1
|
Tang C, Lan R, Ma DR, Zhao M, Zhang Y, Li HY, Liu S, Li BY, Yang JL, Yang HJ, Zhang ZQ. Annexin A1: The dawn of ischemic stroke (Review). Mol Med Rep 2025; 31:62. [PMID: 39749707 PMCID: PMC11726294 DOI: 10.3892/mmr.2024.13427] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2024] [Accepted: 12/05/2024] [Indexed: 01/04/2025] Open
Abstract
Ischemic stroke is a prevalent clinical condition that poses a significant global challenge. Developing innovative strategies to address this issue is crucial. Annexin A1 (ANXA1), a key member of the annexin superfamily, performs various functions, such as inhibiting inflammatory factor release, promoting phagocytosis, and blocking leukocyte migration. Evidence indicates that ANXA1 plays a pivotal role in the pathogenesis of ischemic stroke. The present article reviews involvement of ANXA1 in anti‑atherosclerosis, inflammatory processes, blood‑brain barrier protection, platelet aggregation and anti‑apoptotic mechanisms. The potential applications of ANXA1 in treating ischemic stroke are also explored.
Collapse
Affiliation(s)
- Chen Tang
- Department of Encephalopathy, The First Affiliated Hospital of Henan University of Chinese Medicine, The First Clinical Medical College of The First Affiliated Hospital of Henan University of Chinese Medicine, Zhengzhou, Henan 450000, P.R. China
| | - Rui Lan
- Department of Encephalopathy, The First Affiliated Hospital of Henan University of Chinese Medicine, Zhengzhou, Henan 450000, P.R. China
- Collaborative Innovation Center of Prevention and Treatment of Major Diseases by Chinese and Western Medicine, Zhengzhou, Henan 450000, P.R. China
| | - Dong-Rui Ma
- Academy of Chinese Medical Sciences, Henan University of Chinese Medicine, Zhengzhou, Henan 450000, P.R. China
| | - Min Zhao
- Department of Encephalopathy, The First Affiliated Hospital of Henan University of Chinese Medicine, Zhengzhou, Henan 450000, P.R. China
- Collaborative Innovation Center of Prevention and Treatment of Major Diseases by Chinese and Western Medicine, Zhengzhou, Henan 450000, P.R. China
| | - Yong Zhang
- Department of Traditional Chinese Medicine, The Third Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450000, P.R. China
| | - Hong-Yu Li
- Department of Encephalopathy, The First Affiliated Hospital of Henan University of Chinese Medicine, The First Clinical Medical College of The First Affiliated Hospital of Henan University of Chinese Medicine, Zhengzhou, Henan 450000, P.R. China
| | - Shuang Liu
- Department of Encephalopathy, The First Affiliated Hospital of Henan University of Chinese Medicine, The First Clinical Medical College of The First Affiliated Hospital of Henan University of Chinese Medicine, Zhengzhou, Henan 450000, P.R. China
| | - Bo-Yang Li
- Department of Encephalopathy, The First Affiliated Hospital of Henan University of Chinese Medicine, The First Clinical Medical College of The First Affiliated Hospital of Henan University of Chinese Medicine, Zhengzhou, Henan 450000, P.R. China
| | - Jie-Li Yang
- Department of Encephalopathy, The First Affiliated Hospital of Henan University of Chinese Medicine, The First Clinical Medical College of The First Affiliated Hospital of Henan University of Chinese Medicine, Zhengzhou, Henan 450000, P.R. China
| | - Hui-Jie Yang
- Department of Encephalopathy, The First Affiliated Hospital of Henan University of Chinese Medicine, The First Clinical Medical College of The First Affiliated Hospital of Henan University of Chinese Medicine, Zhengzhou, Henan 450000, P.R. China
| | - Zhen-Qiang Zhang
- Academy of Chinese Medical Sciences, Henan University of Chinese Medicine, Zhengzhou, Henan 450000, P.R. China
| |
Collapse
|
2
|
Wei N, Lu T, Gu J, Cai H. Lipoxin A4 suppresses neutrophil extracellular traps formation through the FPR2-dependent regulation of METTL3 in ischemic stroke. Brain Res Bull 2025; 220:111178. [PMID: 39706534 DOI: 10.1016/j.brainresbull.2024.111178] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2024] [Revised: 12/12/2024] [Accepted: 12/18/2024] [Indexed: 12/23/2024]
Abstract
BACKGROUND This study aimed to clarify whether the neuroprotective effect of LXA4 is associated with the targeting of neutrophil extracellular traps (NETs) in ischemic stroke (IS). METHODS The MCAO rat model was established to assess cerebral infarction, brain water content and neurological deficits. ELISA was employed to examine the activities of MPO, NE, MMP-9. RT-qPCR and western blot was performed to analyze molecular expressions. A luciferase reporter assay was performed to measure the effect of EGR1 on the METTL3 promoter. The formation of NETs and cell viability were evaluated using immunofluorescence staining and CCK8 assay, respectively. RESULTS LXA4 decreased cerebral infarction and brain water content, improved neurological deficits, and reduced the release of NETs-associated indicators (MPO, NE) in MCAO rats. LXA4 reduced NETs formation, MPO and NE levels in vitro. In addition, LXA4 reduced Fe2 + levels while increasing GPX4, SLC7A11 protein expressions, as well as enhancing cell viability in vitro, suggesting the inhibitory effect of LXA4 on ferroptosis. Notably, METTL3 overexpression produced the opposite effects. Furthermore, the effects of METTL3 overexpression on NETs formation and ferroptosis were partially reversed by LXA4 treatment. The inhibition of METTL3 by LXA4 was found to be dependent on FPR2. In vivo experiments verified that LXA4 inhibited NETs formation through inhibition of METTL3 to alleviate brain injury. CONCLUSION This study demonstrates that LXA4 suppresses NETs formation through the FPR2-dependent regulation of METTL3, thereby alleviating brain injury in IS.
Collapse
Affiliation(s)
- Na Wei
- Department of Neurology, Shanghai Tenth People's Hospital Chongming Branch, 2866 Chongming Road, Shanghai 202157, China
| | - Tan Lu
- Department of Orthopaedics, The First Affiliated Hospital of Xinxiang Medical University, 88 Jiankang Road, Weihui, Henan 453100, China.
| | - JianBang Gu
- Department of Neurology, Shanghai Tenth People's Hospital Chongming Branch, 2866 Chongming Road, Shanghai 202157, China
| | - Huan Cai
- Department of Neurology, Shanghai Tenth People's Hospital Chongming Branch, 2866 Chongming Road, Shanghai 202157, China
| |
Collapse
|
3
|
Deng Y, Wang F, Wang T, Zhang X, Chen D, Wang Y, Chen C, Pan G. Research progress in the mechanisms and functions of specialized pro-resolving mediators in neurological diseases. Prostaglandins Other Lipid Mediat 2024; 175:106905. [PMID: 39265777 DOI: 10.1016/j.prostaglandins.2024.106905] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2024] [Revised: 08/30/2024] [Accepted: 09/09/2024] [Indexed: 09/14/2024]
Abstract
The nervous system interacts with the immune system through a variety of cellular regulators, signaling pathways, and molecular mechanisms. Disruptions in these interactions lead to the development of multiple neurological diseases. Recent studies have identified that specialized pro-resolving mediators (SPMs) play a regulatory role in the neuroimmune system. This study reviews recent research on the function of SPMs in the inflammatory process and their association with the nervous system. The review aims to provide new perspectives for studying the pathogenesis of neurological diseases and identify novel targets for clinical therapy.
Collapse
Affiliation(s)
- Yu Deng
- Guangzhou Hospital of Integrated Chinese and Western Medicine Affiliated to Guangzhou University of Chinese Medicine, Guangzhou, Guangdong 510800, China
| | - Fei Wang
- Yancheng TCM Hospital Affiliated to Nanjing University of Chinese Medicine, Yancheng, Jiangsu 224000, China; Yancheng TCM Hospital, Yancheng, Jiangsu 224000, China
| | - Tianle Wang
- Guangzhou Hospital of Integrated Chinese and Western Medicine Affiliated to Guangzhou University of Chinese Medicine, Guangzhou, Guangdong 510800, China
| | - Xu Zhang
- Guangzhou Hospital of Integrated Chinese and Western Medicine Affiliated to Guangzhou University of Chinese Medicine, Guangzhou, Guangdong 510800, China
| | - Du Chen
- Guangzhou Hospital of Integrated Chinese and Western Medicine Affiliated to Guangzhou University of Chinese Medicine, Guangzhou, Guangdong 510800, China
| | - Yuhan Wang
- Hubei University of Chinese Medicine, Wuhan, Hubei 430065, China
| | - Chaojun Chen
- Guangzhou Hospital of Integrated Chinese and Western Medicine Affiliated to Guangzhou University of Chinese Medicine, Guangzhou, Guangdong 510800, China.
| | - Guangtao Pan
- Yancheng TCM Hospital Affiliated to Nanjing University of Chinese Medicine, Yancheng, Jiangsu 224000, China; Yancheng TCM Hospital, Yancheng, Jiangsu 224000, China.
| |
Collapse
|
4
|
Döring Y, van der Vorst EPC, Weber C. Targeting immune cell recruitment in atherosclerosis. Nat Rev Cardiol 2024; 21:824-840. [PMID: 38664575 DOI: 10.1038/s41569-024-01023-z] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 03/26/2024] [Indexed: 10/17/2024]
Abstract
Atherosclerosis is the primary underlying cause of myocardial infarction and stroke. Atherosclerotic cardiovascular disease is characterized by a chronic inflammatory reaction in medium-to-large-sized arteries, with its onset and perpetuation driven by leukocytes infiltrating the subendothelial space. Activation of endothelial cells triggered by hyperlipidaemia and lipoprotein retention in the arterial intima initiates the accumulation of pro-inflammatory leukocytes in the arterial wall, fostering the progression of atherosclerosis. This inflammatory response is coordinated by an array of soluble mediators, namely cytokines and chemokines, that amplify inflammation both locally and systemically and are complemented by tissue-specific molecules that regulate the homing, adhesion and transmigration of leukocytes. Despite abundant evidence from mouse models, only a few therapies targeting leukocytes in atherosclerosis have been assessed in humans. The major challenges for the clinical translation of these therapies include the lack of tissue specificity and insufficient selectivity of inhibition strategies. In this Review, we discuss the latest research on receptor-ligand pairs and interactors that regulate leukocyte influx into the inflamed artery wall, primarily focusing on studies that used pharmacological interventions. We also discuss mechanisms that promote the resolution of inflammation and highlight how major findings from these research areas hold promise as potential therapeutic strategies for atherosclerotic cardiovascular disease.
Collapse
Affiliation(s)
- Yvonne Döring
- Department of Angiology, Swiss Cardiovascular Center, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland.
- Institute for Cardiovascular Prevention (IPEK), Ludwig-Maximilians-University Munich (LMU), Munich, Germany.
- DZHK (German Centre for Cardiovascular Research), Partner Site Munich Heart Alliance, Munich, Germany.
- Department for BioMedical Research (DBMR), University of Bern, Bern, Switzerland.
| | - Emiel P C van der Vorst
- Institute for Cardiovascular Prevention (IPEK), Ludwig-Maximilians-University Munich (LMU), Munich, Germany.
- Institute for Molecular Cardiovascular Research (IMCAR), RWTH Aachen University, Aachen, Germany.
- Aachen-Maastricht Institute for CardioRenal Disease (AMICARE), RWTH Aachen University, Aachen, Germany.
- Interdisciplinary Center for Clinical Research (IZKF), RWTH Aachen University, Aachen, Germany.
| | - Christian Weber
- Institute for Cardiovascular Prevention (IPEK), Ludwig-Maximilians-University Munich (LMU), Munich, Germany.
- DZHK (German Centre for Cardiovascular Research), Partner Site Munich Heart Alliance, Munich, Germany.
- Department of Biochemistry, Cardiovascular Research Institute Maastricht (CARIM), Maastricht University, Maastricht, Netherlands.
- Munich Cluster for Systems Neurology (SyNergy), Munich, Germany.
| |
Collapse
|
5
|
Pajonczyk D, Sternschulte MF, Soehnlein O, Bermudez M, Raabe CA, Rescher U. Comparative analysis of formyl peptide receptor 1 and formyl peptide receptor 2 reveals shared and preserved signalling profiles. Br J Pharmacol 2024. [PMID: 39294930 DOI: 10.1111/bph.17334] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2024] [Revised: 07/03/2024] [Accepted: 08/06/2024] [Indexed: 09/21/2024] Open
Abstract
BACKGROUND AND PURPOSE The pattern recognition receptors, formyl peptide receptors, FPR1 and FPR2, are G protein-coupled receptors that recognize many different pathogen- and host-derived ligands. While FPR1 conveys pro-inflammatory signals, FPR2 is linked with pro-resolving outcomes. To analyse how the two very similar FPRs exert opposite effects in modulating inflammatory responses despite their high homology, a shared expression profile on immune cells and an overlapping ligand repertoire, we questioned whether the signalling profile differs between these two receptors. EXPERIMENTAL APPROACH We deduced EC50 and Emax values for synthetic, pathogen-derived and host-derived peptide agonists for both FPR1 and FPR2 and analysed them within the framework of biased signalling. We furthermore investigated whether FPR isoform-specific agonists affect the ex vivo lifespan of human neutrophils. KEY RESULTS The FPRs share a core signature across signalling pathways. Whereas the synthetic WKYMVm and formylated peptides acted as potent agonists at FPR1, and at FPR2, only WKYMVm was a full agonist. Natural FPR2 agonists, irrespective of N-terminal formylation, displayed lower activity ratios, suggesting an underutilized signalling potential of this receptor. FPR2 agonism did not counteract LPS-induced neutrophil survival, indicating that FPR2 activation per se is not linked with a pro-resolving function. CONCLUSION AND IMPLICATIONS Activation of FPR1 and FPR2 by a representative agonist panel revealed a lack of a receptor-specific signalling texture, challenging assumptions about distinct inflammatory profiles linked to specific receptor isoforms, signalling patterns or agonist classes. These conclusions are restricted to the specific agonists and signalling pathways examined.
