1
|
de Souza T, Monteiro JDC, Curioni CC, Cople-Rodrigues C, Citelli M. Nutrients with Antioxidant Properties and Their Effects on Lower-Limb Ulcers: A Systematic Review. INT J LOW EXTR WOUND 2024; 23:217-230. [PMID: 35072533 DOI: 10.1177/15347346221074861] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
Chronic lower-limb ulcers (LLUs) are ulcers that fail to proceed through an orderly and timely process to produce anatomic and functional integrity. LLUs reduce the quality of life of affected individuals and are a public health problem. The treatment options include medications or surgery. Nutrition therapy is an important adjunct to improve the clinical picture and healing of LLUs. Considering that nutrients with antioxidant properties can improve the process of tissue healing, this systematic review aimed to evaluate the efficacy of antioxidant nutrient supplementation in the treatment of LLUs through randomized clinical trials. This systematic review was performed in accordance with the Preferred Reporting Items for Systematic Reviews and Meta-analyses and the Cochrane Handbook for Systematic Reviews of Interventions. The guiding question was-can antioxidant nutrients help in the treatment of chronic LLUs? In total, 1184 articles were found when searching for antioxidant nutrients associated with the most common causes of LLUs. Fourteen articles were included in this review after removing duplicates, studies with topical and/or venous use of antioxidants, and articles published in other languages, except English. Omega-3 fatty acids, magnesium, zinc, vitamins A, C, D, and resveratrol along with probiotics positively improved the ulcer healing. These effects were more significant when there was initially a deficiency of the respective supplemented nutrients. Therefore, correcting and maintaining an adequate nutritional status can improve ulcer healing and contribute to the clinical treatment of patients with LLUs.
Collapse
Affiliation(s)
- Thamiris de Souza
- Nutrition Institute, Rio de Janeiro State University, Rio de Janeiro, Brazil
| | | | | | | | - Marta Citelli
- Nutrition Institute, Rio de Janeiro State University, Rio de Janeiro, Brazil
| |
Collapse
|
2
|
Mahmoudi Z, Roumi Z, Askarpour SA, Mousavi Z, Shafaei H, Valisoltani N, Shapouri M, Mirshafaei SR, Mirzaee P, Mobarakeh KA, Taghavi Sufiani E, Motiee Bijarpasi Z, Motiei Z, Khosravi M, Doaei S, Gholamalizadeh M. The effects of omega-3 fatty acids supplementation on hemoglobin, hematocrit, and platelet levels of patients with ESRD condition undergoing dialysis. J Transl Autoimmun 2024; 8:100233. [PMID: 38464414 PMCID: PMC10924142 DOI: 10.1016/j.jtauto.2024.100233] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2023] [Revised: 01/30/2024] [Accepted: 02/05/2024] [Indexed: 03/12/2024] Open
Abstract
Background Concomitant inflammation may boost the cardiovascular complications in end-stage renal disease (ESRD) patients undergoing hemodialysis (HD). Omega-3 fatty acids may have certain health benefits in HD patients. The aim of this study was to investigate the effects of omega-3 fatty acids supplementation on hematocrit (HCT), hemoglobin (HB) level and platelet (PLT) counts of HD patients. Methods A randomized controlled trial was conducted on HD patients at a private dialysis center in Rasht, Iran. Three omega-3 fatty acid supplement capsules (3 g/d) were administered daily for two months to patients in the intervention group (n = 55). The control group (n = 60) were given three placebo capsules containing medium chain triglyceride (MCT) oil, similar to the supplemental dose of the intervention group at the same period. Three parameters of HCT, HB and PLT were measured at baseline and after the intervention. Results The PLT count decreased in the intervention group compared to the control group (173.38 ± 74.76 vs. 227.68 ± 86.58 103/mm3, F = 4.83, P = 0.03). No significant change was found on the levels of HCT and HB parameters between the two groups after the intervention. Conclusion Omega-3 supplementation in HD patients may decrease the risk of forming blood clots in the blood vessels. Further studies are warranted.
Collapse
Affiliation(s)
- Zahra Mahmoudi
- Department of Nutrition, Science and Research Branch, Islamic Azad University, Tehran, Iran
| | - Zahra Roumi
- Department of Nutrition, Electronic Health and Statistics Surveillance Research Center, Science and Research Branch. Islamic Azad University, Tehran, Iran
| | - Seyed Ali Askarpour
- Division of Food Safety and Hygiene, Department of Environmental Health Engineering, School of Public Health, Tehran University of Medical Sciences, Tehran, Iran
| | - Zahra Mousavi
- School of Nursing and Midwifery Shahed University, Tehran, Iran
| | - Hanieh Shafaei
- Department of Nursing, School of Nursing and Midwifery, Guilan University of Medical Sciences, Rasht, Iran
| | - Neda Valisoltani
- Department of Medicine, Faculty of Medicine, Semnan University of Medical Sciences, Semnan, Iran
| | - Mahsa Shapouri
- Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Seyed Reza Mirshafaei
- Department of applied mathematics, Roudsar and Amlash branch, Islamic Azad University, Roudsar, Iran
| | - Pouya Mirzaee
- Department of Medicine, Faculty of Medicine, Semnan University of Medical Sciences, Semnan, Iran
| | - Khadijeh Abbasi Mobarakeh
- Department of Community Nutrition, Nutrition and Food Security Research Center, School of Nutrition and Food Science, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Elahe Taghavi Sufiani
- Department of Nursing, School of Nursing and Midwifery, Guilan University of Medical Sciences, Rasht, Iran
| | - Zeinab Motiee Bijarpasi
- Department of Nursing, School of Nursing and Midwifery, Guilan University of Medical Sciences, Rasht, Iran
| | - Zeynab Motiei
- Department of Nursing, School of Nursing and Midwifery, Guilan University of Medical Sciences, Rasht, Iran
| | - Masoud Khosravi
- Urology Research Center, Razi Hospital, School of Medicine, Guilan University of Medical Sciences, Rasht, Iran
| | - Saeid Doaei
- Department of Community Nutrition, School of Nutrition and Food Sciences, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Maryam Gholamalizadeh
- Cancer Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| |
Collapse
|
3
|
Mohammad-Rafiei F, Negahdari S, Tahershamsi Z, Gheibihayat SM. Interface between Resolvins and Efferocytosis in Health and Disease. Cell Biochem Biophys 2024; 82:53-65. [PMID: 37794303 DOI: 10.1007/s12013-023-01187-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2023] [Accepted: 09/25/2023] [Indexed: 10/06/2023]
Abstract
Acute inflammation resolution acts as a vital process for active host response, tissue support, and homeostasis maintenance, during which resolvin D (RvD) and E (RvE) as mediators derived from omega-3 polyunsaturated fatty acids display specific and stereoselective anti-inflammations like restricting neutrophil infiltration and pro-resolving activities. On the other side of the coin, potent macrophage-mediated apoptotic cell clearance, namely efferocytosis, is essential for successful inflammation resolution. Further studies mentioned a linkage between efferocytosis and resolvins. For instance, resolvin D1 (RvD1), which is endogenously formed from docosahexaenoic acid within the inflammation resolution, thereby provoking efferocytosis. There is still limited information regarding the mechanism of action of RvD1-related efferocytosis enhancement at the molecular level. The current review article was conducted to explore recent data on how the efferocytosis process and resolvins relate to each other during the inflammation resolution in illness and health. Understanding different aspects of this connection sheds light on new curative approaches for medical conditions caused by defective efferocytosis and disrupted inflammation resolution.
Collapse
Affiliation(s)
- Fatemeh Mohammad-Rafiei
- Department of Medical Biotechnology, School of Medicine, Shahid Sadoughi University of Medical Sciences, Yazd, Iran
| | - Samira Negahdari
- Legal Medicine Research Center, Legal Medicine Organization, Tehran, Iran
| | - Zahra Tahershamsi
- Department of Biophysics, Faculty of Biological Sciences, Tarbiat Modares University, Tehran, Iran
| | - Seyed Mohammad Gheibihayat
- Department of Medical Biotechnology, School of Medicine, Shahid Sadoughi University of Medical Sciences, Yazd, Iran.
- Network of Immunity in Infection, Malignancy and Autoimmunity (NIIMA), Universal Scientific Education and Research Network (USERN), Munich, Germany.
| |
Collapse
|
4
|
Koudelka A, Buchan GJ, Cechova V, O'Brien JP, Liu H, Woodcock SR, Mullett SJ, Zhang C, Freeman BA, Gelhaus SL. Lipoxin A 4 yields an electrophilic 15-oxo metabolite that mediates FPR2 receptor-independent anti-inflammatory signaling. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.02.06.579101. [PMID: 38370667 PMCID: PMC10871244 DOI: 10.1101/2024.02.06.579101] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/20/2024]
Abstract
The enzymatic oxidation of arachidonic acid is proposed to yield trihydroxytetraene species (termed lipoxins) that resolve inflammation via ligand activation of the formyl peptide receptor, FPR2. While cell and murine models activate signaling responses to synthetic lipoxins, primarily 5S,6R,15S-trihydroxy-7E,9E,11Z,13E-eicosatetraenoic acid (lipoxin A4, LXA4), there are expanding concerns about the biological formation, detection and signaling mechanisms ascribed to LXA4 and related di- and tri-hydroxy ω-6 and ω-3 fatty acids. Herein, the generation and actions of LXA4 and its primary 15-oxo metabolite were assessed in control, LPS-activated and arachidonic acid supplemented RAW 264.7 macrophages. Despite protein expression of all enzymes required for LXA4 synthesis, both LXA4 and its 15-oxo-LXA4 metabolite were undetectable. Moreover, synthetic LXA4 and the membrane permeable 15-oxo-LXA4 methyl ester that is rapidly de-esterified to 15-oxo-LXA4, displayed no ligand activity for the putative LXA4 receptor FPR2, as opposed to the FPR2 ligand WKYMVm. Alternatively, 15-oxo-LXA4, an electrophilic α,β-unsaturated ketone, alkylates nucleophilic amino acids such as cysteine to modulate redox-sensitive transcriptional regulatory protein and enzyme function. 15-oxo-LXA4 activated nuclear factor (erythroid related factor 2)-like 2 (Nrf2)-regulated gene expression of anti-inflammatory and repair genes and inhibited nuclear factor (NF)-κB-regulated pro-inflammatory mediator expression. LXA4 did not impact these macrophage anti-inflammatory and repair responses. In summary, these data show an absence of macrophage LXA4 formation and receptor-mediated signaling actions. Rather, if LXA4 were present in sufficient concentrations, this, and other more abundant mono- and poly-hydroxylated unsaturated fatty acids can be readily oxidized to electrophilic α,β-unsaturated ketone products that modulate the redox-sensitive cysteine proteome via G-protein coupled receptor-independent mechanisms.
Collapse
Affiliation(s)
- Adolf Koudelka
- Department of Pharmacology and Chemical Biology, University of Pittsburgh School of Medicine (Pittsburgh, PA 15213)
| | - Gregory J Buchan
- Department of Pharmacology and Chemical Biology, University of Pittsburgh School of Medicine (Pittsburgh, PA 15213)
| | - Veronika Cechova
- Department of Pharmacology and Chemical Biology, University of Pittsburgh School of Medicine (Pittsburgh, PA 15213)
| | - James P O'Brien
- Department of Pharmacology and Chemical Biology, University of Pittsburgh School of Medicine (Pittsburgh, PA 15213)
| | - Heng Liu
- Department of Pharmacology and Chemical Biology, University of Pittsburgh School of Medicine (Pittsburgh, PA 15213)
| | - Steven R Woodcock
- Department of Pharmacology and Chemical Biology, University of Pittsburgh School of Medicine (Pittsburgh, PA 15213)
| | - Steven J Mullett
- Department of Pharmacology and Chemical Biology, University of Pittsburgh School of Medicine (Pittsburgh, PA 15213)
- Health Sciences Mass Spectrometry Core, University of Pittsburgh (Pittsburgh, PA 15213)
| | - Cheng Zhang
- Department of Pharmacology and Chemical Biology, University of Pittsburgh School of Medicine (Pittsburgh, PA 15213)
| | - Bruce A Freeman
- Department of Pharmacology and Chemical Biology, University of Pittsburgh School of Medicine (Pittsburgh, PA 15213)
| | - Stacy L Gelhaus
- Department of Pharmacology and Chemical Biology, University of Pittsburgh School of Medicine (Pittsburgh, PA 15213)
- Health Sciences Mass Spectrometry Core, University of Pittsburgh (Pittsburgh, PA 15213)
| |
Collapse
|
5
|
Sundaram TS, Addis MF, Giromini C, Rebucci R, Pisanu S, Pagnozzi D, Baldi A. Comprehensive proteomic analysis reveals omega-3 fatty acids to counteract endotoxin-stimulated metabolic dysregulation in porcine enterocytes. Sci Rep 2023; 13:21595. [PMID: 38062040 PMCID: PMC10703801 DOI: 10.1038/s41598-023-48018-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2022] [Accepted: 11/21/2023] [Indexed: 12/18/2023] Open
Abstract
Omega-3 polyunsaturated fatty acids (n-3 PUFA), such as the eicosapentaenoic acid (EPA) and docosahexaenoic acid (DHA), are reported to beneficially affect the intestinal immunity. The biological pathways modulated by n-3 PUFA during an infection, at the level of intestinal epithelial barrier remain elusive. To address this gap, we investigated the proteomic changes induced by n-3 PUFA in porcine enterocyte cell line (IPEC-J2), in the presence and absence of lipopolysaccharide (LPS) stress conditions using shotgun proteomics analysis integrated with RNA-sequencing technology. A total of 33, 85, and 88 differentially abundant proteins (DAPs) were identified in cells exposed to n-3 PUFA (DHA:EPA), LPS, and n-3 PUFA treatment followed by LPS stimulation, respectively. Functional annotation and pathway analysis of DAPs revealed the modulation of central carbon metabolism, including the glycolysis/gluconeogenesis, pentose phosphate pathway, and oxidative phosphorylation processes. Specifically, LPS caused metabolic dysregulation in enterocytes, which was abated upon prior treatment with n-3 PUFA. Besides, n-3 PUFA supplementation facilitated enterocyte development and lipid homeostasis. Altogether, this work for the first time comprehensively described the biological pathways regulated by n-3 PUFA in enterocytes, particularly during endotoxin-stimulated metabolic dysregulation. Additionally, this study may provide nutritional biomarkers in monitoring the intestinal health of human and animals on n-3 PUFA-based diets.
Collapse
Affiliation(s)
- Tamil Selvi Sundaram
- Department of Veterinary Medicine and Animal Sciences, University of Milan, Via Dell'Università 6, 26900, Lodi, Italy.
