1
|
Padovani CM, Wilson RM, Rodriguez A, Spur BW, Yin K. Resolvin D2 attenuates LPS-induced macrophage exhaustion. FASEB J 2024; 38:e23569. [PMID: 38551610 DOI: 10.1096/fj.202302521r] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2023] [Revised: 02/21/2024] [Accepted: 03/07/2024] [Indexed: 04/02/2024]
Abstract
Early in sepsis, a hyperinflammatory response is dominant, but later, an immunosuppressive phase dominates, and the host is susceptible to opportunistic infections. Anti-inflammatory agents may accelerate the host into immunosuppression, and few agents can reverse immunosuppression without causing inflammation. Specialized pro-resolving mediators (SPMs) such as resolvin D2 (RvD2) have been reported to resolve inflammation without being immunosuppressive, but little work has been conducted to examine their effects on immunosuppression. To assess the effects of RvD2 on immunosuppression, we established a model of macrophage exhaustion using two lipopolysaccharide (LPS) treatments or hits. THP-1 monocyte-derived macrophages were first treated with RvD2 or vehicle for 1 h. One LPS hit increased NF-κB activity 11-fold and TNF-α release 60-fold compared to unstimulated macrophages. RvD2 decreased LPS-induced NF-κB activity and TNF-α production but increased bacterial clearance. Two LPS hits reduced macrophage bacterial clearance and decreased macrophage NF-κB activity (45%) and TNF-α release (75%) compared to one LPS hit, demonstrating exhaustion. RvD2 increased NF-κB activity, TNF-α release, and bacterial clearance following two LPS hits compared to controls. TLR2 inhibition abolished RvD2-mediated changes. In a mouse sepsis model, splenic macrophage response to exogenous LPS was reduced compared to controls and was restored by in vivo administration of RvD2, supporting the in vitro results. If RvD2 was added to monocytes before differentiation into macrophages, however, RvD2 reduced LPS responses and increased bacterial clearance following both one and two LPS hits. The results show that RvD2 attenuated macrophage suppression in vitro and in vivo and that this effect was macrophage-specific.
Collapse
Affiliation(s)
- Cristina M Padovani
- Department of Cell Biology and Neuroscience, Rowan-Virtua School of Translational Biomedical Engineering and Sciences, Virtua Health College of Life Sciences of Rowan University, Stratford, New Jersey, USA
| | - Rachael M Wilson
- Department of Cell Biology and Neuroscience, Rowan-Virtua School of Translational Biomedical Engineering and Sciences, Virtua Health College of Life Sciences of Rowan University, Stratford, New Jersey, USA
| | - Ana Rodriguez
- Department of Cell Biology and Neuroscience, Rowan-Virtua School of Translational Biomedical Engineering and Sciences, Virtua Health College of Life Sciences of Rowan University, Stratford, New Jersey, USA
| | - Bernd W Spur
- Department of Cell Biology and Neuroscience, Rowan-Virtua School of Translational Biomedical Engineering and Sciences, Virtua Health College of Life Sciences of Rowan University, Stratford, New Jersey, USA
| | - Kingsley Yin
- Department of Cell Biology and Neuroscience, Rowan-Virtua School of Translational Biomedical Engineering and Sciences, Virtua Health College of Life Sciences of Rowan University, Stratford, New Jersey, USA
| |
Collapse
|
2
|
Shi H, Gao L, Kirby N, Shao B, Shan X, Kudo M, Silasi R, McDaniel JM, Zhou M, McGee S, Jing W, Lupu F, Cleuren A, George JN, Xia L. Clearance of VWF by hepatic macrophages is critical for the protective effect of ADAMTS13 in sickle cell anemia mice. Blood 2024; 143:1293-1309. [PMID: 38142410 PMCID: PMC10997916 DOI: 10.1182/blood.2023021583] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2023] [Revised: 11/21/2023] [Accepted: 12/06/2023] [Indexed: 12/26/2023] Open
Abstract
ABSTRACT Although it is caused by a single-nucleotide mutation in the β-globin gene, sickle cell anemia (SCA) is a systemic disease with complex, incompletely elucidated pathologies. The mononuclear phagocyte system plays critical roles in SCA pathophysiology. However, how heterogeneous populations of hepatic macrophages contribute to SCA remains unclear. Using a combination of single-cell RNA sequencing and spatial transcriptomics via multiplexed error-robust fluorescence in situ hybridization, we identified distinct macrophage populations with diversified origins and biological functions in SCA mouse liver. We previously found that administering the von Willebrand factor (VWF)-cleaving protease ADAMTS13 alleviated vaso-occlusive episode in mice with SCA. Here, we discovered that the ADAMTS13-cleaved VWF was cleared from the circulation by a Clec4f+Marcohigh macrophage subset in a desialylation-dependent manner in the liver. In addition, sickle erythrocytes were phagocytized predominantly by Clec4f+Marcohigh macrophages. Depletion of macrophages not only abolished the protective effect of ADAMTS13 but exacerbated vaso-occlusive episode in mice with SCA. Furthermore, promoting macrophage-mediated VWF clearance reduced vaso-occlusion in SCA mice. Our study demonstrates that hepatic macrophages are important in the pathogenesis of SCA, and efficient clearance of VWF by hepatic macrophages is critical for the protective effect of ADAMTS13 in SCA mice.
Collapse
Affiliation(s)
- Huiping Shi
- Cardiovascular Biology Research Program, Oklahoma Medical Research Foundation, Oklahoma City, OK
- Department of Biochemistry and Molecular Biology, University of Oklahoma Health Sciences Center, Oklahoma City, OK
| | - Liang Gao
- Cardiovascular Biology Research Program, Oklahoma Medical Research Foundation, Oklahoma City, OK
| | - Nicole Kirby
- Cardiovascular Biology Research Program, Oklahoma Medical Research Foundation, Oklahoma City, OK
| | - Bojing Shao
- Cardiovascular Biology Research Program, Oklahoma Medical Research Foundation, Oklahoma City, OK
| | - Xindi Shan
- Cardiovascular Biology Research Program, Oklahoma Medical Research Foundation, Oklahoma City, OK
| | - Mariko Kudo
- Cardiovascular Biology Research Program, Oklahoma Medical Research Foundation, Oklahoma City, OK
| | - Robert Silasi
- Cardiovascular Biology Research Program, Oklahoma Medical Research Foundation, Oklahoma City, OK
| | - John Michael McDaniel
- Cardiovascular Biology Research Program, Oklahoma Medical Research Foundation, Oklahoma City, OK
| | - Meixiang Zhou
- Cardiovascular Biology Research Program, Oklahoma Medical Research Foundation, Oklahoma City, OK
| | - Samuel McGee
- Cardiovascular Biology Research Program, Oklahoma Medical Research Foundation, Oklahoma City, OK
| | - Wei Jing
- Cardiovascular Biology Research Program, Oklahoma Medical Research Foundation, Oklahoma City, OK
| | - Florea Lupu
- Cardiovascular Biology Research Program, Oklahoma Medical Research Foundation, Oklahoma City, OK
| | - Audrey Cleuren
- Cardiovascular Biology Research Program, Oklahoma Medical Research Foundation, Oklahoma City, OK
| | - James N. George
- Hematology-Oncology Section, Department of Medicine, University of Oklahoma Health Sciences Center, Oklahoma City, OK
| | - Lijun Xia
- Cardiovascular Biology Research Program, Oklahoma Medical Research Foundation, Oklahoma City, OK
- Department of Biochemistry and Molecular Biology, University of Oklahoma Health Sciences Center, Oklahoma City, OK
| |
Collapse
|
3
|
Pitchai A, Buhman K, Shannahan JH. Lipid mediators of inhalation exposure-induced pulmonary toxicity and inflammation. Inhal Toxicol 2024; 36:57-74. [PMID: 38422051 PMCID: PMC11022128 DOI: 10.1080/08958378.2024.2318389] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2023] [Accepted: 02/07/2024] [Indexed: 03/02/2024]
Abstract
Many inhalation exposures induce pulmonary inflammation contributing to disease progression. Inflammatory processes are actively regulated via mediators including bioactive lipids. Bioactive lipids are potent signaling molecules involved in both pro-inflammatory and resolution processes through receptor interactions. The formation and clearance of lipid signaling mediators are controlled by multiple metabolic enzymes. An imbalance of these lipids can result in exacerbated and sustained inflammatory processes which may result in pulmonary damage and disease. Dysregulation of pulmonary bioactive lipids contribute to inflammation and pulmonary toxicity following exposures. For example, inhalation of cigarette smoke induces activation of pro-inflammatory bioactive lipids such as sphingolipids, and ceramides contributing to chronic obstructive pulmonary disease. Additionally, exposure to silver nanoparticles causes dysregulation of inflammatory resolution lipids. As inflammation is a common consequence resulting from inhaled exposures and a component of numerous diseases it represents a broadly applicable target for therapeutic intervention. With new appreciation for bioactive lipids, technological advances to reliably identify and quantify lipids have occurred. In this review, we will summarize, integrate, and discuss findings from recent studies investigating the impact of inhaled exposures on pro-inflammatory and resolution lipids within the lung and their contribution to disease. Throughout the review current knowledge gaps in our understanding of bioactive lipids and their contribution to pulmonary effects of inhaled exposures will be presented. New methods being employed to detect and quantify disruption of pulmonary lipid levels following inhalation exposures will be highlighted. Lastly, we will describe how lipid dysregulation could potentially be addressed by therapeutic strategies to address inflammation.
Collapse
Affiliation(s)
- Arjun Pitchai
- School of Health Sciences, College of Health and Human Sciences, Purdue University, West Lafayette, IN, United States
| | - Kimberly Buhman
- Department of Nutrition, College of Health and Human Sciences, Purdue University, West Lafayette, IN, United States
| | - Jonathan H. Shannahan
- School of Health Sciences, College of Health and Human Sciences, Purdue University, West Lafayette, IN, United States
| |
Collapse
|
4
|
Brassolatti P, de Castro CA, dos Santos HL, Simões IT, Almeida-Lopes L, da Silva JV, Duarte FO, Luna GLF, Beck WR, Bossini PS, Anibal FDF. Systemic and local inflammatory response after implantation of biomaterial in critical bone injuries. Acta Cir Bras 2023; 38:e383823. [PMID: 37851783 PMCID: PMC10578104 DOI: 10.1590/acb383823] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2023] [Accepted: 07/25/2023] [Indexed: 10/20/2023] Open
Abstract
PURPOSE To evaluate inflammatory response in critical bone injuries after implantation of the biomaterial composed of hydroxyapatite (HA)/poly (lactic-coglycolic acid) (PLGA)/BLEED. METHODS Forty-eight male Wistar rats (280 ± 20 grams) were divided into two groups: control group (CG), in which the animals do not receive any type of treatment; and biomaterial group (BG), in which the animals received the HA/PLGA/BLEED scaffold. Critical bone injury was induced in the medial region of the skull calotte with the aid of a trephine drill 8 mm in diameter. The biomaterial was implanted in the form of 1.5-mm thick scaffolds. Serum and calotte were collected at one, three and seven days. RESULTS Biomaterial had a significant effect on the morphological structure of the bone, accelerating osteoblast activation within three days, without causing exacerbated systemic inflammation. In addition, quantitative real-time polymerase chain reaction (qRT-PCR) analysis showed that BG induced upregulation of osteogenic genes such as runt-related transcription factor 2, and stimulated genes of inflammatory pathways such as tumor necrosis factor-α, on the first day without overexpressing genes related to bone matrix degradation, such as tissue inhibitor of metalloproteinases-1 and matrix metalloproteinase-9. CONCLUSIONS The HA/PLGA/BLEED® association can be used as a bone graft to aid bone repair, as it is capable of modulating expression of important genes at this stage of the repair process.
Collapse
Affiliation(s)
- Patricia Brassolatti
- Universidade Federal de São Carlos – Postgraduate Program in Evolutionary Genetics and Molecular Biology – Department of Morphology and Pathology – São Carlos (SP) – Brazil
| | - Cynthia Aparecida de Castro
- Universidade Federal de São Carlos – Postgraduate Program in Evolutionary Genetics and Molecular Biology – Department of Morphology and Pathology – São Carlos (SP) – Brazil
| | - Hugo Leonardo dos Santos
- Universidade Federal de São Carlos – Department of Morphology and Pathology – São Carlos (SP) – Brazil
| | - Isabelle Taira Simões
- Universidade Federal de São Carlos – Department of Morphology and Pathology – São Carlos (SP) – Brazil
| | | | | | - Fernanda Oliveira Duarte
- Universidade Federal de São Carlos – Department of Morphology and Pathology – São Carlos (SP) – Brazil
| | - Genoveva Lourdes Flores Luna
- Universidade Federal de São Carlos – Postgraduate Program in Evolutionary Genetics and Molecular Biology – Department of Morphology and Pathology – São Carlos (SP) – Brazil
| | - Wladimir Rafael Beck
- Universidade Federal de São Carlos – Department of Physiological Sciences – São Carlos (SP) – Brazil
| | - Paulo Sergio Bossini
- Institute of Research and Education in the Health Area – São Carlos (SP) – Brazil
| | | |
Collapse
|
5
|
Fredman G, Khan S. Specialized pro-resolving mediators enhance the clearance of dead cells. Immunol Rev 2023; 319:151-157. [PMID: 37787174 DOI: 10.1111/imr.13278] [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: 10/04/2023]
Abstract
The failure to resolve inflammation underpins to several prevalent diseases, like atherosclerosis, and so identifying ways to boost resolution is unmet clinical needs. The resolution of inflammation is governed by several factors such as specialized pro-resolving mediators (SPMs) that counter-regulate pro-inflammatory pathways and promote tissue repair without compromising host defense. A major function of nearly all SPMs is to enhance the clearance of dead cells or efferocytosis. As such, phagocytes, such as macrophages, are essential cellular players in the resolution of inflammation because of their ability to rapidly and efficiently clear dead cells. This review highlights the role of SPMs in the clearance of apoptotic and necroptotic cells and offers insights into how targeting efferocytosis may provide new treatments for non-resolving diseases, like atherosclerosis.