Collapse
Affiliation(s)
- Denise Pajonczyk
- Research Group Cellular Biochemistry - Regulatory Mechanisms of Inflammation, Institute of Molecular Virology, Center of Molecular Biology of Inflammation and "Cells in Motion" Interfaculty Centre, University of Muenster, Muenster, Germany
| | - Merle F Sternschulte
- Research Group Cellular Biochemistry - Regulatory Mechanisms of Inflammation, Institute of Molecular Virology, Center of Molecular Biology of Inflammation and "Cells in Motion" Interfaculty Centre, University of Muenster, Muenster, Germany
- Institute of Experimental Pathology, Center of Molecular Biology of Inflammation, University of Muenster, Muenster, Germany
| | - Oliver Soehnlein
- Institute of Experimental Pathology, Center of Molecular Biology of Inflammation, University of Muenster, Muenster, Germany
| | - Marcel Bermudez
- Institute of Pharmaceutical and Medicinal Chemistry, University of Muenster, Muenster, Germany
| | - Carsten A Raabe
- Research Group Cellular Biochemistry - Regulatory Mechanisms of Inflammation, Institute of Molecular Virology, Center of Molecular Biology of Inflammation and "Cells in Motion" Interfaculty Centre, University of Muenster, Muenster, Germany
| | - Ursula Rescher
- Research Group Cellular Biochemistry - Regulatory Mechanisms of Inflammation, Institute of Molecular Virology, Center of Molecular Biology of Inflammation and "Cells in Motion" Interfaculty Centre, University of Muenster, Muenster, Germany
| |
Collapse
|
6
|
Hardesty JE, Warner JB, Wilkey DW, Phinney BS, Salemi MR, Merchant ML, McClain CJ, Warner DR, Kirpich IA. Hepatic Proteomic Changes Associated with Liver Injury Caused by Alcohol Consumption in Fpr2-/- Mice. Int J Mol Sci 2024; 25:9807. [PMID: 39337294 PMCID: PMC11432144 DOI: 10.3390/ijms25189807] [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: 08/19/2024] [Revised: 09/05/2024] [Accepted: 09/07/2024] [Indexed: 09/30/2024] Open
Abstract
Alcohol-associated liver disease (ALD) is a prevalent medical problem with limited effective treatment strategies. Although many biological processes contributing to ALD have been elucidated, a complete understanding of the underlying mechanisms is still lacking. The current study employed a proteomic approach to identify hepatic changes resulting from ethanol (EtOH) consumption and the genetic ablation of the formyl peptide receptor 2 (FPR2), a G-protein coupled receptor known to regulate multiple signaling pathways and biological processes, in a mouse model of ALD. Since previous research from our team demonstrated a notable reduction in hepatic FPR2 protein levels in patients with alcohol-associated hepatitis (AH), the proteomic changes in the livers of Fpr2-/- EtOH mice were compared to those observed in patients with AH in order to identify common hepatic proteomic alterations. Several pathways linked to exacerbated ALD in Fpr2-/- EtOH mice, as well as hepatic protein changes resembling those found in patients suffering from AH, were identified. These alterations included decreased levels of coagulation factors F2 and F9, as well as reduced hepatic levels of glutamate-cysteine ligase catalytic subunit (GCLC) and total glutathione in Fpr2-/- EtOH compared to WT EtOH mice. In conclusion, the data suggest that FPR2 may play a regulatory role in hepatic blood coagulation and the antioxidant system, both in a pre-clinical model of ALD and in human AH, however further experiments are required to validate these findings.
Collapse
Affiliation(s)
- Josiah E. Hardesty
- Division of Gastroenterology, Hepatology and Nutrition, Department of Medicine, University of Louisville, Louisville, KY 40202, USA; (J.E.H.); (J.B.W.); (C.J.M.); (D.R.W.)
- Department of Pharmacology and Toxicology, School of Medicine, University of Louisville, Louisville, KY 40202, USA;
| | - Jeffrey B. Warner
- Division of Gastroenterology, Hepatology and Nutrition, Department of Medicine, University of Louisville, Louisville, KY 40202, USA; (J.E.H.); (J.B.W.); (C.J.M.); (D.R.W.)
- Department of Pharmacology and Toxicology, School of Medicine, University of Louisville, Louisville, KY 40202, USA;
| | - Daniel W. Wilkey
- The Proteomics Core, School of Medicine, University of Louisville, Louisville, KY 40202, USA;
| | - Brett S. Phinney
- Proteomics Core Facility, University of California Davis, Davis, CA 95616, USA; (B.S.P.); (M.R.S.)
| | - Michelle R. Salemi
- Proteomics Core Facility, University of California Davis, Davis, CA 95616, USA; (B.S.P.); (M.R.S.)
| | - Michael L. Merchant
- Department of Pharmacology and Toxicology, School of Medicine, University of Louisville, Louisville, KY 40202, USA;
- The Proteomics Core, School of Medicine, University of Louisville, Louisville, KY 40202, USA;
| | - Craig J. McClain
- Division of Gastroenterology, Hepatology and Nutrition, Department of Medicine, University of Louisville, Louisville, KY 40202, USA; (J.E.H.); (J.B.W.); (C.J.M.); (D.R.W.)
- Department of Pharmacology and Toxicology, School of Medicine, University of Louisville, Louisville, KY 40202, USA;
- Robley Rex Veterans Medical Center, Louisville, KY 40202, USA
- Alcohol Research Center, School of Medicine, University of Louisville, Louisville, KY 40202, USA
- Hepatobiology & Toxicology Center, School of Medicine, University of Louisville, Louisville, KY 40202, USA
| | - Dennis R. Warner
- Division of Gastroenterology, Hepatology and Nutrition, Department of Medicine, University of Louisville, Louisville, KY 40202, USA; (J.E.H.); (J.B.W.); (C.J.M.); (D.R.W.)
| | - Irina A. Kirpich
- Alcohol Research Center, School of Medicine, University of Louisville, Louisville, KY 40202, USA
- Hepatobiology & Toxicology Center, School of Medicine, University of Louisville, Louisville, KY 40202, USA
- Department of Microbiology and Immunology, School of Medicine, University of Louisville, Louisville, KY 40202, USA
| |
Collapse
|
7
|
Vieira CP, Lelis CA, Ochioni AC, Rosário DKA, Rosario ILS, Vieira IRS, Carvalho APA, Janeiro JM, da Costa MP, Lima FRS, Mariante RM, Alves LA, Foguel D, Junior CAC. Estimating the therapeutic potential of NSAIDs and linoleic acid-isomers supplementation against neuroinflammation. Biomed Pharmacother 2024; 177:116884. [PMID: 38889635 DOI: 10.1016/j.biopha.2024.116884] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2024] [Revised: 06/03/2024] [Accepted: 06/03/2024] [Indexed: 06/20/2024] Open
Abstract
Nonsteroidal anti-inflammatory drugs (NSAIDs) regulate inflammation, which is associated with their role in preventing neurodegenerative diseases in epidemiological studies. It has sparked interest in their unconventional application for reducing neuroinflammation, opening up new avenues in biomedical research. However, given the pharmacological drawbacks of NSAIDs, the development of formulations with naturally antioxidant/anti-inflammatory dietary fatty acids has been demonstrated to be advantageous for the clinical translation of anti-inflammatory-based therapies. It includes improved blood-brain barrier (BBB) permeability and reduced toxicity. It permits us to speculate about the value of linoleic acid (LA)-isomers in preventing and treating neuroinflammatory diseases compared to NSAIDs. Our research delved into the impact of various factors, such as administration route, dosage, timing of intervention, and BBB permeability, on the efficacy of NSAIDs and LA-isomers in preclinical and clinical settings. We conducted a systematic comparison between NSAIDs and LA-isomers regarding their therapeutic effectiveness, BBB compatibility, and side effects. Additionally, we explored their underlying mechanisms in addressing neuroinflammation. Through our analysis, we've identified challenges and drawn conclusions that could propel advancements in treating neurodegenerative diseases and inform the development of future alternative therapeutic strategies.
Collapse
Affiliation(s)
- Carla Paulo Vieira
- Laboratory of Advanced Analysis in Biochemistry and Molecular Biology (LAABBM), Department of Biochemistry, Universidade Federal do Rio de Janeiro (UFRJ), Rio de Janeiro, RJ 21941-909, Brazil; Center for Food Analysis (NAL), Technological Development Support Laboratory (LADETEC), Universidade Federal do Rio de Janeiro (UFRJ), Rio de Janeiro, RJ 21941-598, Brazil; Cellular Communication Laboratory, Instituto Oswaldo Cruz, Fundação Oswaldo Cruz (FIOCRUZ), Rio de Janeiro, RJ 21040-900, Brazil
| | - Carini A Lelis
- Laboratory of Advanced Analysis in Biochemistry and Molecular Biology (LAABBM), Department of Biochemistry, Universidade Federal do Rio de Janeiro (UFRJ), Rio de Janeiro, RJ 21941-909, Brazil; Center for Food Analysis (NAL), Technological Development Support Laboratory (LADETEC), Universidade Federal do Rio de Janeiro (UFRJ), Rio de Janeiro, RJ 21941-598, Brazil
| | - Alan Clavelland Ochioni
- Laboratory of Advanced Analysis in Biochemistry and Molecular Biology (LAABBM), Department of Biochemistry, Universidade Federal do Rio de Janeiro (UFRJ), Rio de Janeiro, RJ 21941-909, Brazil; Center for Food Analysis (NAL), Technological Development Support Laboratory (LADETEC), Universidade Federal do Rio de Janeiro (UFRJ), Rio de Janeiro, RJ 21941-598, Brazil
| | - Denes Kaic A Rosário
- Laboratory of Advanced Analysis in Biochemistry and Molecular Biology (LAABBM), Department of Biochemistry, Universidade Federal do Rio de Janeiro (UFRJ), Rio de Janeiro, RJ 21941-909, Brazil; Center for Food Analysis (NAL), Technological Development Support Laboratory (LADETEC), Universidade Federal do Rio de Janeiro (UFRJ), Rio de Janeiro, RJ 21941-598, Brazil
| | - Iuri L S Rosario
- Center for Food Analysis (NAL), Technological Development Support Laboratory (LADETEC), Universidade Federal do Rio de Janeiro (UFRJ), Rio de Janeiro, RJ 21941-598, Brazil; Laboratory of Inspection and Technology of Milk and Derivatives (LaITLácteos), School of Veterinary Medicine and Zootechnies, Universidade Federal da Bahia (UFBA), Ondina, Salvador, BA 40170-110, Brazil
| | - Italo Rennan S Vieira
- Laboratory of Advanced Analysis in Biochemistry and Molecular Biology (LAABBM), Department of Biochemistry, Universidade Federal do Rio de Janeiro (UFRJ), Rio de Janeiro, RJ 21941-909, Brazil; Center for Food Analysis (NAL), Technological Development Support Laboratory (LADETEC), Universidade Federal do Rio de Janeiro (UFRJ), Rio de Janeiro, RJ 21941-598, Brazil
| | - Anna Paula A Carvalho
- Laboratory of Advanced Analysis in Biochemistry and Molecular Biology (LAABBM), Department of Biochemistry, Universidade Federal do Rio de Janeiro (UFRJ), Rio de Janeiro, RJ 21941-909, Brazil; Center for Food Analysis (NAL), Technological Development Support Laboratory (LADETEC), Universidade Federal do Rio de Janeiro (UFRJ), Rio de Janeiro, RJ 21941-598, Brazil
| | - José Marcos Janeiro
- Glial Cell Biology Laboratory, Institute of Biomedical Sciences, Universidade Federal do Rio de Janeiro (UFRJ), Rio de Janeiro, RJ 21941-590, Brazil
| | - Marion P da Costa
- Laboratory of Inspection and Technology of Milk and Derivatives (LaITLácteos), School of Veterinary Medicine and Zootechnies, Universidade Federal da Bahia (UFBA), Ondina, Salvador, BA 40170-110, Brazil; Graduate Program in Food Science (PGAli), Faculty of Pharmacy, Universidade Federal da Bahia (UFBA), Ondina, Salvador, BA 40170-110, Brazil
| | - Flavia R S Lima
- Glial Cell Biology Laboratory, Institute of Biomedical Sciences, Universidade Federal do Rio de Janeiro (UFRJ), Rio de Janeiro, RJ 21941-590, Brazil
| | - Rafael M Mariante
- Laboratory of Structural Biology, Instituto Oswaldo Cruz, Fundação Oswaldo Cruz (Fiocruz), Rio de Janeiro, RJ 21040-900, Brazil
| | - Luiz Anastácio Alves
- Cellular Communication Laboratory, Instituto Oswaldo Cruz, Fundação Oswaldo Cruz (FIOCRUZ), Rio de Janeiro, RJ 21040-900, Brazil
| | - Debora Foguel
- Laboratory of Protein Aggregation and Amyloidosis, Institute of Medical Biochemistry, Universidade Federal do Rio de Janeiro (UFRJ), Rio de Janeiro, RJ 21941-590, Brazil
| | - Carlos Adam Conte Junior
- Laboratory of Advanced Analysis in Biochemistry and Molecular Biology (LAABBM), Department of Biochemistry, Universidade Federal do Rio de Janeiro (UFRJ), Rio de Janeiro, RJ 21941-909, Brazil; Center for Food Analysis (NAL), Technological Development Support Laboratory (LADETEC), Universidade Federal do Rio de Janeiro (UFRJ), Rio de Janeiro, RJ 21941-598, Brazil.
| |
Collapse
|
8
|
Gerke V, Gavins FNE, Geisow M, Grewal T, Jaiswal JK, Nylandsted J, Rescher U. Annexins-a family of proteins with distinctive tastes for cell signaling and membrane dynamics. Nat Commun 2024; 15:1574. [PMID: 38383560 PMCID: PMC10882027 DOI: 10.1038/s41467-024-45954-0] [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: 01/30/2023] [Accepted: 02/07/2024] [Indexed: 02/23/2024] Open
Abstract
Annexins are cytosolic proteins with conserved three-dimensional structures that bind acidic phospholipids in cellular membranes at elevated Ca2+ levels. Through this they act as Ca2+-regulated membrane binding modules that organize membrane lipids, facilitating cellular membrane transport but also displaying extracellular activities. Recent discoveries highlight annexins as sensors and regulators of cellular and organismal stress, controlling inflammatory reactions in mammals, environmental stress in plants, and cellular responses to plasma membrane rupture. Here, we describe the role of annexins as Ca2+-regulated membrane binding modules that sense and respond to cellular stress and share our view on future research directions in the field.