- University of Veterinary Medicine and Pharmacy in Košice, Komenského 68/73, 04181, Košice, Slovakia.
| | - Maria Filippa Addis
- Department of Veterinary Medicine and Animal Sciences, University of Milan, Via Dell'Università 6, 26900, Lodi, Italy
| | - Carlotta Giromini
- Department of Veterinary Medicine and Animal Sciences, University of Milan, Via Dell'Università 6, 26900, Lodi, Italy
| | - Raffaella Rebucci
- Department of Veterinary Medicine and Animal Sciences, University of Milan, Via Dell'Università 6, 26900, Lodi, Italy
| | - Salvatore Pisanu
- Porto Conte Ricerche S.R.L, S.P. 55 Porto Conte/Capo Caccia, Loc. Tramariglio 15, 07041, Alghero, Italy
| | - Daniela Pagnozzi
- Porto Conte Ricerche S.R.L, S.P. 55 Porto Conte/Capo Caccia, Loc. Tramariglio 15, 07041, Alghero, Italy
| | - Antonella Baldi
- Department of Veterinary Medicine and Animal Sciences, University of Milan, Via Dell'Università 6, 26900, Lodi, Italy
| |
Collapse
|
6
|
Rasquel-Oliveira FS, Silva MDVD, Martelossi-Cebinelli G, Fattori V, Casagrande R, Verri WA. Specialized Pro-Resolving Lipid Mediators: Endogenous Roles and Pharmacological Activities in Infections. Molecules 2023; 28:5032. [PMID: 37446699 DOI: 10.3390/molecules28135032] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2023] [Revised: 06/07/2023] [Accepted: 06/19/2023] [Indexed: 07/15/2023] Open
Abstract
During an infection, inflammation mobilizes immune cells to eliminate the pathogen and protect the host. However, inflammation can be detrimental when exacerbated and/or chronic. The resolution phase of the inflammatory process is actively orchestrated by the specialized pro-resolving lipid mediators (SPMs), generated from omega-3 and -6 polyunsaturated fatty acids (PUFAs) that bind to different G-protein coupled receptors to exert their activity. As immunoresolvents, SPMs regulate the influx of leukocytes to the inflammatory site, reduce cytokine and chemokine levels, promote bacterial clearance, inhibit the export of viral transcripts, enhance efferocytosis, stimulate tissue healing, and lower antibiotic requirements. Metabolomic studies have evaluated SPM levels in patients and animals during infection, and temporal regulation of SPMs seems to be essential to properly coordinate a response against the microorganism. In this review, we summarize the current knowledge on SPM biosynthesis and classifications, endogenous production profiles and their effects in animal models of bacterial, viral and parasitic infections.
Collapse
Affiliation(s)
- Fernanda S Rasquel-Oliveira
- Laboratory of Pain, Inflammation, Neuropathy, and Cancer, Department of Pathology, Center of Biological Sciences, Londrina State University, Londrina 86057-970, Paraná, Brazil
| | - Matheus Deroco Veloso da Silva
- Laboratory of Pain, Inflammation, Neuropathy, and Cancer, Department of Pathology, Center of Biological Sciences, Londrina State University, Londrina 86057-970, Paraná, Brazil
| | - Geovana Martelossi-Cebinelli
- Laboratory of Pain, Inflammation, Neuropathy, and Cancer, Department of Pathology, Center of Biological Sciences, Londrina State University, Londrina 86057-970, Paraná, Brazil
| | - Victor Fattori
- Vascular Biology Program, Boston Children's Hospital, Department of Surgery, Harvard Medical School, Boston, MA 02115, USA
| | - Rubia Casagrande
- Department of Pharmaceutical Sciences, Center of Health Science, Londrina State University, Londrina 86038-440, Paraná, Brazil
| | - Waldiceu A Verri
- Laboratory of Pain, Inflammation, Neuropathy, and Cancer, Department of Pathology, Center of Biological Sciences, Londrina State University, Londrina 86057-970, Paraná, Brazil
| |
Collapse
|
7
|
Zambelli VO, Hösch NG, Farom S, Zychar BC, Spadacci-Morena DD, Carvalho LV, Curi R, Lepsch LB, Scavone C, Sant'Anna OA, Gonçalves LRC, Cury Y, Sampaio SC. Formyl peptide receptors are involved in CTX-induced impairment of lymphocyte functions. Toxicon 2023; 222:106986. [PMID: 36442690 DOI: 10.1016/j.toxicon.2022.106986] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2022] [Revised: 11/18/2022] [Accepted: 11/23/2022] [Indexed: 11/27/2022]
Abstract
Crotoxin (CTX) is a neurotoxin that is isolated from the venom of Crotalus durissus terrificus, which displays immunomodulatory, anti-inflammatory, and anti-tumoral effects. Previous research has demonstrated that CTX promotes the adherence of leukocytes to the endothelial cells in blood microcirculation and the high endothelial venules of lymph nodes, which reduces the number of blood cells and lymphocytes. Studies have also shown that these effects are mediated by lipoxygenase-derived mediators. However, the exact lipoxygenase-derived eicosanoid involved in the CTX effect on lymphocytes is yet to be characterized. As CTX stimulates lipoxin-derived mediators from macrophages and lymphocyte effector functions could be modulated by activating formyl peptide receptors, we aimed to investigate whether these receptors were involved in CTX-induced redistribution and functions of lymphocytes in rats. We used male Wistar rats treated with CTX to demonstrate that Boc2 (butoxycarbonyl-Phe-Leu-Phe-Leu-Phe), an antagonist of formyl peptide receptors, prevented CTX-induced decrease in the number of circulating lymphocytes and increased the expression of the lymphocyte adhesion molecule LFA1. CTX reduced the T and B lymphocyte functions, such as lymphocyte proliferation in response to the mitogen Concanavalin A and antibody production in response to BSA immunization, respectively, which was prevented by the administration of Boc2. Importantly, mesenteric lymph node lymphocytes from CTX-treated rats showed an increased release of 15-epi-LXA4. These results indicate that formyl peptide receptors mediate CTX-induced redistribution of lymphocytes and that 15-epi-LXA4 is a key mediator of the immunosuppressive effects of CTX.
Collapse
Affiliation(s)
- Vanessa O Zambelli
- Laboratory of Pain and Signaling, Butantan Institute, Av. Vital Brazil, 1500, 05503-900, São Paulo, SP, Brazil.
| | - Natália Gabriele Hösch
- Laboratory of Pain and Signaling, Butantan Institute, Av. Vital Brazil, 1500, 05503-900, São Paulo, SP, Brazil
| | - Sarah Farom
- Laboratory of Pain and Signaling, Butantan Institute, Av. Vital Brazil, 1500, 05503-900, São Paulo, SP, Brazil; Laboratory of Pathophysiology, Butantan Institute, Av. Vital Brazil, 1500, 05503-900, São Paulo, SP, Brazil
| | - Bianca C Zychar
- Laboratory of Pathophysiology, Butantan Institute, Av. Vital Brazil, 1500, 05503-900, São Paulo, SP, Brazil
| | - Diva D Spadacci-Morena
- Laboratory of Pathophysiology, Butantan Institute, Av. Vital Brazil, 1500, 05503-900, São Paulo, SP, Brazil
| | - Luciana Vieira Carvalho
- Laboratory of Immunochemistry, Butantan Institute, Av. Vital Brasil, 1500, 05503-900, São Paulo, SP, Brazil
| | - Rui Curi
- Immunobiological Production Section, Bioindustrial Center, Av. Vital Brazil, 1500, 05503-900, São Paulo, SP, Brazil; Interdisciplinary Post-graduate Program in Health Sciences, Cruzeiro of Sul University, São Paulo, SP, Brazil
| | - Lucilia B Lepsch
- Department of Pharmacology, Institute of Biomedical Science, University of São Paulo, São Paulo, 05508-900, Brazil
| | - Cristoforo Scavone
- Department of Pharmacology, Institute of Biomedical Science, University of São Paulo, São Paulo, 05508-900, Brazil
| | - Osvaldo Augusto Sant'Anna
- Laboratory of Immunochemistry, Butantan Institute, Av. Vital Brasil, 1500, 05503-900, São Paulo, SP, Brazil
| | - Luís Roberto C Gonçalves
- Laboratory of Pathophysiology, Butantan Institute, Av. Vital Brazil, 1500, 05503-900, São Paulo, SP, Brazil
| | - Yara Cury
- Laboratory of Pain and Signaling, Butantan Institute, Av. Vital Brazil, 1500, 05503-900, São Paulo, SP, Brazil
| | - Sandra C Sampaio
- Laboratory of Pathophysiology, Butantan Institute, Av. Vital Brazil, 1500, 05503-900, São Paulo, SP, Brazil.
| |
Collapse
|
8
|
Rood KM, Patel N, DeVengencie IM, Quinn JP, Gowdy KM, Costantine MM, Kniss DA. Aspirin modulates production of pro-inflammatory and pro-resolving mediators in endothelial cells. PLoS One 2023; 18:e0283163. [PMID: 37098090 PMCID: PMC10128936 DOI: 10.1371/journal.pone.0283163] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2022] [Accepted: 03/02/2023] [Indexed: 04/26/2023] Open
Abstract
Endothelial cells synthesize biochemical signals to coordinate a response to insults, resolve inflammation and restore barrier integrity. Vascular cells release a variety of vasoactive bioactive lipid metabolites during the inflammatory response and produce pro-resolving mediators (e.g., Lipoxin A4, LXA4) in cooperation with leukocytes and platelets to bring a halt to inflammation. Aspirin, used in a variety of cardiovascular and pro-thrombotic disorders (e.g., atherosclerosis, angina, preeclampsia), potently inhibits proinflammatory eicosanoid formation. Moreover, aspirin stimulates the synthesis of pro-resolving lipid mediators (SPM), so-called Aspirin-Triggered Lipoxins (ATL). We demonstrate that cytokines stimulated a time- and dose-dependent increase in PGI2 (6-ketoPGF1α) and PGE2 formation that is blocked by aspirin. Eicosanoid production was caused by cytokine-induced expression of cyclooxygenase-2 (COX-2). We also detected increased production of pro-resolving LXA4 in cytokine-stimulated endothelial cells. The R-enantiomer of LXA4, 15-epi-LXA4, was enhanced by aspirin, but only in the presence of cytokine challenge, indicating dependence on COX-2 expression. In contrast to previous reports, we detected arachidonate 5-lipoxygenase (ALOX5) mRNA expression and its cognate protein (5-lipoxygenase, 5-LOX), suggesting that endothelial cells possess the enzymatic machinery necessary to synthesize both pro-inflammatory and pro-resolving lipid mediators independent of added leukocytes or platelets. Finally, we observed that, endothelial cells produced LTB4 in the absence of leukocytes. These results indicate that endothelial cells produce both pro-inflammatory and pro-resolving lipid mediators in the absence of other cell types and aspirin exerts pleiotropic actions influencing both COX and LOX pathways.
Collapse
Affiliation(s)
- Kara M Rood
- Division of Maternal-Fetal Medicine, Department of Obstetrics and Gynecology, College of Medicine and Wexner Medical Center, The Ohio State University, Columbus, Ohio, United States of America
| | - Niharika Patel
- Division of Maternal-Fetal Medicine, Department of Obstetrics and Gynecology, Laboratory of Perinatal Research, College of Medicine and Wexner Medical Center, The Ohio State University, Columbus, Ohio, United States of America
| | - Ivana M DeVengencie
- Division of Maternal-Fetal Medicine, Department of Obstetrics and Gynecology, Laboratory of Perinatal Research, College of Medicine and Wexner Medical Center, The Ohio State University, Columbus, Ohio, United States of America
| | - John P Quinn
- Division of Maternal-Fetal Medicine, Department of Obstetrics and Gynecology, Laboratory of Perinatal Research, College of Medicine and Wexner Medical Center, The Ohio State University, Columbus, Ohio, United States of America
| | - Kymberly M Gowdy
- Division of Pulmonary, Critical Care and Sleep Medicine, Department of Internal Medicine, College of Medicine and Wexner Medical Center, Columbus, Ohio, United States of America
- Dorothy Davis Heart and Lung Institute, College of Medicine and Wexner Medical Center, The Ohio State University, Columbus, Ohio, United States of America
| | - Maged M Costantine
- Division of Maternal-Fetal Medicine, Department of Obstetrics and Gynecology, College of Medicine and Wexner Medical Center, The Ohio State University, Columbus, Ohio, United States of America
| | - Douglas A Kniss
- Division of Maternal-Fetal Medicine, Department of Obstetrics and Gynecology, College of Medicine and Wexner Medical Center, The Ohio State University, Columbus, Ohio, United States of America
- Division of Maternal-Fetal Medicine, Department of Obstetrics and Gynecology, Laboratory of Perinatal Research, College of Medicine and Wexner Medical Center, The Ohio State University, Columbus, Ohio, United States of America
- Department of Biomedical Engineering, College of Engineering, Fontana Labs, The Ohio State University, Columbus, Ohio, United States of America
- Infectious Disease Institute, The Ohio State University, Columbus, Ohio, United States of America
| |
Collapse
|
9
|
Hao H, Xie F, Xu F, Wang Q, Wu Y, Zhang D. LipoxinA 4 analog BML-111 protects podocytes cultured in high-glucose medium against oxidative injury via activating Nrf2 pathway. Int Immunopharmacol 2022; 111:109170. [PMID: 36007391 DOI: 10.1016/j.intimp.2022.109170] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2022] [Revised: 08/13/2022] [Accepted: 08/14/2022] [Indexed: 12/14/2022]
Abstract
Numerous studies have shown that the activation of the Nrf2 pathway alleviates oxidative stress and podocyte damage. Emerging evidence indicates that the dual anti-inflammatory and pro-resolution lipid mediator, lipoxin A4 (LXA4), has antioxidant activity. The aim of the present study was to confirm that BML-111, an analog of LXA4, prevents oxidative injury in diabetic podocytes via the regulation of the Nrf2 pathway. Here, we found that BML-111 inhibited high glucose (HG)-induced oxidative injury in the podocyte cell line, MPC5, in vitro, through activating Nrf2. Mechanistically, BML-111 significantly activated Nrf2 and its phase II enzymes, including Nqo1 and Ho-1. Moreover, BML-111 suppressed the migration of MPC5 cells. Additionally, BML-111 decreased the expression of Vcam, Icam and inflammatory cytokines (Il-1α, Il-6, and Tnf) in MPC5 cells. Importantly, BML-111 ameliorated blood glucose levels (approximately 75% of that in the SMZ group) and kidney damage by activating Nrf2, and its phase II enzymes, in diabetic mice. These effects are mainly mediated by Fpr2, a specific LXA4 receptor. Our findings demonstrate that BML-111 alleviates the injury of diabetic podocytes and kidneys by regulating the Nrf2 pathway.
Collapse
Affiliation(s)
- Hua Hao
- Department of Pathology, Yangpu Hospital, School of Medicine, Tongji University, Shanghai 200090, PR China
| | - Fangping Xie
- Department of Pathology, Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi 330006, PR China
| | - Fen Xu
- Medical Examination Center, Yangpu Hospital, School of Medicine, Tongji University, Shanghai 200090, PR China
| | - Qingyu Wang
- Department of Pathology, Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi 330006, PR China
| | - Yun Wu
- Department of Pathology, Yangpu Hospital, School of Medicine, Tongji University, Shanghai 200090, PR China
| | - Dongxin Zhang
- Microbiome Medicine Center, Division of Laboratory Medicine, Zhujiang Hospital, Southern Medical University, Guangzhou, Guangdong 510515, PR China.
| |
Collapse
|
10
|
Spahr A, Divnic‐Resnik T. Impact of health and lifestyle food supplements on periodontal tissues and health. Periodontol 2000 2022; 90:146-175. [PMID: 35916868 PMCID: PMC9804634 DOI: 10.1111/prd.12455] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
According to the new classification, periodontitis is defined as a chronic multifactorial inflammatory disease associated with dysbiotic biofilms and characterized by progressive destruction of the tooth-supporting apparatus. This definition, based on the current scientific evidence, clearly indicates and emphasizes, beside the microbial component dental biofilm, the importance of the inflammatory reaction in the progressive destruction of periodontal tissues. The idea to modulate this inflammatory reaction in order to decrease or even cease the progressive destruction was, therefore, a logical consequence. Attempts to achieve this goal involve various kinds of anti-inflammatory drugs or medications. However, there is also an increasing effort in using food supplements or so-called natural food ingredients to modulate patients' immune responses and maybe even improve the healing of periodontal tissues. The aim of this chapter of Periodontology 2000 is to review the evidence of various food supplements and ingredients regarding their possible effects on periodontal inflammation and wound healing. This review may help researchers and clinicians to evaluate the current evidence and to stimulate further research in this area.
