and Cancer, Department of Pathology, Center of Biological Sciences, Londrina State University, Londrina, Paraná, Brazil.
| |
Collapse
|
262
|
Helsley RN, Varadharajan V, Brown AL, Gromovsky AD, Schugar RC, Ramachandiran I, Fung K, Kabbany MN, Banerjee R, Neumann CK, Finney C, Pathak P, Orabi D, Osborn LJ, Massey W, Zhang R, Kadam A, Sansbury BE, Pan C, Sacks J, Lee RG, Crooke RM, Graham MJ, Lemieux ME, Gogonea V, Kirwan JP, Allende DS, Civelek M, Fox PL, Rudel LL, Lusis AJ, Spite M, Brown JM. Obesity-linked suppression of membrane-bound O-acyltransferase 7 (MBOAT7) drives non-alcoholic fatty liver disease. eLife 2019; 8:e49882. [PMID: 31621579 PMCID: PMC6850774 DOI: 10.7554/elife.49882] [Citation(s) in RCA: 96] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2019] [Accepted: 10/11/2019] [Indexed: 12/14/2022] Open
Abstract
Recent studies have identified a genetic variant rs641738 near two genes encoding membrane bound O-acyltransferase domain-containing 7 (MBOAT7) and transmembrane channel-like 4 (TMC4) that associate with increased risk of non-alcoholic fatty liver disease (NAFLD), non-alcoholic steatohepatitis (NASH), alcohol-related cirrhosis, and liver fibrosis in those infected with viral hepatitis (Buch et al., 2015; Mancina et al., 2016; Luukkonen et al., 2016; Thabet et al., 2016; Viitasalo et al., 2016; Krawczyk et al., 2017; Thabet et al., 2017). Based on hepatic expression quantitative trait loci analysis, it has been suggested that MBOAT7 loss of function promotes liver disease progression (Buch et al., 2015; Mancina et al., 2016; Luukkonen et al., 2016; Thabet et al., 2016; Viitasalo et al., 2016; Krawczyk et al., 2017; Thabet et al., 2017), but this has never been formally tested. Here we show that Mboat7 loss, but not Tmc4, in mice is sufficient to promote the progression of NAFLD in the setting of high fat diet. Mboat7 loss of function is associated with accumulation of its substrate lysophosphatidylinositol (LPI) lipids, and direct administration of LPI promotes hepatic inflammatory and fibrotic transcriptional changes in an Mboat7-dependent manner. These studies reveal a novel role for MBOAT7-driven acylation of LPI lipids in suppressing the progression of NAFLD.
Collapse
Affiliation(s)
- Robert N Helsley
- Department of Cardiovascular and Metabolic SciencesCleveland ClinicClevelandUnited States
- Department of Internal MedicineUniversity of CincinnatiCincinnatiUnited States
| | | | - Amanda L Brown
- Department of Cardiovascular and Metabolic SciencesCleveland ClinicClevelandUnited States
| | - Anthony D Gromovsky
- Department of Cardiovascular and Metabolic SciencesCleveland ClinicClevelandUnited States
| | - Rebecca C Schugar
- Department of Cardiovascular and Metabolic SciencesCleveland ClinicClevelandUnited States
| | - Iyappan Ramachandiran
- Department of Cardiovascular and Metabolic SciencesCleveland ClinicClevelandUnited States
| | - Kevin Fung
- Department of Cardiovascular and Metabolic SciencesCleveland ClinicClevelandUnited States
| | | | - Rakhee Banerjee
- Department of Cardiovascular and Metabolic SciencesCleveland ClinicClevelandUnited States
| | - Chase K Neumann
- Department of Cardiovascular and Metabolic SciencesCleveland ClinicClevelandUnited States
| | - Chelsea Finney
- Department of Cardiovascular and Metabolic SciencesCleveland ClinicClevelandUnited States
| | - Preeti Pathak
- Department of Cardiovascular and Metabolic SciencesCleveland ClinicClevelandUnited States
| | - Danny Orabi
- Department of Cardiovascular and Metabolic SciencesCleveland ClinicClevelandUnited States
| | - Lucas J Osborn
- Department of Cardiovascular and Metabolic SciencesCleveland ClinicClevelandUnited States
| | - William Massey
- Department of Cardiovascular and Metabolic SciencesCleveland ClinicClevelandUnited States
| | - Renliang Zhang
- Department of Cardiovascular and Metabolic SciencesCleveland ClinicClevelandUnited States
| | - Anagha Kadam
- Department of Cardiovascular and Metabolic SciencesCleveland ClinicClevelandUnited States
| | - Brian E Sansbury
- Center for Experimental Therapeutics & Reperfusion Injury, Department of Anesthesiology, Perioperative and Pain MedicineBrigham and Women’s Hospital, Harvard Medical SchoolBostonUnited States
| | - Calvin Pan
- Department of MedicineUniversity of California, Los AngelesLos AngelesUnited States
- Department of MicrobiologyUniversity of California, Los AngelesLos AngelesUnited States
- Department of Human GeneticsUniversity of California, Los AngelesLos AngelesUnited States
| | - Jessica Sacks
- Department of PathobiologyCleveland ClinicClevelandUnited States
| | - Richard G Lee
- Cardiovascular Group, Antisense Drug DiscoveryIonis Pharmaceuticals, IncCarlsbadUnited States
| | - Rosanne M Crooke
- Cardiovascular Group, Antisense Drug DiscoveryIonis Pharmaceuticals, IncCarlsbadUnited States
| | - Mark J Graham
- Cardiovascular Group, Antisense Drug DiscoveryIonis Pharmaceuticals, IncCarlsbadUnited States
| | | | - Valentin Gogonea
- Department of ChemistryCleveland State UniversityClevelandUnited States
| | - John P Kirwan
- Department of PathobiologyCleveland ClinicClevelandUnited States
| | - Daniela S Allende
- Department of Anatomical PathologyCleveland ClinicClevelandUnited States
| | - Mete Civelek
- Department of Biomedical EngineeringUniversity of VirginiaCharlottesvilleUnited States
| | - Paul L Fox
- Department of Cardiovascular and Metabolic SciencesCleveland ClinicClevelandUnited States
| | - Lawrence L Rudel
- Department of Pathology, Section on Lipid SciencesWake Forest University School of MedicineWinston-SalemUnited States
| | - Aldons J Lusis
- Department of MedicineUniversity of California, Los AngelesLos AngelesUnited States
- Department of MicrobiologyUniversity of California, Los AngelesLos AngelesUnited States
- Department of Human GeneticsUniversity of California, Los AngelesLos AngelesUnited States
| | - Matthew Spite
- Center for Experimental Therapeutics & Reperfusion Injury, Department of Anesthesiology, Perioperative and Pain MedicineBrigham and Women’s Hospital, Harvard Medical SchoolBostonUnited States
| | - J Mark Brown
- Department of Cardiovascular and Metabolic SciencesCleveland ClinicClevelandUnited States
| |
Collapse
|
263
|
Docosahexaenoic acid,22:6n-3: Its roles in the structure and function of the brain. Int J Dev Neurosci 2019; 79:21-31. [PMID: 31629800 DOI: 10.1016/j.ijdevneu.2019.10.004] [Citation(s) in RCA: 58] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2019] [Revised: 10/10/2019] [Accepted: 10/11/2019] [Indexed: 12/12/2022] Open
Abstract
Docosahexaenoic acid,22:6n-3 (DHA) and its metabolites are vital for the structure and functional brain development of the fetus and infants, and also for maintenance of healthy brain function of adults. DHA is thought to be an essential nutrient required throughout the life cycle for the maintenance of overall brain health. The mode of actions of DHA and its derivatives at both cellular and molecular levels in the brain are emerging. DHA is the major prevalent fatty acid in the brain membrane. The brain maintains its fatty acid levels mainly via the uptake of plasma free fatty acids. Therefore, circulating plasma DHA is significantly related to cognitive abilities during ageing and is inversely associated with cognitive decline. The signaling pathways of DHA and its metabolites are involved in neurogenesis, antinociceptive effects, anti-apoptotic effect, synaptic plasticity, Ca2+ homeostasis in brain diseases, and the functioning of nigrostriatal activities. Mechanisms of action of DHA metabolites on various processes in the brain are not yet well known. Epidemiological studies support a link between low habitual intake of DHA and a higher risk of brain disorders. A diet characterized by higher intakes of foods containing high in n-3 fatty acids, and/or lower intake of n-6 fatty acids was strongly associated with a lower Alzheimer's Disease and other brain disorders. Supplementation of DHA improves some behaviors associated with attention deficit hyperactivity disorder, bipolar disorder, schizophrenia, and impulsive behavior, as well as cognition. Nevertheless, the outcomes of trials with DHA supplementation have been controversial. Many intervention studies with DHA have shown an apparent benefit in brain function. However, clinical trials are needed for definitive conclusions. Dietary deficiency of n-3 fatty acids during fetal development in utero and the postnatal state has detrimental effects on cognitive abilities. Further research in humans is required to assess a variety of clinical outcomes, including quality of life and mental status, by supplementation of DHA.
Collapse
|
264
|
Gutiérrez S, Svahn SL, Johansson ME. Effects of Omega-3 Fatty Acids on Immune Cells. Int J Mol Sci 2019; 20:ijms20205028. [PMID: 31614433 PMCID: PMC6834330 DOI: 10.3390/ijms20205028] [Citation(s) in RCA: 322] [Impact Index Per Article: 53.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2019] [Revised: 09/27/2019] [Accepted: 10/02/2019] [Indexed: 12/14/2022] Open
Abstract
Alterations on the immune system caused by omega-3 fatty acids have been described for 30 years. This family of polyunsaturated fatty acids exerts major alterations on the activation of cells from both the innate and the adaptive immune system, although the mechanisms for such regulation are diverse. First, as a constitutive part of the cellular membrane, omega-3 fatty acids can regulate cellular membrane properties, such as membrane fluidity or complex assembly in lipid rafts. In recent years, however, a new role for omega-3 fatty acids and their derivatives as signaling molecules has emerged. In this review, we describe the latest findings describing the effects of omega-3 fatty acids on different cells from the immune system and their possible molecular mechanisms.
Collapse
Affiliation(s)
- Saray Gutiérrez
- Department of Physiology, Institute of Neuroscience and Physiology, The Sahlgrenska Academy, University of Gothenburg, 405 30 Gothenburg, Sweden.
| | - Sara L Svahn
- Department of Physiology, Institute of Neuroscience and Physiology, The Sahlgrenska Academy, University of Gothenburg, 405 30 Gothenburg, Sweden.
| | - Maria E Johansson
- Department of Physiology, Institute of Neuroscience and Physiology, The Sahlgrenska Academy, University of Gothenburg, 405 30 Gothenburg, Sweden.
| |
Collapse
|
265
|
Tiwary S, Berzofsky JA, Terabe M. Altered Lipid Tumor Environment and Its Potential Effects on NKT Cell Function in Tumor Immunity. Front Immunol 2019; 10:2187. [PMID: 31620124 PMCID: PMC6759687 DOI: 10.3389/fimmu.2019.02187] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2019] [Accepted: 08/30/2019] [Indexed: 01/26/2023] Open
Abstract
Natural killer T (NKT) cells are CD1d restricted T cells that mostly recognize lipid antigens. These cells share characteristics with both adaptive and innate immune cells and have multiple immunoregulatory roles. In a manner similar to innate immune cells, they respond quickly to stimuli and secrete large amounts of cytokines, amplifying and modulating the immune response. As T cells, they express T cell receptors (TCRs) and respond in an antigen-specific manner like conventional T cells. There are at least two subtypes of NKT cells, type I and type II, that differ in the nature of their TCR, either semi-invariant (type I) or diverse (type II). The two sub-types generally have opposing functions in tumor immunity, with type I promoting and type II suppressing tumor immunity, and they cross-regulate each other, forming an immunoregulatory axis. The tumor has multiple mechanisms by which it can evade immune-surveillance. One such mechanism involves alteration in tumor lipid repertoire and accumulation of lipids and fatty acids that favor tumor growth and evade anti-tumor immunity. Since NKT cells mostly recognize lipid antigens, an altered tumor lipid metabolic profile will also alter the repertoire of lipid antigens that can potentially affect their immune-modulatory function. In this review, we will explore the effects of alterations in the lipid metabolites on tumor growth, antigen cross-presentation, and overall effect on anti-tumor immunity, especially in the context of NKT cells.