Collapse
Affiliation(s)
- Volker Gerke
- Institute of Medical Biochemistry, Center for Molecular Biology of Inflammation (ZMBE), University of Münster, Von-Esmarch-Strasse 56, Münster, Germany.
| | - Felicity N E Gavins
- Department of Life Sciences, Centre for Inflammation Research and Translational Medicine (CIRTM), Brunel University London, Uxbridge, UK
| | - Michael Geisow
- The National Institute for Medical Research, Mill Hill, London, UK
- Delta Biotechnology Ltd, Nottingham, UK
| | - Thomas Grewal
- School of Pharmacy, Faculty of Medicine and Health, University of Sydney, Sydney, NSW, Australia
| | - Jyoti K Jaiswal
- Center for Genetic Medicine Research, Children's National Research Institute, Children's National Research and Innovation Campus, Washington, DC, USA
- Department of Genomics and Precision Medicine, The George Washington University School of Medicine and Health Sciences, Washington, DC, USA
| | - Jesper Nylandsted
- Danish Cancer Institute, Strandboulevarden 49, Copenhagen, Denmark
- Department of Molecular Medicine, University of Southern Denmark, J.B. Winsløws Vej 21-25, Odense, Denmark
| | - Ursula Rescher
- Research Group Cellular Biochemistry, Institute of Molecular Virology, Center for Molecular Biology of Inflammation (ZMBE), University of Münster, Von-Esmarch-Strasse 56, Münster, Germany.
| |
Collapse
|
9
|
Flores J, Tang J. Role of N-formyl peptide receptor 2 in germinal matrix hemorrhage: an intrinsic review of a hematoma resolving pathway. Neural Regen Res 2024; 19:350-354. [PMID: 37488889 PMCID: PMC10503603 DOI: 10.4103/1673-5374.379040] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2023] [Revised: 03/28/2023] [Accepted: 05/04/2023] [Indexed: 07/26/2023] Open
Abstract
Germinal matrix hemorrhage is one of the leading causes of morbidity, mortality, and acquired infantile hydrocephalus in preterm infants in the United States, with little progress made in its clinical management. Blood clots have been shown to elicit secondary brain injury after germinal matrix hemorrhage, by disrupting normal cerebrospinal fluid circulation and absorption after germinal matrix hemorrhage causing post-hemorrhagic hydrocephalus development. Current evidence suggests that rapid hematoma resolution is necessary to improve neurological outcomes after hemorrhagic stroke. Various articles have demonstrated the beneficial effects of stimulating the polarization of microglia cells into the M2 phenotype, as it has been suggested that they play an essential role in the rapid phagocytosis of the blood clot after hemorrhagic models of stroke. N-formyl peptide receptor 2 (FPR2), a G-protein-coupled receptor, has been shown to be neuroprotective after stroke. FPR2 activation has been associated with the upregulation of phagocytic macrophage clearance, yet its mechanism has not been fully explored. Recent literature suggests that FPR2 may play a role in the stimulation of scavenger receptor CD36. Scavenger receptor CD36 plays a vital role in microglia phagocytic blood clot clearance after germinal matrix hemorrhage. FPR2 has been shown to phosphorylate extracellular-signal-regulated kinase 1/2 (ERK1/2), which then promotes the transcription of the dual-specificity protein phosphatase 1 (DUSP1) gene. In this review, we present an intrinsic outline of the main components involved in FPR2 stimulation and hematoma resolution after germinal matrix hemorrhage.
Collapse
Affiliation(s)
- Jerry Flores
- Department of Physiology & Pharmacology, Loma Linda University School of Medicine, Loma Linda, CA, USA
| | - Jiping Tang
- Department of Physiology & Pharmacology, Loma Linda University School of Medicine, Loma Linda, CA, USA
| |
Collapse
|
10
|
Livne-Bar I, Maurya S, Gronert K, Sivak JM. Lipoxins A 4 and B 4 inhibit glial cell activation via CXCR3 signaling in acute retinal neuroinflammation. J Neuroinflammation 2024; 21:18. [PMID: 38212822 PMCID: PMC10782675 DOI: 10.1186/s12974-024-03010-0] [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: 08/02/2023] [Accepted: 01/01/2024] [Indexed: 01/13/2024] Open
Abstract
Lipoxins are small lipids that are potent endogenous mediators of systemic inflammation resolution in a variety of diseases. We previously reported that Lipoxins A4 and B4 (LXA4 and LXB4) have protective activities against neurodegenerative injury. Yet, lipoxin activities and downstream signaling in neuroinflammatory processes are not well understood. Here, we utilized a model of posterior uveitis induced by lipopolysaccharide endotoxin (LPS), which results in rapid retinal neuroinflammation primarily characterized by activation of resident macroglia (astrocytes and Müller glia), and microglia. Using this model, we observed that each lipoxin reduces acute inner retinal inflammation by affecting endogenous glial responses in a cascading sequence beginning with astrocytes and then microglia, depending on the timing of exposure; prophylactic or therapeutic. Subsequent analyses of retinal cytokines and chemokines revealed inhibition of both CXCL9 (MIG) and CXCL10 (IP10) by each lipoxin, compared to controls, following LPS injection. CXCL9 and CXCL10 are common ligands for the CXCR3 chemokine receptor, which is prominently expressed in inner retinal astrocytes and ganglion cells. We found that CXCR3 inhibition reduces LPS-induced neuroinflammation, while CXCR3 agonism alone induces astrocyte reactivity. Together, these data uncover a novel lipoxin-CXCR3 pathway to promote distinct anti-inflammatory and proresolution cascades in endogenous retinal glia.
Collapse
Affiliation(s)
- Izhar Livne-Bar
- Department of Vision Sciences, Donald K Johnson Eye Institute, Krembil Research Institute, University Health Network, Krembil Discovery Tower, 60 Leonard Avenue, Toronto, ON, M5T 0S8, Canada
- Department of Ophthalmology and Vision Science, University of Toronto School of Medicine, Toronto, Canada
| | - Shubham Maurya
- Herbert Wertheim School of Optometry and Vision Science, University of California Berkeley, Berkeley, CA, USA
| | - Karsten Gronert
- Herbert Wertheim School of Optometry and Vision Science, University of California Berkeley, Berkeley, CA, USA
- Vision Science Program, University of California Berkeley, Berkeley, CA, USA
- Infectious Disease and Immunity Program, University of California Berkeley, Berkeley, CA, USA
| | - Jeremy M Sivak
- Department of Vision Sciences, Donald K Johnson Eye Institute, Krembil Research Institute, University Health Network, Krembil Discovery Tower, 60 Leonard Avenue, Toronto, ON, M5T 0S8, Canada.
- Department of Ophthalmology and Vision Science, University of Toronto School of Medicine, Toronto, Canada.
- Department of Laboratory Medicine and Pathobiology, University of Toronto School of Medicine, Toronto, Canada.
| |
Collapse
|
11
|
Meng Y, Lin Y, Zhang JW, Zou WL, Liu YM, Shen XG, Shen QQ, Wang MM, Shao LN, Feng HY, Zhu Y, Yu JT, Lin B, Zhu B. Aspirin intervention before ICU admission reduced the mortality in critically ill patients with acute kidney injury: results from the MIMIC-IV. Front Pharmacol 2023; 14:1292745. [PMID: 38034989 PMCID: PMC10682711 DOI: 10.3389/fphar.2023.1292745] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2023] [Accepted: 10/31/2023] [Indexed: 12/02/2023] Open
Abstract
Background: Aspirin, with its pleiotropic effects such as anti-inflammatory and anti-platelet aggregation, has been widely used for anti-inflammatory, analgesic, and cardiovascular diseases. However, the association between the use of aspirin before the intensive care unit (ICU) and clinical outcomes in critically ill patients with acute kidney injury (AKI) is unknown. Methods: Patients with AKI in this retrospective observational study were selected from the Marketplace for Medical Information in Intensive Care IV (MIMIC-IV). The association between aspirin intervention and 30-day mortality was assessed using Cox proportional hazards model. Logistic regression models were used to assess the association of aspirin intervention with the risks of intracranial hemorrhage, gastrointestinal bleeding and blood transfusion. The propensity score matching (PSM) method was adopted to balance the baseline variables. Sensitivity analysis was performed to validate the results by multiple interpolations for the missing data. Results: The study included 4237 pre-ICU aspirin users and 9745 non-users. In multivariate models, we found a decreased risk of mortality in those who received aspirin before ICU compared to those who did not (30-day:hazard ratio [HR], 0.70; 95% CI, 0.62-0.79; p < 0.001; 90-day:HR, 0.70; 95% CI, 0.63-0.77, p < 0.001; 180-day:HR, 0.72; 95%CI,0.65-0.79, p < 0.001). This benefit was consistent in the post-PSM analyses, sensitivity analyses, and subgroup analyses. Moreover, aspirin intervention was associated with a reduced risk of intracranial hemorrhage and gastrointestinal bleeding (HR, 0.16; 95% CI, 0.10-0.25; p < 0.001; HR, 0.59; 95% CI, 0.38-0.88, p = 0.012) after being adjusted by relating covariates, whereas with a increased risk of blood transfusion (HR, 1.28; 95% CI, 1.16-1.46; p < 0.001). Conclusion: Patients with AKI treated with aspirin before ICU admission might have reduced 30-day, 90-day and 180-day mortality without increasing the risk of intracranial hemorrhage (ICH) or gastrointestinal bleeding, but may increase the risk of transfusion.
Collapse
Affiliation(s)
- Yao Meng
- Urology and Nephrology Center, Department of Nephrology, Zhejiang Provincial People’s Hospital (Affiliated People’s Hospital), Hangzhou Medical College, Hangzhou, Zhejiang, China
- Hangzhou Hospital of Traditional Chinese Medicine, Affiliated to Zhejiang Chinese Medical University, Hangzhou, China
| | - Yi Lin
- Hangzhou Hospital of Traditional Chinese Medicine, Affiliated to Zhejiang Chinese Medical University, Hangzhou, China
| | - Jia-Wei Zhang
- Urology and Nephrology Center, Department of Nephrology, Zhejiang Provincial People’s Hospital (Affiliated People’s Hospital), Hangzhou Medical College, Hangzhou, Zhejiang, China
| | - Wen-Li Zou
- Urology and Nephrology Center, Department of Nephrology, Zhejiang Provincial People’s Hospital (Affiliated People’s Hospital), Hangzhou Medical College, Hangzhou, Zhejiang, China
| | - Yue-Ming Liu
- Urology and Nephrology Center, Department of Nephrology, Zhejiang Provincial People’s Hospital (Affiliated People’s Hospital), Hangzhou Medical College, Hangzhou, Zhejiang, China
| | - Xiao-Gang Shen
- Urology and Nephrology Center, Department of Nephrology, Zhejiang Provincial People’s Hospital (Affiliated People’s Hospital), Hangzhou Medical College, Hangzhou, Zhejiang, China
| | - Quan-Quan Shen
- Urology and Nephrology Center, Department of Nephrology, Zhejiang Provincial People’s Hospital (Affiliated People’s Hospital), Hangzhou Medical College, Hangzhou, Zhejiang, China
| | - Min-Min Wang
- Urology and Nephrology Center, Department of Nephrology, Zhejiang Provincial People’s Hospital (Affiliated People’s Hospital), Hangzhou Medical College, Hangzhou, Zhejiang, China
| | - Li-Na Shao
- Urology and Nephrology Center, Department of Nephrology, Zhejiang Provincial People’s Hospital (Affiliated People’s Hospital), Hangzhou Medical College, Hangzhou, Zhejiang, China
| | - Hong-Yuan Feng
- Hangzhou Hospital of Traditional Chinese Medicine, Affiliated to Zhejiang Chinese Medical University, Hangzhou, China
| | - Yan Zhu
- Hangzhou Hospital of Traditional Chinese Medicine, Affiliated to Zhejiang Chinese Medical University, Hangzhou, China
| | - Jing-Ting Yu
- Hangzhou Hospital of Traditional Chinese Medicine, Affiliated to Zhejiang Chinese Medical University, Hangzhou, China
| | - Bo Lin
- Urology and Nephrology Center, Department of Nephrology, Zhejiang Provincial People’s Hospital (Affiliated People’s Hospital), Hangzhou Medical College, Hangzhou, Zhejiang, China
| | - Bin Zhu
- Urology and Nephrology Center, Department of Nephrology, Zhejiang Provincial People’s Hospital (Affiliated People’s Hospital), Hangzhou Medical College, Hangzhou, Zhejiang, China
| |
Collapse
|
12
|
Sánchez-García S, Jaén RI, Fernández-Velasco M, Delgado C, Boscá L, Prieto P. Lipoxin-mediated signaling: ALX/FPR2 interaction and beyond. Pharmacol Res 2023; 197:106982. [PMID: 37925045 DOI: 10.1016/j.phrs.2023.106982] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/07/2023] [Revised: 11/01/2023] [Accepted: 11/02/2023] [Indexed: 11/06/2023]
Abstract
In the aftermath of tissue injury or infection, an efficient resolution mechanism is crucial to allow tissue healing and preserve appropriate organ functioning. Pro-resolving bioactive lipids prevent uncontrolled inflammation and its consequences. Among these mediators, lipoxins were the first described and their pro-resolving actions have been mainly described in immune cells. They exert their actions mostly through formyl-peptide receptor 2 (ALX/FPR2 receptor), a G-protein-coupled receptor whose biological function is tremendously complex, primarily due to its capacity to mediate variable cellular responses. Moreover, lipoxins can also interact with alternative receptors like the cytoplasmic aryl hydrocarbon receptor, the cysteinyl-leukotrienes receptors or GPR32, triggering different intracellular signaling pathways. The available information about this complex response mediated by lipoxins is addressed in this review, going over the different mechanisms used by these molecules to stop the inflammatory reaction and avoid the development of dysregulated and chronic pathologies.