Collapse
Affiliation(s)
- Gabrielle Fredman
- The Department of Molecular and Cellular Physiology, Albany Medical College, Albany, New York, USA
| | - Sayeed Khan
- The Department of Molecular and Cellular Physiology, Albany Medical College, Albany, New York, USA
| |
Collapse
|
6
|
Thornton JM, Padovani CM, Rodriguez A, Spur BW, Yin K. Lipoxin A 4 promotes antibiotic and monocyte bacterial killing in established Pseudomonas aeruginosa biofilm formed under hydrodynamic conditions. FASEB J 2023; 37:e23098. [PMID: 37462621 PMCID: PMC10694838 DOI: 10.1096/fj.202300619r] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2023] [Revised: 06/30/2023] [Accepted: 07/05/2023] [Indexed: 07/21/2023]
Abstract
Pseudomonas aeruginosa is a gram-negative, opportunistic bacteria commonly found in wounds and in lungs of immunocompromised patients. These bacteria commonly form biofilms which encapsulate the bacteria, making it difficult for antibiotics or immune cells to reach the bacterial cells. We previously reported that Lipoxin A4 (LxA4 ), a Specialized Pro-resolving Mediator, has direct effects on P. aeruginosa where it reduced biofilm formation and promoted ciprofloxacin antibiotic efficacy in a static biofilm-forming system. In the current studies, we examined the actions of LxA4 on established biofilms formed in a biofilm reactor under dynamic conditions with constant flow and shear stress. These conditions allow for biofilm growth with nutrient replenishment and for examination of bacteria within the biofilm structure. We show that LxA4 helped ciprofloxacin reduction of live/dead ratio of bacteria within the biofilm. THP-1 monocytes interacted with the biofilm to increase the number of viable bacteria within the biofilm as well as TNF-α production in the biofilm milieu, suggesting that monocyte interaction with bacterial biofilm exacerbates the inflammatory state. Pre-treatment of the THP-1 monocytes with LxA4 abolished the increase in biofilm bacteria and reduced TNF-α production. The effect of decreased biofilm bacteria was associated with increased LxA4 -induced monocyte adherence to biofilm but not increased bacteria killing suggesting that the mechanism for the reduced biofilm bacteria was due to LxA4 -mediated increase in adherence to biofilm. These results suggest that LxA4 can help antibiotic efficacy and promote monocyte activity against established P. aeruginosa biofilm formed under hydrodynamic conditions.
Collapse
Affiliation(s)
- Julianne M. Thornton
- Department of Cell Biology and Neuroscience, Rowan-Virtua School of Osteopathic Medicine Stratford, NJ, USA 08084
| | - Cristina M. Padovani
- Department of Cell Biology and Neuroscience, Rowan-Virtua School of Osteopathic Medicine Stratford, NJ, USA 08084
| | - Ana Rodriguez
- Department of Cell Biology and Neuroscience, Rowan-Virtua School of Osteopathic Medicine Stratford, NJ, USA 08084
| | - Bernd W. Spur
- Department of Cell Biology and Neuroscience, Rowan-Virtua School of Osteopathic Medicine Stratford, NJ, USA 08084
| | - Kingsley Yin
- Department of Cell Biology and Neuroscience, Rowan-Virtua School of Osteopathic Medicine Stratford, NJ, USA 08084
| |
Collapse
|
7
|
Cartwright IM, Colgan SP. The hypoxic tissue microenvironment as a driver of mucosal inflammatory resolution. Front Immunol 2023; 14:1124774. [PMID: 36742292 PMCID: PMC9890178 DOI: 10.3389/fimmu.2023.1124774] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2022] [Accepted: 01/06/2023] [Indexed: 01/19/2023] Open
Abstract
On the backdrop of all acute inflammatory processes lies the activation of the resolution response. Recent years have witnessed an emerging interest in defining molecular factors that influence the resolution of inflammation. A keystone feature of the mucosal inflammatory microenvironment is hypoxia. The gastrointestinal tract, particularly the colon, exists in a state of physiological hypoxia and during active inflammation, this hypoxic state is enhanced as a result of infiltrating leukocyte oxygen consumption and the activation of oxygen consuming enzymes. Most evidence suggests that mucosal hypoxia promotes the active resolution of inflammation through a variety of mechanisms, including extracellular acidification, purine biosynthesis/salvage, the generation of specialized pro-resolving lipid mediators (ie. resolvins) and altered chemokine/cytokine expression. It is now appreciated that infiltrating innate immune cells (neutrophils, eosinophils, macrophages) have an important role in molding the tissue microenvironment to program an active resolution response. Structural or functional dysregulation of this inflammatory microenvironment can result in the loss of tissue homeostasis and ultimately progression toward chronicity. In this review, we will discuss how inflammatory hypoxia drives mucosal inflammatory resolution and its impact on other microenvironmental factors that influence resolution.
Collapse
Affiliation(s)
- Ian M. Cartwright
- Division of Gastroenterology and Hepatology, University of Colorado Anschutz Medical Campus, Aurora, CO, United States
- Department of Medicine, University of Colorado School of Medicine, Aurora, CO, United States
- Mucosal Inflammation Program, University of Colorado School of Medicine, Aurora, CO, United States
- Rocky Mountain Regional Veterans Affairs Medical Center, Aurora, CO, United States
| | - Sean P. Colgan
- Division of Gastroenterology and Hepatology, University of Colorado Anschutz Medical Campus, Aurora, CO, United States
- Department of Medicine, University of Colorado School of Medicine, Aurora, CO, United States
- Mucosal Inflammation Program, University of Colorado School of Medicine, Aurora, CO, United States
- Rocky Mountain Regional Veterans Affairs Medical Center, Aurora, CO, United States
| |
Collapse
|
8
|
Feng L, Weng J, Yao C, Wang R, Wang N, Zhang Y, Tanaka Y, Su L. Extracellular Vesicles Derived from SIPA1high Breast Cancer Cells Enhance Macrophage Infiltration and Cancer Metastasis through Myosin-9. BIOLOGY 2022; 11:biology11040543. [PMID: 35453742 PMCID: PMC9032110 DOI: 10.3390/biology11040543] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/18/2022] [Revised: 03/28/2022] [Accepted: 03/29/2022] [Indexed: 12/24/2022]
Abstract
Simple Summary The high expression of signal-induced proliferation-associated 1 (SIPA1) in breast cancer could aggravate cancer cell metastasis, but how the tumour microenvironment is involved in this incident is unknown. In this study, we investigated whether breast cancer cells with high SIPA1 expression recruited macrophages into the tumour microenvironment. We also found that extracellular vesicles (EVs) derived from MDA-MB-231 cells significantly enhanced macrophage migration, compared with that from SIPA1-knockdown MDA-MB-231 cells both in vitro and in vivo. In terms of the mechanism, SIPA1 in cancer cells modulated the key protein myosin-9 in EVs and promoted macrophage infiltration via EVs. We confirmed that either down-regulating SIPA1 expression or blocking myosin-9 by its inhibitor, blebbistatin, led to the suppression of macrophage infiltration. These findings contribute to a deep understanding of how SIPA1 regulates the tumour microenvironment in breast cancer to facilitate tumour metastasis and provide a basis for the development of therapeutics against breast cancer metastasis. Abstract Tumour cell metastasis can be genetically regulated by proteins contained in cancer cell-derived extracellular vesicles (EVs) released to the tumour microenvironment. Here, we found that the number of infiltrated macrophages was positively correlated with the expression of signal-induced proliferation-associated 1 (SIPA1) in invasive breast ductal carcinoma tissues and MDA-MB-231 xenograft tumours. EVs derived from MDA-MB-231 cells (231-EVs) significantly enhanced macrophage migration, compared with that from SIPA1-knockdown MDA-MB-231 cells (231/si-EVs) both in vitro and in vivo. We revealed that SIPA1 promoted the transcription of MYH9, which encodes myosin-9, and up-regulated the expression level of myosin-9 in breast cancer cells and their EVs. We also found that blocking myosin-9 by either down-regulating SIPA1 expression or blebbistatin treatment led to the suppression of macrophage infiltration. Survival analysis showed that breast cancer patients with high expression of SIPA1 and MYH9 molecules had worse relapse-free survival (p = 0.028). In summary, SIPA1high breast cancer can enhance macrophage infiltration through EVs enriched with myosin-9, which might aggravate the malignancy of breast cancer.
Collapse
Affiliation(s)
- Lingyun Feng
- Key Laboratory of Molecular Biophysics of Ministry of Education, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan 430074, China; (L.F.); (J.W.); (C.Y.); (R.W.); (N.W.)
| | - Jun Weng
- Key Laboratory of Molecular Biophysics of Ministry of Education, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan 430074, China; (L.F.); (J.W.); (C.Y.); (R.W.); (N.W.)
| | - Chenguang Yao
- Key Laboratory of Molecular Biophysics of Ministry of Education, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan 430074, China; (L.F.); (J.W.); (C.Y.); (R.W.); (N.W.)
| | - Ruyuan Wang
- Key Laboratory of Molecular Biophysics of Ministry of Education, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan 430074, China; (L.F.); (J.W.); (C.Y.); (R.W.); (N.W.)
| | - Ning Wang
- Key Laboratory of Molecular Biophysics of Ministry of Education, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan 430074, China; (L.F.); (J.W.); (C.Y.); (R.W.); (N.W.)
| | - Yilei Zhang
- The Institute of Molecular and Translational Medicine, Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Xi’an Jiaotong University Health Science Center, Xi’an 710061, China;
| | - Yoshimasa Tanaka
- Center for Medical Innovation, Nagasaki University, 1-7-1, Sakamoto, Nagasaki 852-8588, Japan
- Correspondence: (Y.T.); (L.S.); Tel.: +81-95-819-7063 (Y.T.); +86-27-8779-2024 (L.S.); Fax: +81-95-819-2189 (Y.T.); +86-27-8779-2072 (L.S.)
| | - Li Su
- Key Laboratory of Molecular Biophysics of Ministry of Education, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan 430074, China; (L.F.); (J.W.); (C.Y.); (R.W.); (N.W.)
- Correspondence: (Y.T.); (L.S.); Tel.: +81-95-819-7063 (Y.T.); +86-27-8779-2024 (L.S.); Fax: +81-95-819-2189 (Y.T.); +86-27-8779-2072 (L.S.)
| |
Collapse
|
9
|
Nguyen TN, Siddiqui G, Veldhuis NA, Poole DP. Diverse Roles of TRPV4 in Macrophages: A Need for Unbiased Profiling. Front Immunol 2022; 12:828115. [PMID: 35126384 PMCID: PMC8811046 DOI: 10.3389/fimmu.2021.828115] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2021] [Accepted: 12/24/2021] [Indexed: 12/27/2022] Open
Abstract
Transient receptor potential vanilloid 4 (TRPV4) is a non-selective mechanosensitive ion channel expressed by various macrophage populations. Recent reports have characterized the role of TRPV4 in shaping the activity and phenotype of macrophages to influence the innate immune response to pathogen exposure and inflammation. TRPV4 has been studied extensively in the context of inflammation and inflammatory pain. Although TRPV4 activity has been generally described as pro-inflammatory, emerging evidence suggests a more complex role where this channel may also contribute to anti-inflammatory activities. However, detailed understanding of how TRPV4 may influence the initiation, maintenance, and resolution of inflammatory disease remains limited. This review highlights recent insights into the cellular processes through which TRPV4 contributes to pathological conditions and immune processes, with a focus on macrophage biology. The potential use of high-throughput and omics methods as an unbiased approach for studying the functional outcomes of TRPV4 activation is also discussed.
Collapse
Affiliation(s)
- Thanh-Nhan Nguyen
- Drug Discovery Biology Theme, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, VIC, Australia
- Australian Research Council (ARC) Centre of Excellence in Convergent Bio-Nano Science & Technology, Monash University, Parkville, VIC, Australia
| | - Ghizal Siddiqui
- Drug Delivery, Disposition and Dynamics Theme, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, VIC, Australia
| | - Nicholas A. Veldhuis
- Drug Discovery Biology Theme, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, VIC, Australia
- Australian Research Council (ARC) Centre of Excellence in Convergent Bio-Nano Science & Technology, Monash University, Parkville, VIC, Australia
- *Correspondence: Daniel P. Poole, ; Nicholas A. Veldhuis,
| | - Daniel P. Poole
- Drug Discovery Biology Theme, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, VIC, Australia
- Australian Research Council (ARC) Centre of Excellence in Convergent Bio-Nano Science & Technology, Monash University, Parkville, VIC, Australia
- *Correspondence: Daniel P. Poole, ; Nicholas A. Veldhuis,
| |
Collapse
|
10
|
Thornton JM, Yin K. Role of Specialized Pro-Resolving Mediators in Modifying Host Defense and Decreasing Bacterial Virulence. Molecules 2021; 26:molecules26226970. [PMID: 34834062 PMCID: PMC8618792 DOI: 10.3390/molecules26226970] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2021] [Revised: 11/05/2021] [Accepted: 11/15/2021] [Indexed: 12/12/2022] Open
Abstract
Bacterial infection activates the innate immune system as part of the host’s defense against invading pathogens. Host response to bacterial pathogens includes leukocyte activation, inflammatory mediator release, phagocytosis, and killing of bacteria. An appropriate host response requires resolution. The resolution phase involves attenuation of neutrophil migration, neutrophil apoptosis, macrophage recruitment, increased phagocytosis, efferocytosis of apoptotic neutrophils, and tissue repair. Specialized Pro-resolving Mediators (SPMs) are bioactive fatty acids that were shown to be highly effective in promoting resolution of infectious inflammation and survival in several models of infection. In this review, we provide insight into the role of SPMs in active host defense mechanisms for bacterial clearance including a new mechanism of action in which an SPM acts directly to reduce bacterial virulence.