Collapse
Affiliation(s)
- Shweta Tiwary
- Vaccine Branch, Center for Cancer Research, National Cancer Institute, NIH, Bethesda, MD, United States
| | - Jay A. Berzofsky
- Vaccine Branch, Center for Cancer Research, National Cancer Institute, NIH, Bethesda, MD, United States
| | - Masaki Terabe
- Neuro-Oncology Branch, Center for Cancer Research, National Cancer Institute, NIH, Bethesda, MD, United States
| |
Collapse
|
266
|
Quin C, Gibson DL. Dietary Fatty Acids and Host-Microbial Crosstalk in Neonatal Enteric Infection. Nutrients 2019; 11:E2064. [PMID: 31484327 PMCID: PMC6770655 DOI: 10.3390/nu11092064] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2019] [Revised: 08/09/2019] [Accepted: 08/20/2019] [Indexed: 12/11/2022] Open
Abstract
Human milk is the best nutritional choice for infants. However, in instances where breastfeeding is not possible, infant formulas are used as alternatives. While formula manufacturers attempt to mimic the performance of human breast milk, formula-fed babies consistently have higher incidences of infection from diarrheal diseases than those breastfed. Differences in disease susceptibility, progression and severity can be attributed, in part, to nutritional fatty acid differences between breast milk and formula. Despite advances in our understanding of breast milk properties, formulas still present major differences in their fatty acid composition when compared to human breast milk. In this review, we highlight the role of distinct types of dietary fatty acids in modulating host inflammation, both directly and through the microbiome-immune nexus. We present evidence that dietary fatty acids influence enteric disease susceptibility and therefore, altering the fatty acid composition in formula may be a potential strategy to improve infectious outcomes in formula-fed infants.
Collapse
Affiliation(s)
- Candice Quin
- Department of Biology, Okanagan Campus, University of British Columbia, Okanagan Campus ASC 386, 3187 University Way, Kelowna, BC V1V 1V7, Canada
| | - Deanna L Gibson
- Department of Biology, Okanagan Campus, University of British Columbia, Okanagan Campus ASC 386, 3187 University Way, Kelowna, BC V1V 1V7, Canada.
- Department of Medicine, Faculty of Medicine, University of British Columbia, Kelowna, BC V1V 1V7, Canada.
| |
Collapse
|
267
|
Yaribeygi H, Atkin SL, Simental-Mendía LE, Barreto GE, Sahebkar A. Anti-inflammatory effects of resolvins in diabetic nephropathy: Mechanistic pathways. J Cell Physiol 2019; 234:14873-14882. [PMID: 30746696 DOI: 10.1002/jcp.28315] [Citation(s) in RCA: 30] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2018] [Revised: 01/19/2019] [Accepted: 01/24/2019] [Indexed: 01/24/2023]
Abstract
The incidence of diabetes mellitus is growing rapidly. The exact pathophysiology of diabetes is unclear, but there is increasing evidence of the role of the inflammatory response in both developing diabetes as well as its complications. Resolvins are naturally occurring polyunsaturated fatty acids that are found in fish oil and sea food that have been shown to possess anti-inflammatory actions in several tissues including the kidneys. The pathways by which resolvins exert this anti-inflammatory effect are unclear. In this review we discuss the evidence showing that resolvins can suppress inflammatory responses via at least five molecular mechanisms through inhibition of the nucleotide-binding oligomerization domain protein 3 inflammasome, inhibition of nuclear factor κB molecular pathways, improvement of oxidative stress, modulation of nitric oxide synthesis/release and prevention of local and systemic leukocytosis. Complete understanding of these molecular pathways is important as this may lead to the development of new effective therapeutic strategies for diabetes and diabetic nephropathy.
Collapse
Affiliation(s)
- Habib Yaribeygi
- Chronic Kidney Disease Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | | | | | - George E Barreto
- Departamento de Nutrición y Bioquímica, Facultad de Ciencias, Pontificia Universidad Javeriana, Bogotá D.C., Colombia.,Instituto de Ciencias Biomédicas, Universidad Autónoma de Chile, Santiago, Chile
| | - Amirhossein Sahebkar
- Neurogenic Inflammation Research Center, Mashhad University of Medical Sciences, Mashhad, Iran.,Biotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran.,School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran
| |
Collapse
|
268
|
Barden A, Shinde S, Tsai IJ, Croft KD, Beilin LJ, Puddey IB, Mori TA. Effect of weight loss on neutrophil resolvins in the metabolic syndrome. Prostaglandins Leukot Essent Fatty Acids 2019; 148:25-29. [PMID: 31492430 DOI: 10.1016/j.plefa.2019.07.001] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/14/2019] [Revised: 06/26/2019] [Accepted: 07/02/2019] [Indexed: 12/13/2022]
Abstract
BACKGROUND Non-resolving inflammation associates with obesity and insulin resistance, and may be dependent on the balance of inflammatory substances and specialised pro-resolving mediators of inflammation (SPM) that act to halt the inflammatory response. This controlled trial examined the effect of weight loss on neutrophil synthesis of SPM in volunteers with the metabolic syndrome (MetS). METHODS Volunteers with MetS (n = 42) were matched for age and gender and randomly assigned to a 12-wk weight loss program followed by 4-wk weight stabilization or a 16-wk weight maintenance program. At baseline and 16 weeks, isolated neutrophils were stimulated with calcium ionophore and the released SPM were measured by LC-MS/MS. RESULTS At baseline the SPM resolvin (Rv) E1, 18R-RvE3, RvD2 and Maresin-1 (MaR-1) were detected from stimulated neutrophils. The concentration of released RvE1 was at least 6-fold that of other detected SPM. Weight loss of 4.7 ± 0.8 kg, led to a 2-fold increase in RvE1, P = 0.013, relative to the weight maintenance group. The increase in RvE1 after weight loss was related to, but independent of leukotriene B4. CONCLUSION: Following weight loss, human neutrophils from individuals with the metabolic syndrome are capable of releasing larger amounts of RvE1 upon stimulation.
Collapse
Affiliation(s)
- A Barden
- Medical School, University of Western Australia, Perth, Australia.
| | - S Shinde
- Medical School, University of Western Australia, Perth, Australia
| | - I-J Tsai
- Medical School, University of Western Australia, Perth, Australia
| | - K D Croft
- School of Biomedical Sciences, University of Western Australia, Perth, Australia
| | - L J Beilin
- Medical School, University of Western Australia, Perth, Australia
| | - I B Puddey
- Medical School, University of Western Australia, Perth, Australia
| | - T A Mori
- Medical School, University of Western Australia, Perth, Australia
| |
Collapse
|
269
|
Tobón-Arroyave SI, Isaza-Guzmán DM, Gómez-Ortega J, Flórez-Alzate AA. Salivary levels of specialized pro-resolving lipid mediators as indicators of periodontal health/disease status. J Clin Periodontol 2019; 46:978-990. [PMID: 31339183 DOI: 10.1111/jcpe.13173] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2019] [Revised: 07/14/2019] [Accepted: 07/18/2019] [Indexed: 12/31/2022]
Abstract
AIM This cross-sectional case-control study aimed to determine if salivary levels of lipoxin A4 (LXA4), protectin D1 (PD1), resolvin E1 (RvE1) and maresin 1 (MaR1) might constitute a reflection of periodontal health/disease status. MATERIALS AND METHODS One hundred and two periodontitis patients and 61 healthy controls were recruited. Periodontal clinical status was determined by criteria based on full-mouth clinico-radiographical data. Salivary concentration of the analytes was calculated by enzyme-linked immunosorbent assay. The association between the biomarkers with disease status was assessed individually and adjusted for confounding using multivariate binary logistic regression models. RESULTS Significantly decreased LXA4 and increased PD1/MaR1 salivary levels were detected in periodontitis patients in comparison with healthy controls. However, no significant differences were observed for RvE1 levels between clinical groups. Clinical parameters such as probing depth, clinical attachment loss and extent were negatively correlated with LXA4, positively correlated with PD1/MaR1 and not correlated with RvE1 salivary levels. Logistic regression analyses revealed a strong/independent association of LXA4, PD1 and MaR1 salivary levels regarding disease status. Interaction effects between demographic predictor variables and salivary concentration of LXA4, PD1 and MaR1 were also identified. CONCLUSION The results of this study demonstrated a strong/independent association between reduced LXA4 and increased PD1/MaR1 salivary levels with periodontitis suggesting an imbalance in the specialized pro-resolving lipid mediators (SPMs) in periodontal disease.
Collapse
Affiliation(s)
- Sergio Iván Tobón-Arroyave
- Laboratory of Immunodetection and Bioanalysis, Faculty of Dentistry, University of Antioquia, Medellín, Colombia
| | - Diana María Isaza-Guzmán
- Laboratory of Immunodetection and Bioanalysis, Faculty of Dentistry, University of Antioquia, Medellín, Colombia
| | - Juliana Gómez-Ortega
- Laboratory of Immunodetection and Bioanalysis, Faculty of Dentistry, University of Antioquia, Medellín, Colombia
| | | |
Collapse
|
270
|
Kohira T, Kita Y, Tokuoka SM, Shiba M, Satake M, Shimizu T. Characterization of supported liquid extraction as a sample pretreatment method for eicosanoids and related metabolites in biological fluids. J Chromatogr B Analyt Technol Biomed Life Sci 2019; 1124:298-307. [PMID: 31260873 DOI: 10.1016/j.jchromb.2019.06.016] [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: 03/22/2019] [Revised: 05/25/2019] [Accepted: 06/10/2019] [Indexed: 02/01/2023]
Abstract
Sample pretreatment is an important process in liquid chromatography-mass spectrometry-based quantitative lipidomics. Reversed-phase solid phase extraction (RP-SPE) has been widely used for analyzing various types of samples, including aqueous samples such as cell culture media, plasma, serum, urine, and other biological fluids. Because lipid mediators are often protein-bound, prior deproteinization is necessary for their effective recovery. Deproteinization is typically performed by the addition of organic solvents, which requires time-consuming evaporation-reconstitution, or dilution with aqueous solvents before RP-SPE; however, both of these approaches compromise the analytical performance. As a potential alternative, we attempted to utilize supported liquid extraction (SLE), an automation-compatible variant of liquid-liquid extraction, for the determination of eicosanoids and related metabolites in aqueous samples. We screened 81 different sample diluent-eluent conditions and found that the use of 0.1% formic acid-water as the diluent and 0.1% formic acid-methyl acetate as the eluent enabled the optimum recovery of a variety of eicosanoids, except for peptide leukotrienes. The optimized SLE method efficiently removed protein from human plasma, while phospholipids and neutral lipids were modestly recovered. Moreover, the proposed method exhibited a quantitative performance comparable to that of typical ordinary RP-SPE method in the analysis of human platelets stimulated with thrombin receptor-activating peptide 6. Thus, we propose SLE as an attractive option for rapid lipid mediator extraction from aqueous samples.
Collapse
Affiliation(s)
- Takahiro Kohira
- Department of Lipidomics, Graduate School of Medicine, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-0033, Japan; Central Blood Institute, Japanese Red Cross Society, 2-1-67 Tatsumi, Koto-ku, Tokyo 135-8521, Japan
| | - Yoshihiro Kita
- Department of Lipidomics, Graduate School of Medicine, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-0033, Japan; Life Sciences Core Facility, Graduate School of Medicine, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-0033, Japan.
| | - Suzumi M Tokuoka
- Department of Lipidomics, Graduate School of Medicine, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-0033, Japan
| | - Masayuki Shiba
- Central Blood Institute, Japanese Red Cross Society, 2-1-67 Tatsumi, Koto-ku, Tokyo 135-8521, Japan
| | - Masahiro Satake
- Central Blood Institute, Japanese Red Cross Society, 2-1-67 Tatsumi, Koto-ku, Tokyo 135-8521, Japan
| | - Takao Shimizu
- Department of Lipidomics, Graduate School of Medicine, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-0033, Japan; Department of Lipid Signaling, Research Institute, National Center for Global Health and Medicine, 1-21-1 Toyama, Shinjuku-ku, Tokyo 162-8655, Japan
| |
Collapse
|
271
|
Ceciliani F, Lecchi C. The Immune Functions of α 1 Acid Glycoprotein. Curr Protein Pept Sci 2019; 20:505-524. [PMID: 30950347 DOI: 10.2174/1389203720666190405101138] [Citation(s) in RCA: 50] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2018] [Revised: 11/20/2018] [Accepted: 11/27/2018] [Indexed: 12/20/2022]
Abstract
α1-acid glycoprotein (orosomucoid, AGP) is an Acute Phase Protein produced by liver and peripheral tissues in response to systemic reaction to inflammation. AGP functions have been studied mostly in human, cattle and fish, although the protein has been also found in many mammalian species and birds. AGP fulfils at least two set of functions, which are apparently different from each other but in fact intimately linked. On one hand, AGP is an immunomodulatory protein. On the other hand, AGP is one of the most important binding proteins in plasma and, beside modulating pharmacokinetics and pharmacodynamics of many drugs, it is also able to bind and transport several endogen ligands related to inflammation. The focus of this review is the immunomodulatory activity of AGP. This protein regulates every single event related to inflammation, including binding of pathogens and modulating white blood cells activity throughout the entire leukocyte attacking sequence. The regulation of AGP activity is complex: the inflammation induces not only an increase in AGP serum concentration, but also a qualitative change in its carbohydrate moiety, generating a multitude of glycoforms, each of them with different, and sometimes opposite and contradictory, activities. We also present the most recent findings about the relationship between AGP and adipose tissue: AGP interacts with leptin receptor and, given its immunomodulatory function, it may be included among the potential players in the field of immunometabolism.