Collapse
Affiliation(s)
- Sergio Sánchez-García
- Instituto de Investigaciones Biomédicas "Alberto Sols", CSIC-UAM Madrid, Spain; Centro de Investigación Biomédica en Red de Enfermedades Cardiovasculares (CIBER-CV), Instituto de Salud Carlos III, Madrid, Spain
| | - Rafael I Jaén
- Instituto de Investigaciones Biomédicas "Alberto Sols", CSIC-UAM Madrid, Spain; Centro de Investigación Biomédica en Red de Enfermedades Cardiovasculares (CIBER-CV), Instituto de Salud Carlos III, Madrid, Spain
| | - María Fernández-Velasco
- Centro de Investigación Biomédica en Red de Enfermedades Cardiovasculares (CIBER-CV), Instituto de Salud Carlos III, Madrid, Spain; Instituto de Investigación del Hospital La Paz, IdiPaz, Madrid, Spain
| | - Carmen Delgado
- Instituto de Investigaciones Biomédicas "Alberto Sols", CSIC-UAM Madrid, Spain; Centro de Investigación Biomédica en Red de Enfermedades Cardiovasculares (CIBER-CV), Instituto de Salud Carlos III, Madrid, Spain
| | - Lisardo Boscá
- Instituto de Investigaciones Biomédicas "Alberto Sols", CSIC-UAM Madrid, Spain; Centro de Investigación Biomédica en Red de Enfermedades Cardiovasculares (CIBER-CV), Instituto de Salud Carlos III, Madrid, Spain.
| | - Patricia Prieto
- Instituto de Investigaciones Biomédicas "Alberto Sols", CSIC-UAM Madrid, Spain; Centro de Investigación Biomédica en Red de Enfermedades Cardiovasculares (CIBER-CV), Instituto de Salud Carlos III, Madrid, Spain; Departamento de Farmacología, Farmacognosia y Botánica, Facultad de Farmacia, Universidad Complutense de Madrid, Madrid, Spain.
| |
Collapse
|
13
|
Parent S, Vaka R, Risha Y, Ngo C, Kanda P, Nattel S, Khan S, Courtman D, Stewart DJ, Davis DR. Prevention of atrial fibrillation after open-chest surgery with extracellular vesicle therapy. JCI Insight 2023; 8:e163297. [PMID: 37384420 PMCID: PMC10481795 DOI: 10.1172/jci.insight.163297] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2022] [Accepted: 06/28/2023] [Indexed: 07/01/2023] Open
Abstract
Almost half of patients recovering from open-chest surgery experience atrial fibrillation (AF) that results principally from inflammation in the pericardial space surrounding the heart. Given that postoperative AF is associated with increased mortality, effective measures to prevent AF after open-chest surgery are highly desirable. In this study, we tested the concept that extracellular vesicles (EVs) isolated from human atrial explant-derived cells can prevent postoperative AF. Middle-aged female and male rats were randomized to undergo sham operation or induction of sterile pericarditis followed by trans-epicardial injection of human EVs or vehicle into the atrial tissue. Pericarditis increased the probability of inducing AF while EV treatment abrogated this effect in a sex-independent manner. EV treatment reduced infiltration of inflammatory cells and production of pro-inflammatory cytokines. Atrial fibrosis and hypertrophy seen after pericarditis were markedly attenuated by EV pretreatment, an effect attributable to suppression of fibroblast proliferation by EVs. Our study demonstrates that injection of EVs at the time of open-chest surgery shows prominent antiinflammatory effects and prevents AF due to sterile pericarditis. Translation of this finding to patients might provide an effective new strategy to prevent postoperative AF by reducing atrial inflammation and fibrosis.
Collapse
Affiliation(s)
- Sandrine Parent
- University of Ottawa Heart Institute, Division of Cardiology, Department of Medicine, and
- Department of Cellular and Molecular Medicine, Faculty of Medicine, University of Ottawa, Ottawa, Ontario, Canada
| | - Ramana Vaka
- University of Ottawa Heart Institute, Division of Cardiology, Department of Medicine, and
| | - Yousef Risha
- University of Ottawa Heart Institute, Division of Cardiology, Department of Medicine, and
| | - Clarissa Ngo
- University of Ottawa Heart Institute, Division of Cardiology, Department of Medicine, and
| | - Pushpinder Kanda
- University of Ottawa Heart Institute, Division of Cardiology, Department of Medicine, and
- Department of Cellular and Molecular Medicine, Faculty of Medicine, University of Ottawa, Ottawa, Ontario, Canada
| | - Stanley Nattel
- Research Center and Department of Medicine, Montreal Heart Institute, University of Montreal, Montreal, Quebec, Canada
- Department of Pharmacology and Therapeutics, McGill University, Montreal, Quebec, Canada
- Institute of Pharmacology, West German Heart and Vascular Center, Faculty of Medicine, University of Duisburg-Essen, Essen, Germany
| | - Saad Khan
- Ottawa Hospital Research Institute, Division of Regenerative Medicine, Department of Medicine, University of Ottawa, Ottawa, Ontario, Canada
| | - David Courtman
- Department of Cellular and Molecular Medicine, Faculty of Medicine, University of Ottawa, Ottawa, Ontario, Canada
- Ottawa Hospital Research Institute, Division of Regenerative Medicine, Department of Medicine, University of Ottawa, Ottawa, Ontario, Canada
| | - Duncan J. Stewart
- University of Ottawa Heart Institute, Division of Cardiology, Department of Medicine, and
- Department of Cellular and Molecular Medicine, Faculty of Medicine, University of Ottawa, Ottawa, Ontario, Canada
- Ottawa Hospital Research Institute, Division of Regenerative Medicine, Department of Medicine, University of Ottawa, Ottawa, Ontario, Canada
| | - Darryl R. Davis
- University of Ottawa Heart Institute, Division of Cardiology, Department of Medicine, and
- Department of Cellular and Molecular Medicine, Faculty of Medicine, University of Ottawa, Ottawa, Ontario, Canada
| |
Collapse
|
14
|
Becker G, Debatisse J, Rivière M, Crola Da Silva C, Beaudoin-Gobert M, Eker O, Wateau O, Cho TH, Wiart M, Tremblay L, Costes N, Mérida I, Redouté J, Léon C, Langlois JB, Le Bars D, Lancelot S, Nighoghossian N, Mechtouff L, Canet-Soulas E. Spatio-Temporal Characterization of Brain Inflammation in a Non-human Primate Stroke Model Mimicking Endovascular Thrombectomy. Neurotherapeutics 2023; 20:789-802. [PMID: 36976495 PMCID: PMC10275847 DOI: 10.1007/s13311-023-01368-2] [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] [Accepted: 03/13/2023] [Indexed: 03/29/2023] Open
Abstract
Reperfusion therapies in acute ischemic stroke have demonstrated their efficacy in promoting clinical recovery. However, ischemia/reperfusion injury and related inflammation remain a major challenge in patient clinical management. We evaluated the spatio-temporal evolution of inflammation using sequential clinical [11C]PK11195 PET-MRI in a non-human primate (NHP) stroke model mimicking endovascular thrombectomy (EVT) with a neuroprotective cyclosporine A (CsA) treatment. The NHP underwent a 110-min transient endovascular middle cerebral artery occlusion. We acquired [11C]PK11195 dynamic PET-MR imaging at baseline, 7 and 30 days after intervention. Individual voxel-wise analysis was performed thanks to a baseline scan database. We quantified [11C]PK11195 in anatomical regions and in lesioned areas defined on per-occlusion MR diffusion-weighted imaging and perfusion [15O2]H2OPET imaging. [11C]PK11195 parametric maps showed a clear uptake overlapping the lesion core at D7, which further increased at D30. Voxel-wise analysis identified individuals with significant inflammation at D30, with voxels located within the most severe diffusion reduction area during occlusion, mainly in the putamen. The quantitative analysis revealed that thalamic inflammation lasted until D30 and was significantly reduced in the CsA-treated group compared to the placebo. In conclusion, we showed that chronic inflammation matched ADC decrease at occlusion time, a region exposed to an initial burst of damage-associated molecular patterns, in an NHP stroke model mimicking EVT. We described secondary thalamic inflammation and the protective effect of CsA in this region. We propose that major ADC drop in the putamen during occlusion may identify individuals who could benefit from early personalized treatment targeting inflammation.
Collapse
Affiliation(s)
- Guillaume Becker
- CarMeN Laboratory, INRAE U1397, INSERM U1060, Groupement Hospitalier Est, University Claude Bernard Lyon 1, 59 Boulevard Pinel, 69500, Lyon, Bron, France.
| | - Justine Debatisse
- CarMeN Laboratory, INRAE U1397, INSERM U1060, Groupement Hospitalier Est, University Claude Bernard Lyon 1, 59 Boulevard Pinel, 69500, Lyon, Bron, France
| | - Margaux Rivière
- CarMeN Laboratory, INRAE U1397, INSERM U1060, Groupement Hospitalier Est, University Claude Bernard Lyon 1, 59 Boulevard Pinel, 69500, Lyon, Bron, France
| | - Claire Crola Da Silva
- CarMeN Laboratory, INRAE U1397, INSERM U1060, Groupement Hospitalier Est, University Claude Bernard Lyon 1, 59 Boulevard Pinel, 69500, Lyon, Bron, France
| | - Maude Beaudoin-Gobert
- Lyon Neuroscience Research Center, UMR5295, INSERM U1028, CNRS, Université Claude Bernard Lyon 1, Lyon, France
| | - Omer Eker
- UMR-5220, CREATIS, CNRS, INSERM U1206, Université Lyon 1, INSA Lyon, Villeurbanne, France
- , Hospices Civils de Lyon, Lyon, France
| | | | - Tae Hee Cho
- CarMeN Laboratory, INRAE U1397, INSERM U1060, Groupement Hospitalier Est, University Claude Bernard Lyon 1, 59 Boulevard Pinel, 69500, Lyon, Bron, France
- , Hospices Civils de Lyon, Lyon, France
| | - Marlène Wiart
- CarMeN Laboratory, INRAE U1397, INSERM U1060, Groupement Hospitalier Est, University Claude Bernard Lyon 1, 59 Boulevard Pinel, 69500, Lyon, Bron, France
| | - Léon Tremblay
- Cognitive Neuroscience Center, CNRS UMR5229, Université Claude Bernard Lyon 1, Lyon, France
| | | | | | | | - Christelle Léon
- CarMeN Laboratory, INRAE U1397, INSERM U1060, Groupement Hospitalier Est, University Claude Bernard Lyon 1, 59 Boulevard Pinel, 69500, Lyon, Bron, France
| | | | - Didier Le Bars
- , Hospices Civils de Lyon, Lyon, France
- CERMEP, Lyon, France
| | | | - Norbert Nighoghossian
- CarMeN Laboratory, INRAE U1397, INSERM U1060, Groupement Hospitalier Est, University Claude Bernard Lyon 1, 59 Boulevard Pinel, 69500, Lyon, Bron, France
- , Hospices Civils de Lyon, Lyon, France
| | - Laura Mechtouff
- CarMeN Laboratory, INRAE U1397, INSERM U1060, Groupement Hospitalier Est, University Claude Bernard Lyon 1, 59 Boulevard Pinel, 69500, Lyon, Bron, France
- , Hospices Civils de Lyon, Lyon, France
| | - Emmanuelle Canet-Soulas
- CarMeN Laboratory, INRAE U1397, INSERM U1060, Groupement Hospitalier Est, University Claude Bernard Lyon 1, 59 Boulevard Pinel, 69500, Lyon, Bron, France
| |
Collapse
|
15
|
Zharkova O, Salamah MF, Babak MV, Rajan E, Lim LHK, Andrade F, Gil CD, Oliani SM, Moraes LA, Vaiyapuri S. Deletion of Annexin A1 in Mice Upregulates the Expression of Its Receptor, Fpr2/3, and Reactivity to the AnxA1 Mimetic Peptide in Platelets. Int J Mol Sci 2023; 24:ijms24043424. [PMID: 36834844 PMCID: PMC9962723 DOI: 10.3390/ijms24043424] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2023] [Revised: 02/03/2023] [Accepted: 02/06/2023] [Indexed: 02/11/2023] Open
Abstract
Annexin A1 (ANXA1) is an endogenous protein, which plays a central function in the modulation of inflammation. While the functions of ANXA1 and its exogenous peptidomimetics, N-Acetyl 2-26 ANXA1-derived peptide (ANXA1Ac2-26), in the modulation of immunological responses of neutrophils and monocytes have been investigated in detail, their effects on the modulation of platelet reactivity, haemostasis, thrombosis, and platelet-mediated inflammation remain largely unknown. Here, we demonstrate that the deletion of Anxa1 in mice upregulates the expression of its receptor, formyl peptide receptor 2/3 (Fpr2/3, orthologue of human FPR2/ALX). As a result, the addition of ANXA1Ac2-26 to platelets exerts an activatory role in platelets, as characterised by its ability to increase the levels of fibrinogen binding and the exposure of P-selectin on the surface. Moreover, ANXA1Ac2-26 increased the development of platelet-leukocyte aggregates in whole blood. The experiments carried out using a pharmacological inhibitor (WRW4) for FPR2/ALX, and platelets isolated from Fpr2/3-deficient mice ascertained that the actions of ANXA1Ac2-26 are largely mediated through Fpr2/3 in platelets. Together, this study demonstrates that in addition to its ability to modulate inflammatory responses via leukocytes, ANXA1 modulates platelet function, which may influence thrombosis, haemostasis, and platelet-mediated inflammation under various pathophysiological settings.
Collapse
Affiliation(s)
- Olga Zharkova
- Immunology Program, Department of Physiology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117593, Singapore
| | | | - Maria V. Babak
- Department of Chemistry, City University of Hong Kong, Hong Kong, China
| | | | - Lina H. K. Lim
- Immunology Program, Department of Physiology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117593, Singapore
| | - Frans Andrade
- Department of Morphology and Genetics, Federal University of São Paulo (UNIFESP), São Paulo 04023-900, Brazil
| | - Cristiane D. Gil
- Department of Morphology and Genetics, Federal University of São Paulo (UNIFESP), São Paulo 04023-900, Brazil
| | - Sonia M. Oliani
- Department of Morphology and Genetics, Federal University of São Paulo (UNIFESP), São Paulo 04023-900, Brazil
- Department of Biology, Instituto de Biociências, Letras e Ciências Exatas (IBILCE), São Paulo State University (UNESP), São José do Rio Preto, São Paulo 15054-000, Brazil
| | - Leonardo A. Moraes
- Immunology Program, Department of Physiology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117593, Singapore
- School of Pharmacy, University of Reading, Reading RG6 6UB, UK
| | - Sakthivel Vaiyapuri
- School of Pharmacy, University of Reading, Reading RG6 6UB, UK
- Correspondence:
| |
Collapse
|
16
|
Screening of Biomarkers in Liver Tissue after Bariatric Surgery Based on WGCNA and SVM-RFE Algorithms. DISEASE MARKERS 2023; 2023:2970429. [PMID: 36755803 PMCID: PMC9902125 DOI: 10.1155/2023/2970429] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 11/14/2022] [Revised: 01/09/2023] [Accepted: 01/16/2023] [Indexed: 02/03/2023]
Abstract
As the most common chronic liver disease around the world, nonalcoholic fatty liver disease (NAFLD) has a close connection with obesity, diabetes, and metabolic syndrome. Bariatric surgery (BS) is considered to be the most effective treatment for NAFLD. However, the regulatory mechanism of hepatic lipid metabolism after BS remains poorly elucidated. By analyzing two transcriptome datasets regarding liver tissues after BS, namely, GSE83452 and GSE106737, we acquired 110 differentially expressed genes (DEGs). By further analysis of DEGs in terms of the weighted gene coexpression network analysis (WGCNA) and support vector machine-recursive feature elimination (SVM-RFE) algorithms, we identified four crucial genes participating in the regulation of hepatic lipid metabolism: SRGN, THEMIS2, SGK1, and FPR3. In addition, the results of gene set enrichment analysis (GSEA) showed that BS can activate immune-related regulatory pathways and change immune cell infiltration levels. Finally, through cellular level studies, we found that the silencing of SRGN affects the expression of SREBP-1, SIRT1, and FAS during adipogenesis in the liver and the formation of lipid droplets in the liver. In summary, the immune system in the liver is activated after BS, and SRGN participates in the regulation of hepatic lipid metabolism.