Collapse
|
11
|
Yang A, Wu Y, Yu G, Wang H. Role of specialized pro-resolving lipid mediators in pulmonary inflammation diseases: mechanisms and development. Respir Res 2021; 22:204. [PMID: 34261470 PMCID: PMC8279385 DOI: 10.1186/s12931-021-01792-y] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2020] [Accepted: 06/30/2021] [Indexed: 12/12/2022] Open
Abstract
Inflammation is an essential mechanism of various diseases. The development and resolution of inflammation are complex immune-modulation processes which induce the involvement of various types of immune cells. Specialized pro-resolving lipid mediators (SPMs) have been demonstrated to be signaling molecules in inflammation. SPMs are involved in the pathophysiology of different diseases, especially respiratory diseases, including asthma, pneumonia, and chronic obstructive pulmonary disease. All of these diseases are related to the inflammatory response and its persistence. Therefore, a deeper understanding of the mechanisms and development of inflammation in respiratory disease, and the roles of the SPM family in the resolution process, might be useful in the quest for novel therapies and preventive measures for pulmonary diseases.
Collapse
Affiliation(s)
- Ailin Yang
- Department of Respiratory Medicine, Beijing Friendship Hospital, Capital Medical University, No. 95 Yong An Road, Xicheng, , Beijing, 100050, China
| | - Yanjun Wu
- Department of Respiratory Medicine, Beijing Friendship Hospital, Capital Medical University, No. 95 Yong An Road, Xicheng, , Beijing, 100050, China
| | - Ganggang Yu
- Department of Respiratory Medicine, Beijing Friendship Hospital, Capital Medical University, No. 95 Yong An Road, Xicheng, , Beijing, 100050, China.
| | - Haoyan Wang
- Department of Respiratory Medicine, Beijing Friendship Hospital, Capital Medical University, No. 95 Yong An Road, Xicheng, , Beijing, 100050, China.
| |
Collapse
|
12
|
Decker C, Sadhu S, Fredman G. Pro-Resolving Ligands Orchestrate Phagocytosis. Front Immunol 2021; 12:660865. [PMID: 34177900 PMCID: PMC8222715 DOI: 10.3389/fimmu.2021.660865] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2021] [Accepted: 04/06/2021] [Indexed: 12/18/2022] Open
Abstract
The resolution of inflammation is a tissue protective program that is governed by several factors including specialized pro-resolving mediators (SPMs), proteins, gasses and nucleotides. Pro-resolving mediators activate counterregulatory programs to quell inflammation and promote tissue repair in a manner that does not compromise host defense. Phagocytes like neutrophils and macrophages play key roles in the resolution of inflammation because of their ability to remove debris, microbes and dead cells through processes including phagocytosis and efferocytosis. Emerging evidence suggests that failed resolution of inflammation and defective phagocytosis or efferocytosis underpins several prevalent human diseases. Therefore, understanding factors and mechanisms associated with enhancing these processes is a critical need. SPMs enhance phagocytosis and efferocytosis and this review will highlight mechanisms associated with their actions.
Collapse
Affiliation(s)
- Christa Decker
- The Department of Molecular and Cellular Physiology, Albany Medical College, Albany, NY, United States
| | - Sudeshna Sadhu
- The Department of Molecular and Cellular Physiology, Albany Medical College, Albany, NY, United States
| | - Gabrielle Fredman
- The Department of Molecular and Cellular Physiology, Albany Medical College, Albany, NY, United States
| |
Collapse
|
13
|
Thornton JM, Walker JM, Sundarasivarao PYK, Spur BW, Rodriguez A, Yin K. Lipoxin A4 promotes reduction and antibiotic efficacy against Pseudomonas aeruginosa biofilm. Prostaglandins Other Lipid Mediat 2021; 152:106505. [PMID: 33152529 PMCID: PMC7856039 DOI: 10.1016/j.prostaglandins.2020.106505] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2020] [Revised: 09/30/2020] [Accepted: 10/26/2020] [Indexed: 02/07/2023]
Abstract
Pseudomonas aeruginosa (P. aeruginosa) is an opportunistic bacterium commonly found in wound infections and airways of cystic fibrosis patients. P. aeruginosa readily forms biofilms which can reduce the efficacy of antibiotics used to eradicate the pathogen. We have previously shown that a Specialized Pro-resolving Mediator (SPM), Lipoxin A4 (LxA4) is a quorum sensing inhibitor which can reduce P. aeruginosa virulence. In this study, we examined the direct actions of LxA4 and RvD2 on P. aeruginosa biofilm formation and virulence gene expression. The influence of LxA4 on antibiotic efficacy and the combined effects on biofilm formation were also investigated. LxA4 and RvD2 reduced P. aeruginosa biofilm formation and virulence gene expression. LxA4 increased ciprofloxacin inhibition on biofilm formation but did not affect ciprofloxacin's action on non-adherent bacteria. On the other hand, LxA4 increased bacterial killing action of imipenem but did not affect imipenem's action on biofilm. We also found that LxA4 can increase ciprofloxacin's bacterial killing ability in established biofilm. Together these results suggest that LxA4 has direct effects on P. aeruginosa biofilm formation and can increase antibiotic efficacy directly.
Collapse
Affiliation(s)
- J M Thornton
- Department of Cell Biology and Neuroscience, Rowan University School of Osteopathic Medicine, Stratford, NJ, USA
| | - J M Walker
- Department of Cell Biology and Neuroscience, Rowan University School of Osteopathic Medicine, Stratford, NJ, USA
| | - P Y Kadiyam Sundarasivarao
- Department of Cell Biology and Neuroscience, Rowan University School of Osteopathic Medicine, Stratford, NJ, USA
| | - B W Spur
- Department of Cell Biology and Neuroscience, Rowan University School of Osteopathic Medicine, Stratford, NJ, USA
| | - A Rodriguez
- Department of Cell Biology and Neuroscience, Rowan University School of Osteopathic Medicine, Stratford, NJ, USA
| | - K Yin
- Department of Cell Biology and Neuroscience, Rowan University School of Osteopathic Medicine, Stratford, NJ, USA.
| |
Collapse
|
14
|
Galvão I, Melo EM, de Oliveira VLS, Vago JP, Queiroz-Junior C, de Gaetano M, Brennan E, Gahan K, Guiry PJ, Godson C, Teixeira MM. Therapeutic potential of the FPR2/ALX agonist AT-01-KG in the resolution of articular inflammation. Pharmacol Res 2021; 165:105445. [PMID: 33493655 DOI: 10.1016/j.phrs.2021.105445] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/25/2020] [Revised: 01/12/2021] [Accepted: 01/14/2021] [Indexed: 12/16/2022]
Abstract
The resolution of inflammation is a dynamic process, characterized by the biosynthesis of pro-resolving mediators, including the lipid Lipoxin A4 (LXA4). LXA4 acts on the N-formyl peptide receptor 2 (FPR2/ALX) to mediate anti-inflammatory and pro-resolving effects. In order to exploit the therapeutic potential of endogenous LXA4 in the context of inflammation we have recently developed synthetic LXA4 mimetics (sLXms) including a dimethyl-imidazole-containing FPR2/ALX agonist designated AT-01-KG. Here, we have investigated the effect of treatment with AT-01-KG in established models of articular inflammation. In a model of gout, mice were injected with MSU crystals and treated with AT-01-KG at the peak of inflammatory response. The treatment decreased the number of neutrophils in the knee exudate, an effect which was accompanied by low levels of myeloperoxidase, CXCL1 and IL-1β in periarticular tissue. AT-01-KG treatment led to reduced tissue damage and hypernociception. The effects of AT-01-KG on neutrophil accumulation were not observed in MSU treated FPR2/3-/-mice. Importantly, AT-01-KG induced resolution of articular inflammation by increasing neutrophil apoptosis and subsequent efficient efferocytosis. In a model of antigen-induced arthritis, AT-01-KG treatment also attenuated inflammatory responses. These data suggest that AT-01-KG may be a potential new therapy for neutrophilic inflammation of the joints.
Collapse
Affiliation(s)
- Izabela Galvão
- Departamento de Morfologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Eliza M Melo
- Departamento de Bioquímica e Imunologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Vivian L S de Oliveira
- Departamento de Bioquímica e Imunologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Juliana P Vago
- Departamento de Morfologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Celso Queiroz-Junior
- Departamento de Morfologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Monica de Gaetano
- UCD Diabetes Complications Research Centre, UCD Conway Institute School of Medicine, University College Dublin, Dublin, Ireland
| | - Eoin Brennan
- UCD Diabetes Complications Research Centre, UCD Conway Institute School of Medicine, University College Dublin, Dublin, Ireland
| | - Kevin Gahan
- Centre for Synthesis and Chemical Biology, School of Chemistry, University College Dublin, Dublin, Ireland
| | - Patrick J Guiry
- Centre for Synthesis and Chemical Biology, School of Chemistry, University College Dublin, Dublin, Ireland
| | - Catherine Godson
- UCD Diabetes Complications Research Centre, UCD Conway Institute School of Medicine, University College Dublin, Dublin, Ireland
| | - Mauro M Teixeira
- Departamento de Bioquímica e Imunologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil.
| |
Collapse
|
15
|
Lee CH. Role of specialized pro-resolving lipid mediators and their receptors in virus infection: a promising therapeutic strategy for SARS-CoV-2 cytokine storm. Arch Pharm Res 2021; 44:84-98. [PMID: 33398691 PMCID: PMC7781431 DOI: 10.1007/s12272-020-01299-y] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2020] [Accepted: 11/23/2020] [Indexed: 02/07/2023]
Abstract
Unexpected viral infections outbreaks, significantly affect human health, leading to increased mortality and life disruption. Among them is the severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) which emerged as a deadly pandemic, calling for intense research efforts on its pathogenicity mechanism and development of therapeutic strategies. In the SARS-CoV-2 cytokine storm, systemic inflammation has been associated with severe illness and mortality. Recent studies have demonstrated special pro-resolving lipids mediators (SPMs) lipoxins, resolvins, maresins, and protectins as potential therapeutic options for abnormal viral-triggered inflammation. Pro-resolving lipids mediators have shown great promise for the treatment of Herpes simplex virus, respiratory syncytial virus, human immunodeficiency virus, and hepatitis C virus. Based on this, studies are being conducted on their therapeutic effects in SARS-CoV-2 infection. In this review, we discussed SPMs and reviewed evidence from recent studies on SPMs as therapeutic options for viral infections, including SARS-CoV2. Based on our analysis of the previous study, we argue that SPMs are a potential treatment for SARS-CoV-2 infection and other viral infections. We expect further research on how SPMs modulate viral-triggered inflammation through G-protein-coupled receptors (GPCRs), and chemical stability and druggability of SPMs.
Collapse
Affiliation(s)
- Chang Hoon Lee
- College of Pharmacy, Dongguk University, Seoul, 100-715, Republic of Korea.
| |
Collapse
|
16
|
Zhou W, Liu Y, Gao Y, Cheng Y, Chang R, Li X, Zhou Y, Wang S, Liang L, Duan C, Zhang C. MICAL2 is a novel nucleocytoplasmic shuttling protein promoting cancer invasion and growth of lung adenocarcinoma. Cancer Lett 2020; 483:75-86. [PMID: 32360180 DOI: 10.1016/j.canlet.2020.04.019] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2019] [Revised: 04/02/2020] [Accepted: 04/22/2020] [Indexed: 12/16/2022]
Abstract
MICAL2 is a tumor-promoting factor involved in cell migration, invasion, deformation, and proliferation not yet fully explored in lung adenocarcinoma (LUAD). This study demonstrated that MICAL2 was overexpressed and cytoplasm-enriched in LUAD tissues. Moreover, high cytoplasmic MICAL2 and/or total MICAL2 expression levels were positively correlated with lymphatic metastasis and shorter overall survival in LUAD patients. MICAL2 promoted LUAD cell proliferation, migration, invasion, and epithelial to mesenchymal transition-all of which involved the AKT and myosin-9 pathways. Furthermore, MICAL2 was identified as a nucleoplasm shuttling protein dependent on myosin-9 and its C-terminal fragment. MICAL2-ΔC-enriched in the nucleus-had less impact on tumor malignancy in LUAD cells in vitro and in vivo. Tumor promotion by MICAL2 was reduced by nuclear-export inhibitor, myosin-9 inhibitor, or si-myosin-9-all of which effectively inhibited MICAL2's nuclear export. Finally, the expression and subcellular location as well as clinical significance of MICAL2 and myosin-9 were analyzed across TCGA data and LUAD tissue arrays. Our data revealed that MICAL2 overexpression and nuclear export were associated with cancer progression; inhibiting its expression and/or nuclear export may provide a new target for LUAD therapy.
Collapse
Affiliation(s)
- Wolong Zhou
- Department of Thoracic Surgery, Xiangya Hospital, Central South University, Changsha, 410008, PR China
| | - Yuanqi Liu
- Department of Thoracic Surgery, Xiangya Hospital, Central South University, Changsha, 410008, PR China
| | - Yang Gao
- Department of Thoracic Surgery, Xiangya Hospital, Central South University, Changsha, 410008, PR China
| | - Yuanda Cheng
- Department of Thoracic Surgery, Xiangya Hospital, Central South University, Changsha, 410008, PR China
| | - Ruimin Chang
- Department of Thoracic Surgery, Xiangya Hospital, Central South University, Changsha, 410008, PR China
| | - Xizhe Li
- Department of Thoracic Surgery, Xiangya Hospital, Central South University, Changsha, 410008, PR China
| | - Yanwu Zhou
- Department of Thoracic Surgery, Xiangya Hospital, Central South University, Changsha, 410008, PR China
| | - Shaoqiang Wang
- Department of Thoracic Surgery, Affiliated Hospital of Jining Medical College, Jining Medical College, Jining, 272000, PR China
| | - Lubiao Liang
- Department of Thoracic Surgery, Affiliated Hospital of Zunyi Medical University, Zunyi Medical University, Zunyi, 563000, PR China
| | - Chaojun Duan
- Department of Thoracic Surgery, Xiangya Hospital, Central South University, Changsha, 410008, PR China; National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, 410008, PR China.
| | - Chunfang Zhang
- Department of Thoracic Surgery, Xiangya Hospital, Central South University, Changsha, 410008, PR China; National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, 410008, PR China; Hunan Engineering Research Center for Pulmonary Nodules Precise Diagnosis and Treatment, Xiangya Hospital, Central South University, Changsha, 410008, PR China.
| |
Collapse
|
17
|
GSK3: A Kinase Balancing Promotion and Resolution of Inflammation. Cells 2020; 9:cells9040820. [PMID: 32231133 PMCID: PMC7226814 DOI: 10.3390/cells9040820] [Citation(s) in RCA: 67] [Impact Index Per Article: 16.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2020] [Revised: 03/25/2020] [Accepted: 03/26/2020] [Indexed: 12/11/2022] Open
Abstract
GSK3 has been implicated for years in the regulation of inflammation and addressed in a plethora of scientific reports using a variety of experimental (disease) models and approaches. However, the specific role of GSK3 in the inflammatory process is still not fully understood and controversially discussed. Following a detailed overview of structure, function, and various regulatory levels, this review focusses on the immunoregulatory functions of GSK3, including the current knowledge obtained from animal models. Its impact on pro-inflammatory cytokine/chemokine profiles, bacterial/viral infections, and the modulation of associated pro-inflammatory transcriptional and signaling pathways is discussed. Moreover, GSK3 contributes to the resolution of inflammation on multiple levels, e.g., via the regulation of pro-resolving mediators, the clearance of apoptotic immune cells, and tissue repair processes. The influence of GSK3 on the development of different forms of stimulation tolerance is also addressed. Collectively, the role of GSK3 as a kinase balancing the initiation/perpetuation and the amelioration/resolution of inflammation is highlighted.