Collapse
Affiliation(s)
- Fabrizio Ceciliani
- Department of Veterinary Medicine, Universita degli Studi di Milano, Milano, Italy
| | - Cristina Lecchi
- Department of Veterinary Medicine, Universita degli Studi di Milano, Milano, Italy
| |
Collapse
|
272
|
Crouch MJ, Kosaraju R, Guesdon W, Armstrong M, Reisdorph N, Jain R, Fenton J, Shaikh SR. Frontline Science: A reduction in DHA-derived mediators in male obesity contributes toward defects in select B cell subsets and circulating antibody. J Leukoc Biol 2019; 106:241-257. [PMID: 30576001 PMCID: PMC10020993 DOI: 10.1002/jlb.3hi1017-405rr] [Citation(s) in RCA: 41] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2018] [Revised: 11/20/2018] [Accepted: 12/11/2018] [Indexed: 01/25/2023] Open
Abstract
Obesity dysregulates B cell populations, which contributes toward poor immunological outcomes. We previously reported that differing B cell subsets are lowered in the bone marrow of obese male mice. Here, we focused on how lipid metabolites synthesized from docosahexaenoic acid (DHA) known as specialized pro-resolving lipid mediators (SPMs) influence specific B cell populations in obese male mice. Metabololipidomics revealed that splenic SPM precursors 14-hydroxydocosahexaenoic acid (14-HDHA), 17-hydroxydocosahexaenoic acid (17-HDHA), and downstream protectin DX (PDX) were decreased in obese male C57BL/6J mice. Simultaneous administration of these mediators to obese mice rescued major decrements in bone marrow B cells, modest impairments in the spleen, and circulating IgG2c, which is pro-inflammatory in obesity. In vitro studies with B cells, flow cytometry experiments with ALOX5-/- mice, and lipidomic analyses revealed the lowering of 14-HDHA/17-HDHA/PDX and dysregulation of B cell populations in obesity was driven indirectly via B cell extrinsic mechanisms. Notably, the lowering of lipid mediators was associated with an increase in the abundance of n-6 polyunsaturated fatty acids, which have a high affinity for SPM-generating enzymes. Subsequent experiments revealed female obese mice generally maintained the levels of SPM precursors, B cell subsets, and antibody levels. Finally, obese human females had increased circulating plasma cells accompanied by ex vivo B cell TNFα and IL-10 secretion. Collectively, the data demonstrate that DHA-derived mediators of the SPM pathway control the number of B cell subsets and pro-inflammatory antibody levels in obese male but not female mice through a defect that is extrinsic to B cells.
Collapse
Affiliation(s)
- Miranda J Crouch
- Department of Biochemistry & Molecular Biology, Brody School of Medicine, East Carolina University, Greenville, North Carolina, USA.,East Carolina Diabetes & Obesity Institute, East Carolina University, Greenville, North Carolina, USA.,Department of Nutrition, Gillings School of Global Public Health and School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
| | - Rasagna Kosaraju
- Department of Biochemistry & Molecular Biology, Brody School of Medicine, East Carolina University, Greenville, North Carolina, USA.,East Carolina Diabetes & Obesity Institute, East Carolina University, Greenville, North Carolina, USA
| | - William Guesdon
- Department of Biochemistry & Molecular Biology, Brody School of Medicine, East Carolina University, Greenville, North Carolina, USA.,East Carolina Diabetes & Obesity Institute, East Carolina University, Greenville, North Carolina, USA
| | - Michael Armstrong
- Department of Pharmaceutical Sciences, University of Colorado, Denver, Colorado, USA
| | - Nichole Reisdorph
- Department of Pharmaceutical Sciences, University of Colorado, Denver, Colorado, USA
| | - Raghav Jain
- The College of Osteopathic Medicine, Michigan State University, East Lansing, Michigan, USA
| | - Jenifer Fenton
- The College of Osteopathic Medicine, Michigan State University, East Lansing, Michigan, USA
| | - Saame Raza Shaikh
- Department of Nutrition, Gillings School of Global Public Health and School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
| |
Collapse
|
273
|
Lipid mediators in platelet concentrate and extracellular vesicles: Molecular mechanisms from membrane glycerophospholipids to bioactive molecules. Biochim Biophys Acta Mol Cell Biol Lipids 2019; 1864:1168-1182. [DOI: 10.1016/j.bbalip.2019.03.011] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2018] [Revised: 02/15/2019] [Accepted: 03/30/2019] [Indexed: 12/11/2022]
|
274
|
Di Costanzo F, Di Dato V, Ianora A, Romano G. Prostaglandins in Marine Organisms: A Review. Mar Drugs 2019; 17:E428. [PMID: 31340503 PMCID: PMC6669704 DOI: 10.3390/md17070428] [Citation(s) in RCA: 48] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2019] [Revised: 07/15/2019] [Accepted: 07/19/2019] [Indexed: 12/18/2022] Open
Abstract
Prostaglandins (PGs) are lipid mediators belonging to the eicosanoid family. PGs were first discovered in mammals where they are key players in a great variety of physiological and pathological processes, for instance muscle and blood vessel tone regulation, inflammation, signaling, hemostasis, reproduction, and sleep-wake regulation. These molecules have successively been discovered in lower organisms, including marine invertebrates in which they play similar roles to those in mammals, being involved in the control of oogenesis and spermatogenesis, ion transport, and defense. Prostaglandins have also been found in some marine macroalgae of the genera Gracilaria and Laminaria and very recently the PGs pathway has been identified for the first time in some species of marine microalgae. In this review we report on the occurrence of prostaglandins in the marine environment and discuss the anti-inflammatory role of these molecules.
Collapse
Affiliation(s)
- Federica Di Costanzo
- Marine Biotechnology Department, Stazione Zoologica Anton Dohrn Napoli, Villa Comunale, 80121 Napoli, Italy
| | - Valeria Di Dato
- Marine Biotechnology Department, Stazione Zoologica Anton Dohrn Napoli, Villa Comunale, 80121 Napoli, Italy.
| | - Adrianna Ianora
- Marine Biotechnology Department, Stazione Zoologica Anton Dohrn Napoli, Villa Comunale, 80121 Napoli, Italy
| | - Giovanna Romano
- Marine Biotechnology Department, Stazione Zoologica Anton Dohrn Napoli, Villa Comunale, 80121 Napoli, Italy
| |
Collapse
|
275
|
Lemoine S CM, Brigham EP, Woo H, Hanson CK, McCormack MC, Koch A, Putcha N, Hansel NN. Omega-3 fatty acid intake and prevalent respiratory symptoms among U.S. adults with COPD. BMC Pulm Med 2019; 19:97. [PMID: 31122230 PMCID: PMC6533751 DOI: 10.1186/s12890-019-0852-4] [Citation(s) in RCA: 30] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2019] [Accepted: 04/25/2019] [Indexed: 12/21/2022] Open
Abstract
Background Omega-3 fatty acids, including alpha-linolenic acid (ALA), eicosapentaenoic acid (EPA), docosahexaenoic acid (DHA) and derivatives, play a key role in the resolution of inflammation. Higher intake has been linked to decreased morbidity in several diseases, though effects on respiratory diseases like COPD are understudied. Methods The National Health and Nutrition Examination Survey (NHANES), with a focus on dietary assessment, provides a unique opportunity to explore relationships between omega-3 intake and morbidity in respiratory diseases marked by inflammation in the United States (US) population. We investigated relationships between ALA or EPA + DHA intake and respiratory symptoms among US adults with COPD, as well as variation in relationships based on personal characteristics or exposures. Results Of 878 participants, mean age was 60.6 years, 48% were current smokers, and 68% completed high school. Omega-3 intake was, 1.71 ± 0.89 g (ALA), and 0.11 ± 0.21 g (EPA + DHA). Logistic regression models, adjusting for age, gender, race, body mass index, FEV1, education, smoking status, pack-years, total caloric intake, and omega-6 (linoleic acid, LA) intake demonstrated no primary associations between omega-3 intake and respiratory symptoms. Interaction terms were used to determine potential modification of relationships by personal characteristics (race, gender, education) or exposures (LA intake, smoking status), demonstrating that at lower levels of LA intake, increasing ALA intake was associated with reduced odds of chronic cough (pint = 0.015) and wheeze (pint = 0.037). EPA + DHA, but not ALA, was associated with reduced symptoms only among current smokers who did not complete high school. Conclusions Individual factors should be taken into consideration when studying the association of fatty acid intake on respiratory diseases, as differential responses may reveal susceptible subgroups. Electronic supplementary material The online version of this article (10.1186/s12890-019-0852-4) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
| | - Emily P Brigham
- Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Han Woo
- Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | | | - Meredith C McCormack
- Johns Hopkins University School of Medicine, Baltimore, MD, USA.,Johns Hopkins University Bloomberg School of Public Health, Baltimore, MD, USA
| | - Abigail Koch
- Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Nirupama Putcha
- Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Nadia N Hansel
- Johns Hopkins University School of Medicine, Baltimore, MD, USA. .,Johns Hopkins University Bloomberg School of Public Health, Baltimore, MD, USA.
| |
Collapse
|
276
|
Zielen S, Fussbroich D. Impact of secondhand smoke on cystic fibrosis: is there a link to fatty acid metabolism? Thorax 2019; 74:529-530. [PMID: 31048510 DOI: 10.1136/thoraxjnl-2019-213042] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/01/2019] [Indexed: 11/04/2022]
Affiliation(s)
- Stefan Zielen
- University Hospital Frankfurt, Division of Paediatric Pulmonology, Allergy, and Cystic Fibrosis, Goethe University Frankfurt, Frankfurt am Main, Germany
| | - Daniela Fussbroich
- University Hospital Frankfurt, Division of Paediatric Pulmonology, Allergy, and Cystic Fibrosis, Goethe University Frankfurt, Frankfurt am Main, Germany.,Department of Food Technology, Fulda University of Applied Sciences, Fulda, Germany
| |
Collapse
|
277
|
Freitas RDS, Campos MM. Protective Effects of Omega-3 Fatty Acids in Cancer-Related Complications. Nutrients 2019; 11:nu11050945. [PMID: 31035457 PMCID: PMC6566772 DOI: 10.3390/nu11050945] [Citation(s) in RCA: 134] [Impact Index Per Article: 22.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2019] [Revised: 04/22/2019] [Accepted: 04/23/2019] [Indexed: 12/24/2022] Open
Abstract
Omega-3 polyunsaturated fatty acids (PUFAs) are considered immunonutrients and are commonly used in the nutritional therapy of cancer patients due to their ample biological effects. Omega-3 PUFAs play essential roles in cell signaling and in the cell structure and fluidity of membranes. They participate in the resolution of inflammation and have anti-inflammatory and antinociceptive effects. Additionally, they can act as agonists of G protein-coupled receptors, namely, GPR40/FFA1 and GPR120/FFA4. Cancer patients undergo complications, such as anorexia-cachexia syndrome, pain, depression, and paraneoplastic syndromes. Interestingly, the 2017 European Society for Clinical Nutrition and Metabolism (ESPEN) guidelines for cancer patients only discuss the use of omega-3 PUFAs for cancer-cachexia treatment, leaving aside other cancer-related complications that could potentially be managed by omega-3 PUFA supplementation. This critical review aimed to discuss the effects and the possible underlying mechanisms of omega-3 PUFA supplementation in cancer-related complications. Data compilation in this critical review indicates that further investigation is still required to assess the factual benefits of omega-3 PUFA supplementation in cancer-associated illnesses. Nevertheless, preclinical evidence reveals that omega-3 PUFAs and their metabolites might modulate pivotal pathways underlying complications secondary to cancer, indicating that this is a promising field of knowledge to be explored.
Collapse
Affiliation(s)
- Raquel D S Freitas
- Centro de Pesquisa em Toxicologia e Farmacologia, Escola de Ciências da Saúde, PUCRS, Porto Alegre 90619-900, RS, Brazil.
- Programa de Pós-graduação em Medicina e Ciências da Saúde, Escola de Medicina, PUCRS, Porto Alegre 90619-900, RS, Brazil.
| | - Maria M Campos
- Centro de Pesquisa em Toxicologia e Farmacologia, Escola de Ciências da Saúde, PUCRS, Porto Alegre 90619-900, RS, Brazil.
- Programa de Pós-graduação em Medicina e Ciências da Saúde, Escola de Medicina, PUCRS, Porto Alegre 90619-900, RS, Brazil.