Collapse
|
17
|
Classes of Lipid Mediators and Their Effects on Vascular Inflammation in Atherosclerosis. Int J Mol Sci 2023; 24:ijms24021637. [PMID: 36675152 PMCID: PMC9863938 DOI: 10.3390/ijms24021637] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2022] [Revised: 01/06/2023] [Accepted: 01/08/2023] [Indexed: 01/18/2023] Open
Abstract
It is commonly believed that the inactivation of inflammation is mainly due to the decay or cessation of inducers. In reality, in connection with the development of atherosclerosis, spontaneous decay of inducers is not observed. It is now known that lipid mediators originating from polyunsaturated fatty acids (PUFAs), which are important constituents of all cell membranes, can act in the inflamed tissue and bring it to resolution. In fact, PUFAs, such as arachidonic acid (AA), eicosapentaenoic acid (EPA), and docosahexaenoic acid (DHA), are precursors to both pro-inflammatory and anti-inflammatory compounds. In this review, we describe the lipid mediators of vascular inflammation and resolution, and their biochemical activity. In addition, we highlight data from the literature that often show a worsening of atherosclerotic disease in subjects deficient in lipid mediators of inflammation resolution, and we also report on the anti-proteasic and anti-thrombotic properties of these same lipid mediators. It should be noted that despite promising data observed in both animal and in vitro studies, contradictory clinical results have been observed for omega-3 PUFAs. Many further studies will be required in order to clarify the observed conflicts, although lifestyle habits such as smoking or other biochemical factors may often influence the normal synthesis of lipid mediators of inflammation resolution.
Collapse
|
18
|
Li T, Yan Z, Fan Y, Fan X, Li A, Qi Z, Zhang J. Cardiac repair after myocardial infarction: A two-sided role of inflammation-mediated. Front Cardiovasc Med 2023; 9:1077290. [PMID: 36698953 PMCID: PMC9868426 DOI: 10.3389/fcvm.2022.1077290] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2022] [Accepted: 12/19/2022] [Indexed: 01/11/2023] Open
Abstract
Myocardial infarction is the leading cause of death and disability worldwide, and the development of new treatments can help reduce the size of myocardial infarction and prevent adverse cardiovascular events. Cardiac repair after myocardial infarction can effectively remove necrotic tissue, induce neovascularization, and ultimately replace granulation tissue. Cardiac inflammation is the primary determinant of whether beneficial cardiac repair occurs after myocardial infarction. Immune cells mediate inflammatory responses and play a dual role in injury and protection during cardiac repair. After myocardial infarction, genetic ablation or blocking of anti-inflammatory pathways is often harmful. However, enhancing endogenous anti-inflammatory pathways or blocking endogenous pro-inflammatory pathways may improve cardiac repair after myocardial infarction. A deficiency of neutrophils or monocytes does not improve overall cardiac function after myocardial infarction but worsens it and aggravates cardiac fibrosis. Several factors are critical in regulating inflammatory genes and immune cells' phenotypes, including DNA methylation, histone modifications, and non-coding RNAs. Therefore, strict control and timely suppression of the inflammatory response, finding a balance between inflammatory cells, preventing excessive tissue degradation, and avoiding infarct expansion can effectively reduce the occurrence of adverse cardiovascular events after myocardial infarction. This article reviews the involvement of neutrophils, monocytes, macrophages, and regulatory T cells in cardiac repair after myocardial infarction. After myocardial infarction, neutrophils are the first to be recruited to the damaged site to engulf necrotic cell debris and secrete chemokines that enhance monocyte recruitment. Monocytes then infiltrate the infarct site and differentiate into macrophages and they release proteases and cytokines that are harmful to surviving myocardial cells in the pre-infarct period. As time progresses, apoptotic neutrophils are cleared, the recruitment of anti-inflammatory monocyte subsets, the polarization of macrophages toward the repair phenotype, and infiltration of regulatory T cells, which secrete anti-inflammatory factors that stimulate angiogenesis and granulation tissue formation for cardiac repair. We also explored how epigenetic modifications regulate the phenotype of inflammatory genes and immune cells to promote cardiac repair after myocardial infarction. This paper also elucidates the roles of alarmin S100A8/A9, secreted frizzled-related protein 1, and podoplanin in the inflammatory response and cardiac repair after myocardial infarction.
Collapse
Affiliation(s)
- Tingting Li
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin, China
| | - Zhipeng Yan
- Graduate School, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Yajie Fan
- Graduate School, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Xinbiao Fan
- Graduate School, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Aolin Li
- Graduate School, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Zhongwen Qi
- Xiyuan Hospital of China Academy of Chinese Medical Sciences, Beijing, China,*Correspondence: Zhongwen Qi,
| | - Junping Zhang
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin, China,Junping Zhang,
| |
Collapse
|
19
|
Ansari J, Vital SA, Yadav S, Gavins FNE. Regulating Neutrophil PAD4/NOX-Dependent Cerebrovasular Thromboinflammation. Int J Biol Sci 2023; 19:852-864. [PMID: 36778112 PMCID: PMC9910005 DOI: 10.7150/ijbs.77434] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2022] [Accepted: 10/21/2022] [Indexed: 01/11/2023] Open
Abstract
Background: Neutrophil extracellular trap (NET) production has been implicated in the pathogenesis of thromboinflammatory conditions such as Sickle Cell Disease (SCD), contributing to heightened risk for ischemic stroke. NETs are catalyzed by the enzyme Peptidyl Arginine Deiminase 4 (PAD4) and neutrophil derived reactive oxygen species (ROS), especially NADPH oxidase (NOX) which interacts with PAD4 and is therefore critical for neutrophil function. However, the role that NOX-dependent ROS and NETs play in the accelerated cerebral microvascular thrombosis associated with thromboinflammatory conditions, such as SCD, has not been fully elucidated and is the aim of this study. Methods: The in-vitro effects of targeting PAD4 and NOX were examined using physiologically relevant NET assays with neutrophils isolated from healthy volunteers (control) and SCD patients. In addition, in-vivo intravascular effects of targeting PAD4 and NOX in the cerebral microcirculation of C57BL/6 and sickle transgenic mice (STM) were assessed using a photoactivation thrombosis model (light/dye) coupled with real-time fluorescence intravital microscopy. Results: We found that targeting PAD4 and NOX in human neutrophils significantly inhibited ionomycin dependent H3cit+ neutrophils. Targeting PAD4 and NOX in-vivo resulted in prolonged blood flow cessation in cerebrovascular arterioles as well as venules. Moreover, we were able to replicate the effects of PAD4 and NOX targeting in a clinical model of accelerated thromboinflammation by increasing blood flow cessation times in cerebral microvessels in STM. These findings concurred with the clinical setting i.e. neutrophils isolated from SCD patients, which possessed an attenuation of H3cit+ neutrophil production on targeting PAD4 and NOX. Conclusions: Taken together, our compelling data suggests that PAD4 and NOX play a significant role in neutrophil driven thromboinflammation. Targeting PAD4 and NOX limits pathological H3cit+ neutrophils, which may further explain attenuation of cerebral thrombosis. Overall, this study presents a viable pre-clinical model of prevention and management of thromboinflammatory complications such as ischemic stroke.
Collapse
Affiliation(s)
- Junaid Ansari
- Department of Neurology, Louisiana State University Health Sciences Center-Shreveport, Shreveport, LA, 71130, USA
| | - Shantel A Vital
- Department of Molecular & Cellular Physiology, Louisiana State University Health Sciences Center-Shreveport, Shreveport, LA, 71130, USA
| | - Shreya Yadav
- Department of Life Sciences, Centre of Inflammation Research and Translational Medicine (CIRTM), Brunel University London, London. UB8 3PH, UK
| | - Felicity N E Gavins
- Department of Life Sciences, Centre of Inflammation Research and Translational Medicine (CIRTM), Brunel University London, London. UB8 3PH, UK
| |
Collapse
|
20
|
Zheng Y, Li Y, Li S, Hu R, Zhang L. Annexin A1 (Ac2-26)-dependent Fpr2 receptor alleviates sepsis-induced acute kidney injury by inhibiting inflammation and apoptosis in vivo and in vitro. Inflamm Res 2023; 72:347-362. [PMID: 36544058 PMCID: PMC9925514 DOI: 10.1007/s00011-022-01640-9] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2022] [Revised: 09/02/2022] [Accepted: 09/07/2022] [Indexed: 12/24/2022] Open
Abstract
OBJECTIVES Excessive inflammatory responses and apoptosis are critical pathologies that contribute to sepsis-induced acute kidney injury (SI-AKI). Annexin A1 (ANXA1), a member of the calcium-dependent phospholipid-binding protein family, protects against SI-AKI through its anti-inflammatory and antiapoptotic effects, but the underlying mechanisms are still largely unknown. METHODS In vivo, SI-AKI mouse models were established via caecal ligation and puncture (CLP) and were then treated with the Ac2-26 peptide of ANXA1 (ANXA1 (Ac2-26)), WRW4 (Fpr2 antagonist) or both. In vitro, HK-2 cells were induced by lipopolysaccharide (LPS) and then treated with ANXA1 (Ac2-26), Fpr2-siRNA or both. RESULTS In the present study, we found that the expression levels of ANXA1 were decreased, and the expression levels of TNF-α, IL-1β, IL-6, cleaved caspase-3, cleaved caspase-8 and Bax were significantly increased, accompanied by marked kidney tissue apoptosis in vivo. Moreover, we observed that ANXA1 (Ac2-26) significantly reduced the levels of TNF-α, IL-1β and IL-6 and cleaved caspase-3, cleaved caspase-8, FADD and Bax and inhibited apoptosis in kidney tissue and HK-2 cells, accompanied by pathological damage to kidney tissue. Seven-day survival, kidney function and cell viability were significantly improved in vivo and in vitro, respectively. Furthermore, the administration of ANXA1 (Ac2-26) inhibited the CLP- or LPS-induced phosphorylation of PI3K and AKT and downregulated the level of NF-κB in vivo and in vitro. Moreover, our data demonstrate that blocking the Fpr2 receptor by the administration of WRW4 or Fpr2-siRNA reversed the abovementioned regulatory role of ANXA1, accompanied by enhanced phosphorylation of PI3K and AKT and upregulation of the level of NF-κB in vivo and in vitro. CONCLUSIONS Taken together, this study provides evidence that the protective effect of ANXA1 (Ac2-26) on SI-AKI largely depends on the negative regulation of inflammation and apoptosis via the Fpr2 receptor.
Collapse
Affiliation(s)
- Yanlei Zheng
- Department of Critical Care Medicine, Hubei Cancer Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430079 China
| | - Yan Li
- Department of Critical Care Medicine, Xiangyang Central Hospital, Xiangyang, 440121 China
| | - Shi Li
- Department of Critical Care Medicine, Hubei Cancer Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430079 China
| | - Ronghua Hu
- Department of Critical Care Medicine, Hubei Cancer Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430079 China
| | - Li Zhang
- Department of Critical Care Medicine, Hubei Cancer Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430079, China.
| |
Collapse
|
21
|
Jing Y, Hu S, Song J, Dong X, Zhang Y, Sun X, Wang D. Association between polymorphisms in miRNAs and ischemic stroke: A meta-analysis. Medicine (Baltimore) 2022; 101:e32078. [PMID: 36596006 PMCID: PMC9803434 DOI: 10.1097/md.0000000000032078] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
BACKGROUND Atherosclerosis remains a predominant cause of ischemic stroke (IS). Four miRNA polymorphisms associated with arteriosclerosis mechanism were meta-analyzed to explore whether they had predictive significance for IS. METHODS PubMed, Excerpta Medica database, Web of Science, Cochrane Library, Scopus, China National Knowledge Infrastructure, and China Wanfang Database were searched for relevant case-control studies published before September 2022. Two researchers independently reviewed the studies and extracted the data. Data synthesis was carried out on eligible studies. Meta-analysis, subgroup analysis, sensitivity analysis, and publication bias analysis were performed using Stata software 16.0. RESULTS Twenty-two studies were included, comprising 8879 cases and 12,091 controls. The results indicated that there were no significant associations between miR-146a C>G (rs2910164), miR-196a2 T>C (rs11614913) and IS risk in the overall analyses, but miR-149 T>C (rs2292832) and miR-499 A>G (rs3746444) increased IS risk under the allelic model, homozygote model and recessive model. The subgroup analyses based on Trial of Org 101072 in Acute Stroke Treatment classification indicated that rs2910164 increased small artery occlusion (SAO) risk under the allelic model, heterozygote model and dominant model; rs11614913 decreased the risk of SAO under the allelic model, homozygote model, heterozygote model and dominant model. CONCLUSION This Meta-analysis showed that all 4 single nucleotide polymorphisms were associated with the risk of IS or SAO, even though the overall and subgroup analyses were not entirely consistent.