Collapse
|
18
|
Das UN. Molecular pathobiology of scleritis and its therapeutic implications. Int J Ophthalmol 2020; 13:163-175. [PMID: 31956585 DOI: 10.18240/ijo.2020.01.23] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2019] [Accepted: 09/12/2019] [Indexed: 11/23/2022] Open
Abstract
Scleritis and other autoimmune diseases are characterized by an imbalance in the levels of pro-inflammatory and anti-inflammatory molecules with the balance tilted more towards the former due to the failure of recognition of self. The triggering of inflammatory process could be ascribed to the presence of cytoplasmic DNA/chromatin that leads to activation of cytosolic DNA-sensing cGAS-STING (cyclic GMP-AMP synthase linked to stimulator of interferon genes) pathway and enhanced expression of NF-κB that results in an increase in the production of pro-inflammatory bioactive lipids. Bioactive lipids gamma-linolenic acid (GLA), dihomo-GLA (DGLA), prostaglandin E1 (PGE1), prostacyclin (PGI2) and lipoxin A4, resolvins, protectins and maresins have anti-inflammatory actions, bind to DNA to render it non-antigenic and are decreased in autoimmune diseases. These results suggest that efforts designed to enhance the production of anti-inflammatory bioactive lipids may form a new approach to autoimmune diseases. Local injection or infusion of lipoxins, resolvins, protectins and maresins or their precursors such as arachidonic acid may be exploited in the prevention and management of autoimmune diseases including scleritis, uveitis and lupus/rheumatoid arthritis.
Collapse
Affiliation(s)
- Undurti N Das
- UND Life Sciences, Battle Ground, WA 98604, USA.,BioScience Research Centre and Department of Medicine, GVP Medical College and Hospital, Visakhapatnam 530048, India
| |
Collapse
|
19
|
Maruyama M, Rhee C, Utsunomiya T, Zhang N, Ueno M, Yao Z, Goodman SB. Modulation of the Inflammatory Response and Bone Healing. Front Endocrinol (Lausanne) 2020; 11:386. [PMID: 32655495 PMCID: PMC7325942 DOI: 10.3389/fendo.2020.00386] [Citation(s) in RCA: 214] [Impact Index Per Article: 53.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/13/2020] [Accepted: 05/14/2020] [Indexed: 01/08/2023] Open
Abstract
The optimal treatment for complex fractures and large bone defects is an important unsolved issue in orthopedics and related specialties. Approximately 5-10% of fractures fail to heal and develop non-unions. Bone healing can be characterized by three partially overlapping phases: the inflammatory phase, the repair phase, and the remodeling phase. Eventual healing is highly dependent on the initial inflammatory phase, which is affected by both the local and systemic responses to the injurious stimulus. Furthermore, immune cells and mesenchymal stromal cells (MSCs) participate in critical inter-cellular communication or crosstalk to modulate bone healing. Deficiencies in this inter-cellular exchange, inhibition of the natural processes of acute inflammation, and its resolution, or chronic inflammation due to a persistent adverse stimulus can lead to impaired fracture healing. Thus, an initial and optimal transient stage of acute inflammation is one of the key factors for successful, robust bone healing. Recent studies demonstrated the therapeutic potential of immunomodulation for bone healing by the preconditioning of MSCs to empower their immunosuppressive properties. Preconditioned MSCs (also known as "primed/ licensed/ activated" MSCs) are cultured first with pro-inflammatory cytokines (e.g., TNFα and IL17A) or exposed to hypoxic conditions to mimic the inflammatory environment prior to their intended application. Another approach of immunomodulation for bone healing is the resolution of inflammation with anti-inflammatory cytokines such as IL4, IL10, and IL13. In this review, we summarize the principles of inflammation and bone healing and provide an update on cellular interactions and immunomodulation for optimal bone healing.
Collapse
Affiliation(s)
- Masahiro Maruyama
- Department of Orthopaedic Surgery, Stanford University, Stanford, CA, United States
| | - Claire Rhee
- Department of Orthopaedic Surgery, Stanford University, Stanford, CA, United States
| | - Takeshi Utsunomiya
- Department of Orthopaedic Surgery, Stanford University, Stanford, CA, United States
| | - Ning Zhang
- Department of Orthopaedic Surgery, Stanford University, Stanford, CA, United States
| | - Masaya Ueno
- Department of Orthopaedic Surgery, Stanford University, Stanford, CA, United States
| | - Zhenyu Yao
- Department of Orthopaedic Surgery, Stanford University, Stanford, CA, United States
| | - Stuart B. Goodman
- Department of Orthopaedic Surgery, Stanford University, Stanford, CA, United States
- Department of Bioengineering, Stanford University, Stanford, CA, United States
- *Correspondence: Stuart B. Goodman
| |
Collapse
|
20
|
Yang JX, Li M, Chen XO, Lian QQ, Wang Q, Gao F, Jin SW, Zheng SX. Lipoxin A 4 ameliorates lipopolysaccharide-induced lung injury through stimulating epithelial proliferation, reducing epithelial cell apoptosis and inhibits epithelial-mesenchymal transition. Respir Res 2019; 20:192. [PMID: 31438948 PMCID: PMC6704532 DOI: 10.1186/s12931-019-1158-z] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2019] [Accepted: 08/06/2019] [Indexed: 02/07/2023] Open
Abstract
Background Acute respiratory distress syndrome (ARDS) is characterized by alveolar epithelial disruption. Lipoxins (LXs), as so-called “braking signals” of inflammation, are the first mediators identified to have dual anti-inflammatory and inflammatory pro-resolving properties. Methods In vivo, lipoxinA4 was administrated intraperitoneally with 1 μg/per mouse after intra-tracheal LPS administration (10 mg/kg). Apoptosis, proliferation and epithelial–mesenchymal transition of AT II cells were measured by immunofluorescence. In vitro, primary human alveolar type II cells were used to model the effects of lipoxin A4 upon proliferation, apoptosis and epithelial–mesenchymal transition. Results In vivo, lipoxin A4 markedly promoted alveolar epithelial type II cells (AT II cells) proliferation, inhibited AT II cells apoptosis, reduced cleaved caspase-3 expression and epithelial–mesenchymal transition, with the outcome of attenuated LPS-induced lung injury. In vitro, lipoxin A4 increased primary human alveolar epithelial type II cells (AT II cells) proliferation and reduced LPS induced AT II cells apoptosis. LipoxinA4 also inhibited epithelial mesenchymal transition in response to TGF-β1, which was lipoxin receptor dependent. In addition, Smad3 inhibitor (Sis3) and PI3K inhibitor (LY294002) treatment abolished the inhibitory effects of lipoxinA4 on the epithelial mesenchymal transition of primary human AT II cells. Lipoxin A4 significantly downregulated the expressions of p-AKT and p-Smad stimulated by TGF-β1 in primary human AT II cells. Conclusion LipoxinA4 attenuates lung injury via stimulating epithelial cell proliferation, reducing epithelial cell apoptosis and inhibits epithelial–mesenchymal transition. Electronic supplementary material The online version of this article (10.1186/s12931-019-1158-z) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Jing-Xiang Yang
- Department of Anesthesia and Critical Care, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Zhejiang, 325027, China
| | - Ming Li
- Department of Anesthesia and Critical Care, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Zhejiang, 325027, China
| | - Xin-Ou Chen
- Department of Anesthesia and Critical Care, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Zhejiang, 325027, China
| | - Qing-Quan Lian
- Department of Anesthesia and Critical Care, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Zhejiang, 325027, China
| | - Qian Wang
- Department of Anesthesia and Critical Care, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Zhejiang, 325027, China
| | - Fang Gao
- Department of Anesthesia and Critical Care, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Zhejiang, 325027, China. .,Birmingham Acute Care Research Group, Institute of Inflammation and Aging, University of Birmingham, Birmingham, B15 2TT, UK.
| | - Sheng-Wei Jin
- Department of Anesthesia and Critical Care, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Zhejiang, 325027, China.
| | - Sheng-Xing Zheng
- Department of Anesthesia and Critical Care, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Zhejiang, 325027, China.
| |
Collapse
|
21
|
Arienti S, Barth ND, Dorward DA, Rossi AG, Dransfield I. Regulation of Apoptotic Cell Clearance During Resolution of Inflammation. Front Pharmacol 2019; 10:891. [PMID: 31456686 PMCID: PMC6701246 DOI: 10.3389/fphar.2019.00891] [Citation(s) in RCA: 41] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2019] [Accepted: 07/15/2019] [Indexed: 01/17/2023] Open
Abstract
Programmed cell death (apoptosis) has an important role in the maintenance of tissue homeostasis as well as the progression and ultimate resolution of inflammation. During apoptosis, the cell undergoes morphological and biochemical changes [e.g., phosphatidylserine (PtdSer) exposure, caspase activation, changes in mitochondrial membrane potential and DNA cleavage] that act to shut down cellular function and mark the cell for phagocytic clearance. Tissue phagocytes bind and internalize apoptotic cells, bodies, and vesicles, providing a mechanism for the safe disposal of apoptotic material. Phagocytic removal of apoptotic cells before they undergo secondary necrosis reduces the potential for bystander damage to adjacent tissue and importantly initiates signaling pathways within the phagocytic cell that act to dampen inflammation. In a pathological context, excessive apoptosis or failure to clear apoptotic material results in secondary necrosis with the release of pro-inflammatory intracellular contents. In this review, we consider some of the mechanisms by which phagocytosis of apoptotic cells can be controlled. We suggest that matching apoptotic cell load with the capacity for apoptotic cell clearance within tissues may be important for therapeutic strategies that target the apoptotic process for treatment of inflammatory disease.
Collapse
Affiliation(s)
- Simone Arienti
- Centre for Inflammation Research, Queen's Medical Research Institute, University of Edinburgh, Edinburgh, United Kingdom
| | - Nicole D Barth
- Centre for Inflammation Research, Queen's Medical Research Institute, University of Edinburgh, Edinburgh, United Kingdom
| | - David A Dorward
- Centre for Inflammation Research, Queen's Medical Research Institute, University of Edinburgh, Edinburgh, United Kingdom
| | - Adriano G Rossi
- Centre for Inflammation Research, Queen's Medical Research Institute, University of Edinburgh, Edinburgh, United Kingdom
| | - Ian Dransfield
- Centre for Inflammation Research, Queen's Medical Research Institute, University of Edinburgh, Edinburgh, United Kingdom
| |
Collapse
|
22
|
Resolvin D1 promotes the targeting and clearance of necroptotic cells. Cell Death Differ 2019; 27:525-539. [PMID: 31222041 DOI: 10.1038/s41418-019-0370-1] [Citation(s) in RCA: 88] [Impact Index Per Article: 17.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2019] [Revised: 04/30/2019] [Accepted: 05/28/2019] [Indexed: 02/07/2023] Open
Abstract
Inflammation-resolution is a protective response that is mediated by specialized pro-resolving mediators (SPMs). The clearance of dead cells or efferocytosis is a critical cellular program of inflammation-resolution. Impaired efferocytosis can lead to tissue damage in prevalent human diseases, like atherosclerosis. Therefore understanding mechanisms associated with swift clearance of dead cells is of utmost clinical importance. Recently, the accumulation of necroptotic cells (NCs) was observed in human plaques and we postulated that this is due to defective clearance programs. Here we present evidence that NCs are inefficiently taken up by macrophages because they have increased surface expression of a well-known "don't eat me" signal called CD47. High levels of CD47 on NCs stimulated RhoA-pMLC signaling in macrophages that promoted "nibbling", rather than whole-cell engulfment of NCs. Anti-CD47 blocking antibodies limited RhoA-p-MLC signaling and promoted whole-cell NC engulfment. Treatment with anti-CD47 blocking antibodies to Ldlr-/- mice with established atherosclerosis decreased necrotic cores, limited the accumulation of plaque NCs and increased lesional SPMs, including Resolvin D1 (RvD1) compared with IgG controls. Mechanistically, RvD1 promoted whole-cell engulfment of NCs by decreasing RhoA signaling and activating CDC42. RvD1 specifically targeted NCs for engulfment by facilitating the release of the well-known "eat me signal" called calreticulin from macrophages in a CDC42 dependent manner. Lastly, RvD1 enhanced the clearance of NCs in advanced murine plaques. Together, these results suggest new molecules and signaling associated with the clearance of NCs, provide a new paradigm for the regulation of inflammation-resolution, and offer a potential treatment strategy for diseases where NCs underpin the pathology.