- Programa de Pós-graduação em Odontologia, Escola de Ciências da Saúde, PUCRS, Porto Alegre 90619-900, RS, Brazil.
| |
Collapse
|
278
|
Fattori V, Pinho-Ribeiro FA, Staurengo-Ferrari L, Borghi SM, Rossaneis AC, Casagrande R, Verri WA. The specialised pro-resolving lipid mediator maresin 1 reduces inflammatory pain with a long-lasting analgesic effect. Br J Pharmacol 2019; 176:1728-1744. [PMID: 30830967 DOI: 10.1111/bph.14647] [Citation(s) in RCA: 80] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2018] [Revised: 01/30/2019] [Accepted: 02/11/2019] [Indexed: 12/16/2022] Open
Abstract
BACKGROUND AND PURPOSE Maresin 1 (MaR1) is a specialised pro-resolving lipid mediator with anti-inflammatory and analgesic activities. In this study, we addressed the modulation of peripheral and spinal cord cells by MaR1 in the context of inflammatory pain. EXPERIMENTAL APPROACH Mice were treated with MaR1 before intraplantar injection of carrageenan or complete Freund's adjuvant (CFA). Mechanical hyperalgesia was assessed using the electronic von Frey and thermal hyperalgesia using a hot plate. Spinal cytokine production and NF-κB activation were determined by ELISA and astrocytes and microglia activation by RT-qPCR and immunofluorescence. CGRP release by dorsal root ganglia (DRG) neurons was determined by EIA. Neutrophil and macrophage recruitment were determined by immunofluorescence, flow cytometry, and colorimetric methods. Trpv1 and Nav1.8 expression and calcium imaging of DRG neurons were determined by RT-qPCR and Fluo-4AM respectively. KEY RESULTS MaR1 reduced carrageenan- and CFA-induced mechanical and thermal hyperalgesia and neutrophil and macrophage recruitment proximal to CGRP+ fibres in the paw skin. Moreover, MaR1 reduced NF-κB activation, IL-1β and TNF-α production, and spinal cord glial cells activation. In the DRG, MaR1 reduced CFA-induced Nav1.8 and Trpv1 mRNA expression and calcium influx and capsaicin-induced release of CGRP by DRG neurons. CONCLUSIONS AND IMPLICATIONS MaR1 reduced DRG neurons activation and CGRP release explaining, at least in part, its analgesic and anti-inflammatory effects. The enduring analgesic and anti-inflammatory effects and also post-treatment activity of MaR1 suggest that specialised pro-resolving lipid mediators have potential as a new class of drugs for the treatment of inflammatory pain.
Collapse
Affiliation(s)
- Victor Fattori
- Department of Pathology, Centre of Biological Sciences, Londrina State University, Londrina, Brazil
| | - Felipe A Pinho-Ribeiro
- Department of Pathology, Centre of Biological Sciences, Londrina State University, Londrina, Brazil
| | | | - Sergio M Borghi
- Department of Pathology, Centre of Biological Sciences, Londrina State University, Londrina, Brazil
| | - Ana C Rossaneis
- Department of Pathology, Centre of Biological Sciences, Londrina State University, Londrina, Brazil
| | - Rubia Casagrande
- Department of Pharmaceutical Sciences, Centre of Health Science, Londrina State University, Londrina, Brazil
| | - Waldiceu A Verri
- Department of Pathology, Centre of Biological Sciences, Londrina State University, Londrina, Brazil
| |
Collapse
|
279
|
Saika A, Nagatake T, Kunisawa J. Host- and Microbe-Dependent Dietary Lipid Metabolism in the Control of Allergy, Inflammation, and Immunity. Front Nutr 2019; 6:36. [PMID: 31024921 PMCID: PMC6468274 DOI: 10.3389/fnut.2019.00036] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2019] [Accepted: 03/14/2019] [Indexed: 12/14/2022] Open
Abstract
The intestine is the largest immune organ in the body, provides the first line of defense against pathogens, and prevents excessive immune reactions to harmless or beneficial non-self-materials, such as food and intestinal bacteria. Allergic and inflammatory diseases in the intestine occur as a result of dysregulation of immunological homeostasis mediated by intestinal immunity. Several lines of evidence suggest that gut environmental factors, including nutrition and intestinal bacteria, play important roles in controlling host immune responses and maintaining homeostasis. Among nutritional factors, ω3 and ω6 essential polyunsaturated fatty acids (PUFAs) profoundly influence the host immune system. Recent advances in lipidomics technology have led to the identification of lipid mediators derived from ω3- and ω6-PUFAs. In particular, lipid metabolites from ω3-PUFAs (e.g., eicosapentaenoic acid and docosahexaenoic acid) have recently been shown to exert anti-allergic and anti-inflammatory responses; these metabolites include resolvins, protectins, and maresins. Furthermore, a new class of anti-allergic and anti-inflammatory lipid metabolites of 17,18-epoxyeicosatetraenoic acid has recently been identified in the control of allergic and inflammatory diseases in the gut and skin. Although these lipid metabolites were found to be endogenously generated in the host, accumulating evidence indicates that intestinal bacteria also participate in lipid metabolism and thus generate bioactive unique lipid mediators. In this review, we discuss the production machinery of lipid metabolites in the host and intestinal bacteria and the roles of these metabolites in the regulation of host immunity.
Collapse
Affiliation(s)
- Azusa Saika
- Laboratory of Vaccine Materials, Center for Vaccine and Adjuvant Research, and Laboratory of Gut Environmental System, National Institutes of Biomedical Innovation, Health and Nutrition, Osaka, Japan.,Graduate School of Pharmaceutical Sciences, Osaka University, Osaka, Japan
| | - Takahiro Nagatake
- Laboratory of Vaccine Materials, Center for Vaccine and Adjuvant Research, and Laboratory of Gut Environmental System, National Institutes of Biomedical Innovation, Health and Nutrition, Osaka, Japan
| | - Jun Kunisawa
- Laboratory of Vaccine Materials, Center for Vaccine and Adjuvant Research, and Laboratory of Gut Environmental System, National Institutes of Biomedical Innovation, Health and Nutrition, Osaka, Japan.,Graduate School of Pharmaceutical Sciences, Osaka University, Osaka, Japan.,International Research and Development Center for Mucosal Vaccines, Institute of Medical Science, University of Tokyo, Tokyo, Japan.,Graduate School of Medicine, Graduate School of Dentistry, Osaka University, Osaka, Japan.,Graduate School of Medicine, Kobe University, Kobe, Japan
| |
Collapse
|
280
|
Abstract
The tumor immune landscape gained considerable interest based on the knowledge that genetic aberrations in cancer cells alone are insufficient for tumor development. Macrophages are basically supporting all hallmarks of cancer and owing to their tremendous plasticity they may exert a whole spectrum of anti-tumor and pro-tumor activities. As part of the innate immune response, macrophages are armed to attack tumor cells, alone or in concert with distinct T cell subsets. However, in the tumor microenvironment, they sense nutrient and oxygen gradients, receive multiple signals, and respond to this incoming information with a phenotype shift. Often, their functional output repertoire is shifted to become tumor-supportive. Incoming and outgoing signals are chemically heterogeneous but also comprise lipid mediators. Here, we review the current understanding whereby arachidonate metabolites derived from the cyclooxygenase and lipoxygenase pathways shape the macrophage phenotype in a tumor setting. We discuss these findings in the context of cyclooxygenase-2 (COX-2) and microsomal prostaglandin E synthase-1 (mPGES-1) expression and concomitant prostaglandin E2 (PGE2) formation. We elaborate the multiple actions of this lipid in affecting macrophage biology, which are sensors for and generators of this lipid. Moreover, we summarize properties of 5-lipoxygenases (ALOX5) and 15-lipoxygenases (ALOX15, ALOX15B) in macrophages and clarify how these enzymes add to the role of macrophages in a dynamically changing tumor environment. This review will illustrate the potential routes how COX-2/mPGES-1 and ALOX5/-15 in macrophages contribute to the development and progression of a tumor.
Collapse
Affiliation(s)
- Andreas Weigert
- Institute of Biochemistry I/Pathobiochemistry, Faculty of Medicine, Goethe-University Frankfurt, Theodor-Stern-Kai 7, 60590, Frankfurt, Germany
| | - Elisabeth Strack
- Institute of Biochemistry I/Pathobiochemistry, Faculty of Medicine, Goethe-University Frankfurt, Theodor-Stern-Kai 7, 60590, Frankfurt, Germany
| | - Ryan G Snodgrass
- Institute of Biochemistry I/Pathobiochemistry, Faculty of Medicine, Goethe-University Frankfurt, Theodor-Stern-Kai 7, 60590, Frankfurt, Germany
| | - Bernhard Brüne
- Institute of Biochemistry I/Pathobiochemistry, Faculty of Medicine, Goethe-University Frankfurt, Theodor-Stern-Kai 7, 60590, Frankfurt, Germany. .,German Cancer Consortium (DKTK), Partner Site Frankfurt, Frankfurt, Germany.
| |
Collapse
|
281
|
Kutzner L, Rund KM, Ostermann AI, Hartung NM, Galano JM, Balas L, Durand T, Balzer MS, David S, Schebb NH. Development of an Optimized LC-MS Method for the Detection of Specialized Pro-Resolving Mediators in Biological Samples. Front Pharmacol 2019; 10:169. [PMID: 30899221 PMCID: PMC6416208 DOI: 10.3389/fphar.2019.00169] [Citation(s) in RCA: 63] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2018] [Accepted: 02/11/2019] [Indexed: 12/20/2022] Open
Abstract
The cardioprotective and anti-inflammatory effects of long chain omega-3 polyunsaturated fatty acids (n3 PUFA) are believed to be partly mediated by their oxygenated metabolites (oxylipins). In the last two decades interest in a novel group of autacoids termed specialized pro-resolving mediators (SPMs) increased. These are actively involved in the resolution of inflammation. SPMs are multiple hydroxylated fatty acids including resolvins, maresins, and protectins derived from the n3 PUFA eicosapentaenoic acid (EPA) and docosahexaenoic acid (DHA) as well as lipoxins derived from arachidonic acid (ARA). In the present paper, we developed an LC-MS/MS method for a comprehensive set of 18 SPMs derived from ARA, EPA, and DHA and integrated it into our targeted metabolomics platform. Quantification was based on external calibration utilizing five deuterated internal standards in combination with a second internal standard for quality assessment of sample preparation in each sample. The tandem mass spectrometric parameters were carefully optimized for sensitive and specific detection. The influence of source parameters of the used AB Sciex 6500 QTRAP instrument as well as electronic parameters and the selection of transitions are discussed. The method was validated/characterized based on the criteria listed in the European Medicines Agency (EMA) guideline on bioanalytical method validation and method performance is demonstrated regarding recovery of internal standards (between 78 ± 4% and 87 ± 3% from 500 μL of human serum) as well as extraction efficacy of SPMs in spiked plasma (intra-day accuracy within ±20 and ±15% at 0.1 and 0.3 nM in plasma, respectively). Based on the lower limit of quantification of 0.02-0.2 nM, corresponding to 0.18-2.7 pg on column, SPMs were generally not detectable/quantifiable in plasma and serum supporting that circulating levels of SPMs are very low, i.e., <0.1 nM in healthy subjects. Following septic shock or peritonitis, SPMs could be quantified in the samples of several patients. However, in these studies with a small number of patients no clear correlation with severity of inflammation could be observed.
Collapse
Affiliation(s)
- Laura Kutzner
- Chair of Food Chemistry, Faculty of Mathematics and Natural Sciences, University of Wuppertal, Wuppertal, Germany
| | - Katharina M Rund
- Chair of Food Chemistry, Faculty of Mathematics and Natural Sciences, University of Wuppertal, Wuppertal, Germany
| | - Annika I Ostermann
- Chair of Food Chemistry, Faculty of Mathematics and Natural Sciences, University of Wuppertal, Wuppertal, Germany
| | - Nicole M Hartung
- Chair of Food Chemistry, Faculty of Mathematics and Natural Sciences, University of Wuppertal, Wuppertal, Germany
| | - Jean-Marie Galano
- Institut des Biomolécules Max Mousseron (IBMM), UMR 5247 CNRS, ENSCM, Université de Montpellier, Montpellier, France
| | - Laurence Balas
- Institut des Biomolécules Max Mousseron (IBMM), UMR 5247 CNRS, ENSCM, Université de Montpellier, Montpellier, France
| | - Thierry Durand
- Institut des Biomolécules Max Mousseron (IBMM), UMR 5247 CNRS, ENSCM, Université de Montpellier, Montpellier, France
| | - Michael S Balzer
- Division of Nephrology and Hypertension, Department of Medicine, Hannover Medical School, Hannover, Germany
| | - Sascha David
- Division of Nephrology and Hypertension, Department of Medicine, Hannover Medical School, Hannover, Germany
| | - Nils Helge Schebb
- Chair of Food Chemistry, Faculty of Mathematics and Natural Sciences, University of Wuppertal, Wuppertal, Germany
| |
Collapse
|
282
|
Sugimoto MA, Vago JP, Perretti M, Teixeira MM. Mediators of the Resolution of the Inflammatory Response. Trends Immunol 2019; 40:212-227. [DOI: 10.1016/j.it.2019.01.007] [Citation(s) in RCA: 141] [Impact Index Per Article: 23.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2018] [Revised: 01/13/2019] [Accepted: 01/14/2019] [Indexed: 02/06/2023]
|
283
|
Treatment with maresin 1, a docosahexaenoic acid-derived pro-resolution lipid, protects skin from inflammation and oxidative stress caused by UVB irradiation. Sci Rep 2019; 9:3062. [PMID: 30816324 PMCID: PMC6395735 DOI: 10.1038/s41598-019-39584-6] [Citation(s) in RCA: 56] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2018] [Accepted: 01/22/2019] [Indexed: 12/19/2022] Open
Abstract
Acute exposure to UVB irradiation causes skin inflammation and oxidative stress, and long-term exposure to UVB irradiation may lead to carcinogenesis. Our organism has endogenous mechanisms to actively limit inflammation. Maresin 1 (MaR1; 7R,14S-dihydroxy-docosa-4Z,8E,10E,12Z,16Z,19Z-hexaenoic acid) is a pro-resolution lipid mediator derived from the docosahexaenoic acid, which presents anti-inflammatory and pro-resolution effects. However, it remains to be determined if treatment with MaR1 can inhibit inflammatory and oxidative alterations in the skin triggered by UVB. The treatment with MaR1 (0.1-10 ng/mice at -10 min relative to the UVB irradiation protocol) reduced UVB-induced skin edema, neutrophil recruitment (MPO; myeloperoxidase activity, and migration of LysM-eGFP+ cells), cytokine production, matrix metalloproteinase-9 activity, keratinocyte apoptosis, epidermal thickening, mast cells counts and degradation of skin collagen in hairless mice. UVB irradiation caused a decrease of GSH (reduced glutathione) levels, activity of the enzyme catalase, ferric reducing ability (FRAP), and ABTS radical scavenging capacity as well as induced lipid hydroperoxide, superoxide anion production, and gp91phox mRNA expression. These parameters that indicate oxidative stress were inhibited by MaR1 treatment. Therefore, these data suggest MaR1 as a promising pharmacological tool in controlling the deleterious effects related to UVB irradiation.