Collapse
Affiliation(s)
- Yunnan Jing
- Heilongjiang University of Chinese Medicine, Harbin, China
- Department of Acupuncture and Moxibustion, The Second Affiliated Hospital of Heilongjiang University of Chinese Medicine, Harbin, China
| | - Siya Hu
- Heilongjiang University of Chinese Medicine, Harbin, China
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Heilongjiang University of Chinese Medicine, Harbin, China
| | - Jing Song
- Department of Acupuncture and Moxibustion, The Second Affiliated Hospital of Heilongjiang University of Chinese Medicine, Harbin, China
| | - Xu Dong
- Department of Acupuncture and Moxibustion, The Second Affiliated Hospital of Heilongjiang University of Chinese Medicine, Harbin, China
| | - Ying Zhang
- Heilongjiang University of Chinese Medicine, Harbin, China
- Department of Acupuncture and Moxibustion, The Second Affiliated Hospital of Heilongjiang University of Chinese Medicine, Harbin, China
| | - Xiaowei Sun
- Heilongjiang University of Chinese Medicine, Harbin, China
- Department of Acupuncture and Moxibustion, The First Affiliated Hospital of Heilongjiang University of Chinese Medicine, Harbin, China
| | - Dongyan Wang
- Department of Acupuncture and Moxibustion, The Second Affiliated Hospital of Heilongjiang University of Chinese Medicine, Harbin, China
- * Correspondence: Dongyan Wang, Department of Acupuncture and Moxibustion, The Second Affiliated Hospital of Heilongjiang University of Chinese Medicine, 411 Gogoli Dajie, Nangang District, Harbin City, Heilongjiang Province 150000, China (e-mail: )
| |
Collapse
|
22
|
Huang K, Wang Z, He Z, Li Y, Li S, Shen K, Zhu G, Liu Z, Lv S, Zhang C, Yang H, Yang X, Liu S. Downregulated formyl peptide receptor 2 expression in the epileptogenic foci of patients with focal cortical dysplasia type IIb and tuberous sclerosis complex. Immun Inflamm Dis 2022; 10:e706. [PMID: 36301030 PMCID: PMC9597500 DOI: 10.1002/iid3.706] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2022] [Revised: 08/30/2022] [Accepted: 09/01/2022] [Indexed: 11/07/2022] Open
Abstract
BACKGROUND Focal cortical dysplasia type IIb (FCDIIb) and tuberous sclerosis complex (TSC) show persistent neuroinflammation, which promotes epileptogenesis and epilepsy progression, suggesting that endogenous resolution of inflammation is inadequate to relieve neuronal network hyperexcitability. To explore the potential roles of formyl peptide receptor 2 (FPR2), which is a key regulator of inflammation resolution, in epilepsy caused by FCDIIb and TSC, we examined the expression and cellular distribution of FPR2. METHOD The expression of FPR2 and nuclear factor-κB (NF-κB) signaling pathway was examined by real-time PCR, western blots, and analyzed via one-way analysis of variance. The distribution of FPR2 was detected using immunostaining. The expression of resolvin D1 (RvD1, the endogenous ligand of FPR2) was observed via enzyme-linked immunosorbent assay. Pearson's correlation test was used to evaluate the correlation between the expression levels of FPR2 and RvD1 and the clinical variants. RESULTS The expression of FPR2 was significantly lower in FCDIIb (p = .0146) and TSC (p = .0006) cortical lesions than in controls, as was the expression of RvD1 (FCDIIb: p = .00431; TSC: p = .0439). Weak FPR2 immunoreactivity was observed in dysmorphic neurons (DNs), balloon cells (BCs), and giant cells (GCs) in FCDIIb and TSC tissues. Moreover, FPR2 was mainly distributed in dysplastic neurons; it was sparse in microglia and nearly absent in astrocytes. The NF-κB pathway was significantly activated in patients with FCDIIb and TSC, and the protein level of NF-κB was negatively correlated with the protein level of FPR2 (FCDIIb: p = .00395; TSC: p = .0399). In addition, the protein level of FPR2 was negatively correlated with seizure frequency in FCDIIb (p = .0434) and TSC (p = .0351) patients. CONCLUSION In summary, these results showed that the expression and specific distribution of FPR2 may be involved in epilepsy caused by FCDIIb and TSC, indicating that downregulation of FPR2 mediated the dysfunction of neuroinflammation resolution in FCDIIb and TSC.
Collapse
Affiliation(s)
- Kaixuan Huang
- Department of Neurosurgery, Epilepsy Research Center of PLA, Xinqiao HospitalArmy Medical University (Third Military Medical University)ChongqingChina
| | - Zhongke Wang
- Department of NeurosurgeryArmed Police Hospital of ChongqingChongqingChina
| | - Zeng He
- Department of Neurosurgery, Epilepsy Research Center of PLA, Xinqiao HospitalArmy Medical University (Third Military Medical University)ChongqingChina
| | - Yang Li
- Department of Neurosurgery, Epilepsy Research Center of PLA, Xinqiao HospitalArmy Medical University (Third Military Medical University)ChongqingChina
| | - Shujing Li
- Department of Neurosurgery, Epilepsy Research Center of PLA, Xinqiao HospitalArmy Medical University (Third Military Medical University)ChongqingChina
| | - Kaifeng Shen
- Department of Neurosurgery, Epilepsy Research Center of PLA, Xinqiao HospitalArmy Medical University (Third Military Medical University)ChongqingChina
| | - Gang Zhu
- Department of Neurosurgery, Epilepsy Research Center of PLA, Xinqiao HospitalArmy Medical University (Third Military Medical University)ChongqingChina
| | - Zhonghong Liu
- Department of NeurosurgeryArmed Police Hospital of ChongqingChongqingChina
| | - Shengqing Lv
- Department of Neurosurgery, Epilepsy Research Center of PLA, Xinqiao HospitalArmy Medical University (Third Military Medical University)ChongqingChina
| | - Chunqing Zhang
- Department of Neurosurgery, Epilepsy Research Center of PLA, Xinqiao HospitalArmy Medical University (Third Military Medical University)ChongqingChina
| | - Hui Yang
- Department of Neurosurgery, Epilepsy Research Center of PLA, Xinqiao HospitalArmy Medical University (Third Military Medical University)ChongqingChina
| | - Xiaolin Yang
- Department of Neurosurgery, Epilepsy Research Center of PLA, Xinqiao HospitalArmy Medical University (Third Military Medical University)ChongqingChina
| | - Shiyong Liu
- Department of Neurosurgery, Epilepsy Research Center of PLA, Xinqiao HospitalArmy Medical University (Third Military Medical University)ChongqingChina
| |
Collapse
|
23
|
WKYMVm/FPR2 Alleviates Spinal Cord Injury by Attenuating the Inflammatory Response of Microglia. Mediators Inflamm 2022; 2022:4408099. [PMID: 35935810 PMCID: PMC9348919 DOI: 10.1155/2022/4408099] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2022] [Accepted: 07/11/2022] [Indexed: 11/17/2022] Open
Abstract
Spinal cord injury (SCI) is a common traumatic disease of the nervous system. The pathophysiological process of SCI includes primary injury and secondary injuries. An excessive inflammatory response leads to secondary tissue damage, which in turn exacerbates cellular and organ dysfunction. Due to the irreversibility of primary injury, current research on SCI mainly focuses on secondary injury, and the inflammatory response is considered the primary target. Thus, modulating the inflammatory response has been suggested as a new strategy for the treatment of SCI. In this study, microglial cell lines, primary microglia, and a rat SCI model were used, and we found that WKYMVm/FPR2 plays an anti-inflammatory role and reduces tissue damage after SCI by suppressing the extracellular signal-regulated kinases 1 and 2 (ERK1/2) and nuclear factor-κB (NF-κB) signaling pathways. FPR2 was activated by WKYMVm, suppressing the secretion of tumor necrosis factor-α (TNF-α), interleukin-6 (IL-6), and interleukin-1β (IL-1β) by inhibiting M1 microglial polarization. Moreover, FPR2 activation by WKYMVm could reduce structural disorders and neuronal loss in SCI rats. Overall, this study illustrated that the activation of FPR2 by WKYMVm repressed M1 microglial polarization by suppressing the ERK1/2 and NF-κB signaling pathways to alleviate tissue damage and locomotor decline after SCI. These findings provide further insight into SCI and help identify novel treatment strategies.
Collapse
|
24
|
Hao J, Feng Y, Xu X, Li L, Yang K, Dai G, Gao W, Zhang M, Fan Y, Yin T, Wang J, Yang B, Jiao L, Zhang L. Plasma Lipid Mediators Associate With Clinical Outcome After Successful Endovascular Thrombectomy in Patients With Acute Ischemic Stroke. Front Immunol 2022; 13:917974. [PMID: 35865524 PMCID: PMC9295711 DOI: 10.3389/fimmu.2022.917974] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2022] [Accepted: 06/14/2022] [Indexed: 11/13/2022] Open
Abstract
BackgroundNeuroinflammatory response contributes to early neurological deterioration (END) and unfavorable long-term functional outcome in patients with acute ischemic stroke (AIS) who recanalized successfully by endovascular thrombectomy (EVT), but there are no reliable biomarkers for their accurate prediction. Here, we sought to determine the temporal plasma profiles of the bioactive lipid mediators lipoxin A4 (LXA4), resolvin D1 (RvD1), and leukotriene B4 (LTB4) for their associations with clinical outcome.MethodsWe quantified levels of LXA4, RvD1, and LTB4 in blood samples retrospectively and longitudinally collected from consecutive AIS patients who underwent complete angiographic recanalization by EVT at admission (pre-EVT) and 24 hrs post-EVT. The primary outcome was unfavorable long-term functional outcome, defined as a 90-day modified Rankin Scale score of 3-6. Secondary outcome was END, defined as an increase in National Institutes of Health Stroke Scale (NIHSS) score ≥4 points at 24 hrs post-EVT.ResultsEighty-one consecutive AIS patients and 20 healthy subjects were recruited for this study. Plasma levels of LXA4, RvD1, and LTB4 were significantly increased in post-EVT samples from AIS patients, as compared to those of healthy controls. END occurred in 17 (20.99%) patients, and 38 (46.91%) had unfavorable 90-day functional outcome. Multiple logistic regression analyses demonstrated that post-EVT levels of LXA4 (adjusted odd ratio [OR] 0.992, 95% confidence interval [CI] 0.987-0.998), ΔLXA4 (adjusted OR 0.995, 95% CI 0.991-0.999), LTB4 (adjusted OR 1.003, 95% CI 1.001-1.005), ΔLTB4 (adjusted OR 1.004, 95% CI 1.002-1.006), and post-EVT LXA4/LTB4 (adjusted OR 0.023, 95% CI 0.001-0.433) and RvD1/LTB4 (adjusted OR 0.196, 95% CI 0.057-0.682) ratios independently predicted END, and post-EVT LXA4 levels (adjusted OR 0.995, 95% CI 0.992-0.999), ΔLXA4 levels (adjusted OR 0.996, 95% CI 0.993-0.999), and post-EVT LXA4/LTB4 ratio (adjusted OR 0.285, 95% CI 0.096-0.845) independently predicted unfavorable 90-day functional outcome. These were validated using receiver operating characteristic curve analyses.ConclusionsPlasma lipid mediators measured 24 hrs post-EVT were independent predictors for early and long-term outcomes. Further studies are needed to determine their causal-effect relationship, and whether the imbalance between anti-inflammatory/pro-resolving and pro-inflammatory lipid mediators could be a potential adjunct therapeutic target.
Collapse
Affiliation(s)
- Jiheng Hao
- Department of Neurosurgery, Liaocheng People’s hospital, Liaocheng, China
- Department of Neurosurgery, Xuanwu Hospital, Capital Medical University, Beijing, China
| | - Yao Feng
- Department of Neurosurgery, Xuanwu Hospital, Capital Medical University, Beijing, China
- China International Neuroscience Institute (China-INI), Beijing, China
| | - Xin Xu
- Department of Neurosurgery, Xuanwu Hospital, Capital Medical University, Beijing, China
- China International Neuroscience Institute (China-INI), Beijing, China
- *Correspondence: Xin Xu, ; Liqun Jiao, ; Liyong Zhang,
| | - Long Li
- Department of Neurosurgery, Xuanwu Hospital, Capital Medical University, Beijing, China
- China International Neuroscience Institute (China-INI), Beijing, China
| | - Kun Yang
- Department of Evidence-based Medicine, Xuanwu Hospital, Capital Medical University, Beijing, China
| | - Gaolei Dai
- Department of Intervention, Liaocheng People’s hospital, Liaocheng, China
| | - Weiwei Gao
- Department of Neurology, Tianjin Huanhu Hospital, Tianjin, China
| | - Meng Zhang
- Department of Neurosurgery, Liaocheng People’s hospital, Liaocheng, China
| | - Yaming Fan
- Department of Neurosurgery, Liaocheng People’s hospital, Liaocheng, China
| | - Tengkun Yin
- Department of Neurosurgery, Liaocheng People’s hospital, Liaocheng, China
| | - Jiyue Wang
- Department of Neurosurgery, Liaocheng People’s hospital, Liaocheng, China
| | - Bin Yang
- Department of Neurosurgery, Xuanwu Hospital, Capital Medical University, Beijing, China
- China International Neuroscience Institute (China-INI), Beijing, China
| | - Liqun Jiao
- Department of Neurosurgery, Xuanwu Hospital, Capital Medical University, Beijing, China
- China International Neuroscience Institute (China-INI), Beijing, China
- Department of Interventional Neuroradiology, Xuanwu Hospital, Capital Medical Universit, Beijing, China
- *Correspondence: Xin Xu, ; Liqun Jiao, ; Liyong Zhang,
| | - Liyong Zhang
- Department of Neurosurgery, Liaocheng People’s hospital, Liaocheng, China
- *Correspondence: Xin Xu, ; Liqun Jiao, ; Liyong Zhang,
| |
Collapse
|
25
|
Oda H, Tanaka S, Shinohara M, Morimura Y, Yokoyama Y, Kayawake H, Yamada Y, Yutaka Y, Ohsumi A, Nakajima D, Hamaji M, Menju T, Date H. Specialized Proresolving Lipid Meditators Agonistic to Formyl Peptide Receptor Type 2 Attenuate Ischemia-reperfusion Injury in Rat Lung. Transplantation 2022; 106:1159-1169. [PMID: 34873128 DOI: 10.1097/tp.0000000000003987] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
BACKGROUND Lung ischemia-reperfusion injury (IRI) is a form of acute lung injury characterized by nonspecific alveolar damage and lung edema due to robust inflammation. Little is known about the roles of specialized proresolving lipid mediators (SPMs) in lung IRI. Therefore, we aimed to evaluate the dynamic changes in endogenous SPMs during the initiation and resolution of lung IRI and to determine the effects of SPM supplementation on lung IRI. METHODS We used a rat left hilar clamp model with 90 min of ischemia, followed by reperfusion. Dynamic changes in endogenous SPMs were evaluated using liquid chromatography-tandem mass spectrometry. RESULTS Endogenous SPMs in the left lung showed a decreasing trend after 1 h of reperfusion. Oxygenation improved between 3 and 7 d following reperfusion; however, the level of endogenous SPMs remained low compared with that in the naïve lung. Among SPM receptors, only formyl peptide receptor type 2 (ALX/FPR2) gene expression in the left lung was increased 3 h after reperfusion, and the inflammatory cells were immunohistochemically positive for ALX/FPR2. Administration of aspirin-triggered (AT) resolvin D1 (AT-RvD1) and AT lipoxin A4 (AT-LXA4), which are agonistic to ALX/FPR2, immediately after reperfusion improved lung function, reduced inflammatory cytokine levels, attenuated lung edema, and decreased neutrophil infiltration 3 h after reperfusion. The effects of AT-RvD1 and AT-LXA4 were not observed after pretreatment with the ALX/FPR2 antagonist. CONCLUSIONS The level of intrapulmonary endogenous SPMs decreased during lung IRI process and the administration of AT-RvD1 and AT-LXA4 prevented the exacerbation of lung injury via ALX/FPR2.