Collapse
Affiliation(s)
- Axel Spahr
- Discipline of Periodontics, School of Dentistry, Faculty of Medicine and HealthThe University of SydneySydneyNew South WalesAustralia
| | - Tihana Divnic‐Resnik
- Discipline of Periodontics, School of Dentistry, Faculty of Medicine and HealthThe University of SydneySydneyNew South WalesAustralia
| |
Collapse
|
11
|
Effect of resolvin D1 on experimental bacterial keratitis to prevent corneal scar. Graefes Arch Clin Exp Ophthalmol 2022; 260:3293-3302. [PMID: 35522296 DOI: 10.1007/s00417-022-05686-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2022] [Revised: 04/14/2022] [Accepted: 04/22/2022] [Indexed: 11/04/2022] Open
Abstract
PURPOSE The study aims to investigate the role of the lipid mediator resolvin D1 (RvD1) in bacterial keratitis in a murine model. METHODS The effect of RvD1 on Pseudomonas aeruginosa-stimulated human corneal epithelial cells (HCECs) and mouse macrophages and dendritic cells (DCs) was assessed. C57BL/6 mouse corneas were abraded and treated with RvD1 after stimulation with P. aeruginosa, following which cytokine production level in the cornea and drainage lymph nodes was compared with that in controls. Corneal opacity and thickness were assessed using anterior segment photographs, and optical coherence tomography and corneal infiltrates were analyzed using immunohistochemistry for neutrophils. RESULTS RvD1 significantly inhibited pro-inflammatory cytokine production in HCECs, mouse macrophages, and DCs. Corneal opacity and corneal thickness were reduced, and the development of corneal infiltrates, specifically neutrophils, was also significantly inhibited by RvD1 in response to stimulation with P. aeruginosa. CONCLUSIONS RvD1 inhibits P. aeruginosa-induced corneal inflammation. This finding supports a potential therapeutic approach for patients with bacterial keratitis.
Collapse
|
12
|
Sundaram TS, Giromini C, Rebucci R, Pistl J, Bhide M, Baldi A. Role of omega-3 polyunsaturated fatty acids, citrus pectin, and milk-derived exosomes on intestinal barrier integrity and immunity in animals. J Anim Sci Biotechnol 2022; 13:40. [PMID: 35399093 PMCID: PMC8996583 DOI: 10.1186/s40104-022-00690-7] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2021] [Accepted: 02/07/2022] [Indexed: 11/10/2022] Open
Abstract
The gastrointestinal tract of livestock and poultry is prone to challenge by feedborne antigens, pathogens, and other stress factors in the farm environment. Excessive physiological inflammation and oxidative stress that arises firstly disrupts the intestinal epithelial barrier followed by other components of the gastrointestinal tract. In the present review, the interrelationship between intestinal barrier inflammation and oxidative stress that contributes to the pathogenesis of inflammatory bowel disease was described. Further, the role of naturally existing immunomodulatory nutrients such as the omega-3 polyunsaturated fatty acids, citrus pectin, and milk-derived exosomes in preventing intestinal barrier inflammation was discussed. Based on the existing evidence, the possible molecular mechanism of these bioactive nutrients in the intestinal barrier was outlined for application in animal diets.
Collapse
Affiliation(s)
- Tamil Selvi Sundaram
- Department of Veterinary Science for Health, Animal Production and Food Safety, University of Milan, Via Trentacoste 2, 20134, Milan, Italy.
- University of Veterinary Medicine and Pharmacy in Košice, Komenského 68/73, 04181, Košice, Slovakia.
| | - Carlotta Giromini
- Department of Veterinary Science for Health, Animal Production and Food Safety, University of Milan, Via Trentacoste 2, 20134, Milan, Italy
| | - Raffaella Rebucci
- Department of Veterinary Science for Health, Animal Production and Food Safety, University of Milan, Via Trentacoste 2, 20134, Milan, Italy
| | - Juraj Pistl
- University of Veterinary Medicine and Pharmacy in Košice, Komenského 68/73, 04181, Košice, Slovakia
| | - Mangesh Bhide
- University of Veterinary Medicine and Pharmacy in Košice, Komenského 68/73, 04181, Košice, Slovakia
| | - Antonella Baldi
- Department of Veterinary Science for Health, Animal Production and Food Safety, University of Milan, Via Trentacoste 2, 20134, Milan, Italy
| |
Collapse
|
13
|
Lipoxin A 4 and its analog attenuate high fat diet-induced atherosclerosis via Keap1/Nrf2 pathway. Exp Cell Res 2022; 412:113025. [PMID: 35026282 DOI: 10.1016/j.yexcr.2022.113025] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2021] [Revised: 01/03/2022] [Accepted: 01/09/2022] [Indexed: 01/07/2023]
Abstract
Excessive oxidative stress and decreased antioxidant capacity of macrophages are initial factors which cause macrophages to transform to foam cells, which represents a key event in the progression of atherosclerosis (AS). BML-111, the analog of lipoxin A4 (LXA4) strongly attenuated high fat (HF) diet-induced atherosclerosis by activating NF-E2 related factor 2 (Nrf2). However, the effect was not through a specific LXA4 receptor (formyl peptide receptor 2, FPR2). BML-111 also strongly inhibited HF diet-induced promotion of MDA level, increased HDL level and decreased IL-1, MCP-1, IL-6, VCAM, ICAM and TNF-α level in aorta. In the in vitro experiments, LXA4 inhibited THP-1 cells to transform to foam cells via Nrf2 pathway. Our findings demonstrated that LXA4 and its analog prevented AS induced by HF diet in SD rats, under which the possible mechanism is through Keap1/Nrf2 pathway.
Collapse
|
14
|
Han M, Lai S, Ge Y, Zhou X, Zhao J. Changes of Lipoxin A4 and the Anti-inflammatory Role During Parturition. Reprod Sci 2021; 29:1332-1342. [PMID: 34786659 DOI: 10.1007/s43032-021-00800-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2021] [Accepted: 11/06/2021] [Indexed: 11/29/2022]
Abstract
Parturition is the physiological process of newborn birth; more and more evidences show that parturition is closely related to the occurrence and resolution of inflammation. However, the inflammatory media and the mechanism are not very clear during parturition. Here, we investigate the inflammatory event during human parturition and in mouse model. We found that the pro-inflammatory cytokines (IL-6, IL-8, and IL-1β) and cells (neutrophil and macrophage) are decreased in pregnant women in labor and in mouse labor model. Mechanistically, increased stress stimulates the high-level adrenaline production in labor. Then, adrenaline upregulates the expression of 12/15-LOX (lipoxygenase) to produce more lipoxin A4 (LXA4), which is an inflammation inhibitor. Thus, LXA4 promotes the elimination of inflammation during labor to protect the body from excessive inflammatory damages. In addition, using BOC-2, the inhibitor of LXA4 receptor could reboot the pro-inflammatory cytokines. Our study indicates that LXA4 is induced by adrenaline in labor and appropriate interference of this pathway may be a potential strategy to regulate the inflammatory process in parturition.
Collapse
Affiliation(s)
- Mei Han
- Chongqing Key Laboratory of Translational Research for Cancer Metastasis and Individualized Treatment, Chongqing University Cancer Hospital, Chongqing, 400030, China
| | - Shaoyang Lai
- The Department of Obstetrics & Gynecology, Tongji Hospital, Tongji Medical College of Huazhong University of Science & Technology, Wuhan, 430030, Hubei Province, China
| | - Yimeng Ge
- Center for Reproductive Medicine, Department of Obstetrics and Gynecology, Peking University Third Hospital, 49 North Garden Rd., Haidian District, Beijing, 100191, China
| | - Xuan Zhou
- The Department of Obstetrics & Gynecology, Tongji Hospital, Tongji Medical College of Huazhong University of Science & Technology, Wuhan, 430030, Hubei Province, China
| | - Jie Zhao
- The Department of Obstetrics & Gynecology, Tongji Hospital, Tongji Medical College of Huazhong University of Science & Technology, Wuhan, 430030, Hubei Province, China. .,Center for Reproductive Medicine, Department of Obstetrics and Gynecology, Peking University Third Hospital, 49 North Garden Rd., Haidian District, Beijing, 100191, China. .,National Clinical Research Center for Obstetrics and Gynecology, Beijing, 100191, China. .,Key Laboratory of Assisted Reproduction (Peking University), Ministry of Education, Beijing, 100191, China. .,Beijing Key Laboratory of Reproductive Endocrinology and Assisted Reproductive Technology, Beijing, 100191, China.
| |
Collapse
|
15
|
Domínguez-Rivas E, Ávila-Muñoz E, Schwarzacher SW, Zepeda A. Adult hippocampal neurogenesis in the context of lipopolysaccharide-induced neuroinflammation: A molecular, cellular and behavioral review. Brain Behav Immun 2021; 97:286-302. [PMID: 34174334 DOI: 10.1016/j.bbi.2021.06.014] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/20/2020] [Revised: 06/17/2021] [Accepted: 06/19/2021] [Indexed: 12/17/2022] Open
Abstract
The continuous generation of new neurons occurs in at least two well-defined niches in the adult rodent brain. One of these areas is the subgranular zone of the dentate gyrus (DG) in the hippocampus. While the DG is associated with contextual and spatial learning and memory, hippocampal neurogenesis is necessary for pattern separation. Hippocampal neurogenesis begins with the activation of neural stem cells and culminates with the maturation and functional integration of a portion of the newly generated glutamatergic neurons into the hippocampal circuits. The neurogenic process is continuously modulated by intrinsic factors, one of which is neuroinflammation. The administration of lipopolysaccharide (LPS) has been widely used as a model of neuroinflammation and has yielded a body of evidence for unveiling the detrimental impact of inflammation upon the neurogenic process. This work aims to provide a comprehensive overview of the current knowledge on the effects of the systemic and central administration of LPS upon the different stages of neurogenesis and discuss their effects at the molecular, cellular, and behavioral levels.
Collapse
Affiliation(s)
- Eduardo Domínguez-Rivas
- Departamento de Medicina Genómica y Toxicología Ambiental, Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México, Mexico City, Mexico
| | - Evangelina Ávila-Muñoz
- Departamento de Medicina Genómica y Toxicología Ambiental, Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México, Mexico City, Mexico
| | - Stephan W Schwarzacher
- Institute of Clinical Neuroanatomy, Neuroscience Center, Goethe University Frankfurt, Frankfurt am Main, Germany
| | - Angélica Zepeda
- Departamento de Medicina Genómica y Toxicología Ambiental, Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México, Mexico City, Mexico; Institute of Clinical Neuroanatomy, Neuroscience Center, Goethe University Frankfurt, Frankfurt am Main, Germany.
| |
Collapse
|
16
|
Zhao S, Xu Z. Lipoxin A4 inhibits the development of endometriosis in a mouse model by suppressing local estradiol synthesis. Prostaglandins Other Lipid Mediat 2021; 153:106521. [PMID: 33279654 DOI: 10.1016/j.prostaglandins.2020.106521] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2020] [Revised: 11/12/2020] [Accepted: 11/30/2020] [Indexed: 10/22/2022]
Abstract
We investigated whether lipoxin A4 (LXA4) inhibits the development of endometriosis by suppressing local estradiol synthesis. An endometriosis mouse model was constructed by surgical transplantation to subcutanous tissue sites. The treatment group received daily injections of LXA4 (10 μg/Kg) for 21days after which lesions were recovered. We measured 17β-HSD1, 17β-HSD2, CYP11A1, CYP19A1, CYP17A1, and estrogen receptor mRNA expression levels using real-time RT-PCR. In addition, immunohistochemistry was performed to determine protein expression and localization. After LXA4 administration, the volume of endometrial lesions was significantly reduced. Administration of LXA4 resulted in a more rudimentary architecture with a reduced number of developed glands surrounded by a small amount of stroma. LXA4 downregulated the mRNA and protein expression levels of 17β-HSD1, CYP11A1, CYP19A1, CYP17A1, ERα, and ERβ. Furthermore, LXA4 downregulated the expression of ERβ, aromatase expression, and 17β-HSD1 enzyme activity, which affected local estradiol production, resulting in reduced endometriosis. Results from our endometriosis mouse model showed that treatment with LXA4 reduced expression of enzymes and receptors associated or implicated with estrogen-dependent regulation of extra-uterine tissue. We believe that LXA4 has a potential therapeutic value for the treatment of endometriosis.
Collapse
Affiliation(s)
- Shenzhi Zhao
- Wenzhou Medical University, Wenzhou, 325027, People's Republic of China
| | - Zhangye Xu
- Department of Gynecology and Obstetrics, The Second Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325027, People's Republic of China.
| |
Collapse
|
17
|
Effect of Omega-3 on Recurrent Aphthous Stomatitis and Improvement Quality of Life. Int J Dent 2021; 2021:6617575. [PMID: 33628246 PMCID: PMC7889386 DOI: 10.1155/2021/6617575] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2020] [Revised: 01/17/2021] [Accepted: 01/27/2021] [Indexed: 11/18/2022] Open
Abstract
Objective Recurrent aphthous stomatitis is one of the most common chronic inflammatory diseases in oral mucosa. Beneficial effects of omega-3 supplements on some inflammatory diseases have been proved. The aim of present study was to evaluate the effect of omega-3 supplements in recurrent aphthous stomatitis management and improve oral health-related quality of life. Methods In this double-blind clinical trial, 40 patients with minor recurrent aphthous stomatitis were randomly divided into case and control groups. The case group received 1000 mg capsules of omega-3, while the control group received placebo capsules for 6 months. The questionnaires of the ulcer severity score and the chronic oral mucosal disease questionnaire were filled by the patients in three steps, at the baseline session, after 3 months, and after 6 months. The data were analyzed by SPSS 22 software through ANOVA, Mann-Whitney, and chi-square tests. P < 0.05 was considered as significant. Results In the omega-3 group, the ulcer severity score showed significant reduction by three-month and six-month follow-ups (P < 0.0001, P < 0.0001, respectively). The mean score of the chronic oral mucosal disease questionnaire significantly improved by three-month and six-month follow-ups in the omega-3-receiving group. Conclusion Use of omega-3 oral supplements decreased the severity of aphthous ulcer and improved oral health-related quality of life.
Collapse
|
18
|
Cioccari L, Luethi N, Masoodi M. Lipid Mediators in Critically Ill Patients: A Step Towards Precision Medicine. Front Immunol 2020; 11:599853. [PMID: 33324417 PMCID: PMC7724037 DOI: 10.3389/fimmu.2020.599853] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2020] [Accepted: 10/26/2020] [Indexed: 12/15/2022] Open
Abstract
A dysregulated response to systemic inflammation is a common pathophysiological feature of most conditions encountered in the intensive care unit (ICU). Recent evidence indicates that a dysregulated inflammatory response is involved in the pathogenesis of various ICU-related disorders associated with high mortality, including sepsis, acute respiratory distress syndrome, cerebral and myocardial ischemia, and acute kidney injury. Moreover, persistent or non-resolving inflammation may lead to the syndrome of persistent critical illness, characterized by acquired immunosuppression, catabolism and poor long-term functional outcomes. Despite decades of research, management of many disorders in the ICU is mostly supportive, and current therapeutic strategies often do not take into account the heterogeneity of the patient population, underlying chronic conditions, nor the individual state of the immune response. Fatty acid-derived lipid mediators are recognized as key players in the generation and resolution of inflammation, and their signature provides specific information on patients' inflammatory status and immune response. Lipidomics is increasingly recognized as a powerful tool to assess lipid metabolism and the interaction between metabolic changes and the immune system via profiling lipid mediators in clinical studies. Within the concept of precision medicine, understanding and characterizing the individual immune response may allow for better stratification of critically ill patients as well as identification of diagnostic and prognostic biomarkers. In this review, we provide an overview of the role of fatty acid-derived lipid mediators as endogenous regulators of the inflammatory, anti-inflammatory and pro-resolving response and future directions for use of clinical lipidomics to identify lipid mediators as diagnostic and prognostic markers in critical illness.