Collapse
|
23
|
Cartwright JA, Lucas CD, Rossi AG. Inflammation Resolution and the Induction of Granulocyte Apoptosis by Cyclin-Dependent Kinase Inhibitor Drugs. Front Pharmacol 2019; 10:55. [PMID: 30886578 PMCID: PMC6389705 DOI: 10.3389/fphar.2019.00055] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2018] [Accepted: 01/18/2019] [Indexed: 12/14/2022] Open
Abstract
Inflammation is a necessary dynamic tissue response to injury or infection and it's resolution is essential to return tissue homeostasis and function. Defective or dysregulated inflammation resolution contributes significantly to the pathogenesis of many, often common and challenging to treat human conditions. The transition of inflammation to resolution is an active process, involving the clearance of inflammatory cells (granulocytes), a change of mediators and their receptors, and prevention of further inflammatory cell infiltration. This review focuses on the use of cyclin dependent kinase inhibitor drugs to pharmacologically target this inflammatory resolution switch, specifically through inducing granulocyte apoptosis and phagocytic clearance of apoptotic cells (efferocytosis). The key processes and pathways required for granulocyte apoptosis, recruitment of phagocytes and mechanisms of engulfment are discussed along with the cumulating evidence for cyclin dependent kinase inhibitor drugs as pro-resolution therapeutics.
Collapse
Affiliation(s)
- Jennifer A. Cartwright
- Queen's Medical Research Institute, University of Edinburgh Centre for Inflammation Research, Edinburgh BioQuarter, Edinburgh, United Kingdom
- MRC Centre for Regenerative Medicine, University of Edinburgh, Edinburgh BioQuarter, Edinburgh, United Kingdom
| | - Christopher D. Lucas
- Queen's Medical Research Institute, University of Edinburgh Centre for Inflammation Research, Edinburgh BioQuarter, Edinburgh, United Kingdom
| | - Adriano G. Rossi
- Queen's Medical Research Institute, University of Edinburgh Centre for Inflammation Research, Edinburgh BioQuarter, Edinburgh, United Kingdom
| |
Collapse
|
24
|
The genetics and molecular pathogenesis of systemic lupus erythematosus (SLE) in populations of different ancestry. Gene 2018; 668:59-72. [DOI: 10.1016/j.gene.2018.05.041] [Citation(s) in RCA: 72] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2018] [Accepted: 05/13/2018] [Indexed: 01/21/2023]
|
25
|
Chen L, Deng H, Cui H, Fang J, Zuo Z, Deng J, Li Y, Wang X, Zhao L. Inflammatory responses and inflammation-associated diseases in organs. Oncotarget 2018; 9:7204-7218. [PMID: 29467962 PMCID: PMC5805548 DOI: 10.18632/oncotarget.23208] [Citation(s) in RCA: 2399] [Impact Index Per Article: 399.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2017] [Accepted: 11/03/2017] [Indexed: 02/07/2023] Open
Abstract
Inflammation is a biological response of the immune system that can be triggered by a variety of factors, including pathogens, damaged cells and toxic compounds. These factors may induce acute and/or chronic inflammatory responses in the heart, pancreas, liver, kidney, lung, brain, intestinal tract and reproductive system, potentially leading to tissue damage or disease. Both infectious and non-infectious agents and cell damage activate inflammatory cells and trigger inflammatory signaling pathways, most commonly the NF-κB, MAPK, and JAK-STAT pathways. Here, we review inflammatory responses within organs, focusing on the etiology of inflammation, inflammatory response mechanisms, resolution of inflammation, and organ-specific inflammatory responses.
Collapse
Affiliation(s)
- Linlin Chen
- College of Veterinary Medicine, Sichuan Agricultural University, Wenjiang, Chengdu 611130, China
| | - Huidan Deng
- College of Veterinary Medicine, Sichuan Agricultural University, Wenjiang, Chengdu 611130, China
| | - Hengmin Cui
- College of Veterinary Medicine, Sichuan Agricultural University, Wenjiang, Chengdu 611130, China
- Key Laboratory of Animal Diseases and Environmental Hazards of Sichuan Province, Sichuan Agriculture University, Wenjiang, Chengdu 611130, China
| | - Jing Fang
- College of Veterinary Medicine, Sichuan Agricultural University, Wenjiang, Chengdu 611130, China
- Key Laboratory of Animal Diseases and Environmental Hazards of Sichuan Province, Sichuan Agriculture University, Wenjiang, Chengdu 611130, China
| | - Zhicai Zuo
- College of Veterinary Medicine, Sichuan Agricultural University, Wenjiang, Chengdu 611130, China
- Key Laboratory of Animal Diseases and Environmental Hazards of Sichuan Province, Sichuan Agriculture University, Wenjiang, Chengdu 611130, China
| | - Junliang Deng
- College of Veterinary Medicine, Sichuan Agricultural University, Wenjiang, Chengdu 611130, China
- Key Laboratory of Animal Diseases and Environmental Hazards of Sichuan Province, Sichuan Agriculture University, Wenjiang, Chengdu 611130, China
| | - Yinglun Li
- College of Veterinary Medicine, Sichuan Agricultural University, Wenjiang, Chengdu 611130, China
- Key Laboratory of Animal Diseases and Environmental Hazards of Sichuan Province, Sichuan Agriculture University, Wenjiang, Chengdu 611130, China
| | - Xun Wang
- College of Veterinary Medicine, Sichuan Agricultural University, Wenjiang, Chengdu 611130, China
- Key Laboratory of Animal Diseases and Environmental Hazards of Sichuan Province, Sichuan Agriculture University, Wenjiang, Chengdu 611130, China
| | - Ling Zhao
- College of Veterinary Medicine, Sichuan Agricultural University, Wenjiang, Chengdu 611130, China
- Key Laboratory of Animal Diseases and Environmental Hazards of Sichuan Province, Sichuan Agriculture University, Wenjiang, Chengdu 611130, China
| |
Collapse
|
26
|
Abstract
The engulfment of apoptotic cells by phagocytes, a process referred to as efferocytosis, is essential for maintenance of normal tissue homeostasis and a prerequisite for the resolution of inflammation. Neutrophils are the predominant circulating white blood cell in humans, and contain an arsenal of toxic substances that kill and degrade microbes. Neutrophils are short-lived and spontaneously die by apoptosis. This review will highlight how the engulfment of apoptotic neutrophils by human phagocytes occurs, how heterogeneity of phagocyte populations influences efferocytosis signaling, and downstream consequences of efferocytosis. The efferocytosis of apoptotic neutrophils by macrophages promotes anti-inflammatory signaling, prevents neutrophil lysis, and dampens immune responses. Given the immunomodulatory properties of efferocytosis, understanding pathways that regulate and enhance efferocytosis could be harnessed to combat infection and chronic inflammatory conditions.
Collapse
Affiliation(s)
- Mallary C Greenlee-Wacker
- Inflammation Program, Department of Internal Medicine, Roy J. and Lucille A. Carver College of Medicine University of Iowa, Veterans Administration Medical Center, Iowa City, IA, USA
| |
Collapse
|
27
|
Kalinec GM, Lomberk G, Urrutia RA, Kalinec F. Resolution of Cochlear Inflammation: Novel Target for Preventing or Ameliorating Drug-, Noise- and Age-related Hearing Loss. Front Cell Neurosci 2017; 11:192. [PMID: 28736517 PMCID: PMC5500902 DOI: 10.3389/fncel.2017.00192] [Citation(s) in RCA: 83] [Impact Index Per Article: 11.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2017] [Accepted: 06/20/2017] [Indexed: 12/11/2022] Open
Abstract
A significant number of studies support the idea that inflammatory responses are intimately associated with drug-, noise- and age-related hearing loss (DRHL, NRHL and ARHL). Consequently, several clinical strategies aimed at reducing auditory dysfunction by preventing inflammation are currently under intense scrutiny. Inflammation, however, is a normal adaptive response aimed at restoring tissue functionality and homeostasis after infection, tissue injury and even stress under sterile conditions, and suppressing it could have unintended negative consequences. Therefore, an appropriate approach to prevent or ameliorate DRHL, NRHL and ARHL should involve improving the resolution of the inflammatory process in the cochlea rather than inhibiting this phenomenon. The resolution of inflammation is not a passive response but rather an active, highly controlled and coordinated process. Inflammation by itself produces specialized pro-resolving mediators with critical functions, including essential fatty acid derivatives (lipoxins, resolvins, protectins and maresins), proteins and peptides such as annexin A1 and galectins, purines (adenosine), gaseous mediators (NO, H2S and CO), as well as neuromodulators like acetylcholine and netrin-1. In this review article, we describe recent advances in the understanding of the resolution phase of inflammation and highlight therapeutic strategies that might be useful in preventing inflammation-induced cochlear damage. In particular, we emphasize beneficial approaches that have been tested in pre-clinical models of inflammatory responses induced by recognized ototoxic drugs such as cisplatin and aminoglycoside antibiotics. Since these studies suggest that improving the resolution process could be useful for the prevention of inflammation-associated diseases in humans, we discuss the potential application of similar strategies to prevent or mitigate DRHL, NRHL and ARHL.
Collapse
Affiliation(s)
- Gilda M Kalinec
- Laboratory of Auditory Cell Biology, Department of Head and Neck Surgery, David Geffen School of Medicine, University of CaliforniaLos Angeles, Los Angeles, CA, United States
| | - Gwen Lomberk
- Epigenetics and Chromatin Dynamics Laboratory, Translational Epigenomic Program, Center for Individualized Medicine (CIM) Mayo ClinicRochester, MN, United States
| | - Raul A Urrutia
- Epigenetics and Chromatin Dynamics Laboratory, Translational Epigenomic Program, Center for Individualized Medicine (CIM) Mayo ClinicRochester, MN, United States
| | - Federico Kalinec
- Laboratory of Auditory Cell Biology, Department of Head and Neck Surgery, David Geffen School of Medicine, University of CaliforniaLos Angeles, Los Angeles, CA, United States
| |
Collapse
|
28
|
Brennan EP, Cacace A, Godson C. Specialized pro-resolving mediators in renal fibrosis. Mol Aspects Med 2017; 58:102-113. [PMID: 28479307 DOI: 10.1016/j.mam.2017.05.001] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2017] [Revised: 04/28/2017] [Accepted: 05/01/2017] [Indexed: 12/31/2022]
Abstract
Inflammation and its timely resolution play a critical role in effective host defence and wound healing. Unresolved inflammatory responses underlie the pathology of many prevalent diseases resulting in tissue fibrosis and eventual organ failure as typified by kidney, lung and liver fibrosis. The role of autocrine and paracrine mediators including cytokines, prostaglandins and leukotrienes in initiating and sustaining inflammation is well established. More recently a physiological role for specialized pro-resolving lipid mediators [SPMs] in modulating inflammatory responses and promoting the resolution of inflammation has been appreciated. As will be discussed in this review, SPMs not only attenuate the development of fibrosis through promoting the resolution of inflammation but may also directly suppress fibrotic responses. These findings suggest novel therapeutic paradigms to treat intractable life-limiting diseases such as renal fibrosis.
Collapse
Affiliation(s)
- Eoin P Brennan
- UCD Diabetes Complications Research Centre, UCD Conway Institute & UCD School of Medicine, University College Dublin, Belfield, Dublin 4, Ireland.
| | - Antonino Cacace
- UCD Diabetes Complications Research Centre, UCD Conway Institute & UCD School of Medicine, University College Dublin, Belfield, Dublin 4, Ireland.
| | - Catherine Godson
- UCD Diabetes Complications Research Centre, UCD Conway Institute & UCD School of Medicine, University College Dublin, Belfield, Dublin 4, Ireland.
| |
Collapse
|
29
|
Bonjoch L, Casas V, Carrascal M, Closa D. Involvement of exosomes in lung inflammation associated with experimental acute pancreatitis. J Pathol 2017; 240:235-45. [PMID: 27447723 DOI: 10.1002/path.4771] [Citation(s) in RCA: 54] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2015] [Revised: 06/21/2016] [Accepted: 07/07/2016] [Indexed: 12/17/2022]
Abstract
A frequent complication of acute pancreatitis is the lung damage associated with the systemic inflammatory response. Although various pro-inflammatory mediators generated at both local and systemic levels have been identified, the pathogenic mechanisms of the disease are still poorly understood. In recent years, exosomes have emerged as a new intercellular communication system able to transfer encapsulated proteins and small RNAs and protect them from degradation. Using an experimental model of taurocholate-induced acute pancreatitis in rats, we aimed to evaluate the role of exosomes in the extent of the systemic inflammatory response. Induction of pancreatitis increased the concentration of circulating exosomes, which showed a different proteomic profile to those obtained from control animals. A series of tracking experiments using PKH26-stained exosomes revealed that circulating exosomes effectively reached the alveolar compartment and were internalized by macrophages. In vitro experiments revealed that exosomes obtained under inflammatory conditions activate and polarize these alveolar macrophages towards a pro-inflammatory phenotype. Interestingly, the proteomic analysis of circulating exosomes during acute pancreatitis suggested a multi-organ origin with a relevant role for the liver as a source of these vesicles. Tracking experiments also revealed that the liver retains the majority of exosomes from the peritoneal cavity. We conclude that exosomes are involved in the lung damage associated with experimental acute pancreatitis and could be relevant mediators in the systemic effects of pancreatitis. Copyright © 2016 Pathological Society of Great Britain and Ireland. Published by John Wiley & Sons, Ltd.