Collapse
Affiliation(s)
- Hiromi Oda
- Department of Thoracic Surgery, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Satona Tanaka
- Department of Thoracic Surgery, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Masakazu Shinohara
- The Integrated Center for Mass Spectrometry, Kobe University Graduate School of Medicine, Kobe, Japan
- Division of Epidemiology, Kobe University Graduate School of Medicine, Kobe, Japan
| | - Yuki Morimura
- Department of Thoracic Surgery, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Yuhei Yokoyama
- Department of Thoracic Surgery, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Hidenao Kayawake
- Department of Thoracic Surgery, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Yoshito Yamada
- Department of Thoracic Surgery, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Yojiro Yutaka
- Department of Thoracic Surgery, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Akihiro Ohsumi
- Department of Thoracic Surgery, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Daisuke Nakajima
- Department of Thoracic Surgery, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Masatsugu Hamaji
- Department of Thoracic Surgery, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Toshi Menju
- Department of Thoracic Surgery, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Hiroshi Date
- Department of Thoracic Surgery, Kyoto University Graduate School of Medicine, Kyoto, Japan
| |
Collapse
|
26
|
Li YZ, Wang YY, Huang L, Zhao YY, Chen LH, Zhang C. Annexin A Protein Family in Atherosclerosis. Clin Chim Acta 2022; 531:406-417. [PMID: 35562096 DOI: 10.1016/j.cca.2022.05.009] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2022] [Revised: 05/06/2022] [Accepted: 05/06/2022] [Indexed: 12/25/2022]
Abstract
Atherosclerosis, a silent chronic vascular pathology, is the cause of the majority of cardiovascular ischaemic events. Atherosclerosis is characterized by a series of deleterious changes in cellularity, including endothelial dysfunction, transmigration of circulating inflammatory cells into the arterial wall, pro-inflammatory cytokines production, lipid accumulation in the intima, vascular local inflammatory response, atherosclerosis-related cells apoptosis and autophagy. Proteins of Annexin A (AnxA) family, the well-known Ca2+ phospholipid-binding protein, have many functions in regulating inflammation-related enzymes and cell signaling transduction, thus influencing cell adhesion, migration, differentiation, proliferation and apoptosis. There is now accumulating evidence that some members of the AnxA family, such as AnxA1, AnxA2, AnxA5 and AnxA7, play major roles in the development of atherosclerosis. This article discusses the major roles of AnxA1, AnxA2, AnxA5 and AnxA7, and the multifaceted mechanisms of the main biological process in which they are involved in atherosclerosis. Considering these evidences, it has been proposed that AnxA are drivers- and not merely participator- on the road to atherosclerosis, thus the progression of atherosclerosis may be prevented by targeting the expression or function of the AnxA family proteins.
Collapse
Affiliation(s)
- Yong-Zhen Li
- Institute of Cardiovascular Disease, Key Lab for Arteriosclerology of Hunan province, Hengyang Medical College, University of South China, Hengyang, Hunan 421001, People's Republic of China
| | - Yan-Yue Wang
- Institute of Cardiovascular Disease, Key Lab for Arteriosclerology of Hunan province, Hengyang Medical College, University of South China, Hengyang, Hunan 421001, People's Republic of China
| | - Liang Huang
- Research Laboratory of Translational Medicine, Hengyang Medical College, University of South China, Hengyang, Hunan 421001, People's Republic of China
| | - Yu-Yan Zhao
- Institute of Cardiovascular Disease, Key Lab for Arteriosclerology of Hunan province, Hengyang Medical College, University of South China, Hengyang, Hunan 421001, People's Republic of China
| | - Lin-Hui Chen
- Institute of Cardiovascular Disease, Key Lab for Arteriosclerology of Hunan province, Hengyang Medical College, University of South China, Hengyang, Hunan 421001, People's Republic of China
| | - Chi Zhang
- Institute of Cardiovascular Disease, Key Lab for Arteriosclerology of Hunan province, Hengyang Medical College, University of South China, Hengyang, Hunan 421001, People's Republic of China.
| |
Collapse
|
27
|
Schebb NH, Kühn H, Kahnt AS, Rund KM, O’Donnell VB, Flamand N, Peters-Golden M, Jakobsson PJ, Weylandt KH, Rohwer N, Murphy RC, Geisslinger G, FitzGerald GA, Hanson J, Dahlgren C, Alnouri MW, Offermanns S, Steinhilber D. Formation, Signaling and Occurrence of Specialized Pro-Resolving Lipid Mediators-What is the Evidence so far? Front Pharmacol 2022; 13:838782. [PMID: 35308198 PMCID: PMC8924552 DOI: 10.3389/fphar.2022.838782] [Citation(s) in RCA: 96] [Impact Index Per Article: 32.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2021] [Accepted: 02/02/2022] [Indexed: 12/14/2022] Open
Abstract
Formation of specialized pro-resolving lipid mediators (SPMs) such as lipoxins or resolvins usually involves arachidonic acid 5-lipoxygenase (5-LO, ALOX5) and different types of arachidonic acid 12- and 15-lipoxygenating paralogues (15-LO1, ALOX15; 15-LO2, ALOX15B; 12-LO, ALOX12). Typically, SPMs are thought to be formed via consecutive steps of oxidation of polyenoic fatty acids such as arachidonic acid, eicosapentaenoic acid or docosahexaenoic acid. One hallmark of SPM formation is that reported levels of these lipid mediators are much lower than typical pro-inflammatory mediators including the monohydroxylated fatty acid derivatives (e.g., 5-HETE), leukotrienes or certain cyclooxygenase-derived prostaglandins. Thus, reliable detection and quantification of these metabolites is challenging. This paper is aimed at critically evaluating i) the proposed biosynthetic pathways of SPM formation, ii) the current knowledge on SPM receptors and their signaling cascades and iii) the analytical methods used to quantify these pro-resolving mediators in the context of their instability and their low concentrations. Based on current literature it can be concluded that i) there is at most, a low biosynthetic capacity for SPMs in human leukocytes. ii) The identity and the signaling of the proposed G-protein-coupled SPM receptors have not been supported by studies in knock-out mice and remain to be validated. iii) In humans, SPM levels were neither related to dietary supplementation with their ω-3 polyunsaturated fatty acid precursors nor were they formed during the resolution phase of an evoked inflammatory response. iv) The reported low SPM levels cannot be reliably quantified by means of the most commonly reported methodology. Overall, these questions regarding formation, signaling and occurrence of SPMs challenge their role as endogenous mediators of the resolution of inflammation.
Collapse
Affiliation(s)
- Nils Helge Schebb
- Chair of Food Chemistry, Faculty of Mathematics and Natural Sciences, University of Wuppertal, Wuppertal, Germany
| | - Hartmut Kühn
- Department of Biochemistry, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
| | - Astrid S. Kahnt
- Institute of Pharmaceutical Chemistry, Goethe University Frankfurt, Frankfurt, Germany
| | - Katharina M. Rund
- Chair of Food Chemistry, Faculty of Mathematics and Natural Sciences, University of Wuppertal, Wuppertal, Germany
| | - Valerie B. O’Donnell
- School of Medicine, Systems Immunity Research Institute, School of Medicine, Cardiff University, Cardiff, United Kingdom
| | - Nicolas Flamand
- Département de Médecine, Faculté de Médecine, Centre de Recherche de l’Institut Universitaire de Cardiologie et de Pneumologie de Québec, Canada Excellence Research Chair on the Microbiome-Endocannabinoidome Axis in Metabolic Health (CERC-MEND), Université Laval, Québec, QC, Canada
| | - Marc Peters-Golden
- Division of Pulmonary and Critical Care Medicine, Department of Internal Medicine, University of Michigan Medical School, Ann Arbor, MI, United States
| | - Per-Johan Jakobsson
- Rheumatology Unit, Department of Medicine, Karolinska Institutet, Karolinska University Hospital, Stockholm, Sweden
| | - Karsten H. Weylandt
- Division of Medicine, Department of Gastroenterology, Metabolism and Oncology, Ruppin General Hospital, Brandenburg Medical School, Neuruppin, Germany
| | - Nadine Rohwer
- Division of Medicine, Department of Gastroenterology, Metabolism and Oncology, Ruppin General Hospital, Brandenburg Medical School, Neuruppin, Germany
- Department of Molecular Toxicology, German Institute of Human Nutrition Potsdam-Rehbruecke, Nuthetal, Germany
| | - Robert C. Murphy
- Department of Pharmacology, University of Colorado-Denver, Aurora, CO, United States
| | - Gerd Geisslinger
- Institute of Clinical Pharmacology, Pharmazentrum Frankfurt, University Hospital of Goethe-University, Frankfurt, Germany
- Fraunhofer Institute for Translational Medicine and Pharmacology, ITMP and Fraunhofer Cluster of Excellence for Immune Mediated Diseases, CIMD, Frankfurt, Germany
| | - Garret A. FitzGerald
- Institute for Translational Medicine and Therapeutics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, United States
| | - Julien Hanson
- Laboratory of Molecular Pharmacology, GIGA-Molecular Biology of Diseases, University of Liège, Liège, Belgium
- Laboratory of Medicinal Chemistry, Centre for Interdisciplinary Research on Medicines (CIRM), University of Liège, Liège, Belgium
| | - Claes Dahlgren
- Department of Rheumatology and Inflammation Research, Institute of Medicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Mohamad Wessam Alnouri
- Department of Pharmacology, Max Planck Institute for Heart and Lung Research, Bad Nauheim, Germany
| | - Stefan Offermanns
- Department of Pharmacology, Max Planck Institute for Heart and Lung Research, Bad Nauheim, Germany
- Center for Molecular Medicine, Goethe University Frankfurt, Frankfurt, Germany
| | - Dieter Steinhilber
- Institute of Pharmaceutical Chemistry, Goethe University Frankfurt, Frankfurt, Germany
- Fraunhofer Institute for Translational Medicine and Pharmacology, ITMP and Fraunhofer Cluster of Excellence for Immune Mediated Diseases, CIMD, Frankfurt, Germany
| |
Collapse
|
28
|
Zhang J, Li Z, Fan M, Jin W. Lipoxins in the Nervous System: Brighter Prospects for Neuroprotection. Front Pharmacol 2022; 13:781889. [PMID: 35153778 PMCID: PMC8826722 DOI: 10.3389/fphar.2022.781889] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2021] [Accepted: 01/07/2022] [Indexed: 12/28/2022] Open
Abstract
Lipoxins (LXs) are generated from arachidonic acid and are involved in the resolution of inflammation and confer protection in a variety of pathological processes. In the nervous system, LXs exert an array of protective effects against neurological diseases, including ischemic or hemorrhagic stroke, neonatal hypoxia-ischemia encephalopathy, brain and spinal cord injury, Alzheimer's disease, multiple sclerosis, and neuropathic pain. Lipoxin administration is a potential therapeutic strategy in neurological diseases due to its notable efficiency and unique superiority regarding safety. Here, we provide an overview of LXs in terms of their synthesis, signaling pathways and neuroprotective evidence. Overall, we believe that, along with advances in lipoxin-related drug design, LXs will bring brighter prospects for neuroprotection.
Collapse
Affiliation(s)
- Jiayu Zhang
- Graduate School of Hebei Medical University, Shijiazhuang, China.,Department of Neurology, Hebei General Hospital, Shijiazhuang, China
| | - Zhe Li
- Department of Neurology, Hebei General Hospital, Shijiazhuang, China
| | - Mingyue Fan
- Department of Neurology, Hebei General Hospital, Shijiazhuang, China
| | - Wei Jin
- Department of Neurology, Hebei General Hospital, Shijiazhuang, China
| |
Collapse
|
29
|
Lipoxin A 4 and its analog attenuate high fat diet-induced atherosclerosis via Keap1/Nrf2 pathway. Exp Cell Res 2022; 412:113025. [PMID: 35026282 DOI: 10.1016/j.yexcr.2022.113025] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2021] [Revised: 01/03/2022] [Accepted: 01/09/2022] [Indexed: 01/07/2023]
Abstract
Excessive oxidative stress and decreased antioxidant capacity of macrophages are initial factors which cause macrophages to transform to foam cells, which represents a key event in the progression of atherosclerosis (AS). BML-111, the analog of lipoxin A4 (LXA4) strongly attenuated high fat (HF) diet-induced atherosclerosis by activating NF-E2 related factor 2 (Nrf2). However, the effect was not through a specific LXA4 receptor (formyl peptide receptor 2, FPR2). BML-111 also strongly inhibited HF diet-induced promotion of MDA level, increased HDL level and decreased IL-1, MCP-1, IL-6, VCAM, ICAM and TNF-α level in aorta. In the in vitro experiments, LXA4 inhibited THP-1 cells to transform to foam cells via Nrf2 pathway. Our findings demonstrated that LXA4 and its analog prevented AS induced by HF diet in SD rats, under which the possible mechanism is through Keap1/Nrf2 pathway.
Collapse
|
30
|
Ansari J, Gavins FNE. Neutrophils and Platelets: Immune Soldiers Fighting Together in Stroke Pathophysiology. Biomedicines 2021; 9:biomedicines9121945. [PMID: 34944761 PMCID: PMC8698717 DOI: 10.3390/biomedicines9121945] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2021] [Revised: 12/12/2021] [Accepted: 12/14/2021] [Indexed: 12/31/2022] Open
Abstract
Neutrophils and platelets exhibit a diverse repertoire of functions in thromboinflammatory conditions such as stroke. Most cerebral ischemic events result from longstanding chronic inflammation secondary to underlying pathogenic conditions, e.g., hypertension, diabetes mellitus, obstructive sleep apnea, coronary artery disease, atrial fibrillation, morbid obesity, dyslipidemia, and sickle cell disease. Neutrophils can enable, as well as resolve, cerebrovascular inflammation via many effector functions including neutrophil extracellular traps, serine proteases and reactive oxygen species, and pro-resolving endogenous molecules such as Annexin A1. Like neutrophils, platelets also engage in pro- as well as anti-inflammatory roles in regulating cerebrovascular inflammation. These anucleated cells are at the core of stroke pathogenesis and can trigger an ischemic event via adherence to the hypoxic cerebral endothelial cells culminating in aggregation and clot formation. In this article, we review and highlight the evolving role of neutrophils and platelets in ischemic stroke and discuss ongoing preclinical and clinical strategies that may produce viable therapeutics for prevention and management of stroke.