Collapse
Affiliation(s)
- Luca Cioccari
- Department of Intensive Care Medicine, Inselspital, Bern University Hospital, Bern, Switzerland.,Australian and New Zealand Intensive Care Research Centre, School of Public Health and Preventive Medicine, Monash University, Prahran, VIC, Australia
| | - Nora Luethi
- Australian and New Zealand Intensive Care Research Centre, School of Public Health and Preventive Medicine, Monash University, Prahran, VIC, Australia.,Department of Emergency Medicine, Inselspital, Bern University Hospital, Bern, Switzerland
| | - Mojgan Masoodi
- Institute of Clinical Chemistry, Inselspital, Bern University Hospital, Bern, Switzerland
| |
Collapse
|
19
|
Abstract
Resolvins, belonging to the group of specialized proresolving mediators (SPMs), are metabolic products of omega-3 polyunsaturated fatty acids (ω-3 PUFAs) and are synthesized during the initial phases of acute inflammatory responses to promote the resolution of inflammation. Resolvins are produced for termination of neutrophil infiltration, stimulation of the clearance of apoptotic cells by macrophages, and promotion of tissue remodeling and homeostasis. Metabolic dysregulation due to either uncontrolled activity of pro-inflammatory responses or to inefficient resolution of inflammation results in chronic inflammation and may also lead to atherosclerosis or other chronic autoimmune diseases such as rheumatoid arthritis, psoriasis, systemic lupus erythematosus, vasculitis, inflammatory bowel diseases, and type 1 diabetes mellitus. The pathogenesis of such diseases involves a complex interplay between the immune system and, environmental factors (non-infectious or infectious), and critically depends on individual susceptibility to such factors. In the present review, resolvins and their roles in the resolution of inflammation, as well as the role of these mediators as potential therapeutic agents to counteract specific chronic autoimmune and inflammatory diseases are discussed.
Collapse
|
20
|
Boff D, Oliveira VLS, Queiroz Junior CM, Galvão I, Batista NV, Gouwy M, Menezes GB, Cunha TM, Verri Junior WA, Proost P, Teixeira MM, Amaral FA. Lipoxin A 4 impairs effective bacterial control and potentiates joint inflammation and damage caused by Staphylococcus aureus infection. FASEB J 2020; 34:11498-11510. [PMID: 32741032 DOI: 10.1096/fj.201802830rr] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2018] [Revised: 05/12/2020] [Accepted: 05/15/2020] [Indexed: 02/07/2023]
Abstract
Staphylococcus aureus is the main cause of septic arthritis in humans, a disease associated with high morbidity and mortality. Inflammation triggered in response to infection is fundamental to control bacterial growth but may cause permanent tissue damage. Here, we evaluated the role of Lipoxin A4 (LXA4 ) in S aureus-induced arthritis in mice. Septic arthritis was induced by S aureus injection into tibiofemoral joints. At different time points, we evaluated cell recruitment and bacterial load in the joint, the production of pro-inflammatory molecules, and LXA4 in inflamed tissue and analyzed joint damage and dysfunction. LXA4 was investigated using genetically modified mice or by pharmacological blockade of its synthesis and receptor. CD11c+ cells were evaluated in lymph nodes by confocal microscopy and flow cytometry and dendritic cell chemotaxis using the Boyden chamber. Absence or pharmacological blockade of 5-lipoxygenase (5-LO) reduced joint inflammation and dysfunction and was associated with better control of infection at 4 to 7 days after the infection. There was an increase in LXA4 in joints of S aureus-infected mice and administration of LXA4 reversed the phenotype in 5-LO-/- mice. Blockade or absence of the LXA4 receptor FPR2 has a phenotype similar to 5-LO-/- mice. Mechanistically, LXA4 appeared to control migration and function of dendritic cells, cells shown to be crucial for adequate protective responses in the model. Thus, after the first days of infection when symptoms become evident therapies that inhibit LXA4 synthesis or action could be useful for treatment of S aureus-induced arthritis.
Collapse
Affiliation(s)
- Daiane Boff
- Immunopharmacology, Department of Biochemistry and Immunology, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil.,Laboratory of Molecular Immunology, Department of Microbiology and Immunology, Rega Institute for Medical Research, KU Leuven, Leuven, Belgium
| | - Vivian Louise Soares Oliveira
- Immunopharmacology, Department of Biochemistry and Immunology, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Celso M Queiroz Junior
- Department of Morphology, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Izabela Galvão
- Immunopharmacology, Department of Biochemistry and Immunology, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Nathalia Vieira Batista
- Immunopharmacology, Department of Biochemistry and Immunology, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Mieke Gouwy
- Laboratory of Molecular Immunology, Department of Microbiology and Immunology, Rega Institute for Medical Research, KU Leuven, Leuven, Belgium
| | - Gustavo Batista Menezes
- Departamento de Morfologia, Centro de Biologia Gastrointestinal, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Thiago Mattar Cunha
- Department of Pharmacology, Faculdade de Medicina de Ribeirao Preto, Center for Research in Inflammatory Diseases, Universidade de São Paulo, São Paulo, Brazil
| | | | - Paul Proost
- Laboratory of Molecular Immunology, Department of Microbiology and Immunology, Rega Institute for Medical Research, KU Leuven, Leuven, Belgium
| | - Mauro Martins Teixeira
- Immunopharmacology, Department of Biochemistry and Immunology, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Flávio Almeida Amaral
- Immunopharmacology, Department of Biochemistry and Immunology, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| |
Collapse
|
21
|
Zhang Y, Eser BE, Kristensen P, Guo Z. Fatty acid hydratase for value-added biotransformation: A review. Chin J Chem Eng 2020. [DOI: 10.1016/j.cjche.2020.02.008] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
|
22
|
Kurtoğlu EL, Kayhan B, Gül M, Kayhan B, Akdoğan Kayhan M, Karaca ZM, Yeşilada E, Yılmaz S. A bioactive product lipoxin A4 attenuates liver fibrosis in an experimental model by regulating immune response and modulating the expression of regeneration genes. TURKISH JOURNAL OF GASTROENTEROLOGY 2020; 30:745-757. [PMID: 31418419 DOI: 10.5152/tjg.2019.18276] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
BACKGROUND/AIMS Lipoxin A4 (LXA4), an anti-inflammatory lipid mediator, regulates leukocyte cellular activity and activates gene transcription. The therapeutic effect of LXA4 on liver fibrosis and its mechanism on the immune system are largely unknown. Because the regenerative capacity of hepatocytes in acute and chronic liver failure models of mouse increases by silencing MKK4, we aimed to investigate the effect of parenteral administration of LXA4 on the genes responsible for regeneration of liver, namely MKK4, MKK7, and ATF2, and visualize the therapeutic effects in an experimental model. MATERIALS AND METHODS Fibrosis was induced in mice by administration of thioacetamide (TAA). LXA4 was administered during the last two weeks of fibrosis induction. The fibrosis level was measured by Knodell scoring. The liver function was measured by analyzing serum ALT, AST, and AP levels. Expression levels of genes responsible for liver fibrosis (TGF-α) and cell regeneration (MKK4, MKK7, and ATF2) have been measured by RT-PCR analysis. Inflammatory and anti-inflammatory cytokine levels were measured in serum samples and liver homogenates by Enzyme Linked Immunosorbent Assay (ELISA). Ultrathin sections were examined using a transmission electron microscope and analyzed. RESULTS We observed significant healing in liver of the LXA4-treated group, histologically. This finding was in parallel with reduction of serum ALT, AST, but not AP levels. TGF-α and MKK4 expressions were significantly reduced in the LXA4-treated group. Administration of LXA4 caused significant elevation of IL-10 in systemic circulation; however, that elevation was not detected in liver homogenates. Nevertheless, significant reductions in TNF-α and IL-17 have been observed. CONCLUSION The anti-inflammatory effect of LXA4 maintains the regenerative capacity of liver during fibrosis in an experimental liver fibrosis model. LXA4 may be therapeutically beneficial in liver fibrosis.
Collapse
Affiliation(s)
- Elçin Latife Kurtoğlu
- Department of Medical Biology and Genetics, İnönü University School of Medicine, Malatya, Turkey
| | - Başak Kayhan
- Department of Medical Biology and Genetics, İnönü University School of Medicine, Malatya, Turkey;Transplantation Immunology Laboratory, Department of General Surgery, Institute of Liver Transplantation, İnönü University School of Medicine, Malatya, Turkey
| | - Mehmet Gül
- Department of Histology and Embryology, İnönü University School of Medicine, Malatya, Turkey
| | - Burçak Kayhan
- Division of Gastroenterology, Department of Internal Medicine, Karabük University School of Medicine, Karabük, Turkey
| | - Meral Akdoğan Kayhan
- Division of Gastroenterology, Department of Internal Medicine, Abant İzzet Baysal School of Medicine, Bolu, Turkey
| | - Zeynal Mete Karaca
- Department of Medical Biology and Genetics, İnönü University School of Medicine, Malatya, Turkey
| | - Elif Yeşilada
- Department of Medical Biology and Genetics, İnönü University School of Medicine, Malatya, Turkey
| | - Sezai Yılmaz
- Department of Surgery, Liver Transplantation Institute, İnönü University School of Medicine, Malatya, Turkey
| |
Collapse
|
23
|
MacKnight HP, Stephenson DJ, Hoeferlin LA, Benusa SD, DeLigio JT, Maus KD, Ali AN, Wayne JS, Park MA, Hinchcliffe EH, Brown RE, Ryan JJ, Diegelmann RF, Chalfant CE. The interaction of ceramide 1-phosphate with group IVA cytosolic phospholipase A 2 coordinates acute wound healing and repair. Sci Signal 2019; 12:12/610/eaav5918. [PMID: 31796632 DOI: 10.1126/scisignal.aav5918] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
The sphingolipid ceramide 1-phosphate (C1P) directly binds to and activates group IVA cytosolic phospholipase A2 (cPLA2α) to stimulate the production of eicosanoids. Because eicosanoids are important in wound healing, we examined the repair of skin wounds in knockout (KO) mice lacking cPLA2α and in knock-in (KI) mice in which endogenous cPLA2α was replaced with a mutant form having an ablated C1P interaction site. Wound closure rate was not affected in the KO or KI mice, but wound maturation was enhanced in the KI mice compared to that in wild-type controls. Wounds in KI mice displayed increased infiltration of dermal fibroblasts into the wound environment, increased wound tensile strength, and a higher ratio of type I:type III collagen. In vitro, primary dermal fibroblasts (pDFs) from KI mice showed substantially increased collagen deposition and migration velocity compared to pDFs from wild-type and KO mice. KI mice also showed an altered eicosanoid profile of reduced proinflammatory prostaglandins (PGE2 and TXB2) and an increased abundance of certain hydroxyeicosatetraenoic acid (HETE) species. Specifically, an increase in 5-HETE enhanced dermal fibroblast migration and collagen deposition. This gain-of-function role for the mutant cPLA2α was also linked to the relocalization of cPLA2α and 5-HETE biosynthetic enzymes to the cytoplasm and cytoplasmic vesicles. These findings demonstrate the regulation of key wound-healing mechanisms in vivo by a defined protein-lipid interaction and provide insights into the roles that cPLA2α and eicosanoids play in orchestrating wound repair.
Collapse
Affiliation(s)
- H Patrick MacKnight
- Department of Cell Biology, Microbiology, and Molecular Biology, University of South Florida, Tampa, FL 33620, USA.,Department of Biochemistry and Molecular Biology, Virginia Commonwealth University, Richmond, VA 23298, USA
| | - Daniel J Stephenson
- Department of Cell Biology, Microbiology, and Molecular Biology, University of South Florida, Tampa, FL 33620, USA.,Department of Biochemistry and Molecular Biology, Virginia Commonwealth University, Richmond, VA 23298, USA
| | - L Alexis Hoeferlin
- Department of Biochemistry and Molecular Biology, Virginia Commonwealth University, Richmond, VA 23298, USA
| | - Savannah D Benusa
- Department of Anatomy and Neurobiology, Virginia Commonwealth University, Richmond, Virginia, VA 23298, USA
| | - James T DeLigio
- Department of Cell Biology, Microbiology, and Molecular Biology, University of South Florida, Tampa, FL 33620, USA.,Department of Biochemistry and Molecular Biology, Virginia Commonwealth University, Richmond, VA 23298, USA
| | - Kenneth D Maus
- Department of Cell Biology, Microbiology, and Molecular Biology, University of South Florida, Tampa, FL 33620, USA
| | - Anika N Ali
- Department of Cell Biology, Microbiology, and Molecular Biology, University of South Florida, Tampa, FL 33620, USA
| | - Jennifer S Wayne
- Department of Biomedical Engineering, Virginia Commonwealth University, Richmond, VA 23284, USA
| | - Margaret A Park
- Department of Cell Biology, Microbiology, and Molecular Biology, University of South Florida, Tampa, FL 33620, USA.,Department of Biochemistry and Molecular Biology, Virginia Commonwealth University, Richmond, VA 23298, USA.,Moffitt Cancer Center, Tampa, FL 33620, USA
| | | | | | - John J Ryan
- Department of Biology, Virginia Commonwealth University, Richmond, VA 23298, USA
| | - Robert F Diegelmann
- Department of Biochemistry and Molecular Biology, Virginia Commonwealth University, Richmond, VA 23298, USA
| | - Charles E Chalfant
- Department of Cell Biology, Microbiology, and Molecular Biology, University of South Florida, Tampa, FL 33620, USA. .,Department of Biochemistry and Molecular Biology, Virginia Commonwealth University, Richmond, VA 23298, USA.,Moffitt Cancer Center, Tampa, FL 33620, USA.,Research Service, James A. Haley Veterans Hospital, Tampa, FL 33612, USA
| |
Collapse
|
24
|
Davies CL, Patir A, McColl BW. Myeloid Cell and Transcriptome Signatures Associated With Inflammation Resolution in a Model of Self-Limiting Acute Brain Inflammation. Front Immunol 2019; 10:1048. [PMID: 31156629 PMCID: PMC6533855 DOI: 10.3389/fimmu.2019.01048] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2018] [Accepted: 04/24/2019] [Indexed: 12/17/2022] Open
Abstract
Inflammation contributes to tissue repair and restoration of function after infection or injury. However, some forms of inflammation can cause tissue damage and disease, particularly if inappropriately activated, excessive, or not resolved adequately. The mechanisms that prevent excessive or chronic inflammation are therefore important to understand. This is particularly important in the central nervous system where some effects of inflammation can have particularly harmful consequences, including irreversible damage. An increasing number of neurological disorders, both acute and chronic, and their complications are associated with aberrant neuroinflammatory activity. Here we describe a model of self-limiting acute brain inflammation optimized to study mechanisms underlying inflammation resolution. Inflammation was induced by intracerebral injection of lipopolysaccharide (LPS) and the temporal profile of key cellular and molecular changes were defined during the progression of the inflammatory response. The kinetics of accumulation and loss of neutrophils in the brain enabled well-demarcated phases of inflammation to be operatively defined, including induction and resolution phases. Microglial reactivity and accumulation of monocyte-derived macrophages were maximal at the onset of and during the resolution phase. We profiled the transcriptome-wide gene expression changes at representative induction and resolution timepoints and used gene coexpression network analysis to identify gene clusters. This revealed a distinct cluster of genes associated with inflammation resolution that were induced selectively or maximally during this phase and indicated an active programming of gene expression that may drive resolution as has been described in other tissues. Induction of gene networks involved in lysosomal function, lipid metabolism, and a comparative switch to MHC-II antigen presentation (relative to MHC-I during induction) were prominent during the resolution phase. The restoration and/or further induction of microglial homeostatic signature genes was notable during the resolution phase. We propose the current model as a tractable reductionist system to complement more complex models for further understanding how inflammation resolution in the brain is regulated and as a platform for in vivo testing/screening of candidate resolution-modifying interventions. Our data highlight how resolution involves active cellular and transcriptome reprogramming and identify candidate gene networks associated with resolution-phase adaptations that warrant further study.