Collapse
|
284
|
Körner A, Schlegel M, Kaussen T, Gudernatsch V, Hansmann G, Schumacher T, Giera M, Mirakaj V. Sympathetic nervous system controls resolution of inflammation via regulation of repulsive guidance molecule A. Nat Commun 2019; 10:633. [PMID: 30733433 PMCID: PMC6367413 DOI: 10.1038/s41467-019-08328-5] [Citation(s) in RCA: 46] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2018] [Accepted: 01/04/2019] [Indexed: 12/27/2022] Open
Abstract
The bidirectional communication between the immune and nervous system is important in regulating immune responses. Here we show that the adrenergic nerves of sympathetic nervous system orchestrate inflammation resolution and regenerative programs by modulating repulsive guidance molecule A (RGM-A). In murine peritonitis, adrenergic nerves and RGM-A show bidirectional activation by stimulating the mutual expression and exhibit a higher potency for the cessation of neutrophil infiltration; this reduction is accompanied by increased pro-resolving monocyte or macrophage recruitment, polymorphonucleocyte clearance and specialized pro-resolving lipid mediators production at sites of injury. Chemical sympathectomy results in hyperinflammation and ineffective resolution in mice, while RGM-A treatments reverse these phenotypes. Signalling network analyses imply that RGM-A and β2AR agonist regulate monocyte activation by suppressing NF-κB activity but activating RICTOR and PI3K/AKT signalling. Our results thus illustrate the function of sympathetic nervous system and RGM-A in regulating resolution and tissue repair in a murine acute peritonitis model. Diverse interactions between the nervous and immune systems have been shown, but specific mechanistic insights are still lacking. Here the authors show, using both mouse inflammation models and clinical correlation, that adrenergic nerve may ameliorate inflammation by inducing repulsive guidance molecule A signalling.
Collapse
Affiliation(s)
- Andreas Körner
- Department of Anesthesiology and Intensive Care Medicine, Molecular Intensive Care Medicine, University Hospital Tübingen, Eberhard-Karls University, Hoppe-Seyler-Str. 3, 72076, Tübingen, Germany
| | - Martin Schlegel
- Department of Anesthesiology and Intensive Care Medicine, Molecular Intensive Care Medicine, University Hospital Tübingen, Eberhard-Karls University, Hoppe-Seyler-Str. 3, 72076, Tübingen, Germany
| | - Torsten Kaussen
- Department of Pediatric Cardiology and Critical Care, Carl-Neuberg-Str. 1, Hannover Medical School, 30625, Hannover, Germany
| | - Verena Gudernatsch
- Department of Anesthesiology and Intensive Care Medicine, Molecular Intensive Care Medicine, University Hospital Tübingen, Eberhard-Karls University, Hoppe-Seyler-Str. 3, 72076, Tübingen, Germany
| | - Georg Hansmann
- Department of Pediatric Cardiology and Critical Care, Carl-Neuberg-Str. 1, Hannover Medical School, 30625, Hannover, Germany
| | - Timo Schumacher
- Department of Pediatric Cardiology and Critical Care, Carl-Neuberg-Str. 1, Hannover Medical School, 30625, Hannover, Germany
| | - Martin Giera
- Center for Proteomics and Metabolomics, Leiden University Medical Center (LUMC), Albinusdreef 2, 2333 ZA, Leiden, The Netherlands
| | - Valbona Mirakaj
- Department of Anesthesiology and Intensive Care Medicine, Molecular Intensive Care Medicine, University Hospital Tübingen, Eberhard-Karls University, Hoppe-Seyler-Str. 3, 72076, Tübingen, Germany.
| |
Collapse
|
285
|
Coppey L, Davidson E, Shevalye H, Obrosov A, Yorek M. Effect of Early and Late Interventions with Dietary Oils on Vascular and Neural Complications in a Type 2 Diabetic Rat Model. J Diabetes Res 2019; 2019:5020465. [PMID: 31485451 PMCID: PMC6702827 DOI: 10.1155/2019/5020465] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/19/2019] [Revised: 05/28/2019] [Accepted: 07/08/2019] [Indexed: 02/07/2023] Open
Abstract
AIMS Determine the effect of dietary oils enriched in different mono- or polyunsaturated fatty acids, i.e., olive oil (18 : 1, oleic acid), safflower oil (18 : 2 n-6, linoleic acid), flaxseed oil (18 : 3 n-3, alpha linolenic acid), evening primrose oil (18 : 3 n-6, gamma linolenic acid), or menhaden oil (20:5/22 : 6 n-3 eicosapentaenoic/docosahexaenoic acids), on vascular and neural complications in high-fat-fed low-dose streptozotocin-treated Sprague-Dawley rats, an animal model for late-stage type 2 diabetes. MATERIALS AND METHODS Rats were fed a high-fat diet (45% kcal as fat primarily derived from lard) for 8 weeks and then treated with a low dose of streptozotocin (30 mg/kg) in order to induce hyperglycemia. After an additional 8 (early intervention) or 20 (late intervention) weeks, the different groups of rats were fed diets with 1/2 of the kcal of fat derived from lard replaced by the different dietary oils. In addition, a control group fed a standard diet (4.25% kcal as fat) and a diabetic group maintained on the high-fat diet were maintained. The treatment period was approximately 16 weeks. The endpoints evaluated included vascular reactivity of epineurial arterioles, motor and sensory nerve conduction velocity, thermal and corneal sensitivity, and innervation of sensory nerves in the cornea and skin. RESULTS Our findings show that menhaden and flaxseed oil provided the greatest benefit for correcting peripheral nerve damage caused by diabetes, whereas enriching the high-fat diet with menhaden oil provided the most benefit to acetylcholine-mediated vascular relaxation of epineurial arterioles of the sciatic nerve. Enriching the diets with fatty acids derived from the other oils provided none to partial improvements. CONCLUSIONS These studies imply that long-chain n-6 and n-3 polyunsaturated fatty acids could be an effective treatment for diabetic peripheral neuropathy with n-3 polyunsaturated fatty acids derived from fish oil being the most effective.
Collapse
MESH Headings
- Animals
- Diabetes Mellitus, Experimental/chemically induced
- Diabetes Mellitus, Experimental/diet therapy
- Diabetes Mellitus, Experimental/metabolism
- Diabetes Mellitus, Experimental/physiopathology
- Diabetes Mellitus, Type 2/chemically induced
- Diabetes Mellitus, Type 2/diet therapy
- Diabetes Mellitus, Type 2/metabolism
- Diabetes Mellitus, Type 2/physiopathology
- Diabetic Angiopathies/diet therapy
- Diabetic Angiopathies/metabolism
- Diabetic Angiopathies/physiopathology
- Diabetic Neuropathies/diet therapy
- Diabetic Neuropathies/metabolism
- Diabetic Neuropathies/physiopathology
- Diet, High-Fat
- Dietary Fats, Unsaturated/administration & dosage
- Dietary Fats, Unsaturated/pharmacology
- Drug Administration Schedule
- Fatty Liver/metabolism
- Fatty Liver/pathology
- Fatty Liver/physiopathology
- Lipid Metabolism/drug effects
- Male
- Rats
- Rats, Sprague-Dawley
- Sciatic Nerve/drug effects
- Sciatic Nerve/physiopathology
- Streptozocin
- Time Factors
Collapse
Affiliation(s)
- Lawrence Coppey
- Department of Internal Medicine, University of Iowa, Iowa City, IA, USA
| | - Eric Davidson
- Department of Internal Medicine, University of Iowa, Iowa City, IA, USA
| | - Hanna Shevalye
- Department of Internal Medicine, University of Iowa, Iowa City, IA, USA
| | - Alexander Obrosov
- Department of Internal Medicine, University of Iowa, Iowa City, IA, USA
| | - Mark Yorek
- Department of Internal Medicine, University of Iowa, Iowa City, IA, USA
- Department of Veterans Affairs Iowa City Health Care System, Iowa City, IA, USA
- Veterans Affairs Center for the Prevention and Treatment of Visual Loss, Iowa City, IA, USA
- Fraternal Order of Eagles Diabetes Research Center, University of Iowa, Iowa City, IA, USA
| |
Collapse
|
286
|
Pino AM, Rodríguez JP. Is fatty acid composition of human bone marrow significant to bone health? Bone 2019; 118:53-61. [PMID: 29258874 DOI: 10.1016/j.bone.2017.12.014] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/28/2017] [Accepted: 12/15/2017] [Indexed: 12/31/2022]
Abstract
The bone marrow adipose tissue (BMAT) is a conserved component of the marrow microenvironment, providing storage and release of energy and stabilizing the marrow extent. Also, it is recognized both the amount and quality of BMAT are relevant to preserve the functional relationships between BMAT, bone, and blood cell production. In this article we ponder the information supporting the tenet that the quality of BMAT is relevant to bone health. In the human adult the distribution of BMAT is heterogeneous over the entire skeleton, and both BMAT accumulation and bone loss come about with aging in healthy populations. But some pathological conditions which increase BMAT formation lead to bone impairment and fragility. Analysis in vivo of the relative content of saturated and unsaturated fatty acids (FA) in BMAT indicates site-related bone marrow fat composition and an association between increased unsaturation index (UI) and bone health. With aging some impairment ensues in the regulation of bone marrow cells and systemic signals leading to local chronic inflammation. Most of the bone loss diseases which evolve altered BMAT composition have as common factors aging and/or chronic inflammation. Both saturated and unsaturated FAs originate lipid species which are active mediators in the inflammation process. Increased free saturated FAs may lead to lipotoxicity of bone marrow cells. The pro-inflammatory, anti-inflammatory or resolving actions of compounds derived from long chain poly unsaturated FAs (PUFA) on bone cells is varied, and depending on the metabolism of the parent n:3 or n:6 PUFAs series. Taking together the evidence substantiate that marrow adipocyte function is fundamental for an efficient link between systemic and marrow fatty acids to accomplish specific energy or regulatory needs of skeletal and marrow cells. Further, they reveal marrow requirements of PUFAs.
Collapse
Affiliation(s)
- Ana María Pino
- Laboratorio de Biología Celular, INTA, Universidad de Chile, Chile
| | | |
Collapse
|
287
|
Sancéau JY, Maltais R, Poirier D, Marette A. Total Synthesis of the Antidiabetic (Type 2) Lipid Mediator Protectin DX/PDX. J Org Chem 2018; 84:495-505. [PMID: 30586310 DOI: 10.1021/acs.joc.8b01973] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
The first total synthesis of a lipid mediator derived from natural ω-3-fatty acid docosahexaenoic acid (DHA), 10 S,17 S-diHDHA (also referred to as protectin DX/PDX), was achieved in a convergent route (29 steps). The two chiral hydroxyl groups at C-10 and C-17 were derived from readily available ( S)-1,2,4-butanetriol and ( R)-glycidol, respectively. The two stereodefined E-double bonds were generated by a Takai olefination, and the skipped diene side chain was introduced with a stereocontrolled Wittig olefination. Importantly, the sensitive conjugated E, Z, E-triene intermediate was generated by a Boland reduction of the central triple bond of a E, E-dienyne. Overall, this synthetic strategy should allow the preparation of a larger quantity of PDX, which is inaccessible via previously reported biosynthetic approaches.