Collapse
Affiliation(s)
- Laia Bonjoch
- Department of Experimental Pathology, Institut d'Investigacions Biomèdiques de Barcelona, Consejo Superior de Investigaciones Científicas (IIBB-CSIC), Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain
| | - Vanessa Casas
- Consejo Superior de Investigaciones Científicas/Universitat Autònoma de Barcelona (CSIC/UAB) Proteomics Facility, Institut d'Investigacions Biomèdiques de Barcelona (IIBB), Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain
| | - Montserrat Carrascal
- Consejo Superior de Investigaciones Científicas/Universitat Autònoma de Barcelona (CSIC/UAB) Proteomics Facility, Institut d'Investigacions Biomèdiques de Barcelona (IIBB), Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain
| | - Daniel Closa
- Department of Experimental Pathology, Institut d'Investigacions Biomèdiques de Barcelona, Consejo Superior de Investigaciones Científicas (IIBB-CSIC), Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain.
| |
Collapse
|
30
|
Sansbury BE, Spite M. Resolution of Acute Inflammation and the Role of Resolvins in Immunity, Thrombosis, and Vascular Biology. Circ Res 2017; 119:113-30. [PMID: 27340271 DOI: 10.1161/circresaha.116.307308] [Citation(s) in RCA: 143] [Impact Index Per Article: 20.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/02/2016] [Accepted: 05/26/2016] [Indexed: 12/11/2022]
Abstract
Acute inflammation is a host-protective response that is mounted in response to tissue injury and infection. Initiated and perpetuated by exogenous and endogenous mediators, acute inflammation must be resolved for tissue repair to proceed and for homeostasis to be restored. Resolution of inflammation is an actively regulated process governed by an array of mediators as diverse as those that initiate inflammation. Among these, resolvins have emerged as a genus of evolutionarily conserved proresolving mediators that act on specific cellular receptors to regulate leukocyte trafficking and blunt production of inflammatory mediators, while also promoting clearance of dead cells and tissue repair. Given that chronic unresolved inflammation is emerging as a central causative factor in the development of cardiovascular diseases, an understanding of the endogenous processes that govern normal resolution of acute inflammation is critical for determining why sterile maladaptive cardiovascular inflammation perpetuates. Here, we provide an overview of the process of resolution with a focus on the enzymatic biosynthesis and receptor-dependent actions of resolvins and related proresolving mediators in immunity, thrombosis, and vascular biology. We discuss how nutritional and current therapeutic approaches modulate resolution and propose that harnessing resolution concepts could potentially lead to the development of new approaches for treating chronic cardiovascular inflammation in a manner that is not host disruptive.
Collapse
Affiliation(s)
- Brian E Sansbury
- From the Center for Experimental Therapeutics and Reperfusion Injury, Department of Anesthesiology, Perioperative and Pain Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA
| | - Matthew Spite
- From the Center for Experimental Therapeutics and Reperfusion Injury, Department of Anesthesiology, Perioperative and Pain Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA.
| |
Collapse
|
31
|
Zheng S, D'Souza VK, Bartis D, Dancer RCA, Parekh D, Naidu B, Gao-Smith F, Wang Q, Jin S, Lian Q, Thickett DR. Lipoxin A 4 promotes lung epithelial repair whilst inhibiting fibroblast proliferation. ERJ Open Res 2016; 2:00079-2015. [PMID: 27957484 PMCID: PMC5140017 DOI: 10.1183/23120541.00079-2015] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2015] [Accepted: 06/05/2016] [Indexed: 11/29/2022] Open
Abstract
Therapy that promotes epithelial repair whilst protecting against fibroproliferation is critical for restoring lung function in acute and chronic respiratory diseases. Primary human alveolar type II cells were used to model the effects of lipoxin A4in vitro upon wound repair, proliferation, apoptosis and transdifferention. Effects of lipoxin A4 upon primary human lung fibroblast proliferation, collagen production, and myofibroblast differentiation were also assessed. Lipoxin A4 promoted type II cell wound repair and proliferation, blocked the negative effects of soluble Fas ligand/tumour necrosis factor α upon cell proliferation, viability and apoptosis, and augmented the epithelial cell proliferative response to bronchoaveolar lavage fluid (BALF) from acute respiratory distress syndrome (ARDS). In contrast, Lipoxin A4 reduced fibroblast proliferation, collagen production and myofibroblast differentiation induced by transforming growth factor β and BALF from ARDS. The effects of Lipoxin A4 were phosphatidylinositol 3′-kinase dependent and mediated via the lipoxin A4 receptor. Lipoxin A4 appears to promote alveolar epithelial repair by stimulating epitheial cell wound repair, proliferation, reducing apoptosis and promoting trans-differentiation of alveolar type II cells into type I cells. Lipoxin A4 reduces fibroblast proliferation, collagen production and myofibroblast differentiation. These data suggest that targeting lipoxin actions may be a therapeutic strategy for treating the resolution phase of ARDS. Lipoxin A4 promotes epithelial repair while inhibiting fibroproliferation in vitro in human alveolar epithelial cellshttp://ow.ly/SxMu301cBRP
Collapse
Affiliation(s)
- Shengxing Zheng
- Dept of Anaesthesia and Critical Care, Second Affiliated Hospital of Wenzhou Medical University, Zhejiang , China; Centre for Translational Inflammation and Fibrosis Research, Institute of Inflammation and Ageing, University of Birmingham, Birmingham, UK; Both authors contributed equally
| | - Vijay K D'Souza
- Centre for Translational Inflammation and Fibrosis Research, Institute of Inflammation and Ageing, University of Birmingham, Birmingham, UK; Both authors contributed equally
| | - Domokos Bartis
- Centre for Translational Inflammation and Fibrosis Research, Institute of Inflammation and Ageing, University of Birmingham, Birmingham, UK
| | - Rachel C A Dancer
- Centre for Translational Inflammation and Fibrosis Research, Institute of Inflammation and Ageing, University of Birmingham, Birmingham, UK
| | - Dhruv Parekh
- Centre for Translational Inflammation and Fibrosis Research, Institute of Inflammation and Ageing, University of Birmingham, Birmingham, UK
| | - Babu Naidu
- Centre for Translational Inflammation and Fibrosis Research, Institute of Inflammation and Ageing, University of Birmingham, Birmingham, UK
| | - Fang Gao-Smith
- Dept of Anaesthesia and Critical Care, Second Affiliated Hospital of Wenzhou Medical University, Zhejiang , China; Centre for Translational Inflammation and Fibrosis Research, Institute of Inflammation and Ageing, University of Birmingham, Birmingham, UK
| | - Qian Wang
- Dept of Anaesthesia and Critical Care, Second Affiliated Hospital of Wenzhou Medical University, Zhejiang , China; Centre for Translational Inflammation and Fibrosis Research, Institute of Inflammation and Ageing, University of Birmingham, Birmingham, UK
| | - Shengwei Jin
- Dept of Anaesthesia and Critical Care, Second Affiliated Hospital of Wenzhou Medical University, Zhejiang , China
| | - Qingquan Lian
- Dept of Anaesthesia and Critical Care, Second Affiliated Hospital of Wenzhou Medical University, Zhejiang , China
| | - David R Thickett
- Centre for Translational Inflammation and Fibrosis Research, Institute of Inflammation and Ageing, University of Birmingham, Birmingham, UK
| |
Collapse
|
32
|
|
33
|
The resolution of inflammation: Principles and challenges. Semin Immunol 2015; 27:149-60. [PMID: 25911383 DOI: 10.1016/j.smim.2015.03.014] [Citation(s) in RCA: 263] [Impact Index Per Article: 29.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/11/2015] [Revised: 03/27/2015] [Accepted: 03/30/2015] [Indexed: 12/11/2022]
Abstract
The concept that chemokines, cytokines and pro-inflammatory mediators act in a co-ordinated fashion to drive the initiation of the inflammatory reaction is well understood. The significance of such networks acting during the resolution of inflammation however is poorly appreciated. In recent years, specific pro-resolving mediators were discovered which activate resolution pathways to return tissues to homeostasis. These mediators are diverse in nature, and include specialized lipid mediators (lipoxins, resolvins, protectins and maresins) proteins (annexin A1, galectins) and peptides, gaseous mediators including hydrogen sulphide, a purine (adenosine), as well as neuromodulator release under the control of the vagus nerve. Functionally, they can act to limit further leukocyte recruitment, induce neutrophil apoptosis and enhance efferocytosis by macrophages. They can also switch macrophages from classical to alternatively activated cells, promote the return of non-apoptotic cells to the lymphatics and help initiate tissue repair mechanisms and healing. Within this review we highlight the essential cellular aspects required for successful tissue resolution, briefly discuss the pro-resolution mediators that drive these processes and consider potential challenges faced by researchers in the quest to discover how inflammation resolves and why chronic inflammation persists.
Collapse
|
34
|
Higgins G, Buchanan P, Perriere M, Al-Alawi M, Costello RW, Verriere V, McNally P, Harvey BJ, Urbach V. Activation of P2RY11 and ATP release by lipoxin A4 restores the airway surface liquid layer and epithelial repair in cystic fibrosis. Am J Respir Cell Mol Biol 2014; 51:178-90. [PMID: 24588705 DOI: 10.1165/rcmb.2012-0424oc] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
In cystic fibrosis (CF), the airway surface liquid (ASL) height is reduced as a result of impaired ion transport, which favors bacterial colonization and inflammation of the airway and leads to progressive lung destruction. Lipoxin (LX)A4, which promotes resolution of inflammation, is inadequately produced in the airways of patients with CF. We previously demonstrated that LXA4 stimulates an ASL height increase and epithelial repair. Here we report the molecular mechanisms involved in these processes. We found that LXA4 (1 nM) induced an apical ATP release from non-CF (NuLi-1) and CF (CuFi-1) airway epithelial cell lines and CF primary cultures. The ATP release induced by LXA4 was completely inhibited by antagonists of the ALX/FPR2 receptor and Pannexin-1 channels. LXA4 induced an increase in intracellular cAMP and calcium, which were abolished by the selective inhibition of the P2RY11 purinoreceptor. Pannexin-1 and ATP hydrolysis inhibition and P2RY11 purinoreceptor knockdown all abolished the increase of ASL height induced by LXA4. Inhibition of the A2b adenosine receptor did not affect the ASL height increase induced by LXA4, whereas the PKA inhibitor partially inhibited this response. The stimulation of NuLi-1 and CuFi-1 cell proliferation, migration, and wound repair by LXA4 was inhibited by the antagonists of Pannexin-1 channel and P2RY11 purinoreceptor. Taken together, our results provide evidence for a novel role of LXA4 in stimulating apical ATP secretion via Pannexin-1 channels and P2RY11 purinoreceptors activation leading to an ASL height increase and epithelial repair.
Collapse
|
35
|
Abstract
Acute inflammation in the lung is essential to health. So too is its resolution. In response to invading microbes, noxious stimuli, or tissue injury, an acute inflammatory response is mounted to protect the host. To limit inflammation and prevent collateral injury of healthy, uninvolved tissue, the lung orchestrates the formation of specialized proresolving mediators, specifically lipoxins, resolvins, protectins, and maresins. These immunoresolvents are agonists for resolution that interact with specific receptors on leukocytes and structural cells to blunt further inflammation and promote catabasis. This process appears to be defective in several common lung diseases that are characterized by excess or chronic inflammation. Here, we review the molecular and cellular effectors of resolution of acute inflammation in the lung.
Collapse
Affiliation(s)
- Bruce D Levy
- Pulmonary and Critical Care Medicine, Department of Internal Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts 02115;
| | | |
Collapse
|
36
|
Gong JH, Gong JP, Li JZ, He K, Li PZ, Jiang XW. Glycogen synthase kinase 3 inhibitor attenuates endotoxin-induced liver injury. J Surg Res 2013; 184:1035-44. [PMID: 23721934 DOI: 10.1016/j.jss.2013.04.051] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2013] [Revised: 02/05/2013] [Accepted: 04/22/2013] [Indexed: 12/01/2022]
Abstract
BACKGROUND/AIMS Endotoxin (lipopolysaccharide, LPS)-induced acute liver injury was attenuated by endotoxin tolerance (ET), which is characterized by phosphatidylinositol 3-kinase pathway/Akt signaling. Glycogen synthase kinase 3 (GSK-3) acts downstream of phosphatidylinositol 3-kinase pathway/Akt and GSK-3 inhibitor protects against organic injury. This study evaluates the hypothesis that ET attenuated LPS-induced liver injury through inhibiting GSK-3 functional activity and downstream signaling. METHODS Sprague-Dawley rats with or without low-dose LPS pretreatment were challenged with or without large dose of LPS and subsequently received studies. Serum tumor necrosis factor-alpha, interleukin-10, alanine aminotransferase, lactate dehydrogenase, and total bilirubin levels were analyzed, morphology of liver tissue was performed, glycogen content, myeloperoxidase content, phagocytosis activity of Kupffer cells, and the expression and inhibitory phosphorylation as well as kinase activity of GSK-3 were examined. Survival after LPS administration was also determined. RESULTS LPS induced significant increases of serum TNF-α, alanine aminotransferase, lactate dehydrogenase, and total bilirubin (P < 0.05), which were companied by obvious alterations in liver: the injury of liver tissue, the decrease of glycogen, the infiltration of neutrophils, and the enhancement of phagocytosis of Kupffer cells (P < 0.05). LPS pretreatment significantly attenuated these alterations, promoted the inhibitory phosphorylation of GSK-3 and inhibited its kinase activity, and improved the survival rate (P < 0.05). CONCLUSIONS ET attenuated LPS-induced acute liver injury through inhibiting GSK-3 functional activity and its downstream signaling.
Collapse
Affiliation(s)
- Jun-hua Gong
- Department of Hepatobiliary Surgery, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | | | | | | | | | | |
Collapse
|
37
|
Medeiros R, Kitazawa M, Passos GF, Baglietto-Vargas D, Cheng D, Cribbs DH, LaFerla FM. Aspirin-triggered lipoxin A4 stimulates alternative activation of microglia and reduces Alzheimer disease-like pathology in mice. THE AMERICAN JOURNAL OF PATHOLOGY 2013; 182:1780-9. [PMID: 23506847 DOI: 10.1016/j.ajpath.2013.01.051] [Citation(s) in RCA: 125] [Impact Index Per Article: 11.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/25/2012] [Revised: 01/15/2013] [Accepted: 01/17/2013] [Indexed: 12/29/2022]
Abstract
Microglia play an essential role in innate immunity, homeostasis, and neurotropic support in the central nervous system. In Alzheimer disease (AD), these cells may affect disease progression by modulating the buildup of β-amyloid (Aβ) or releasing proinflammatory cytokines and neurotoxic substances. Discovering agents capable of increasing Aβ uptake by phagocytic cells is of potential therapeutic interest for AD. Lipoxin A4 (LXA4) is an endogenous lipid mediator with potent anti-inflammatory properties directly involved in inflammatory resolution, an active process essential for appropriate host responses, tissue protection, and the return to homeostasis. Herein, we demonstrate that aspirin-triggered LXA4 (15 μg/kg) s.c., twice a day, reduced NF-κB activation and levels of proinflammatory cytokines and chemokines, as well as increased levels of anti-inflammatory IL-10 and transforming growth factor-β. Such changes in the cerebral milieu resulted in recruitment of microglia in an alternative phenotype, as characterized by the up-regulation of YM1 and arginase-1 and the down-regulation of inducible nitric oxide synthase expression. Microglia in an alternative phenotype-positive cells demonstrated improved phagocytic function, promoting clearance of Aβ deposits and ultimately leading to reduction in synaptotoxicity and improvement in cognition. Our data indicate that activating LXA4 signaling may represent a novel therapeutic approach for AD.