Collapse
Affiliation(s)
- Claire L. Davies
- Centre for Reproductive Health, The Queen's Medical Research Institute, University of Edinburgh, Edinburgh, United Kingdom
| | - Anirudh Patir
- The Roslin Institute, University of Edinburgh, Easter Bush, Midlothian, United Kingdom
| | - Barry W. McColl
- Centre for Discovery Brain Sciences, UK Dementia Research Institute at The University of Edinburgh, The Chancellor's Building, Edinburgh BioQuarter, Edinburgh, United Kingdom
| |
Collapse
|
25
|
Xu Z, Zhao S, Zhou T, Liao T, Huang X, Xiang H, Zhang Q, Huang Y, Lin F, Ye D, Huang Y. Lipoxin A4 interferes with embryo implantation via suppression of epithelial-mesenchymal transition. Am J Reprod Immunol 2019; 81:e13107. [PMID: 30811719 DOI: 10.1111/aji.13107] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2018] [Revised: 01/24/2019] [Accepted: 01/31/2019] [Indexed: 12/11/2022] Open
Abstract
PROBLEM To test whether lipoxin A4 (LXA4) interferes with embryo implantation via suppression of epithelial-mesenchymal transition (EMT). METHOD OF STUDY We developed a mouse model of LXA4 blocking embryo implantation and detected the indicators of EMT to confirm that LXA4 inhibits EMT might be a mechanism of interfering with the embryo implantation. We detected integrin-linked kinase (ILK), N-formylpeptide receptor 2 (FPR2), vascular endothelial growth factor, matrix metalloproteinases (MMPs), Akt, GSK3β, NF-ĸB, twist, vimentin, fibronectin, and β-catenin mRNA expression using reverse transcriptase-polymerase chain reaction (RT-PCR) and real-time RT-PCR; localized protein expression using immunohistochemistry and Western blotting assay; MMPs activity assay by gelatin zymography; and the status of implantation in pregnant animals assessed by pontamine blue reaction test. RESULTS Preimplantation administration of LXA4 resulted in implantation failure. LXA4 has a time- and dose-dependent effect on embryo implantation. Day 0.5 after fertilization is the most effective time to use LXA4 to block embryo implantation. (a) LXA4 reduced endometrial stroma edema; (b) LXA4 inhibited the activity of MMP9 and significantly upregulated the expression of β-catenin, and downregulated the expression of vimentin, fibronectin, twist, NF-κB, Akt, and Gsk-3β in the endometrium and TEV-1 cells; (c) LXA4 upregulated the expression of FPR2, and downregulated the expression of ILK; FPR2-overexpressing had an inhibitory effect on ILK in TEV-1 cells. CONCLUSION LXA4 inhibits EMT which attenuates ILK action by enhancing FPR2; therefore, this might be a mechanism of interfering with embryo implantation.
Collapse
Affiliation(s)
- Zhangye Xu
- Department of Gynecology and Obstetrics, The Second Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Shenzhi Zhao
- Department of Gynecology and Obstetrics, The Second Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Tong Zhou
- Department of Gynecology and Obstetrics, The Second Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Tingting Liao
- Department of Gynecology and Obstetrics, The Second Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Xianping Huang
- Department of Gynecology and Obstetrics, The Second Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Huiqiu Xiang
- Department of Gynecology and Obstetrics, The Second Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Qiong Zhang
- Department of Gynecology and Obstetrics, The Second Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Yanjun Huang
- Department of Gynecology and Obstetrics, The First Affiliated Hospital, Wenzhou Medical College, Wenzhou, China
| | - Feng Lin
- Department of Gynecology and Obstetrics, The First Affiliated Hospital, Wenzhou Medical College, Wenzhou, China
| | - Duyun Ye
- Department of Pathophysiology, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yinping Huang
- Department of Gynecology and Obstetrics, The First Affiliated Hospital, Wenzhou Medical College, Wenzhou, China
| |
Collapse
|
26
|
Sima C, Viniegra A, Glogauer M. Macrophage immunomodulation in chronic osteolytic diseases-the case of periodontitis. J Leukoc Biol 2019; 105:473-487. [PMID: 30452781 PMCID: PMC6386606 DOI: 10.1002/jlb.1ru0818-310r] [Citation(s) in RCA: 61] [Impact Index Per Article: 12.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2018] [Revised: 10/26/2018] [Accepted: 10/29/2018] [Indexed: 12/12/2022] Open
Abstract
Periodontitis (PD) is a chronic osteolytic disease that shares pathogenic inflammatory features with other conditions associated with nonresolving inflammation. A hallmark of PD is inflammation-mediated alveolar bone loss. Myeloid cells, in particular polymorphonuclear neutrophils (PMN) and macrophages (Mac), are essential players in PD by control of gingival biofilm pathogenicity, activation of adaptive immunity, as well as nonresolving inflammation and collateral tissue damage. Despite mounting evidence of significant innate immune implications to PD progression and healing after therapy, myeloid cell markers and targets for immune modulation have not been validated for clinical use. The remarkable plasticity of monocytes/Mac in response to local activation factors enables these cells to play central roles in inflammation and restoration of tissue homeostasis and provides opportunities for biomarker and therapeutic target discovery for management of chronic inflammatory conditions, including osteolytic diseases such as PD and arthritis. Along a wide spectrum of activation states ranging from proinflammatory to pro-resolving, Macs respond to environmental changes in a site-specific manner in virtually all tissues. This review summarizes the existing evidence on Mac immunomodulation therapies for osteolytic diseases in the broader context of conditions associated with nonresolving inflammation, and discusses osteoimmune implications of Macs in PD.
Collapse
Affiliation(s)
- Corneliu Sima
- Department of Oral Medicine, Infection, and Immunity, Harvard School of Dental Medicine, Harvard Medical School, Boston, Massachusetts, USA
| | - Ana Viniegra
- Dental Research Institute, Faculty of Dentistry, University of Toronto, Toronto, ON, Canada
| | - Michael Glogauer
- Dental Research Institute, Faculty of Dentistry, University of Toronto, Toronto, ON, Canada
| |
Collapse
|
27
|
Thatcher TH, Woeller CF, McCarthy CE, Sime PJ. Quenching the fires: Pro-resolving mediators, air pollution, and smoking. Pharmacol Ther 2019; 197:212-224. [PMID: 30759375 DOI: 10.1016/j.pharmthera.2019.02.001] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Exposure to air pollution and other environmental inhalation hazards, such as occupational exposures to dusts and fumes, aeroallergens, and tobacco smoke, is a significant cause of chronic lung inflammation leading to respiratory disease. It is now recognized that resolution of inflammation is an active process controlled by a novel family of small lipid mediators termed "specialized pro-resolving mediators" or SPMs, derived mainly from dietary omega-3 polyunsaturated fatty acids. Chronic inflammation results from an imbalance between pro-inflammatory and pro-resolution pathways. Research is ongoing to develop SPMs, and the pro-resolution pathway more generally, as a novel therapeutic approach to diseases characterized by chronic inflammation. Here, we will review evidence that the resolution pathway is dysregulated in chronic lung inflammatory diseases, and that SPMs and related molecules have exciting therapeutic potential to reverse or prevent chronic lung inflammation, with a focus on lung inflammation due to inhalation of environmental hazards including urban particulate matter, organic dusts and tobacco smoke.
Collapse
Affiliation(s)
- Thomas H Thatcher
- Division of Pulmonary and Critical Care Medicine, University of Rochester School of Medicine and Dentistry Rochester, NY 14642, United States; Lung Biology and Disease Program, University of Rochester School of Medicine and Dentistry, Rochester, NY 14642, United States
| | - Collynn F Woeller
- Lung Biology and Disease Program, University of Rochester School of Medicine and Dentistry, Rochester, NY 14642, United States; Department of Environmental Medicine, University of Rochester School of Medicine and Dentistry, Rochester, NY 14642, United States
| | - Claire E McCarthy
- National Cancer Institute, Division of Cancer Biology, 9609 Medical Center Drive, Rockville, MD 20850, United States
| | - Patricia J Sime
- Division of Pulmonary and Critical Care Medicine, University of Rochester School of Medicine and Dentistry Rochester, NY 14642, United States; Lung Biology and Disease Program, University of Rochester School of Medicine and Dentistry, Rochester, NY 14642, United States; Department of Environmental Medicine, University of Rochester School of Medicine and Dentistry, Rochester, NY 14642, United States.
| |
Collapse
|
28
|
Omega-3 Fatty Acid Supplementation, Pro-Resolving Mediators, and Clinical Outcomes in Maternal-Infant Pairs. Nutrients 2019; 11:nu11010098. [PMID: 30621269 PMCID: PMC6356980 DOI: 10.3390/nu11010098] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2018] [Revised: 12/21/2018] [Accepted: 01/03/2019] [Indexed: 12/31/2022] Open
Abstract
Omega (n)-3 fatty acids are vital to neonatal maturation, and recent investigations reveal n-3 fatty acids serve as substrates for the biosynthesis of specialized pro-resolving lipid mediators (SPM) that have anti-inflammatory and immune-stimulating effects. The role SPM play in the protection against negative maternal-fetal health outcomes is unclear, and there are no current biomarkers of n-3 fatty acid sufficiency. We sought to ascertain the relationships between n-3 fatty acid intake, SPM levels, and maternal-fetal health outcomes. We obtained n-3 fatty acid intake information from 136 mothers admitted for delivery using a food frequency questionnaire and measured docosahexaenoic acid (DHA)-derived SPMs resolvin D1 (RvD1) and RvD2 in maternal and cord plasma. We found significantly elevated SPM in maternal versus cord plasma, and increased SPM levels were associated with at-risk outcomes. We also identified that increased DHA intake was associated with elevated maternal plasma RvD1 (p = 0.03; R² = 0.18) and RvD2 (p = 0.04; R² = 0.20) in the setting of neonatal intensive care unit (NICU) admission. These findings indicate that increased n-3 fatty acid intake may provide increased substrate for the production of SPM during high-risk pregnancy/delivery conditions, and that increased maternal plasma SPM could serve as a biomarker for negative neonatal outcomes.
Collapse
|
29
|
Jarosz AC, Badawi A. Metabolites of prostaglandin synthases as potential biomarkers of Lyme disease severity and symptom resolution. Inflamm Res 2018; 68:7-17. [PMID: 30121835 PMCID: PMC6314976 DOI: 10.1007/s00011-018-1180-5] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2018] [Accepted: 08/08/2018] [Indexed: 01/09/2023] Open
Abstract
Background Lyme disease or Lyme borreliosis (LB) is the commonest vector-borne disease in the North America. It is an inflammatory disease caused by the bacterium Borrelia burgdorferi. The role of the inflammatory processes mediated by prostaglandins (PGs), thromboxanes and leukotrienes (LTs) in LB severity and symptoms resolution is yet to be elucidated. Objectives We aim to systematically review and evaluate the role of PGs and related lipid mediators in the induction and resolution of inflammation in LB. Methods We conducted a comprehensive search in PubMed, Ovid MEDLINE(R), Embase and Embase Classic to identify cell-culture, animal and human studies reporting the changes in PGs and related lipid mediators of inflammation during the course of LB. Results We identified 18 studies to be included into this systematic review. The selected reports consisted of seven cell-culture studies, seven animal studies, and four human studies (from three patient populations). Results from cell-culture and animal studies suggest that PGs and other lipid mediators of inflammation are elevated in LB and may contribute to disease development. The limited number of human studies showed that subjects with Lyme meningitis, Lyme arthritis (LA) and antibiotic-refractory LA had increased levels of an array of PGs and lipid mediators (e.g., LTB4, 8-isoPGF2α, and phospholipases A2 activity). Levels of these markers were significantly reduced following the treatment with antibiotics or non-steroidal anti-inflammatory drugs. Conclusion Dysregulation of prostaglandins and related lipid mediators may play a role in the etiology of LB and persistence of inflammation that may lead to long-term complications. Further investigation into the precise levels of a wide range of PGs and related factors is critical as it may propose novel markers that can be used for early diagnosis. Electronic supplementary material The online version of this article (10.1007/s00011-018-1180-5) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Alicia Caroline Jarosz
- Department of Nutritional Sciences, Faculty of Medicine, University of Toronto, FitzGerald Building, 150 College Street, Toronto, ON, M5S3E2, Canada
| | - Alaa Badawi
- Department of Nutritional Sciences, Faculty of Medicine, University of Toronto, FitzGerald Building, 150 College Street, Toronto, ON, M5S3E2, Canada.
- Public Health Risk Sciences Division, Public Health Agency of Canada, 180 Queen Street West, Toronto, ON, M5V 3L7, Canada.
| |
Collapse
|
30
|
de Medeiros MCS, Medeiros JCA, de Medeiros HJ, Leitão JCGDC, Knackfuss MI. Dietary intervention and health in patients with systemic lupus erythematosus: A systematic review of the evidence. Crit Rev Food Sci Nutr 2018; 59:2666-2673. [PMID: 29648479 DOI: 10.1080/10408398.2018.1463966] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/24/2023]
Abstract
Objective: The aim of this study was to evaluate the scientific evidence of dietary intervention, either through diet or supplementation, and its effects on the health of patients with systemic lupus erythematosus. Methods: Literature searches were conducted using Scopus, PubMed, BioMed Central and Science Direct databases. The terms used for the search were diet, nutritional support, nutrition therapy and systemic lupus erythematosus. Results: Eleven studies with interventions related to supplementation of omega-3 fatty acids, vitamin D and turmeric, as well as changes in diet composition, such as low glycaemic index diet were identified. Conclusions: The studies evidenced that omega-3 supplementation reduced inflammation, disease activity, endothelial dysfunction and oxidative stress; vitamin D supplementation increased serum levels, reduced inflammatory and hemostatic markers; turmeric supplementation reduced proteinuria, hematuria and systolic blood pressure; and low glycaemic index diet caused weight loss and reduced fatigue.