Collapse
Affiliation(s)
- Jean-Yves Sancéau
- Organic Synthesis Service, Medicinal Chemistry Platform , Centre Hospitalier Universitaire (CHU) de Québec-Research Center , Québec , QC , G1V 4G2 , Canada
| | - René Maltais
- Organic Synthesis Service, Medicinal Chemistry Platform , Centre Hospitalier Universitaire (CHU) de Québec-Research Center , Québec , QC , G1V 4G2 , Canada
| | - Donald Poirier
- Organic Synthesis Service, Medicinal Chemistry Platform , Centre Hospitalier Universitaire (CHU) de Québec-Research Center , Québec , QC , G1V 4G2 , Canada.,Department of Molecular Medicine, Faculty of Medicine , Université Laval , Québec , QC , G1V 0A6 , Canada
| | - André Marette
- Department of Medicine , Québec Heart and Lung Institute , Laval Hospital, Québec , QC G1V 4G5 , Canada
| |
Collapse
|
288
|
Gorjao R, Dos Santos CMM, Serdan TDA, Diniz VLS, Alba-Loureiro TC, Cury-Boaventura MF, Hatanaka E, Levada-Pires AC, Sato FT, Pithon-Curi TC, Fernandes LC, Curi R, Hirabara SM. New insights on the regulation of cancer cachexia by N-3 polyunsaturated fatty acids. Pharmacol Ther 2018; 196:117-134. [PMID: 30521881 DOI: 10.1016/j.pharmthera.2018.12.001] [Citation(s) in RCA: 47] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Cancer cachexia is a multifactorial syndrome that develops during malignant tumor growth. Changes in plasma levels of several hormones and inflammatory factors result in an intense catabolic state, decreased activity of anabolic pathways, anorexia, and marked weight loss, leading to cachexia development and/or accentuation. Inflammatory mediators appear to be related to the control of a highly regulated process of muscle protein degradation that accelerates the process of cachexia. Several mediators have been postulated to participate in this process, including TNF-α, myostatin, and activated protein degradation pathways. Some interventional therapies have been proposed, including nutritional (dietary, omega-3 fatty acid supplementation), hormonal (insulin), pharmacological (clenbuterol), and nonpharmacological (physical exercise) therapies. Omega-3 (n-3) polyunsaturated fatty acids (PUFAs), especially eicosapentaenoic acid (EPA) and docosahexaenoic acid, are recognized for their anti-inflammatory properties and have been used in therapeutic approaches to treat or attenuate cancer cachexia. In this review, we discuss recent findings on cellular and molecular mechanisms involved in inflammation in the cancer cachexia syndrome and the effectiveness of n-3 PUFAs to attenuate or prevent cancer cachexia.
Collapse
Affiliation(s)
- Renata Gorjao
- Institute of Physical Activity Sciences and Sports, Cruzeiro do Sul University, Sao Paulo, Brazil
| | | | | | | | | | | | - Elaine Hatanaka
- Institute of Physical Activity Sciences and Sports, Cruzeiro do Sul University, Sao Paulo, Brazil
| | | | - Fábio Takeo Sato
- Institute of Biology, State University of Campinas, Campinas, Brazil; School of Biomedical Sciences, Monash University, Melbourne, Australia
| | | | | | - Rui Curi
- Institute of Physical Activity Sciences and Sports, Cruzeiro do Sul University, Sao Paulo, Brazil; Institute of Biomedical Sciences, University of Sao Paulo, Sao Paulo, Brazil
| | - Sandro Massao Hirabara
- Institute of Physical Activity Sciences and Sports, Cruzeiro do Sul University, Sao Paulo, Brazil; Institute of Biomedical Sciences, University of Sao Paulo, Sao Paulo, Brazil.
| |
Collapse
|
289
|
Photobiomodulation modulates the resolution of inflammation during acute lung injury induced by sepsis. Lasers Med Sci 2018; 34:191-199. [DOI: 10.1007/s10103-018-2688-1] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2018] [Accepted: 11/08/2018] [Indexed: 12/28/2022]
|
290
|
Borges FT, Convento MB, Schor N. Bone marrow-derived mesenchymal stromal cell: what next? STEM CELLS AND CLONING-ADVANCES AND APPLICATIONS 2018; 11:77-83. [PMID: 30510433 PMCID: PMC6231430 DOI: 10.2147/sccaa.s147804] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
Bone marrow mesenchymal stromal cell (MSC) is a potential alternative in regenerative medicine and has great potential in many pathologic conditions including kidney disease. Although most of the studies demonstrate MSC efficiency, the regenerative potential may not be efficient in all diseases and patients. Stem cell feasibility is modified by donor characteristics as gender, age, diet, and health status, producing both positive and negative results. The conditioning of MSC can potentiate its effects and modify its culture medium (CM). In current practices, the cell-free treatment is gaining notable attention, while MSC-conditioned CM is being applied and studied in many experimental diseases, including, but not limited to, certain kidney diseases. This may be the next step for clinical trials. Studies in stem cell CM have focused mainly on extracellular vesicles, nucleic acids (mRNA and microRNA), lipids, and proteins presented in this CM. They mediate regenerative effects of MSC in a harmonic manner. In this review, we will analyze the regenerative potential of MSC and its CM as well as discuss some effective techniques for modifying its fractions and improving its therapeutic potential. CM fractions may be modified by hypoxic conditions, inflammation, lipid exposition, and protein growth factors. Other possible mechanisms of action of stem cells are also suggested. In the future, the MSC paracrine effect may be modified to more closely meet each patient’s needs.
Collapse
Affiliation(s)
- Fernanda T Borges
- Nephrology Division, Department of Medicine, Universidade Federal de São Paulo, São Paulo, SP, Brazil, .,Interdisciplinary Postgraduate Program in Health Sciences, Universidade Cruzeiro do Sul, São Paulo, SP, Brazil,
| | - Marcia Bastos Convento
- Nephrology Division, Department of Medicine, Universidade Federal de São Paulo, São Paulo, SP, Brazil,
| | - Nestor Schor
- Nephrology Division, Department of Medicine, Universidade Federal de São Paulo, São Paulo, SP, Brazil,
| |
Collapse
|
291
|
Hornedo-Ortega R, Cerezo AB, de Pablos RM, Krisa S, Richard T, García-Parrilla MC, Troncoso AM. Phenolic Compounds Characteristic of the Mediterranean Diet in Mitigating Microglia-Mediated Neuroinflammation. Front Cell Neurosci 2018; 12:373. [PMID: 30405355 PMCID: PMC6206263 DOI: 10.3389/fncel.2018.00373] [Citation(s) in RCA: 77] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2018] [Accepted: 10/01/2018] [Indexed: 12/21/2022] Open
Abstract
Neuroinflammation is a pathological feature of quite a number of Central Nervous System diseases such as Alzheimer and Parkinson's disease among others. The hallmark of brain neuroinflammation is the activation of microglia, which are the immune resident cells in the brain and represents the first line of defense when injury or disease occur. Microglial activated cells can adopt different phenotypes to carry out its diverse functions. Thus, the shift into pro-inflammatory/neurotoxic or anti-inflammatory/neuroprotective phenotypes, depending of the brain environment, has totally changed the understanding of microglia in neurodegenerative disease. For this reason, novel therapeutic strategies which aim to modify the microglia polarization are being developed. Additionally, the understanding of how nutrition may influence the prevention and/or treatment of neurodegenerative diseases has grown greatly in recent years. The protective role of Mediterranean diet (MD) in preventing neurodegenerative diseases has been reported in a number of studies. The Mediterranean dietary pattern includes as distinctive features the moderate intake of red wine and extra virgin olive oil, both of them rich in polyphenolic compounds, such as resveratrol, oleuropein and hydroxytyrosol and their derivatives, which have demonstrated anti-inflammatory effects on microglia on in vitro studies. This review summarizes our understanding of the role of dietary phenolic compounds characteristic of the MD in mitigating microglia-mediated neuroinflammation, including explanation regarding their bioavailability, metabolism and blood-brain barrier.
Collapse
Affiliation(s)
- Ruth Hornedo-Ortega
- MIB, Unité de Recherche Oenologie, EA4577, USC 1366 INRA, ISVV, Unive. de Bordeaux, Bordeaux, France
| | - Ana B. Cerezo
- Departamento de Nutrición y Bromatología, Toxicología y Medicina Legal, Área de Nutrición y Bromatología, Facultad de Farmacia, Universidad de Sevilla, Seville, Spain
| | - Rocío M. de Pablos
- Departamento de Bioquímica y Biología Molecular, Facultad de Farmacia, Universidad de Sevilla, Seville, Spain
| | - Stéphanie Krisa
- MIB, Unité de Recherche Oenologie, EA4577, USC 1366 INRA, ISVV, Unive. de Bordeaux, Bordeaux, France
| | - Tristan Richard
- MIB, Unité de Recherche Oenologie, EA4577, USC 1366 INRA, ISVV, Unive. de Bordeaux, Bordeaux, France
| | - M. Carmen García-Parrilla
- Departamento de Nutrición y Bromatología, Toxicología y Medicina Legal, Área de Nutrición y Bromatología, Facultad de Farmacia, Universidad de Sevilla, Seville, Spain
| | - Ana M. Troncoso
- Departamento de Nutrición y Bromatología, Toxicología y Medicina Legal, Área de Nutrición y Bromatología, Facultad de Farmacia, Universidad de Sevilla, Seville, Spain
| |
Collapse
|
292
|
Proto JD, Doran AC, Gusarova G, Yurdagul A, Sozen E, Subramanian M, Islam MN, Rymond CC, Du J, Hook J, Kuriakose G, Bhattacharya J, Tabas I. Regulatory T Cells Promote Macrophage Efferocytosis during Inflammation Resolution. Immunity 2018; 49:666-677.e6. [PMID: 30291029 PMCID: PMC6192849 DOI: 10.1016/j.immuni.2018.07.015] [Citation(s) in RCA: 290] [Impact Index Per Article: 41.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2017] [Revised: 04/05/2018] [Accepted: 07/25/2018] [Indexed: 12/13/2022]
Abstract
Regulatory T (Treg) cell responses and apoptotic cell clearance (efferocytosis) represent critical arms of the inflammation resolution response. We sought to determine whether these processes might be linked through Treg-cell-mediated enhancement of efferocytosis. In zymosan-induced peritonitis and lipopolysaccharide-induced lung injury, Treg cells increased early in resolution, and Treg cell depletion decreased efferocytosis. In advanced atherosclerosis, where defective efferocytosis drives disease progression, Treg cell expansion improved efferocytosis. Mechanistic studies revealed the following sequence: (1) Treg cells secreted interleukin-13 (IL-13), which stimulated IL-10 production in macrophages; (2) autocrine-paracrine signaling by IL-10 induced Vav1 in macrophages; and (3) Vav1 activated Rac1 to promote apoptotic cell engulfment. In summary, Treg cells promote macrophage efferocytosis during inflammation resolution via a transcellular signaling pathway that enhances apoptotic cell internalization. These findings suggest an expanded role of Treg cells in inflammation resolution and provide a mechanistic basis for Treg-cell-enhancement strategies for non-resolving inflammatory diseases.
Collapse
Affiliation(s)
- Jonathan D Proto
- Department of Medicine, Columbia University, New York, NY 10032, USA
| | - Amanda C Doran
- Department of Medicine, Columbia University, New York, NY 10032, USA
| | - Galina Gusarova
- Department of Medicine, Columbia University, New York, NY 10032, USA
| | - Arif Yurdagul
- Department of Medicine, Columbia University, New York, NY 10032, USA
| | - Erdi Sozen
- Department of Medicine, Columbia University, New York, NY 10032, USA; Department of Biochemistry, Marmara University, Istanbul, Turkey
| | | | - Mohammad N Islam
- Department of Medicine, Columbia University, New York, NY 10032, USA
| | | | - Jasper Du
- Department of Medicine, Columbia University, New York, NY 10032, USA
| | - Jaime Hook
- Department of Medicine, Columbia University, New York, NY 10032, USA
| | - George Kuriakose
- Department of Medicine, Columbia University, New York, NY 10032, USA
| | - Jahar Bhattacharya
- Department of Medicine, Columbia University, New York, NY 10032, USA; Department of Physiology and Cellular Biophysics, Columbia University, New York, NY 10032, USA
| | - Ira Tabas
- Department of Medicine, Columbia University, New York, NY 10032, USA; Department of Physiology and Cellular Biophysics, Columbia University, New York, NY 10032, USA; Department of Pathology and Cell Biology, Columbia University, New York, NY 10032, USA.
| |
Collapse
|
293
|
Ambrożewicz E, Wójcik P, Wroński A, Łuczaj W, Jastrząb A, Žarković N, Skrzydlewska E. Pathophysiological Alterations of Redox Signaling and Endocannabinoid System in Granulocytes and Plasma of Psoriatic Patients. Cells 2018; 7:cells7100159. [PMID: 30301214 PMCID: PMC6210326 DOI: 10.3390/cells7100159] [Citation(s) in RCA: 62] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2018] [Revised: 10/03/2018] [Accepted: 10/04/2018] [Indexed: 02/01/2023] Open
Abstract
Inflammatory granulocytes are characterized by an oxidative burst, which may promote oxidative stress and lipid modification both in affected tissues and on a systemic level. On the other hand, redox signaling involving lipid peroxidation products acting as second messengers of free radicals play important yet not fully understood roles in the pathophysiology of inflammation and various stress-associated disorders. Therefore, the aim of this study was to evaluate the onset of oxidative stress and alterations of enzyme-dependent lipid metabolism resulting from redox imbalance in granulocytes and plasma obtained from patients with psoriasis vulgaris or psoriatic arthritis in comparison to the healthy subjects. The results obtained revealed enhanced activity of pro-oxidant enzymes nicotinamide adenine dinucleotide phosphate (NADPH) and xanthine oxidases in granulocytes with a decrease of enzymatic and non-enzymatic antioxidants in the plasma of psoriatic patients. The nuclear factor erythroid 2–related factor 2 (Nrf2) and its regulators were increased in both forms of psoriasis while heme oxygenase 1 levels were increased only in psoriasis vulgaris. The redox imbalance was associated with decreased levels of phospholipids and of free polyunsaturated fatty acids but with enhanced activity of enzymes involved in lipid metabolism (phospholipase A2, acetylhydrolase PAF, cyclooxygenases 1 and 2) and increased lipid peroxidation products 4-hydroxynonenal, isoprostanes, and neuroprostanes. Increased endocannabinoids and G protein-coupled receptor 55 were observed in both forms of the disease while expression of the cannabinoid type 1 receptor (CB1) was increased only in patients with psoriatic arthritis, which is opposite to the cannabinoid type 2 receptor. This receptor was increased only in psoriasis vulgaris. Changes in protein expression promoted the apoptosis of granulocytes by increased caspases mainly in psoriasis vulgaris. This study indicates that inhibition of the Nrf2 pathway in psoriatic arthritis promotes a redox imbalance. In addition, increased expression of CB1 receptors leads to increased oxidative stress, lipid modifications, and inflammation, which, in turn, may promote the progression of psoriasis into the advanced, arthritic form of the disease.