Collapse
Affiliation(s)
- Rodrigo Medeiros
- Institute for Memory Impairments and Neurological Disorders, University of California, Irvine, Irvine, CA 92697-4545, USA
| | | | | | | | | | | | | |
Collapse
|
38
|
Recchiuti A, Serhan CN. Pro-Resolving Lipid Mediators (SPMs) and Their Actions in Regulating miRNA in Novel Resolution Circuits in Inflammation. Front Immunol 2012; 3:298. [PMID: 23093949 PMCID: PMC3477628 DOI: 10.3389/fimmu.2012.00298] [Citation(s) in RCA: 100] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2012] [Accepted: 09/07/2012] [Indexed: 12/12/2022] Open
Abstract
Unresolved inflammation is associated with several widely occurring diseases such as arthritis, periodontal diseases, cancer, and atherosclerosis. Endogenous mechanisms that curtail excessive inflammation and prompt its timely resolution are of considerable interest. In recent years, previously unrecognized chemical mediators derived from polyunsaturated fatty acids were identified that control the acute inflammatory response by activating local resolution programs. Among these are the so-called specialized pro-resolving lipid mediators (SPMs) that include lipoxins (LX), resolvins (Rv), protectins (PD), and maresins (MaR), because they are enzymatically biosynthesized during resolution of self-limited inflammation. They each possess distinct chemical structures and regulate cellular pathways by their ability to activate pro-resolving G-protein coupled receptors (GPCRs) in a stereospecific manner. For instance, RvD1 controls several miRNAs of interest in self-limited acute inflammation that counter-regulate the mediators and proteins that are involved in inflammation. Here, we overview some of the biosynthesis and mechanisms of SPM actions with focus on the recently reported miR involved in their pro-resolving responses that underscore their beneficial actions in the regulation of acute inflammation and its timely resolution. The elucidation of these mechanisms operating in vivo to keep acute inflammation within physiologic boundaries as well as stimulate resolution have opened resolution pharmacology and many new opportunities to target inflammation-related human pathologies via activating resolution mechanisms.
Collapse
Affiliation(s)
- Antonio Recchiuti
- Center for Experimental Therapeutics and Reperfusion Injury, Department of Anesthesiology, Perioperative and Pain Medicine, Brigham and Women's Hospital, Harvard Medical School, Harvard Institutes of Medicine Boston, MA, USA
| | | |
Collapse
|
39
|
Börgeson E, Godson C. Resolution of inflammation: therapeutic potential of pro-resolving lipids in type 2 diabetes mellitus and associated renal complications. Front Immunol 2012; 3:318. [PMID: 23087692 PMCID: PMC3474937 DOI: 10.3389/fimmu.2012.00318] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2012] [Accepted: 09/29/2012] [Indexed: 01/04/2023] Open
Abstract
The role of inflammation in the pathogenesis of type 2 diabetes mellitus (T2DM) and its associated complications is increasingly recognized. The resolution of inflammation is actively regulated by endogenously produced lipid mediators such as lipoxins, resolvins, protectins, and maresins. Here we review the potential role of these lipid mediators in diabetes-associated pathologies, specifically focusing on adipose inflammation and diabetic kidney disease, i.e., diabetic nephropathy (DN). DN is one of the major complications of T2DM and we propose that pro-resolving lipid mediators may have therapeutic potential in this context. Adipose inflammation is also an important component of T2DM-associated insulin resistance and altered adipokine secretion. Promoting the resolution of adipose inflammation would therefore likely be a beneficial therapeutic approach in T2DM.
Collapse
Affiliation(s)
- Emma Börgeson
- UCD Diabetes Research Centre, UCD Conway Institute, School of Medicine and Medical Sciences, University College Dublin Dublin, Ireland
| | | |
Collapse
|
40
|
Börgeson E, McGillicuddy FC, Harford KA, Corrigan N, Higgins DF, Maderna P, Roche HM, Godson C. Lipoxin A4 attenuates adipose inflammation. FASEB J 2012; 26:4287-94. [PMID: 22700871 DOI: 10.1096/fj.12-208249] [Citation(s) in RCA: 88] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
Aging and adiposity are associated with chronic low-grade inflammation, which underlies the development of obesity-associated complications, including type 2 diabetes mellitus (T2DM). The mechanisms underlying adipose inflammation may include macrophage infiltration and activation, which, in turn, affect insulin sensitivity of adipocytes. There is a growing appreciation that specific lipid mediators (including lipoxins, resolvins, and protectins) can promote the resolution of inflammation. Here, we investigated the effect of lipoxin A4 (LXA4), the predominant endogenously generated lipoxin, on adipose tissue inflammation. Using adipose tissue explants from perigonadal depots of aging female C57BL/6J mice (Animalia, Chordata, Mus musculus) as a model of age-associated adipose inflammation, we report that LXA4 (1 nM) attenuates adipose inflammation, decreasing IL-6 and increasing IL-10 expression (P<0.05). The altered cytokine milieu correlated with increased GLUT-4 and IRS-1 expression, suggesting improved insulin sensitivity. Further investigations revealed the ability of LXA4 to rescue macrophage-induced desensitization to insulin-stimulated signaling and glucose uptake in cultured adipocytes, using vehicle-stimulated cells as controls. This was associated with preservation of Akt activation and reduced secretion of proinflammatory cytokines, including TNF-α. We therefore propose that LXA4 may represent a potentially useful and novel therapeutic strategy to subvert adipose inflammation and insulin resistance, key components of T2DM.
Collapse
Affiliation(s)
- Emma Börgeson
- UCD Diabetes Research Centre, UCD Conway Institute, School of Medicine and Medical Science, University College Dublin, Dublin, Ireland
| | | | | | | | | | | | | | | |
Collapse
|
41
|
Chiu CY, Gomolka B, Dierkes C, Huang NR, Schroeder M, Purschke M, Manstein D, Dangi B, Weylandt KH. Omega-6 docosapentaenoic acid-derived resolvins and 17-hydroxydocosahexaenoic acid modulate macrophage function and alleviate experimental colitis. Inflamm Res 2012; 61:967-76. [PMID: 22618200 DOI: 10.1007/s00011-012-0489-8] [Citation(s) in RCA: 59] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2012] [Revised: 04/28/2012] [Accepted: 05/02/2012] [Indexed: 12/11/2022] Open
Abstract
OBJECTIVE Enzymatically oxygenated lipid products derived from omega-3 and omega-6 fatty acids play an important role in inflammation dampening. This study examined the anti-inflammatory effects of n-6 docosapentaenoic acid-derived (17S)-hydroxy-docosapentaenoic acid (17-HDPAn-6) and (10,17S)-dihydroxy-docosapentaenoic acid (10,17-HDPAn-6) as well as n-3 docosahexaenoic acid-derived 17(R/S)-hydroxy-docosahexaenoic acid (17-HDHA). MATERIALS AND METHODS The effects of 17-HDPAn-6, 10,17-HDPAn-6 or 17-HDHA on activity and M1/M2 polarization of murine macrophage cell line RAW 264.7 were examined by phagocytosis assay and real-time PCR. To assess anti-inflammatory effects in vivo, dextran sodium sulfate (DSS) colitis was induced in mice treated with 17-HDPAn-6, 10,17-HDPAn-6, 17-HDHA or NaCl. RESULTS Our results show that 17-HDPAn-6, 10,17-HDPAn-6 and 17-HDHA increase phagocytosis in macrophages in vitro and promote polarization towards the anti-inflammatory M2 phenotype with decreased gene expression of TNF-α and inducible Nitric oxide synthase and increased expression of the chemokine IL-1 receptor antagonist and the Scavenger receptor Type A. Intraperitoneal treatment with 17-HDPAn-6, 10,17-HDPAn-6, or 17-HDHA alleviated DSS-colitis and significantly improved body weight loss, colon epithelial damage, and macrophage infiltration. CONCLUSION These results suggest that DPAn-6-derived 17-HDPAn-6 and 10,17-HDPAn-6 as well as the DHA-derived 17-HDHA have inflammation-dampening and resolution-promoting effects that could be used to treat inflammatory conditions such as inflammatory bowel disease.
Collapse
Affiliation(s)
- Cheng-Ying Chiu
- Department of Hepatology, Gastroenterology and Endocrinology, Charité University Medicine Berlin, Campus Virchow Klinikum, Berlin, Germany
| | | | | | | | | | | | | | | | | |
Collapse
|
42
|
Abstract
Sepsis is characterized by systemic inflammation with release of a large amount of inflammatory mediators. If sustained, this inflammatory response can lead to multiple organ failure and/or immunoparalysis. In the latter condition, the host may be susceptible to opportunistic infections or be unable to clear existing infections. Therefore, it is potentially beneficial to resolve inflammation by reducing inflammation without compromising host defense. We examined the effect of lipoxin A4 (LXA4), a compound with inflammatory resolution properties, in the cecal ligation and puncture (CLP) model of sepsis. Cecal ligation and puncture rats were given either saline or LXA4 (40 μg/kg, i.p.) 5 h after surgery. Lipoxin A4 administration increased 8-day survival of CLP rats, which lived longer than 48 h, and attenuated tissue injury after 8 days. Therefore, we investigated the effects of LXA4 on systemic inflammation and bacterial load 48 h after CLP sepsis. Plasma IL-6, monocyte chemotactic protein 1, and IL-10 levels were reduced in LXA4-treated rats compared with CLP rats given saline vehicle. Lipoxin A4 reduced phosphorylation of the p65 subunit of nuclear factor κB (NF-κB) at serines 536 and 468 in peritoneal macrophages, suggesting that LXA4 reduced production of proinflammatory mediators through an NF-κB-mediated mechanism. Lipoxin A4 reduced blood bacterial load and increased peritoneal macrophage number without affecting phagocytic ability, suggesting that LXA4 reduced blood bacterial load by enhancing macrophage recruitment. It also suggests that LXA4 reduced systemic inflammation and NF-κB activation without compromising host defense. Increased macrophage recruitment was in part due to a direct effect of LXA4 as LXA4 increased peritoneal macrophage recruitment in sham controls and partly due to reduced production of IL-10 as LXA4 decreased macrophage IL-10 release (a known inhibitor of macrophage migration) after CLP. The results suggest that LXA4 increased survival in sepsis by simultaneously reducing systemic inflammation as well as bacterial spread.
Collapse
|
43
|
Lin CP, Adrianto I, Lessard CJ, Kelly JA, Kaufman KM, Guthridge JM, Freedman BI, Anaya JM, Alarcón-Riquelme ME, Pons-Estel BA, Martin J, Glenn S, Adler A, Bae SC, Park SY, Bang SY, Song YW, Boackle SA, Brown EE, Edberg JC, Alarcón GS, Petri MA, Criswell LA, Ramsey-Goldman R, Reveille JD, Vila LM, Gilkeson GS, Kamen DL, Ziegler J, Jacob CO, Rasmussen A, James JA, Kimberly RP, Merrill JT, Niewold TB, Scofield RH, Stevens AM, Tsao BP, Vyse TJ, Langefeld CD, Moser KL, Harley JB, Gaffney PM, Montgomery CG. Role of MYH9 and APOL1 in African and non-African populations with lupus nephritis. Genes Immun 2012; 13:232-8. [PMID: 22189356 PMCID: PMC3330160 DOI: 10.1038/gene.2011.82] [Citation(s) in RCA: 46] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2011] [Revised: 11/04/2011] [Accepted: 11/14/2011] [Indexed: 01/31/2023]
Abstract
Systemic lupus erythematosus (SLE) is a complex autoimmune disease characterized by autoantibody production and organ damage. Lupus nephritis (LN) is one of the most severe manifestations of SLE. Multiple studies reported associations between renal diseases and variants in the non-muscle myosin heavy chain 9 (MYH9) and the neighboring apolipoprotein L 1 (APOL1) genes. We evaluated 167 variants spanning MYH9 for association with LN in a multiethnic sample. The two previously identified risk variants in APOL1 were also tested for association with LN in European-Americans (EAs) (N = 579) and African-Americans (AAs) (N = 407). Multiple peaks of association exceeding a Bonferroni corrected P-value of P < 2.03 × 10(-3) were observed between LN and MYH9 in EAs (N = 4620), with the most pronounced association at rs2157257 (P = 4.7 × 10(-4), odds ratio (OR) = 1.205). A modest effect with MYH9 was also detected in Gullah (rs8136069, P = 0.0019, OR = 2.304). No association between LN and MYH9 was found in AAs, Asians, Amerindians or Hispanics. This study provides the first investigation of MYH9 in LN in non-Africans and of APOL1 in LN in any population, and presents novel insight into the potential role of MYH9 in LN in EAs.
Collapse
Affiliation(s)
- C P Lin
- Arthritis and Clinical Immunology Research Program, Oklahoma Medical Research Foundation, Oklahoma City, OK 73104, USA
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
44
|
Prescott D, McKay DM. Aspirin-triggered lipoxin enhances macrophage phagocytosis of bacteria while inhibiting inflammatory cytokine production. Am J Physiol Gastrointest Liver Physiol 2011; 301:G487-97. [PMID: 21659618 DOI: 10.1152/ajpgi.00042.2011] [Citation(s) in RCA: 46] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
The macrophage plays a major role in the induction and resolution phases of inflammation; however, how lipid mediator-derived signals may modulate macrophage function in the resolution of inflammation driven by microbes (e.g., in inflammatory bowel disease) is not well understood. We examined the effects of aspirin-triggered lipoxin (ATL), a stable analog of lipoxin A(4), on the antimicrobial responses of human peripheral blood mononuclear cell-derived macrophages and the monocytic THP-1 cell line. Additionally, we assessed the expression and localization of the lipoxin receptor, formyl peptide receptor 2 (FPR2), in colonic mucosal biopsies from patients with Crohn's disease to determine whether the capacity for lipoxin signaling is altered in inflammatory bowel disease. We found that THP-1 cells treated with ATL (100 nM) displayed increased phagocytosis of inert fluorescent beads and Escherichia coli in a scavenger receptor- and PI3K-dependent, opsonization-independent manner. This ATL-induced increase in phagocytosis was also observed in primary human macrophages, where it was associated with an inhibition of E. coli-induced IL-1β and IL-8 production. Finally, we found that FPR2 gene expression was increased approximately sixfold in the colon of patients with Crohn's disease, a finding reproduced in vitro by the treatment of THP-1 cells with interferon-γ or lipopolysaccharide. These results suggest that lipoxin signaling is upregulated in inflammatory environments, and, in addition to their known role in tissue resolution following injury, lipoxins can enhance macrophage clearance of invading microbes.