Collapse
Affiliation(s)
| | | | | | - José Carlos Gomes de Carvalho Leitão
- Universidade de Trás-os-Montes e Alto Douro , Vila Real , Portugal.,CIDESD-Research Center in Sports Sciences, Health Sciences and Human Development
| | | |
Collapse
|
31
|
Maile MD, Standiford TJ, Engoren MC, Stringer KA, Jewell ES, Rajendiran TM, Soni T, Burant CF. Associations of the plasma lipidome with mortality in the acute respiratory distress syndrome: a longitudinal cohort study. Respir Res 2018; 19:60. [PMID: 29636049 PMCID: PMC5894233 DOI: 10.1186/s12931-018-0758-3] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2017] [Accepted: 03/22/2018] [Indexed: 12/15/2022] Open
Abstract
Background It is unknown if the plasma lipidome is a useful tool for improving our understanding of the acute respiratory distress syndrome (ARDS). Therefore, we measured the plasma lipidome of individuals with ARDS at two time-points to determine if changes in the plasma lipidome distinguished survivors from non-survivors. We hypothesized that both the absolute concentration and change in concentration over time of plasma lipids are associated with 28-day mortality in this population. Methods Samples for this longitudinal observational cohort study were collected at multiple tertiary-care academic medical centers as part of a previous multicenter clinical trial. A mass spectrometry shot-gun lipidomic assay was used to quantify the lipidome in plasma samples from 30 individuals. Samples from two different days were analyzed for each subject. After removing lipids with a coefficient of variation > 30%, differences between cohorts were identified using repeated measures analysis of variance. The false discovery rate was used to adjust for multiple comparisons. Relationships between significant compounds were explored using hierarchical clustering of the Pearson correlation coefficients and the magnitude of these relationships was described using receiver operating characteristic curves. Results The mass spectrometry assay reliably measured 359 lipids. After adjusting for multiple comparisons, 90 compounds differed between survivors and non-survivors. Survivors had higher levels for each of these lipids except for five membrane lipids. Glycerolipids, particularly those containing polyunsaturated fatty acid side-chains, represented many of the lipids with higher concentrations in survivors. The change in lipid concentration over time did not differ between survivors and non-survivors. Conclusions The concentration of multiple plasma lipids is associated with mortality in this group of critically ill patients with ARDS. Absolute lipid levels provided more information than the change in concentration over time. These findings support future research aimed at integrating lipidomics into critical care medicine. Electronic supplementary material The online version of this article (10.1186/s12931-018-0758-3) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Michael D Maile
- Department of Anesthesiology, Division of Critical Care Medicine, University of Michigan Medical School, 4172 Cardiovascular Center, 1500 East Medical Center Drive, SPC 5861, Ann Arbor, MI, 48109, USA. .,Michigan Center for Integrative Research in Critical Care, University of Michigan, Ann Arbor, Michigan, USA.
| | - Theodore J Standiford
- Department of Internal Medicine, Division of Pulmonary and Critical Care Medicine, University of Michigan Medical School, Ann Arbor, Michigan, USA.,Michigan Center for Integrative Research in Critical Care, University of Michigan, Ann Arbor, Michigan, USA
| | - Milo C Engoren
- Department of Anesthesiology, Division of Critical Care Medicine, University of Michigan Medical School, 4172 Cardiovascular Center, 1500 East Medical Center Drive, SPC 5861, Ann Arbor, MI, 48109, USA.,Michigan Center for Integrative Research in Critical Care, University of Michigan, Ann Arbor, Michigan, USA
| | - Kathleen A Stringer
- Department of Clinical Pharmacy, College of Pharmacy, University of Michigan, Ann Arbor, Michigan, USA.,Michigan Center for Integrative Research in Critical Care, University of Michigan, Ann Arbor, Michigan, USA
| | - Elizabeth S Jewell
- Department of Anesthesiology, Division of Critical Care Medicine, University of Michigan Medical School, 4172 Cardiovascular Center, 1500 East Medical Center Drive, SPC 5861, Ann Arbor, MI, 48109, USA
| | - Thekkelnaycke M Rajendiran
- Michigan Center for Translational Pathology, Department of Pathology, University of Michigan, Ann Arbor, Michigan, USA.,Michigan Regional Comprehensive Metabolomics Resource Core, University of Michigan, Ann Arbor, Michigan, USA
| | - Tanu Soni
- Michigan Regional Comprehensive Metabolomics Resource Core, University of Michigan, Ann Arbor, Michigan, USA
| | - Charles F Burant
- Division of Metabolism, Endocrinology, and Diabetes, University of Michigan, Ann Arbor, MI, USA
| |
Collapse
|
32
|
Boff D, Crijns H, Teixeira MM, Amaral FA, Proost P. Neutrophils: Beneficial and Harmful Cells in Septic Arthritis. Int J Mol Sci 2018; 19:E468. [PMID: 29401737 PMCID: PMC5855690 DOI: 10.3390/ijms19020468] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2017] [Revised: 01/30/2018] [Accepted: 02/01/2018] [Indexed: 12/17/2022] Open
Abstract
Septic arthritis is an inflammatory joint disease that is induced by pathogens such as Staphylococcus aureus. Infection of the joint triggers an acute inflammatory response directed by inflammatory mediators including microbial danger signals and cytokines and is accompanied by an influx of leukocytes. The recruitment of these inflammatory cells depends on gradients of chemoattractants including formylated peptides from the infectious agent or dying cells, host-derived leukotrienes, complement proteins and chemokines. Neutrophils are of major importance and play a dual role in the pathogenesis of septic arthritis. On the one hand, these leukocytes are indispensable in the first-line defense to kill invading pathogens in the early stage of disease. However, on the other hand, neutrophils act as mediators of tissue destruction. Since the elimination of inflammatory neutrophils from the site of inflammation is a prerequisite for resolution of the acute inflammatory response, the prolonged stay of these leukocytes at the inflammatory site can lead to irreversible damage to the infected joint, which is known as an important complication in septic arthritis patients. Thus, timely reduction of the recruitment of inflammatory neutrophils to infected joints may be an efficient therapy to reduce tissue damage in septic arthritis.
Collapse
Affiliation(s)
- Daiane Boff
- Imunofarmacologia, Department of Biochemistry and Immunology, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte 31270-901, Brazil.
- Laboratory of Molecular Immunology, Department of Microbiology and Immunology, Rega Institute for Medical Research, KU Leuven, B-3000 Leuven, Belgium.
| | - Helena Crijns
- Imunofarmacologia, Department of Biochemistry and Immunology, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte 31270-901, Brazil.
- Laboratory of Molecular Immunology, Department of Microbiology and Immunology, Rega Institute for Medical Research, KU Leuven, B-3000 Leuven, Belgium.
| | - Mauro M Teixeira
- Imunofarmacologia, Department of Biochemistry and Immunology, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte 31270-901, Brazil.
| | - Flavio A Amaral
- Imunofarmacologia, Department of Biochemistry and Immunology, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte 31270-901, Brazil.
| | - Paul Proost
- Laboratory of Molecular Immunology, Department of Microbiology and Immunology, Rega Institute for Medical Research, KU Leuven, B-3000 Leuven, Belgium.
| |
Collapse
|
33
|
Thao NP, Lee YS, Luyen BTT, Oanh HV, Ali I, Arooj M, Koh YS, Yang SY, Kim YH. Chemicals from Cimicifuga dahurica and Their Inhibitory Effects on Pro-inflammatory Cytokine Production by LPS-stimulated Bone Marrow-derived Dendritic Cells. ACTA ACUST UNITED AC 2018. [DOI: 10.20307/nps.2018.24.3.194] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Affiliation(s)
- Nguyen Phuong Thao
- Institute of Marine Biochemistry (IMBC), Vietnam Academy of Science and Technology (VAST), 18-Hoang Quoc Viet, Hanoi, Vietnam
| | - Young Suk Lee
- College of Pharmacy, Chungnam National University, Daejeon 34134, Republic of Korea
| | | | - Ha Van Oanh
- Hanoi University of Pharmacy, 13-15 Le Thanh Tong, Hanoi, Vietnam
| | - Irshad Ali
- School of Medicine and Jeju Research Center for Natural Medicine, Jeju National University, Jeju 63243, Republic of Korea
| | - Madeeha Arooj
- School of Medicine and Jeju Research Center for Natural Medicine, Jeju National University, Jeju 63243, Republic of Korea
| | - Young Sang Koh
- School of Medicine and Jeju Research Center for Natural Medicine, Jeju National University, Jeju 63243, Republic of Korea
| | - Seo Young Yang
- College of Pharmacy, Chungnam National University, Daejeon 34134, Republic of Korea
| | - Young Ho Kim
- College of Pharmacy, Chungnam National University, Daejeon 34134, Republic of Korea
| |
Collapse
|
34
|
Shearer GC, Borkowski K, Puumala SL, Harris WS, Pedersen TL, Newman JW. Abnormal lipoprotein oxylipins in metabolic syndrome and partial correction by omega-3 fatty acids. Prostaglandins Leukot Essent Fatty Acids 2018; 128:1-10. [PMID: 29413356 DOI: 10.1016/j.plefa.2017.10.006] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/02/2017] [Revised: 10/10/2017] [Accepted: 10/19/2017] [Indexed: 12/31/2022]
Abstract
Metabolic syndrome (MetSyn) is characterized by chronic inflammation which mediates the associated high risk for cardiovascular and other diseases. Oxylipins are a superclass of lipid mediators with potent bioactivities in inflammation, vascular biology, and more. While their role as locally produced agents is appreciated, most oxylipins in plasma are found in lipoproteins suggesting defective regulation of inflammation could be mediated by the elevated VLDL and low HDL levels characteristic of MetSyn. Our objective was to compare the oxylipin composition of VLDL, LDL, and HDL in 14 optimally healthy individuals and 31 MetSyn patients, and then to determine the effects of treating MetSyn subjects with 4g/day of prescription omega-3 fatty acids (P-OM3) on lipoprotein oxylipin profiles. We compared oxylipin compositions of healthy (14) and MetSyn (31) subjects followed by randomization and assignment to 4g/d P-OM3 for 16 weeks using LC/MS/MS. Compared to healthy subjects, MetSyn is characterized by abnormalities of (1) pro-inflammatory, arachidonate-derived oxylipins from the lipoxygenase pathway in HDL; and (2) oxylipins mostly not derived from arachidonate in VLDL. P-OM3 treatment corrected many components of these abnormalities, reducing the burden of inflammatory mediators within peripherally circulating lipoproteins that could interfere with, or enhance, local effectors of inflammatory stress. We conclude that MetSyn is associated with a disruption of lipoprotein oxylipin patterns consistent with greater inflammatory stress, and the partial correction of these dysoxylipinemias by treatment with omega-3 fatty acids could explain some of their beneficial effects.
Collapse
Affiliation(s)
- Gregory C Shearer
- Sanford Research, Sioux Falls, SD, USA; Sanford School of Medicine, University of South Dakota, Sioux Falls, SD, USA; The Pennsylvania State University, Department of Nutritional Sciences, University Park, PA, USA.
| | - Kamil Borkowski
- The Pennsylvania State University, Department of Nutritional Sciences, University Park, PA, USA; West Coast Metabolomics Center, UC Davis Genome Center, University of California Davis, CA, USA
| | | | - William S Harris
- Sanford Research, Sioux Falls, SD, USA; Sanford School of Medicine, University of South Dakota, Sioux Falls, SD, USA
| | - Theresa L Pedersen
- Obesity and Metabolism Research Unit, USDA, ARS, Western Human Nutrition Research Center, Davis, CA, USA
| | - John W Newman
- Obesity and Metabolism Research Unit, USDA, ARS, Western Human Nutrition Research Center, Davis, CA, USA; Department of Nutrition, University of California, Davis, CA, USA
| |
Collapse
|
35
|
Kurgan S, Kantarci A. Molecular basis for immunohistochemical and inflammatory changes during progression of gingivitis to periodontitis. Periodontol 2000 2017; 76:51-67. [DOI: 10.1111/prd.12146] [Citation(s) in RCA: 50] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/14/2016] [Indexed: 12/22/2022]
|
36
|
Natural killer cells play an essential role in resolution of antigen-induced inflammation in mice. Mol Immunol 2017; 93:1-8. [PMID: 29112834 DOI: 10.1016/j.molimm.2017.10.019] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2017] [Revised: 10/19/2017] [Accepted: 10/25/2017] [Indexed: 12/29/2022]
Abstract
This study examined whether NK cells are important for resolution of antigen-induced inflammation. C57BL/6 mice were immunized twice with methylated BSA (mBSA) and inflammation induced by intraperitoneal injection of mBSA. Mice were injected intravenously with anti-asialo GM1 (αASGM1) or a control antibody 24h prior to peritonitis induction and peritoneal exudate collected at different time points. Expression of surface molecules and apoptosis on peritoneal cells was determined by flow cytometry and concentration of chemokines, cytokines, soluble cytokine receptors and lipid mediators by ELISA and LC-MS/MS. Apoptosis in parathymic lymph nodes and spleens was determined by TUNEL staining. Mice administered αASGM1 had lower peritoneal NK cell numbers and a higher number of peritoneal neutrophils 12h after induction of inflammation than control mice. The number of neutrophils was still high in the αASGM1 treated mice when their number had returned to baseline levels in the control mice, 48h after induction of inflammation. Peritoneal concentrations of the neutrophil regulators G-CSF and IL-12p40 were higher at 12h in the αASGM1 treated mice than in the control mice, whereas concentrations of lipid mediators implicated in resolution of inflammation, i.e. LXA4 and PGE2, were lower. Reduced apoptosis was detected in peritoneal neutrophils as well as in draining lymph nodes and spleens from the αASGM1 treated mice compared with that in the control mice. In addition, αASGM1 treated mice had lower number of peritoneal NK cells expressing NKp46 and NKG2D, receptors implicated in NK cell-induced neutrophil apoptosis. Furthermore, αASGM1 treatment completely blocked the increase in CD27+ NK cells that occurred in control mice following induction of inflammation, but CD27+ NK cells have been suggested to have a regulatory role. These results indicate a crucial role for NK cells in resolution of antigen-induced inflammation and suggest their importance in tempering neutrophil recruitment and maintaining neutrophil apoptosis.
Collapse
|
37
|
Nordgren TM, Heires AJ, Bailey KL, Katafiasz DM, Toews ML, Wichman CS, Romberger DJ. Docosahexaenoic acid enhances amphiregulin-mediated bronchial epithelial cell repair processes following organic dust exposure. Am J Physiol Lung Cell Mol Physiol 2017; 314:L421-L431. [PMID: 29097425 DOI: 10.1152/ajplung.00273.2017] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
Injurious dust exposures in the agricultural workplace involve the release of inflammatory mediators and activation of epidermal growth factor receptor (EGFR) in the respiratory epithelium. Amphiregulin (AREG), an EGFR ligand, mediates tissue repair and wound healing in the lung epithelium. Omega-3 fatty acids such as docosahexaenoic acid (DHA) are also known modulators of repair and resolution of inflammatory injury. This study investigated how AREG, DHA, and EGFR modulate lung repair processes following dust-induced injury. Primary human bronchial epithelial (BEC) and BEAS-2B cells were treated with an aqueous extract of swine confinement facility dust (DE) in the presence of DHA and AREG or EGFR inhibitors. Mice were exposed to DE intranasally with or without EGFR inhibition and DHA. Using a decellularized lung scaffolding tissue repair model, BEC recolonization of human lung scaffolds was analyzed in the context of DE, DHA, and AREG treatments. Through these investigations, we identified an important role for AREG in mediating BEC repair processes. DE-induced AREG release from BEC, and DHA treatment following DE exposure, enhanced this release. Both DHA and AREG also enhanced BEC repair capacities and rescued DE-induced recellularization deficits. In vivo, DHA treatment enhanced AREG production following DE exposure, whereas EGFR inhibitor-treated mice exhibited reduced AREG in their lung homogenates. These data indicate a role for AREG in the process of tissue repair after inflammatory lung injury caused by environmental dust exposure and implicate a role for DHA in regulating AREG-mediated repair signaling in BEC.