Collapse
Affiliation(s)
- Ewa Ambrożewicz
- Department of Analytical Chemistry, Medical University of Bialystok, 15-222 Bialystok, Poland.
| | - Piotr Wójcik
- Department of Analytical Chemistry, Medical University of Bialystok, 15-222 Bialystok, Poland.
| | - Adam Wroński
- Dermatological Specialized Center "DERMAL" NZOZ in Bialystok, 15-453 Bialystok Poland.
| | - Wojciech Łuczaj
- Department of Analytical Chemistry, Medical University of Bialystok, 15-222 Bialystok, Poland.
| | - Anna Jastrząb
- Department of Analytical Chemistry, Medical University of Bialystok, 15-222 Bialystok, Poland.
| | - Neven Žarković
- LabOS, Rudjer Boskovic Institute, Laboratory for Oxidative Stress, 10000 Zagreb, Croatia.
| | - Elżbieta Skrzydlewska
- Department of Analytical Chemistry, Medical University of Bialystok, 15-222 Bialystok, Poland.
| |
Collapse
|
294
|
Lu Y, Xu Q, Yin G, Xu W, Jiang H. Resolvin D1 inhibits the proliferation of lipopolysaccharide-treated HepG2 hepatoblastoma and PLC/PRF/5 hepatocellular carcinoma cells by targeting the MAPK pathway. Exp Ther Med 2018; 16:3603-3610. [PMID: 30233715 PMCID: PMC6143846 DOI: 10.3892/etm.2018.6651] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2017] [Accepted: 10/20/2017] [Indexed: 12/22/2022] Open
Abstract
Hepatocellular carcinoma (HCC) and hepatoblastoma are common malignant tumor types in China. The aim of the present study was to evaluate the effects of resolvin D1 (RvD1) on inflammatory factor levels and mitogen-activated protein kinase (MAPK) signaling in lipopolysaccharide (LPS)-treated liver cancer cells. First, HepG2 hepatoblastoma and PLC/PRF/5 HCC cells were cultured and treated with LPS with or without various concentrations of RvD1 (0, 0.025, 0.05, 0.1 and 0.2%). Subsequently, ELISA was performed to measure the protein levels of tumor necrosis factor (TNF)-α, interleukin (IL)-1β and IL-6 in the culture medium. In addition, cell proliferation of the liver cancer cells was assessed by MTT assay. Reverse transcription-quantitative polymerase chain reaction and western blotting were performed to detect the expression of TNF-α, IL-1β and IL-6 in the cultured cells. Western blotting was also performed to assess the protein expression of phosphorylated extracellular signal-related kinase (p-ERK), p-c-Jun N-terminal kinase (p-JNK) and p-p38. Compared with the control group, LPS treatment increased the protein levels of TNF-α, IL-1β and IL-6 in the culture medium, and RvD1 inhibited this increase in a concentration-dependent manner. RvD1 also reduced the LPS-induced increase in TNF-α, IL-1β, IL-6, p-ERK, p-JNK and p-p38 expression levels in liver cancer cells. LPS promoted the proliferation of liver cancer cells, while RvD1 attenuated this effect. In summary, the current findings suggest that RvD1 inhibits cell proliferation and the expression of inflammatory cytokines in LPS-treated liver cancer cells by targeting the MAPK pathway.
Collapse
Affiliation(s)
- You Lu
- Department of Interventional Radiology, Jiangsu Cancer Hospital, Nanjing, Jiangsu 210009, P.R. China
| | - Qingyu Xu
- Department of Interventional Radiology, Jiangsu Cancer Hospital, Nanjing, Jiangsu 210009, P.R. China
| | - Guowen Yin
- Department of Interventional Radiology, Jiangsu Cancer Hospital, Nanjing, Jiangsu 210009, P.R. China
| | - Weidong Xu
- Department of Interventional Radiology, Jiangsu Cancer Hospital, Nanjing, Jiangsu 210009, P.R. China
| | - Hao Jiang
- Department of Interventional Radiology, Jiangsu Cancer Hospital, Nanjing, Jiangsu 210009, P.R. China
| |
Collapse
|
295
|
Ruiz-Miyazawa KW, Staurengo-Ferrari L, Pinho-Ribeiro FA, Fattori V, Zaninelli TH, Badaro-Garcia S, Borghi SM, Andrade KC, Clemente-Napimoga JT, Alves-Filho JC, Cunha TM, Fraceto LF, Cunha FQ, Napimoga MH, Casagrande R, Verri WA. 15d-PGJ 2-loaded nanocapsules ameliorate experimental gout arthritis by reducing pain and inflammation in a PPAR-gamma-sensitive manner in mice. Sci Rep 2018; 8:13979. [PMID: 30228306 PMCID: PMC6143605 DOI: 10.1038/s41598-018-32334-0] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2018] [Accepted: 09/05/2018] [Indexed: 12/13/2022] Open
Abstract
Gout arthritis (GA) is a painful inflammatory disease in response to monosodium urate (MSU) crystals in the joints. 15deoxy-Δ12,14-prostaglandin J2 (15d-PGJ2) is a natural activator of PPAR-γ with analgesic, anti-inflammatory, and pro-resolution properties. Thus, we aimed to evaluate the effect and mechanisms of action of 15d-PGJ2 nanocapsules (NC) in the model of GA in mice, since a reduction of 33-fold in the dose of 15d-PGJ2 has been reported. Mice were treated with 15d-PGJ2-loaded NC, inert NC, free 15d-PGJ2 (without NC), or 15d-PGJ2-loaded NC+ GW9662, a PPAR-γ inhibitor. We show that 15d-PGJ2-loaded NC provided analgesic effect in a dose that the free 15d-PGJ2 failed to inhibiting pain and inflammation. Hence, 15d-PGJ2-loaded NC reduced MSU-induced IL-1β, TNF-α, IL-6, IL-17, and IL-33 release and oxidative stress. Also, 15d-PGJ2-loaded NC decreased the maturation of IL-1β in LPS-primed BMDM triggered by MSU. Further, 15d-PGJ2-loaded NC decreased the expression of the components of the inflammasome Nlrp3, Asc, and Pro-caspase-1, as consequence of inhibiting NF-κB activation. All effects were PPAR-γ-sensitive. Therefore, we demonstrated that 15d-PGJ2-loaded NC present analgesic and anti-inflammatory properties in a PPAR-γ-dependent manner inhibiting IL-1β release and NF-κB activation in GA. Concluding, 15d-PGJ2-loaded NC ameliorates MSU-induced GA in a PPAR-γ-sensitive manner.
Collapse
Affiliation(s)
- Kenji W Ruiz-Miyazawa
- Departamento de Ciências Patológicas, Universidade Estadual de Londrina-UEL, Rod. Celso Garcia Cid, Km 380, PR445, 86057-970, Cx. Postal 10.011, Londrina, Paraná, Brazil
| | - Larissa Staurengo-Ferrari
- Departamento de Ciências Patológicas, Universidade Estadual de Londrina-UEL, Rod. Celso Garcia Cid, Km 380, PR445, 86057-970, Cx. Postal 10.011, Londrina, Paraná, Brazil
| | - Felipe A Pinho-Ribeiro
- Departamento de Ciências Patológicas, Universidade Estadual de Londrina-UEL, Rod. Celso Garcia Cid, Km 380, PR445, 86057-970, Cx. Postal 10.011, Londrina, Paraná, Brazil
| | - Victor Fattori
- Departamento de Ciências Patológicas, Universidade Estadual de Londrina-UEL, Rod. Celso Garcia Cid, Km 380, PR445, 86057-970, Cx. Postal 10.011, Londrina, Paraná, Brazil
| | - Tiago H Zaninelli
- Departamento de Ciências Patológicas, Universidade Estadual de Londrina-UEL, Rod. Celso Garcia Cid, Km 380, PR445, 86057-970, Cx. Postal 10.011, Londrina, Paraná, Brazil
| | - Stephanie Badaro-Garcia
- Departamento de Ciências Patológicas, Universidade Estadual de Londrina-UEL, Rod. Celso Garcia Cid, Km 380, PR445, 86057-970, Cx. Postal 10.011, Londrina, Paraná, Brazil
| | - Sergio M Borghi
- Departamento de Ciências Patológicas, Universidade Estadual de Londrina-UEL, Rod. Celso Garcia Cid, Km 380, PR445, 86057-970, Cx. Postal 10.011, Londrina, Paraná, Brazil
| | - Ketlem C Andrade
- Departamento de Ciências Patológicas, Universidade Estadual de Londrina-UEL, Rod. Celso Garcia Cid, Km 380, PR445, 86057-970, Cx. Postal 10.011, Londrina, Paraná, Brazil
| | - Juliana T Clemente-Napimoga
- Laboratory of Immunology and Molecular Biology, São Leopoldo Mandic Institute and Researcher Center, Campinas, Brazil
| | - Jose C Alves-Filho
- Department of Pharmacology, Ribeirão Preto Medical School, University of São Paulo, Avenida Bandeirantes s/n, 14050-490, Ribeirão Preto, São Paulo, Brazil
| | - Thiago M Cunha
- Department of Pharmacology, Ribeirão Preto Medical School, University of São Paulo, Avenida Bandeirantes s/n, 14050-490, Ribeirão Preto, São Paulo, Brazil
| | - Leonardo F Fraceto
- Department of Environmental Engineering, São Paulo State University, Sorocaba, Brazil
| | - Fernando Q Cunha
- Department of Pharmacology, Ribeirão Preto Medical School, University of São Paulo, Avenida Bandeirantes s/n, 14050-490, Ribeirão Preto, São Paulo, Brazil
| | - Marcelo H Napimoga
- Laboratory of Immunology and Molecular Biology, São Leopoldo Mandic Institute and Researcher Center, Campinas, Brazil
| | - Rubia Casagrande
- Departamento de Ciências Farmacêuticas, Universidade Estadual de Londrina-UEL, Avenida Robert Koch, 60, Hospital Universitário, 86038-350, Londrina, Paraná, Brazil
| | - Waldiceu A Verri
- Departamento de Ciências Patológicas, Universidade Estadual de Londrina-UEL, Rod. Celso Garcia Cid, Km 380, PR445, 86057-970, Cx. Postal 10.011, Londrina, Paraná, Brazil.
| |
Collapse
|
296
|
Schuster S, Johnson CD, Hennebelle M, Holtmann T, Taha AY, Kirpich IA, Eguchi A, Ramsden CE, Papouchado BG, McClain CJ, Feldstein AE. Oxidized linoleic acid metabolites induce liver mitochondrial dysfunction, apoptosis, and NLRP3 activation in mice. J Lipid Res 2018; 59:1597-1609. [PMID: 30084831 PMCID: PMC6121934 DOI: 10.1194/jlr.m083741] [Citation(s) in RCA: 73] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2018] [Revised: 06/30/2018] [Indexed: 01/03/2023] Open
Abstract
Circulating oxidized linoleic acid (LA) metabolites (OXLAMs) are increased in patients with nonalcoholic steatohepatitis (NASH) and their levels correlate with disease severity. However, the mechanisms by which OXLAMs contribute to NASH development are incompletely understood. We tested the hypothesis that LA or OXLAMs provided directly through the diet are involved in the development of hepatic injury. C57BL/6 mice were fed an isocaloric high-fat diet containing low LA, high LA, or OXLAMs for 8 weeks. The livers of OXLAM-fed mice showed lower triglyceride concentrations, but higher FA oxidation and lipid peroxidation in association with increased oxidative stress. OXLAM-induced mitochondrial dysfunction was associated with reduced Complex I protein and hepatic ATP levels, as well as increased mitochondrial biogenesis and cytoplasmic mitochondrial DNA. Oxidative stress increased thioredoxin-interacting protein (TXNIP) in the liver and stimulated the activation of mitochondrial apoptosis signal-regulating kinase 1 (ASK1) leading to apoptosis. We also found increased levels of NOD-like receptor protein 3 (NLRP3) inflammasome components and Caspase-1 activation in the livers of OXLAM-fed mice. In vitro, OXLAMs induced hepatocyte cell death, which was partly dependent on Caspase-1 activation. This study identified key mechanisms by which dietary OXLAMs contribute to NASH development, including mitochondrial dysfunction, hepatocyte cell death, and NLRP3 inflammasome activation.