Collapse
Affiliation(s)
- David Prescott
- Gastrointestinal Research Group, Department of Physiology and Pharmacology, Calvin, Phoebe and Joan Snyder Institute of Infection, Immunity and Inflammation, University of Calgary, Calgary, Alberta, Canada
| | | |
Collapse
|
45
|
Palmer CD, Mancuso CJ, Weiss JP, Serhan CN, Guinan EC, Levy O. 17(R)-Resolvin D1 differentially regulates TLR4-mediated responses of primary human macrophages to purified LPS and live E. coli. J Leukoc Biol 2011; 90:459-70. [PMID: 21653234 DOI: 10.1189/jlb.0311145] [Citation(s) in RCA: 46] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023] Open
Abstract
Detection and clearance of bacterial infection require balanced effector and resolution signals to avoid chronic inflammation. Detection of GNB LPS by TLR4 on m induces inflammatory responses, contributing to chronic inflammation and tissue injury. LXs and Rvs are endogenous lipid mediators that enhance resolution of inflammation, and their actions on primary human m responses toward GNB are largely uncharacterized. Here, we report that LXA(4), LXB(4), and RvD1, tested at 0.1-1 μM, inhibited LPS-induced TNF production from primary human m, with ATL and 17(R)-RvD1, demonstrating potent inhibition at 0.1 μM. In addition, 17(R)-RvD1 inhibited LPS-induced primary human m production of IL-7, IL-12p70, GM-CSF, IL-8, CCL2, and MIP-1α without reducing that of IL-6 or IL-10. Remarkably, when stimulated with live Escherichia coli, m treated with 17(R)-RvD1 demonstrated increased TNF production and enhanced internalization and killing of the bacteria. 17(R)-RvD1-enhanced TNF, internalization, and killing were not evident for an lpxM mutant of E. coli expressing hypoacylated LPS with reduced inflammatory activity. Furthermore, 17(R)-RvD1-enhanced, E. coli-induced TNF production was evident in WT but not TLR4-deficient murine m. Thus, Rvs differentially modulate primary human m responses to E. coli in an LPS- and TLR4-dependent manner, such that this Rv could promote resolution of GNB/LPS-driven inflammation by reducing m proinflammatory responses to isolated LPS and increasing m responses important for clearance of infection.
Collapse
|
46
|
Serhan CN, Krishnamoorthy S, Recchiuti A, Chiang N. Novel anti-inflammatory--pro-resolving mediators and their receptors. Curr Top Med Chem 2011; 11:629-47. [PMID: 21261595 DOI: 10.2174/1568026611109060629] [Citation(s) in RCA: 211] [Impact Index Per Article: 16.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2009] [Accepted: 06/02/2010] [Indexed: 01/04/2023]
Abstract
Resolution of inflammation, an actively coordinated program, is essential to maintain host health. It involves effective removal of inflammatory stimuli and the spatio-temporal control of leukocyte trafficking as well as chemical mediator generation. During the active resolution process, new classes of small, local acting endogenous autacoids, namely the lipoxins, D and E series resolvins, (neuro)protectins, and maresins have been identified. These specialized pro-resolving lipid mediators (SPM) prevent excessive inflammation and promote removal of microbes and apoptotic cells, thereby expediting resolution and return to tissue homeostasis. As part of their molecular mechanism, SPM exert their potent actions via activating specific pro-resolving G-protein coupled receptors. Together these SPM and their receptors provide new concepts and opportunities for therapeutics, namely promoting active resolution as opposed to the conventionally used enzyme inhibitors and receptor antagonists. This approach may offer new targets suitable for drug design for treating inflammation related diseases, for the new terrain of resolution pharmacology.
Collapse
Affiliation(s)
- Charles N Serhan
- Center for Experimental Therapeutics and Reperfusion Injury, Department of Anesthesia, Perioperative and Pain Medicine, Harvard Institute of Medicine, 77 Avenue Louis Pasteur, Boston, MA 02115,USA.
| | | | | | | |
Collapse
|
47
|
Lipoxin A₄ inhibits porphyromonas gingivalis-induced aggregation and reactive oxygen species production by modulating neutrophil-platelet interaction and CD11b expression. Infect Immun 2011; 79:1489-97. [PMID: 21263017 DOI: 10.1128/iai.00777-10] [Citation(s) in RCA: 54] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
Porphyromonas gingivalis is an etiological agent that is strongly associated with periodontal disease, and it correlates with numerous inflammatory disorders, such as cardiovascular disease. Circulating bacteria may contribute to atherogenesis by promoting CD11b/CD18-mediated interactions between neutrophils and platelets, causing reactive oxygen species (ROS) production and aggregation. Lipoxin A₄ (LXA₄) is an endogenous anti-inflammatory and proresolving mediator that is protective of inflammatory disorders. The aim of this study was to investigate the effect of LXA₄ on the P. gingivalis-induced activation of neutrophils and platelets and the possible involvement of Rho GTPases and CD11b/CD18 integrins. Platelet/leukocyte aggregation and ROS production was examined by lumiaggregometry and fluorescence microscopy. Integrin activity was studied by flow cytometry, detecting the surface expression of CD11b/CD18 as well as the exposure of the high-affinity integrin epitope, whereas the activation of Rac2/Cdc42 was examined using a glutathione S-transferase pulldown assay. The study shows that P. gingivalis activates Rac2 and Cdc42 and upregulates CD11b/CD18 and its high-affinity epitope on neutrophils, and that these effects are diminished by LXA₄. Furthermore, we found that LXA₄ significantly inhibits P. gingivalis-induced aggregation and ROS generation in whole blood. However, in platelet-depleted blood and in isolated neutrophils and platelets, LXA₄ was unable to inhibit either aggregation or ROS production, respectively. In conclusion, this study suggests that LXA₄ antagonizes P. gingivalis-induced cell activation in a manner that is dependent on leukocyte-platelet interaction, likely via the inhibition of Rho GTPase signaling and the downregulation of CD11b/CD18. These findings may contribute to new strategies in the prevention and treatment of periodontitis-induced inflammatory disorders, such as atherosclerosis.
Collapse
|
48
|
Bannenberg G, Serhan CN. Specialized pro-resolving lipid mediators in the inflammatory response: An update. BIOCHIMICA ET BIOPHYSICA ACTA 2010; 1801:1260-73. [PMID: 20708099 PMCID: PMC2994245 DOI: 10.1016/j.bbalip.2010.08.002] [Citation(s) in RCA: 309] [Impact Index Per Article: 22.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/06/2010] [Revised: 07/23/2010] [Accepted: 08/02/2010] [Indexed: 12/25/2022]
Abstract
A new genus of specialized pro-resolving mediators (SPM) which include several families of distinct local mediators (lipoxins, resolvins, protectins, and maresins) are actively involved in the clearance and regulation of inflammatory exudates to permit restoration of tissue homeostasis. Classic lipid mediators that are temporally regulated are formed from arachidonic acid, and novel local mediators were uncovered that are biosynthesized from ω-3 poly-unsaturated fatty acids, such as eicosapentaenoic acid, docosapentaenoic acid and docosahexaenoic acid. The biosynthetic pathways for resolvins are constituted by fatty acid lipoxygenases and cyclooxygenase-2 via transcellular interactions established by innate immune effector cells which migrate from the vasculature to inflamed tissue sites. SPM provide local control over the execution of an inflammatory response towards resolution, and include recently recognized actions of SPM such as tissue protection and host defense. The structural families of the SPM do not resemble classic eicosanoids (PG or LT) and are novel structures that function uniquely via pro-resolving cellular and molecular targets. The extravasation of inflammatory cells expressing SPM biosynthetic routes are matched by the temporal provision of essential fatty acids from circulation needed as substrate for the formation of SPM. The present review provides an update and overview of the biosynthetic pathways and actions of SPM, and examines resolution as an integrated component of the inflammatory response and its return to homeostasis via biochemically active resolution mechanisms.
Collapse
Affiliation(s)
- Gerard Bannenberg
- Department of Plant Molecular Genetics, Centro Nacional de Biotecnología/CSIC, Madrid, Spain.
| | | |
Collapse
|
49
|
Maderna P, Cottell DC, Toivonen T, Dufton N, Dalli J, Perretti M, Godson C. FPR2/ALX receptor expression and internalization are critical for lipoxin A4 and annexin-derived peptide-stimulated phagocytosis. FASEB J 2010; 24:4240-9. [PMID: 20570963 PMCID: PMC4338542 DOI: 10.1096/fj.10-159913] [Citation(s) in RCA: 142] [Impact Index Per Article: 10.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
Lipoxins (LXs) are endogenously produced eicosanoids with well-described anti-inflammatory and proresolution activities, stimulating nonphlogistic phagocytosis of apoptotic cells by macrophages. LXA(4) and the glucocorticoid-derived annexin A1 peptide (Ac2-26) bind to a common G-protein-coupled receptor, termed FPR2/ALX. However, direct evidence of the involvement of FPR2/ALX in the anti-inflammatory and proresolution activity of LXA(4) is still to be investigated. Here we describe FPR2/ALX trafficking in response to LXA(4) and Ac2-26 stimulation. We have transfected cells with HA-tagged FPR2/ALX and studied receptor trafficking in unstimulated, LXA(4) (1-10 nM)- and Ac2-26 (30 μM)-treated cells using multiple approaches that include immunofluorescent confocal microscopy, immunogold labeling of cryosections, and ELISA and investigated receptor trafficking in agonist-stimulated phagocytosis. We conclude that PKC-dependent internalization of FPR2/ALX is required for phagocytosis. Using bone marrow-derived macrophages (BMDMs) from mice in which the FPR2/ALX ortholog Fpr2 had been deleted, we observed the nonredundant function for this receptor in LXA(4) and Ac2-26 stimulated phagocytosis of apoptotic neutrophils. LXA(4) stimulated phagocytosis 1.7-fold above basal (P<0.001) by BMDMs from wild-type mice, whereas no effect was found on BMDMs from Fpr2(-/-) mice. Similarly, Ac2-26 stimulates phagocytosis by BMDMs from wild-type mice 1.5-fold above basal (P<0.05). However, Ac2-26 failed to stimulate phagocytosis by BMDMs isolated from Fpr2(-/-) mice relative to vehicle. These data reveal novel and complex mechanisms of the FPR2/ALX receptor trafficking and functionality in the resolution of inflammation.
Collapse
Affiliation(s)
- Paola Maderna
- UCD Diabetes Research Centre, UCD School of Medicine and Medical Science, University College Dublin, Dublin, Ireland
| | - David C. Cottell
- The Electron Microscopy Laboratory, UCD Conway Institute of Biomolecular and Biomedical Research, University College Dublin, Dublin, Ireland
| | - Tiina Toivonen
- The Electron Microscopy Laboratory, UCD Conway Institute of Biomolecular and Biomedical Research, University College Dublin, Dublin, Ireland
| | - Neil Dufton
- The William Harvey Research Institute, Barts and The London School of Medicine, Queen Mary University of London, London, UK
| | - Jesmond Dalli
- The William Harvey Research Institute, Barts and The London School of Medicine, Queen Mary University of London, London, UK
| | - Mauro Perretti
- The William Harvey Research Institute, Barts and The London School of Medicine, Queen Mary University of London, London, UK
| | - Catherine Godson
- UCD Diabetes Research Centre, UCD School of Medicine and Medical Science, University College Dublin, Dublin, Ireland
| |
Collapse
|
50
|
Maldonado-Pérez D, Golightly E, Denison FC, Jabbour HN, Norman JE. A role for lipoxin A4 as anti-inflammatory and proresolution mediator in human parturition. FASEB J 2010; 25:569-75. [PMID: 20959513 DOI: 10.1096/fj.10-170340] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/27/2023]
Abstract
The purpose of this study was to investigate the role of lipoxin A(4), an anti-inflammatory and proresolution modulator, during human parturition. We measured serum levels of lipoxin A(4) and myometrial protein release using ELISA, quantified lipoxin receptor (FPR2/ALX) mRNA expression using qRT-PCR, and localized protein expression using immunohistochemstry in myometrial biopsies from pregnant women. In addition, we compared the effects of lipoxin A(4) (100 nM) with vehicle on basal and LPS-stimulated expression of proinflammatory cytokines from samples of myometrium from pregnant women. Mean ± SE circulating level of lipoxin A(4) was 5.89 ± 0.63 nM at 24-wk gestation, with a further modest increase during pregnancy (P<0.05), but no differences in gestation matched women before and after labor (P>0.05). Levels of lipoxin A(4) in nonpregnant women were 0.48 ± 0.04 nM, significantly lower than in pregnant women (P<0.001). FPR2/ALX localized to myocytes and neutrophils, with a 9-fold increase in mRNA expression in labor (P<0.001). Lipoxin A(4) significantly reduced LPS-induced but not basal expression of the proinflammatory cytokines IL-6 and IL-8 in cultured myometrium (P<0.05), compared to vehicle-treated controls. We demonstrate for the first time a potential role for lipoxin A(4) and its receptor in the resolution of the inflammatory events of both physiological and pathological labor.
Collapse
Affiliation(s)
- David Maldonado-Pérez
- Division of Reproductive and Developmental Sciences, University of Edinburgh, Centre for Reproductive Biology, 47 Little France Crescent, Edinburgh EH16 4TJ, UK
| | | | | | | | | |
Collapse
|