Collapse
Affiliation(s)
- Tara M Nordgren
- Pulmonary, Critical Care, Sleep and Allergy Division, Department of Internal Medicine, University of Nebraska Medical Center , Omaha, Nebraska.,Division of Biomedical Sciences, School of Medicine, University of California Riverside , Riverside, California
| | - Art J Heires
- Pulmonary, Critical Care, Sleep and Allergy Division, Department of Internal Medicine, University of Nebraska Medical Center , Omaha, Nebraska
| | - Kristina L Bailey
- Veterans Affairs Nebraska-Western Iowa Health Care System, Omaha, Nebraska.,Pulmonary, Critical Care, Sleep and Allergy Division, Department of Internal Medicine, University of Nebraska Medical Center , Omaha, Nebraska
| | - Dawn M Katafiasz
- Pulmonary, Critical Care, Sleep and Allergy Division, Department of Internal Medicine, University of Nebraska Medical Center , Omaha, Nebraska
| | - Myron L Toews
- Department of Pharmacology and Experimental Neuroscience, University of Nebraska Medical Center , Omaha, Nebraska
| | - Christopher S Wichman
- Department of Biostatistics, University of Nebraska Medical Center , Omaha, Nebraska
| | - Debra J Romberger
- Veterans Affairs Nebraska-Western Iowa Health Care System, Omaha, Nebraska.,Pulmonary, Critical Care, Sleep and Allergy Division, Department of Internal Medicine, University of Nebraska Medical Center , Omaha, Nebraska
| |
Collapse
|
38
|
Elliott E, Hanson CK, Anderson-Berry AL, Nordgren TM. The role of specialized pro-resolving mediators in maternal-fetal health. Prostaglandins Leukot Essent Fatty Acids 2017; 126:98-104. [PMID: 29031403 PMCID: PMC5647871 DOI: 10.1016/j.plefa.2017.09.017] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/12/2017] [Revised: 09/20/2017] [Accepted: 09/21/2017] [Indexed: 12/17/2022]
Abstract
Infants developing in a pro-inflammatory intrauterine environment have a significant risk for severe complications after birth. It has been shown that omega-3 fatty acids reduce inflammation, and also reduce early preterm births and decrease risk of infant admission to the neonatal intensive care unit. However, the mechanism for omega-3 fatty acids exerting these effects was previously unknown. Recent evidence has shown that downstream products of polyunsaturated fatty acids called specialized pro-resolving mediators may mediate inflammatory physiology, thus playing an important role in maternal-fetal health. In this review, current knowledge relating to specialized pro-resolving mediators in pregnancy, delivery, and perinatal disease states will be summarized.
Collapse
Affiliation(s)
- E Elliott
- University of Nebraska Medical Center, Pediatrics, 981205 Nebraska Medical Center, Omaha, NE 68198-1205, United States
| | - C K Hanson
- University of Nebraska Medical Center, College of Allied Health Professions, Medical Nutrition Education, 984045 Nebraska Medical Center, Omaha, NE 68198-4045, United States
| | - A L Anderson-Berry
- University of Nebraska Medical Center, Pediatrics, 981205 Nebraska Medical Center, Omaha, NE 68198-1205, United States
| | - T M Nordgren
- University of Nebraska Medical Center, Internal Medicine, Pulmonary, Critical Care, Sleep & Allergy Division, 985910 Nebraska Medicine, Omaha, NE 68198-5910, United States; University of California Riverside, Division of Biomedical Sciences, School of Medicine, 900 University Avenue, Riverside, CA 92521, United States.
| |
Collapse
|
39
|
Ácidos graxos ômega‐3, estado inflamatório e marcadores bioquímicos de pacientes com lúpus eritematoso sistêmico: estudo piloto. REVISTA BRASILEIRA DE REUMATOLOGIA 2017. [DOI: 10.1016/j.rbr.2016.08.002] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023] Open
|
40
|
Changing dietary n-6:n-3 ratio using different oil sources affects performance, behavior, cytokines mRNA expression and meat fatty acid profile of broiler chickens. ACTA ACUST UNITED AC 2017; 4:44-51. [PMID: 30167483 PMCID: PMC6112305 DOI: 10.1016/j.aninu.2017.08.003] [Citation(s) in RCA: 36] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2017] [Revised: 07/25/2017] [Accepted: 08/05/2017] [Indexed: 12/22/2022]
Abstract
Typical formulated broiler diets are deficient in n-3 poly-unsaturated fatty acids (PUFA) due to widening n-6:n-3 PUFA ratio which could greatly affect performance, immune system of birds and, more importantly, meat quality. This study was conducted to evaluate the effect of modifying dietary n-6:n-3 PUFA ratio from plant and animal oil sources on performance, behavior, cytokine mRNA expression, antioxidative status and meat fatty acid profile of broiler chickens. Birds (n = 420) were fed 7 diets enriched with different dietary oil sources and ratios as follows: sunflower oil in control diet (C); fish oil (FO); 1:1 ratio of sunflower oil to FO (C1FO1); 3:1 ratio of sunflower oil to fish oil (C3FO1); linseed oil (LO); 1:1 ratio of sunflower oil to linseed oil (C1LO1); 3:1 ratio of sunflower oil to linseed oil (C3LO1), resulting in dietary n-6:n-3 ratios of approximately 40:1, 1.5:1, 4:1, 8:1, 1:1, 2.5:1 and 5:1, respectively. The best final body weight, feed conversion ratio as well as protein efficiency ratio of broilers were recorded in the C1FO1 and C1LO1 groups. Compared with the control group, the dressing percentage and breast and thigh yield were highest in the C1FO1 and C1LO1 groups. Narrowing the dietary n-6:n-3 ratio increased (P < 0.05) n-3 PUFA content of breast meat. Moreover, the breast meat contents of eicosapentaenoic acid and docosahexaenoic acid increased (P < 0.05) with increasing dietary FO whereas α-linolenic acid content was higher with LO supplementation. Also, enriching the diets with n-3 PUFA from FO and LO clearly decreased (P < 0.05) serum total cholesterol, triglycerides and very low-density lipoproteins and enhanced antioxidative status. The feeding frequency was decreased (P < 0.05) in the C1FO1 and C1LO1 groups. Likewise, n-3 PUFA-enriched diets enhanced the frequency of preening, wing flapping and flightiness. Animal oil source addition, compared to plant oil, to broiler diets enhanced the relative mRNA expression of interferon gamma, interleukin-1 beta, interleukin-2 and interleukin-6 genes, especially at low n-6:n-3 ratios. This study has clearly shown that narrowing n-6:n-3 ratio through the addition of FO or LO improved performance and immune response of broilers and resulted in healthy chicken meat, enriched with long chain n-3 PUFA.
Collapse
|
41
|
Perucci LO, Sugimoto MA, Gomes KB, Dusse LM, Teixeira MM, Sousa LP. Annexin A1 and specialized proresolving lipid mediators: promoting resolution as a therapeutic strategy in human inflammatory diseases. Expert Opin Ther Targets 2017; 21:879-896. [PMID: 28786708 DOI: 10.1080/14728222.2017.1364363] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
INTRODUCTION The timely resolution of inflammation is essential to restore tissue homeostasis and to avoid chronic inflammatory diseases. Resolution of inflammation is an active process modulated by various proresolving mediators, including annexin A1 (AnxA1) and specialized proresolving lipid mediators (SPMs), which counteract excessive inflammatory responses and stimulate proresolving mechanisms. Areas covered: The protective effects of AnxA1 and SPMs have been extensively explored in pre-clinical animal models. However, studies investigating the function of these molecules in human diseases are just emerging. This review highlights recent advances on the role of proresolving mediators, and pharmacological opportunities of promoting resolution pathways in preclinical models and patients with various human diseases. Expert opinion: Dysregulation or 'failure' in proresolving mechanisms might be involved in the pathogenesis of chronic inflammatory diseases. Altered levels of proresolving mediators were found in a wide range of human diseases. In some cases, AnxA1 and SPMs are up-regulated in human blood and tissues but fail to engage in proresolving signaling and, hence, to regulate excessive inflammation. Thus, the new concept of 'resolution pharmacology' could be applied to compensate deficiency of endogenous proresolving mediators' generation and/or possible failures in the engagement of resolution pathways observed in many chronic inflammatory diseases.
Collapse
Affiliation(s)
- Luiza Oliveira Perucci
- a Departamento de Análises Clínicas e Toxicológicas, Faculdade de Farmácia , Universidade Federal de Minas Gerais , Belo Horizonte , Minas Gerais , Brazil.,b Programa de Pós-Graduação em Análises Clínicas e Toxicológicas , Universidade Federal de Minas Gerais , Belo Horizonte , Minas Gerais , Brazil
| | - Michelle Amantéa Sugimoto
- a Departamento de Análises Clínicas e Toxicológicas, Faculdade de Farmácia , Universidade Federal de Minas Gerais , Belo Horizonte , Minas Gerais , Brazil.,c Programa de Pós-Graduação em Ciências Farmacêuticas , Universidade Federal de Minas Gerais , Belo Horizonte , Minas Gerais , Brazil
| | - Karina Braga Gomes
- a Departamento de Análises Clínicas e Toxicológicas, Faculdade de Farmácia , Universidade Federal de Minas Gerais , Belo Horizonte , Minas Gerais , Brazil.,b Programa de Pós-Graduação em Análises Clínicas e Toxicológicas , Universidade Federal de Minas Gerais , Belo Horizonte , Minas Gerais , Brazil
| | - Luci Maria Dusse
- a Departamento de Análises Clínicas e Toxicológicas, Faculdade de Farmácia , Universidade Federal de Minas Gerais , Belo Horizonte , Minas Gerais , Brazil.,b Programa de Pós-Graduação em Análises Clínicas e Toxicológicas , Universidade Federal de Minas Gerais , Belo Horizonte , Minas Gerais , Brazil.,c Programa de Pós-Graduação em Ciências Farmacêuticas , Universidade Federal de Minas Gerais , Belo Horizonte , Minas Gerais , Brazil
| | - Mauro Martins Teixeira
- d Departamento de Bioquímica e Imunologia, Instituto de Ciências Biológicas , Universidade Federal de Minas Gerais , Belo Horizonte , Minas Gerais , Brazil
| | - Lirlândia Pires Sousa
- a Departamento de Análises Clínicas e Toxicológicas, Faculdade de Farmácia , Universidade Federal de Minas Gerais , Belo Horizonte , Minas Gerais , Brazil.,b Programa de Pós-Graduação em Análises Clínicas e Toxicológicas , Universidade Federal de Minas Gerais , Belo Horizonte , Minas Gerais , Brazil.,c Programa de Pós-Graduação em Ciências Farmacêuticas , Universidade Federal de Minas Gerais , Belo Horizonte , Minas Gerais , Brazil
| |
Collapse
|
42
|
Botchlett R, Woo SL, Liu M, Pei Y, Guo X, Li H, Wu C. Nutritional approaches for managing obesity-associated metabolic diseases. J Endocrinol 2017; 233:R145-R171. [PMID: 28400405 PMCID: PMC5511693 DOI: 10.1530/joe-16-0580] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/15/2017] [Accepted: 04/11/2017] [Indexed: 01/10/2023]
Abstract
Obesity is an ongoing pandemic and serves as a causal factor of a wide spectrum of metabolic diseases including diabetes, fatty liver disease, and cardiovascular disease. Much evidence has demonstrated that nutrient overload/overnutrition initiates or exacerbates inflammatory responses in tissues/organs involved in the regulation of systemic metabolic homeostasis. This obesity-associated inflammation is usually at a low-grade and viewed as metabolic inflammation. When it exists continuously, inflammation inappropriately alters metabolic pathways and impairs insulin signaling cascades in peripheral tissues/organs such as adipose tissue, the liver and skeletal muscles, resulting in local fat deposition and insulin resistance and systemic metabolic dysregulation. In addition, inflammatory mediators, e.g., proinflammatory cytokines, and excessive nutrients, e.g., glucose and fatty acids, act together to aggravate local insulin resistance and form a vicious cycle to further disturb the local metabolic pathways and exacerbate systemic metabolic dysregulation. Owing to the critical role of nutrient metabolism in controlling the initiation and progression of inflammation and insulin resistance, nutritional approaches have been implicated as effective tools for managing obesity and obesity-associated metabolic diseases. Based on the mounting evidence generated from both basic and clinical research, nutritional approaches are commonly used for suppressing inflammation, improving insulin sensitivity, and/or decreasing fat deposition. Consequently, the combined effects are responsible for improvement of systemic insulin sensitivity and metabolic homeostasis.
Collapse
Affiliation(s)
- Rachel Botchlett
- Department of Nutrition and Food ScienceTexas A&M University, College Station, USA
- Pinnacle Clinical ResearchLive Oak, USA
| | - Shih-Lung Woo
- Department of Nutrition and Food ScienceTexas A&M University, College Station, USA
| | - Mengyang Liu
- Department of Nutrition and Food ScienceTexas A&M University, College Station, USA
| | - Ya Pei
- Department of Nutrition and Food ScienceTexas A&M University, College Station, USA
| | - Xin Guo
- Department of Nutrition and Food ScienceTexas A&M University, College Station, USA
- Baylor College of MedicineHouston, USA
| | - Honggui Li
- Department of Nutrition and Food ScienceTexas A&M University, College Station, USA
| | - Chaodong Wu
- Department of Nutrition and Food ScienceTexas A&M University, College Station, USA
| |
Collapse
|
43
|
Lodewijkx PJ, Besselink MG, Witteman BJ, Schepers NJ, Gooszen HG, van Santvoort HC, Bakker OJ. Nutrition in acute pancreatitis: a critical review. Expert Rev Gastroenterol Hepatol 2017; 10:571-80. [PMID: 26823272 DOI: 10.1586/17474124.2016.1141048] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Severe acute pancreatitis poses unique nutritional challenges. The optimal nutritional support in patients with severe acute pancreatitis has been a subject of debate for decades. This review provides a critical review of the available literature. According to current literature, enteral nutrition is superior to parenteral nutrition, although several limitations should be taken into account. The optimal route of enteral nutrition remains unclear, but normal or nasogastric tube feeding seems safe when tolerated. In patients with predicted severe acute pancreatitis an on-demand feeding strategy is advised and when patients do not tolerate an oral diet after 72 hours, enteral nutrition can be started. The use of supplements, both parenteral as enteral, are not recommended. Optimal nutritional support in severe cases often requires a tailor-made approach with day-to-day evaluation of its effectiveness.
Collapse
Affiliation(s)
- Piet J Lodewijkx
- a Department of Surgery , Jeroen Bosch hospital , s-Hertogenbosch , The Netherlands
| | - Marc G Besselink
- b Department of Surgery , Academic Medical Center , Amsterdam , The Netherlands
| | - Ben J Witteman
- c Department of Gastroenterology and Hepatology , Hospital Gelderse Vallei Ede , Ede , The Netherlands
| | - Nicolien J Schepers
- d Department of Gastroenterology and Hepatology , Erasmus MC University Medical Center , Rotterdam , The Netherlands.,e Department of Gastroenterology and Hepatology , St. Antonius Hospital , Nieuwegein , The Netherlands
| | - Hein G Gooszen
- f Department of Operating Theatres and Evidence Based Surgery , Radboud University Medical Center , Nijmegen , The Netherlands
| | | | - Olaf J Bakker
- g Department of Surgery , University Medical Center Utrecht , Utrecht , The Netherlands
| | | |
Collapse
|
44
|
Freire MO, Dalli J, Serhan CN, Van Dyke TE. Neutrophil Resolvin E1 Receptor Expression and Function in Type 2 Diabetes. THE JOURNAL OF IMMUNOLOGY 2016; 198:718-728. [PMID: 27994073 DOI: 10.4049/jimmunol.1601543] [Citation(s) in RCA: 60] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Subscribe] [Scholar Register] [Received: 09/08/2016] [Accepted: 11/15/2016] [Indexed: 12/31/2022]
Abstract
Unresolved inflammation is key in linking metabolic dysregulation and the immune system in type 2 diabetes. Successful regulation of acute inflammation requires biosynthesis of specialized proresolving lipid mediators, such as E-series resolvin (RvE) 1, and activation of cognate G protein-coupled receptors. RvE1 binds to leukotriene B4 (BLT-1) on neutrophils and to ERV-1/ChemR23 on monocyte/macrophages. We show novel actions of RvE1 and expression patterns of neutrophil receptors in type 2 diabetes. Neutrophils from healthy subjects express functional BLT-1, low levels of minimally functional ERV-1, and inversed coexpression when compared to neutrophils from type 2 diabetes subjects. Stimulation with TNF-α or LPS increased the expression of ERV-1 by healthy and diabetic neutrophils. RvE1 counteracted LPS and TNF-α induction of ERV-1 overexpression and endogenous diabetic overexpression, activating phagocytosis and resolution signals. Functional ERV-1 was determined by phosphorylation of the signaling protein ribosomal S6. Receptor-antagonism experiments revealed that the increase in phosphorylation of ribosomal S6 was mediated by BLT-1 in healthy subject neutrophils and by ERV-1 in diabetes. Metabololipidomics reveal a proinflammatory profile in diabetic serum. Cell phagocytosis is impaired in type 2 diabetes and requires RvE1 for activation. The dose of RvE1 required to activate resolution signals in type 2 diabetic neutrophils was significantly higher than in healthy controls. RvE1 rescues the dysregulation seen on neutrophil receptor profile and, following a therapeutic dosage, activates phagocytosis and resolution signals in type 2 diabetes. These findings reveal the importance of resolution receptors in health, disease, and dysregulation of inflammation in type 2 diabetes.