Collapse
Affiliation(s)
- Susanne Schuster
- Department of Pediatrics, University of California San Diego, La Jolla, CA
| | - Casey D Johnson
- Department of Pediatrics, University of California San Diego, La Jolla, CA
| | - Marie Hennebelle
- Department of Food Science and Technology, University of California, Davis, CA University of Louisville, Louisville, KY
| | - Theresa Holtmann
- Department of Pediatrics, University of California San Diego, La Jolla, CA
| | - Ameer Y Taha
- Department of Food Science and Technology, University of California, Davis, CA University of Louisville, Louisville, KY
| | - Irina A Kirpich
- Division of Gastroenterology, Hepatology, and Nutrition University of Louisville, Louisville, KY; Hepatobiology and Toxicology Program, University of Louisville, Louisville, KY; Department of Pharmacology and Toxicology and University of Louisville Alcohol Center, University of Louisville School of Medicine, Louisville, KY
| | - Akiko Eguchi
- Department of Gastroenterology and Hepatology, Mie University Graduate School of Medicine, Tsu, Japan
| | - Christopher E Ramsden
- Intramural Programs of the National Institute on Aging and the National Institute on Alcohol Abuse and Alcoholism, National Institutes of Health, Bethesda, MD, and FOODplus Research Center, School of Agriculture, Food, and Wine, University of Adelaide, Adelaide, Australia
| | - Bettina G Papouchado
- Department of Pathology, Veterans Affairs San Diego Healthcare System, San Diego, CA
| | - Craig J McClain
- Division of Gastroenterology, Hepatology, and Nutrition University of Louisville, Louisville, KY; Hepatobiology and Toxicology Program, University of Louisville, Louisville, KY; Department of Pharmacology and Toxicology and University of Louisville Alcohol Center, University of Louisville School of Medicine, Louisville, KY; Robley Rex Veterans Medical Center, Louisville, KY
| | - Ariel E Feldstein
- Department of Pediatrics, University of California San Diego, La Jolla, CA.
| |
Collapse
|
297
|
Rosenthal MD, Patel J, Staton K, Martindale RG, Moore FA, Upchurch GR. Can Specialized Pro-resolving Mediators Deliver Benefit Originally Expected from Fish Oil? Curr Gastroenterol Rep 2018; 20:40. [PMID: 30078085 DOI: 10.1007/s11894-018-0647-4] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
PURPOSE OF THE REVIEW Fish oil (FO) supplementation has historically been used by individuals suffering from cardiovascular disease and other inflammatory processes. However, a meta-analysis of several large randomized control trials (RCTs) suggested FO conferred no benefit in reducing cardiovascular risk. Skeptics surmised that the lack of benefit was related to FO dose or drug interactions; therefore, the widely accepted practice of FO consumption was brought into question. RECENT FINDINGS Thereafter, Serhan et al. identified specialized pro-resolving mediators (SPMs) to be one of the bioactive components and mechanisms of action of FO. SPMs are thought to enhance resolution of inflammation, as opposed to classic anti-inflammatory agents which inhibit inflammatory pathways. Numerous diseases, including persistent Inflammation, immunosuppression, and catabolic syndrome (PICS), are rooted in a burden of chronic inflammation. SPMs are gaining traction as potential therapeutic agents used to resolve inflammation in cardiovascular disorders, inflammatory bowel disease, sepsis, pancreatitis, and acute respiratory distress syndrome (ARDS). This narrative reviews the history of FO and the various studies that made the health benefits of FO inconclusive, as well as an overview of SPMs and their use in specific disease states.
Collapse
Affiliation(s)
- Martin D Rosenthal
- Department of Surgery, Division of Trauma and Acute Care Surgery, University of Florida College of Medicine, PO Box 10019, Gainesville, FL, 32610-0019, USA.
| | - Jayshil Patel
- Department of Medicine, Division of Pulmonary Critical Care, Medical College of Wisconsin, Milwaukee, WI, USA
| | - Kyle Staton
- Department of Surgery, Division of Trauma and Acute Care Surgery, University of Florida College of Medicine, PO Box 10019, Gainesville, FL, 32610-0019, USA
| | - Robert G Martindale
- Department of Surgery, Division Gastroenterology Surgery, Oregon Health Science University, Portland, OR, USA
| | - Frederick A Moore
- Department of Surgery, Division of Trauma and Acute Care Surgery, University of Florida College of Medicine, PO Box 10019, Gainesville, FL, 32610-0019, USA
| | - Gilbert R Upchurch
- Department of Surgery, Division of Vascular Surgery, University of Florida College of Medicine, Gainesville, FL, USA
| |
Collapse
|
298
|
Lippestad M, Hodges RR, Utheim TP, Serhan CN, Dartt DA. Signaling pathways activated by resolvin E1 to stimulate mucin secretion and increase intracellular Ca 2+ in cultured rat conjunctival goblet cells. Exp Eye Res 2018; 173:64-72. [PMID: 29702100 PMCID: PMC6488018 DOI: 10.1016/j.exer.2018.04.015] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2018] [Revised: 04/18/2018] [Accepted: 04/23/2018] [Indexed: 12/31/2022]
Abstract
Glycoconjugate mucin secretion from conjunctival goblet cells is tightly regulated by nerves and specialized pro-resolving mediators (SPMs) to maintain ocular surface health. Here we investigated the actions of the SPM resolvin E1 (RvE1) on cultured rat conjunctival goblet cell glycoconjugate secretion and intracellular [Ca2+] ([Ca2+]i) and the signaling pathways used by RvE1. Goblet cells were cultured from rat conjunctiva in RPMI medium. The amount of RvE1-stimulated glycoconjugate mucin secretion was determined using an enzyme-linked lectin assay with Ulex Europaeus Agglutinin 1 lectin. Cultured goblet cells were also incubated with the Ca2+ indicator dye fura 2/AM and [Ca2+]i was measured. Cultured goblet cells were incubated with inhibitors to phospholipase (PL-) C, D, and A2 signaling pathways. RvE1 stimulated glycoconjugate secretion in a concentration dependent manner and was inhibited with the Ca2+ chelator BAPTA. The Ca2+i response was also increased in a concentration manner when stimulated by RvE1. Inhibition of PLC, PLD, and PLA2, but not Ca2+/calmodulin-dependent kinase blocked RvE1-stimulated increase in [Ca2+]i and glycoconjugate secretion. We conclude that under normal, physiological conditions RvE1 stimulates multiple pathways to increase glycoconjugate secretion and [Ca2+]i. RvE1 could be an important regulator of goblet cell glycoconjugate mucin secretion to maintain ocular surface health.
Collapse
Affiliation(s)
- Marit Lippestad
- Schepens Eye Research Institute, Massachusetts Eye and Ear Infirmary, Department of Ophthalmology, Harvard Medical School, Boston, MA, USA; Faculty of Medicine, University of Oslo, Oslo, Norway; Department of Medical Biochemistry, Oslo University Hospital, Oslo, Norway
| | - Robin R Hodges
- Schepens Eye Research Institute, Massachusetts Eye and Ear Infirmary, Department of Ophthalmology, Harvard Medical School, Boston, MA, USA
| | - Tor P Utheim
- Schepens Eye Research Institute, Massachusetts Eye and Ear Infirmary, Department of Ophthalmology, Harvard Medical School, Boston, MA, USA; Department of Medical Biochemistry, Oslo University Hospital, Oslo, Norway; Department of Plastic and Reconstructive Surgery, University of Oslo, Oslo, Norway
| | - Charles N Serhan
- Center for Experimental Therapeutics and Reperfusion Injury, Department of Anesthesiology, Perioperative and Pain Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
| | - Darlene A Dartt
- Schepens Eye Research Institute, Massachusetts Eye and Ear Infirmary, Department of Ophthalmology, Harvard Medical School, Boston, MA, USA; Department of Plastic and Reconstructive Surgery, University of Oslo, Oslo, Norway.
| |
Collapse
|
299
|
Çolakoğlu M, Tunçer S, Banerjee S. Emerging cellular functions of the lipid metabolizing enzyme 15-Lipoxygenase-1. Cell Prolif 2018; 51:e12472. [PMID: 30062726 DOI: 10.1111/cpr.12472] [Citation(s) in RCA: 55] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2018] [Accepted: 04/22/2018] [Indexed: 02/06/2023] Open
Abstract
The oxygenation of polyunsaturated fatty acids such as arachidonic and linoleic acid through lipoxygenases (LOXs) and cyclooxygenases (COXs) leads to the production of bioactive lipids that are important both in the induction of acute inflammation and its resolution. Amongst the several isoforms of LOX that are expressed in mammals, 15-LOX-1 was shown to be important both in the context of inflammation, being expressed in cells of the immune system, and in epithelial cells where the enzyme has been shown to crosstalk with a number of important signalling pathways. This review looks into the latest developments in understanding the role of 15-LOX-1 in different disease states with emphasis on the emerging role of the enzyme in the tumour microenvironment as well as a newly re-discovered form of cell death called ferroptosis. We also discuss future perspectives on the feasibility of use of this protein as a target for therapeutic interventions.
Collapse
Affiliation(s)
- Melis Çolakoğlu
- Department of Biological Sciences, Middle East Technical University, Ankara, Turkey
| | - Sinem Tunçer
- Department of Biological Sciences, Middle East Technical University, Ankara, Turkey
| | - Sreeparna Banerjee
- Department of Biological Sciences, Middle East Technical University, Ankara, Turkey
| |
Collapse
|
300
|
Djelmis J, Ivaniševic M, Desoye G, van Poppel M, Berberovic E, Soldo D, Oreskovic S. Higher Cord Blood Levels of Fatty Acids in Pregnant Women With Type 1 Diabetes Mellitus. J Clin Endocrinol Metab 2018; 103:2620-2629. [PMID: 29722816 DOI: 10.1210/jc.2018-00272] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/02/2018] [Accepted: 04/25/2018] [Indexed: 01/11/2023]
Abstract
CONTEXT Type 1 diabetes mellitus (T1DM) is associated with a disturbance of carbohydrate and lipid metabolism. OBJECTIVE To determine whether T1DM alters maternal and neonatal fatty acid (FA) levels. DESIGN Observational study. SETTING Academic hospital. PATIENTS Sixty pregnant women (30 women with T1DM with good glycemic control and 30 healthy women) were included in the study. Maternal blood, umbilical vein, and artery blood samples were collected immediately upon delivery. Following lipid extraction, the FA profiles of the total FA pool of maternal serum and umbilical vein and artery serum were determined by gas chromatography. RESULTS Total FA concentration in maternal serum did not differ between the study groups; it was significantly higher in umbilical vein serum of the T1DM group compared with that in the control group [median (interquartile range)]: T1DM 2126.2 (1446.4 to 3181.3) and control 1073.8 (657.5 to 2226.0; P < 0.001), and in umbilical artery vein serum: T1DM 1805.7 (1393.1 to 2125.0) and control 990.0 (643.3 to 1668.0; P < 0.001). Composition of FAs in umbilical vein serum showed significantly higher concentrations of saturated, monounsaturated, and polyunsaturated FAs (SFAs, MUFAs, and PUFAs, respectively) in the T1DM group than compared with those in the control group (P = 0.001). Furthermore, cord blood levels of leptin (P < 0.001), C-peptide (P < 0.001), and insulin resistance (P = 0.015) were higher in the T1DM group compared with controls. CONCLUSION The neonates born to mothers with T1DM had higher concentrations of total FAs, SFAs and MUFAs, as well as PUFAs, compared with control newborns.
Collapse
Affiliation(s)
- Josip Djelmis
- Referral Center for Diabetes in Pregnancy, Ministry of Health Republic of Croatia, Clinical Department of Obstetrics and Gynecology, Zagreb University Hospital Center, School of Medicine, University of Zagreb, Zagreb, Croatia
| | - Marina Ivaniševic
- Referral Center for Diabetes in Pregnancy, Ministry of Health Republic of Croatia, Clinical Department of Obstetrics and Gynecology, Zagreb University Hospital Center, School of Medicine, University of Zagreb, Zagreb, Croatia
| | - Gernot Desoye
- Department of Obstetrics and Gynecology, Medical University of Graz, Graz, Austria
| | | | - Edina Berberovic
- Clinical Department of Obstetrics and Gynecology, Holy Spirit University Hospital, Zagreb, Croatia
| | - Dragan Soldo
- Department of Obstetrics and Gynecology, Mostar Clinical Hospital Center, School of Medicine, University of Mostar, Mostar, Bosnia and Herzegovina
| | - Slavko Oreskovic
- Referral Center for Diabetes in Pregnancy, Ministry of Health Republic of Croatia, Clinical Department of Obstetrics and Gynecology, Zagreb University Hospital Center, School of Medicine, University of Zagreb, Zagreb, Croatia
| |
Collapse
|