Collapse
Affiliation(s)
- Marcelo O Freire
- Department of Applied Oral Sciences, Center for Periodontology, The Forsyth Institute, Cambridge, MA 02142.,Department of Infection and Immunity, Harvard School of Dental Medicine, Boston, MA 02115; and
| | - Jesmond Dalli
- Department of Anesthesiology, Perioperative and Pain Medicine, Center for Experimental Therapeutics and Reperfusion Injury, Harvard Institutes of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA 02115
| | - Charles N Serhan
- Department of Anesthesiology, Perioperative and Pain Medicine, Center for Experimental Therapeutics and Reperfusion Injury, Harvard Institutes of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA 02115
| | - Thomas E Van Dyke
- Department of Applied Oral Sciences, Center for Periodontology, The Forsyth Institute, Cambridge, MA 02142; .,Department of Infection and Immunity, Harvard School of Dental Medicine, Boston, MA 02115; and
| |
Collapse
|
45
|
Omega-3 fatty acids, inflammatory status and biochemical markers of patients with systemic lupus erythematosus: a pilot study. REVISTA BRASILEIRA DE REUMATOLOGIA 2016; 57:526-534. [PMID: 29173690 DOI: 10.1016/j.rbre.2016.09.014] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2016] [Accepted: 08/30/2016] [Indexed: 12/29/2022] Open
Abstract
BACKGROUND Studies have shown that omega-3 fatty acids reduce the concentrations of eicosanoids, cytokines, chemokines, C-reactive protein (CRP) and other inflammatory mediators. OBJECTIVE To investigate the effects of omega-3 fatty acids on circulating levels of inflammatory mediators and biochemical markers in women with systemic lupus erythematosus (SLE). METHODS Experimental clinical study (clinical trial: NCT02524795); 49 women with SLE (ACR1982/1997) were randomized: 22 to the omega-3 group (daily intake of 1080mg EPA+200mg DHA, for 12 weeks) and 27 to the control group. The inflammatory mediators and biochemical markers at T0 and T1 in omega-3 group were compared using Wilcoxon test. U-Mann-Whitney test was used to compare variations of measured variables [ΔV=pre-treatment (T0)-post-treatment (T1) concentrations] between groups. p<0.05 was considered significant. RESULTS The median (interquartile range - IQR) of age was 37 (29-48) years old, of disease duration was 7 (4-13) years, and of SLEDAI-2K was 1 (0-2). The median (IQR) of variation in CRP levels between the two groups showed a decrease in omega-3 group while there was an increase in control group (p=0.008). The serum concentrations of IL-6 and IL-10, leptin and adiponectin did not change after a 12 week treatment. CONCLUSIONS Supplementation with omega-3 had no impact on serum concentrations of IL-6, IL-10, leptin and adiponectin in women with SLE and low disease activity. There was a significant decrease of CRP levels as well as evidence that omega-3 may impact total and LDL-cholesterol.
Collapse
|
46
|
Perucci LO, Santos PC, Ribeiro LS, Souza DG, Gomes KB, Dusse LMS, Sousa LP. Lipoxin A4 Is Increased in the Plasma of Preeclamptic Women. Am J Hypertens 2016; 29:1179-85. [PMID: 27179254 DOI: 10.1093/ajh/hpw053] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2016] [Accepted: 05/03/2016] [Indexed: 12/21/2022] Open
Abstract
BACKGROUND Excessive inflammation is involved in preeclampsia (PE) pathogenesis. Lipoxin A4 (LXA4) is an eicosanoid that counter-regulates inflammation. The main objective of this study was to determine LXA4 plasma levels in PE women. The correlations among LXA4 levels, ultrasensitive C-reactive protein (us-CRP) levels, and clinical/laboratory parameters of the studied participants were also investigated. METHODS LXA4 plasma levels were determined by ELISA in 23 nonpregnant, 26 normotensive pregnant, and 27 PE women (early PE (N = 10) and late PE (N = 17)), according to gestational age (GA) at clinical symptoms onset). The clinical/laboratory parameters included in Spearman's correlation analysis were: systolic and diastolic blood pressure (SBP and DBP, respectively), lactate dehydrogenase (LDH) activity, platelet count, proteinuria, and white blood cell count (WBC). RESULTS LXA4 levels were higher in PE women than in nonpregnant and normotensive pregnant women, and similar between nonpregnant and normotensive pregnant women. LXA4 plasma levels were higher in early PE vs. normotensive pregnancy (GA < 34 weeks) and in late PE vs. normotensive pregnancy (GA ≥ 34 weeks). No significant differences were detected between early and late PE. LXA4 levels were positively correlated with us-CRP levels, SBP, DBP, and WBC. No significant correlation was detected between LXA4 levels and the other laboratory parameters. CONCLUSIONS Chronic inflammation in PE, in spite of increased levels of LXA4, points to a possible failure in this regulatory pathway. Further studies are necessary to clarify this issue and to evaluate the role of LXA4 and other proresolving mediators of inflammation in the pathogenesis of PE.
Collapse
Affiliation(s)
- Luiza O Perucci
- Departamento de Análises Clínicas e Toxicológicas, Faculdade de Farmácia, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil; Programa de Pós-Graduação em Análises Clínicas e Toxicológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Patrícia C Santos
- Departamento de Microbiologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Lucas S Ribeiro
- Departamento de Microbiologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Danielle G Souza
- Departamento de Microbiologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Karina B Gomes
- Departamento de Análises Clínicas e Toxicológicas, Faculdade de Farmácia, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil; Programa de Pós-Graduação em Análises Clínicas e Toxicológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Luci M S Dusse
- Departamento de Análises Clínicas e Toxicológicas, Faculdade de Farmácia, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil; Programa de Pós-Graduação em Análises Clínicas e Toxicológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Lirlândia P Sousa
- Departamento de Análises Clínicas e Toxicológicas, Faculdade de Farmácia, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil; Programa de Pós-Graduação em Análises Clínicas e Toxicológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil;
| |
Collapse
|
47
|
Effects of n-3 PUFAs on Intestinal Mucosa Innate Immunity and Intestinal Microbiota in Mice after Hemorrhagic Shock Resuscitation. Nutrients 2016; 8:nu8100609. [PMID: 27690096 PMCID: PMC5083997 DOI: 10.3390/nu8100609] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2016] [Revised: 09/05/2016] [Accepted: 09/07/2016] [Indexed: 12/19/2022] Open
Abstract
n-3 polyunsaturated fatty acids (PUFAs) can improve the function of the intestinal barrier after damage from ischemia-reperfusion or hemorrhagic shock resuscitation (HSR). However, the effects of n-3 PUFAs on intestinal microbiota and the innate immunity of the intestinal mucosa after HSR remain unclear. In the present study, 40 C57BL/6J mice were randomly assigned to five groups: control, sham, HSR, HSR + n-3 PUFAs and HSR + n-6 PUFAs. Mice were sacrificed 12 h after HSR. Liver, spleen, mesenteric lymph nodes and terminal ileal tissues were collected. Intestinal mucosae were scraped aseptically. Compared with the HSR group, the number of goblet cells increased, expression of mucin 2 was restored and disturbed intestinal microbiota were partly stabilized in the PUFA-administered groups, indicating that both n-3 and n-6 PUFAs reduced overproliferation of Gammaproteobacteria while promoting the growth of Bacteroidetes. Notably, n-3 PUFAs had an advantage over n-6 PUFAs in improving ileal tissue levels of lysozyme after HSR. Thus, PUFAs, especially n-3 PUFAs, partly improved the innate immunity of intestinal mucosa in mice after HSR. These findings suggest a clinical rationale for providing n-3 PUFAs to patients recovering from ischemia-reperfusion.
Collapse
|
48
|
Shenoy PA, Kuo A, Vetter I, Smith MT. The Walker 256 Breast Cancer Cell- Induced Bone Pain Model in Rats. Front Pharmacol 2016; 7:286. [PMID: 27630567 PMCID: PMC5005431 DOI: 10.3389/fphar.2016.00286] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2016] [Accepted: 08/18/2016] [Indexed: 12/19/2022] Open
Abstract
The majority of patients with terminal breast cancer show signs of bone metastasis, the most common cause of pain in cancer. Clinically available drug treatment options for the relief of cancer-associated bone pain are limited due to either inadequate pain relief and/or dose-limiting side-effects. One of the major hurdles in understanding the mechanism by which breast cancer causes pain after metastasis to the bones is the lack of suitable preclinical models. Until the late twentieth century, all animal models of cancer induced bone pain involved systemic injection of cancer cells into animals, which caused severe deterioration of animal health due to widespread metastasis. In this mini-review we have discussed details of a recently developed and highly efficient preclinical model of breast cancer induced bone pain: Walker 256 cancer cell- induced bone pain in rats. The model involves direct localized injection of cancer cells into a single tibia in rats, which avoids widespread metastasis of cancer cells and hence animals maintain good health throughout the experimental period. This model closely mimics the human pathophysiology of breast cancer induced bone pain and has great potential to aid in the process of drug discovery for treating this intractable pain condition.
Collapse
Affiliation(s)
- Priyank A Shenoy
- School of Biomedical Sciences, The University of QueenslandBrisbane, QLD, Australia; Centre for Integrated Preclinical Drug Development, The University of QueenslandBrisbane, QLD, Australia
| | - Andy Kuo
- Centre for Integrated Preclinical Drug Development, The University of Queensland Brisbane, QLD, Australia
| | - Irina Vetter
- Institute for Molecular Bioscience, The University of QueenslandBrisbane, QLD, Australia; School of Pharmacy, The University of QueenslandBrisbane, QLD, Australia
| | - Maree T Smith
- Centre for Integrated Preclinical Drug Development, The University of QueenslandBrisbane, QLD, Australia; School of Pharmacy, The University of QueenslandBrisbane, QLD, Australia
| |
Collapse
|
49
|
Perttula K, Edmands WMB, Grigoryan H, Cai X, Iavarone AT, Gunter MJ, Naccarati A, Polidoro S, Hubbard A, Vineis P, Rappaport SM. Evaluating Ultra-long-Chain Fatty Acids as Biomarkers of Colorectal Cancer Risk. Cancer Epidemiol Biomarkers Prev 2016; 25:1216-23. [PMID: 27257090 PMCID: PMC6319388 DOI: 10.1158/1055-9965.epi-16-0204] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2016] [Accepted: 05/20/2016] [Indexed: 12/21/2022] Open
Abstract
BACKGROUND Cross-sectional studies reported a novel set of hydroxylated ultra-long-chain fatty acids (ULCFA) that were present at significantly lower levels in colorectal cancer cases than controls. Follow-up studies suggested that these molecules were potential biomarkers of protective exposure for colorectal cancer. To test the hypothesis that ULCFAs reflect causal pathways, we measured their levels in prediagnostic serum from incident colorectal cancer cases and controls. METHODS Serum from 95 colorectal cancer patients and 95 matched controls was obtained from the Italian arm of the European Prospective Investigation into Cancer and Nutrition cohort and analyzed by liquid chromatography-high-resolution mass spectrometry. Levels of 8 ULCFAs were compared between cases and controls with paired t tests and a linear model that used time to diagnosis (TTD) to determine whether case-control differences were influenced by disease progression. RESULTS Although paired t tests detected significantly lower levels of four ULCFAs in colorectal cancer cases, confirming earlier reports, the case-control differences diminished significantly with increasing TTD (7 days-14 years). CONCLUSION Levels of several ULCFAs were lower in incident colorectal cancer cases than controls. However, because case-control differences decreased with increasing TTD, we conclude that these molecules were likely consumed by processes related to cancer progression rather than causal pathways. IMPACT ULCFA levels are unlikely to represent exposures that protect individuals from colorectal cancer. Future research should focus on the diagnostic potential and origins of these molecules. Our use of TTD as a covariate in a linear model provides an efficient method for distinguishing causal and reactive biomarkers in biospecimens from prospective cohorts. Cancer Epidemiol Biomarkers Prev; 25(8); 1216-23. ©2016 AACR.
Collapse
Affiliation(s)
- Kelsi Perttula
- School of Public Health, University of California, Berkeley, California
| | | | - Hasmik Grigoryan
- School of Public Health, University of California, Berkeley, California
| | - Xiaoming Cai
- School of Public Health, University of California, Berkeley, California
| | - Anthony T Iavarone
- California Institute for Quantitative Biosciences, University of California, Berkeley, California
| | - Marc J Gunter
- International Agency for Research on Cancer, Lyon, France
| | | | | | - Alan Hubbard
- School of Public Health, University of California, Berkeley, California
| | - Paolo Vineis
- HuGeF Foundation, Torino, Italy. MRC-PHE Centre for Environment and Health, Imperial College, London, United Kingdom
| | | |
Collapse
|
50
|
Considerations for the Conduct of Clinical Trials with Antiinflammatory Agents in Cystic Fibrosis. A Cystic Fibrosis Foundation Workshop Report. Ann Am Thorac Soc 2016; 12:1398-406. [PMID: 26146892 DOI: 10.1513/annalsats.201506-361ot] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
Inflammation leads to lung destruction and loss of pulmonary function in patients with cystic fibrosis (CF). Drugs that modulate the cystic fibrosis transmembrane conductance regulator (CFTR) have recently been approved. Although the impact of CFTR modulators on sweat chloride and lung function are exciting, they have not yet demonstrated an effect on inflammation. Therefore, CF antiinflammatory drug development must continue. Unfortunately, the lack of clarity with this process has left investigators and industry sponsors frustrated. The Cystic Fibrosis Foundation established a working group in early 2014 to address this issue. There are many inflammatory processes disrupted in CF, and, therefore, there are many potential targets amenable to antiinflammatory therapy. Regardless of a drug's specific mechanism of action, it must ultimately affect the neutrophil or its products to impact CF. The working group concluded that before bringing new antiinflammatory drugs to clinical trial, preclinical safety studies must be conducted in disease-relevant models to assuage safety concerns. Furthermore, although studies of antiinflammatory therapies must first establish safety in adults, subsequent studies must involve children, as they are most likely to reap the most benefit. The working group also recommended that pharmacokinetic-pharmacodynamic studies and early-phase safety studies be performed before proceeding to larger studies of longer duration. In addition, innovative study designs may improve the likelihood of adequately assessing treatment response and mitigating risk before conducting multiyear studies. Learning from past experiences and incorporating this knowledge into new drug development programs will be instrumental in bringing new antiinflammatory therapies to patients.
Collapse
|