Collapse
Affiliation(s)
- Junaid Ansari
- Department of Neurology, Louisiana State University Health Shreveport, Shreveport, LA 71130, USA
- Correspondence: (J.A.); (F.N.E.G.); Tel.: +1-318-626-4282 (J.A.); Tel.: +44-(0)1895-267-151 (F.N.E.G.)
| | - Felicity N. E. Gavins
- The Centre for Inflammation Research and Translational Medicine (CIRTM), Department of Life Sciences, Brunel University London, Uxbridge, Middlesex UB8 3PH, UK
- Correspondence: (J.A.); (F.N.E.G.); Tel.: +1-318-626-4282 (J.A.); Tel.: +44-(0)1895-267-151 (F.N.E.G.)
| |
Collapse
|
31
|
Kalia N. A historical review of experimental imaging of the beating heart coronary microcirculation in vivo. J Anat 2021; 242:3-16. [PMID: 34905637 PMCID: PMC9773169 DOI: 10.1111/joa.13611] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2021] [Revised: 11/25/2021] [Accepted: 12/03/2021] [Indexed: 12/25/2022] Open
Abstract
Following a myocardial infarction (MI), the prognosis of patients is highly dependent upon the re-establishment of perfusion not only in the occluded coronary artery, but also within the coronary microcirculation. However, our fundamental understanding of the pathophysiology of the tiniest blood vessels of the heart is limited primarily because no current clinical imaging tools can directly visualise them. Moreover, in vivo experimental studies of the beating heart using intravital imaging have also been hampered due to obvious difficulties related to significant inherent contractile motion, movement of the heart brought about by nearby lungs and its location in an anatomically challenging position for microscopy. However, recent advances in microscopy techniques, and the development of fluorescent reporter mice and fluorescently conjugated antibodies allowing visualisation of vascular structures, thromboinflammatory cells and blood flow, have allowed us to overcome some of these challenges and increase our basic understanding of cardiac microvascular pathophysiology. In this review, the elegant attempts of the pioneers in intravital imaging of the beating heart will be discussed, which focussed on providing new insights into the anatomy and physiology of the healthy heart microvessels. The reviews end with the more recent studies that focussed on disease pathology and increasing our understanding of myocardial thromboinflammatory cell recruitment and flow disturbances, particularly in the setting of diseases such as MI.
Collapse
Affiliation(s)
- Neena Kalia
- Microcirculation Research GroupInstitute of Cardiovascular SciencesCollege of Medical and Dental SciencesUniversity of BirminghamBirminghamUK
| |
Collapse
|
32
|
Zhu J, Li L, Ding J, Huang J, Shao A, Tang B. The Role of Formyl Peptide Receptors in Neurological Diseases via Regulating Inflammation. Front Cell Neurosci 2021; 15:753832. [PMID: 34650406 PMCID: PMC8510628 DOI: 10.3389/fncel.2021.753832] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2021] [Accepted: 09/02/2021] [Indexed: 01/02/2023] Open
Abstract
Formyl peptide receptors (FPRs) are a group of G protein-coupled cell surface receptors that play important roles in host defense and inflammation. Owing to the ubiquitous expression of FPRs throughout different cell types and since they interact with structurally diverse chemotactic agonists, they have a dual function in inflammatory processes, depending on binding with different ligands so that accelerate or inhibit key intracellular kinase-based regulatory pathways. Neuroinflammation is closely associated with the pathogenesis of neurodegenerative diseases, neurogenic tumors and cerebrovascular diseases. From recent studies, it is clear that FPRs are important biomarkers for neurological diseases as they regulate inflammatory responses by monitoring glial activation, accelerating neural differentiation, regulating angiogenesis, and controlling blood brain barrier (BBB) permeability, thereby affecting neurological disease progression. Given the complex mechanisms of neurological diseases and the difficulty of healing, we are eager to find new and effective therapeutic targets. Here, we review recent research about various mechanisms of the effects generated after FPR binding to different ligands, role of FPRs in neuroinflammation as well as the development and prognosis of neurological diseases. We summarize that the FPR family has dual inflammatory functional properties in central nervous system. Emphasizing that FPR2 acts as a key molecule that mediates the active resolution of inflammation, which binds with corresponding receptors to reduce the expression and activation of pro-inflammatory composition, govern the transport of immune cells to inflammatory tissues, and restore the integrity of the BBB. Concurrently, FPR1 is essentially related to angiogenesis, cell proliferation and neurogenesis. Thus, treatment with FPRs-modulation may be effective for neurological diseases.
Collapse
Affiliation(s)
- Jiahui Zhu
- The Fourth School of Clinical Medicine, Zhejiang Chinese Medical University, Hangzhou, China
| | - Lingfei Li
- Department of Neurology, The Affiliated Hangzhou First People's Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Jiao Ding
- The Fourth School of Clinical Medicine, Zhejiang Chinese Medical University, Hangzhou, China
| | - Jinyu Huang
- Department of Cardiology, The Affiliated Hangzhou First People's Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Anwen Shao
- Department of Neurosurgery, The Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Bo Tang
- Department of Neurology, The Affiliated Hangzhou First People's Hospital, Zhejiang University School of Medicine, Hangzhou, China
| |
Collapse
|
33
|
Busch L, Vieten S, Brödel S, Endres K, Bufe B. Emerging contributions of formyl peptide receptors to neurodegenerative diseases. Biol Chem 2021; 403:27-41. [PMID: 34505459 DOI: 10.1515/hsz-2021-0258] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2021] [Accepted: 08/23/2021] [Indexed: 12/28/2022]
Abstract
Inflammation is a central element of many neurodegenerative diseases. Formyl peptide receptors (FPRs) can trigger several receptor-dependent signal transduction pathways that play a key role in neuroinflammation and neurodegeneration. They are chemotactic receptors that help to regulate pro- and anti-inflammatory responses in most mammals. FPRs are primarily expressed in the immune and nervous systems where they interact with a complex pattern of pathogen-derived and host-endogenous molecules. Mounting evidence points towards a contribution of FPRs - via neuropathological ligands such as Amyloid beta, and neuroprotective ligands such as Humanin, Lipoxin A4, and Annexin A1 - to multiple pathological aspects of neurodegenerative diseases. In this review, we aim to summarize the interplay of FPRs with neuropathological and neuroprotective ligands. Next, we depict their capability to trigger a number of ligand-dependent cell signaling pathways and their potential to interact with additional intracellular cofactors. Moreover, we highlight first studies, demonstrating that a pharmacological inhibition of FPRs helps to ameliorate neuroinflammation, which may pave the way towards novel therapeutic strategies.
Collapse
Affiliation(s)
- Lukas Busch
- Department of Informatics and Microsystems Technology, University of Applied Sciences Kaiserslautern, D-66482 Zweibrücken, Germany
| | - Stefan Vieten
- Department of Informatics and Microsystems Technology, University of Applied Sciences Kaiserslautern, D-66482 Zweibrücken, Germany
| | - Susan Brödel
- Department of Informatics and Microsystems Technology, University of Applied Sciences Kaiserslautern, D-66482 Zweibrücken, Germany
| | - Kristina Endres
- Department of Psychiatry and Psychotherapy, University Medical Centre of the Johannes Gutenberg University, D-55131 Mainz, Germany
| | - Bernd Bufe
- Department of Informatics and Microsystems Technology, University of Applied Sciences Kaiserslautern, D-66482 Zweibrücken, Germany
| |
Collapse
|
34
|
Tylek K, Trojan E, Leśkiewicz M, Regulska M, Bryniarska N, Curzytek K, Lacivita E, Leopoldo M, Basta-Kaim A. Time-Dependent Protective and Pro-Resolving Effects of FPR2 Agonists on Lipopolysaccharide-Exposed Microglia Cells Involve Inhibition of NF-κB and MAPKs Pathways. Cells 2021; 10:cells10092373. [PMID: 34572022 PMCID: PMC8472089 DOI: 10.3390/cells10092373] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2021] [Revised: 09/01/2021] [Accepted: 09/07/2021] [Indexed: 02/07/2023] Open
Abstract
Prolonged or excessive microglial activation may lead to disturbances in the resolution of inflammation (RoI). The importance of specialized pro-resolving lipid mediators (SPMs) in RoI has been highlighted. Among them, lipoxins (LXA4) and aspirin-triggered lipoxin A4 (AT-LXA4) mediate beneficial responses through the activation of N-formyl peptide receptor-2 (FPR2). We aimed to shed more light on the time-dependent protective and anti-inflammatory impact of the endogenous SPMs, LXA4, and AT-LXA4, and of a new synthetic FPR2 agonist MR-39, in lipopolysaccharide (LPS)-exposed rat microglial cells. Our results showed that LXA4, AT-LXA4, and MR-39 exhibit a protective and pro-resolving potential in LPS-stimulated microglia, even if marked differences were apparent regarding the time dependency and efficacy of inhibiting particular biomarkers. The LXA4 action was found mainly after 3 h of LPS stimulation, and the AT-LXA4 effect was varied in time, while MR-39′s effect was mainly observed after 24 h of stimulation by endotoxin. MR-39 was the only FPR2 ligand that attenuated LPS-evoked changes in the mitochondrial membrane potential and diminished the ROS and NO release. Moreover, the LPS-induced alterations in the microglial phenotype were modulated by LXA4, AT-LXA4, and MR-39. The anti-inflammatory effect of MR-39 on the IL-1β release was mediated through FPR2. All tested ligands inhibited TNF-α production, while AT-LXA4 and MR-39 also diminished IL-6 levels in LPS-stimulated microglia. The favorable action of LXA4 and MR-39 was mediated through the inhibition of ERK1/2 phosphorylation. AT-LXA4 and MR39 diminished the phosphorylation of the transcription factor NF-κB, while AT-LXA4 also affected p38 kinase phosphorylation. Our results suggest that new pro-resolving synthetic mediators can represent an attractive treatment option for the enhancement of RoI, and that FPR2 can provide a perspective as a target in immune-related brain disorders.
Collapse
Affiliation(s)
- Kinga Tylek
- Laboratory of Immunoendocrinology, Department of Experimental Neuroendocrinology, Maj Institute of Pharmacology, Polish Academy of Sciences, 12 Smętna St., 31-343 Kraków, Poland; (K.T.); (E.T.); (M.L.); (M.R.); (N.B.); (K.C.)
| | - Ewa Trojan
- Laboratory of Immunoendocrinology, Department of Experimental Neuroendocrinology, Maj Institute of Pharmacology, Polish Academy of Sciences, 12 Smętna St., 31-343 Kraków, Poland; (K.T.); (E.T.); (M.L.); (M.R.); (N.B.); (K.C.)
| | - Monika Leśkiewicz
- Laboratory of Immunoendocrinology, Department of Experimental Neuroendocrinology, Maj Institute of Pharmacology, Polish Academy of Sciences, 12 Smętna St., 31-343 Kraków, Poland; (K.T.); (E.T.); (M.L.); (M.R.); (N.B.); (K.C.)
| | - Magdalena Regulska
- Laboratory of Immunoendocrinology, Department of Experimental Neuroendocrinology, Maj Institute of Pharmacology, Polish Academy of Sciences, 12 Smętna St., 31-343 Kraków, Poland; (K.T.); (E.T.); (M.L.); (M.R.); (N.B.); (K.C.)
| | - Natalia Bryniarska
- Laboratory of Immunoendocrinology, Department of Experimental Neuroendocrinology, Maj Institute of Pharmacology, Polish Academy of Sciences, 12 Smętna St., 31-343 Kraków, Poland; (K.T.); (E.T.); (M.L.); (M.R.); (N.B.); (K.C.)
| | - Katarzyna Curzytek
- Laboratory of Immunoendocrinology, Department of Experimental Neuroendocrinology, Maj Institute of Pharmacology, Polish Academy of Sciences, 12 Smętna St., 31-343 Kraków, Poland; (K.T.); (E.T.); (M.L.); (M.R.); (N.B.); (K.C.)
| | - Enza Lacivita
- Department of Pharmacy—Drug Sciences, University of Bari, Via Orabona 4, 70125 Bari, Italy; (E.L.); (M.L.)
| | - Marcello Leopoldo
- Department of Pharmacy—Drug Sciences, University of Bari, Via Orabona 4, 70125 Bari, Italy; (E.L.); (M.L.)
| | - Agnieszka Basta-Kaim
- Laboratory of Immunoendocrinology, Department of Experimental Neuroendocrinology, Maj Institute of Pharmacology, Polish Academy of Sciences, 12 Smętna St., 31-343 Kraków, Poland; (K.T.); (E.T.); (M.L.); (M.R.); (N.B.); (K.C.)
- Correspondence: ; Tel.: +48-12-662-32-73
| |
Collapse
|
35
|
Liu W, Hsu AY, Wang Y, Lin T, Sun H, Pachter JS, Groisman A, Imperioli M, Yungher FW, Hu L, Wang P, Deng Q, Fan Z. Mitofusin-2 regulates leukocyte adhesion and β2 integrin activation. J Leukoc Biol 2021; 111:771-791. [PMID: 34494308 DOI: 10.1002/jlb.1a0720-471r] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023] Open
Abstract
Neutrophils are critical for inflammation and innate immunity, and their adhesion to vascular endothelium is a crucial step in neutrophil recruitment. Mitofusin-2 (MFN2) is required for neutrophil adhesion, but molecular details are unclear. Here, we demonstrated that β2 -integrin-mediated slow-rolling and arrest, but not PSGL-1-mediated cell rolling, are defective in MFN2-deficient neutrophil-like HL60 cells. This adhesion defect is associated with reduced expression of fMLP (N-formylmethionyl-leucyl-phenylalanine) receptor FPR1 as well as the inhibited β2 integrin activation, as assessed by conformation-specific monoclonal antibodies. MFN2 deficiency also leads to decreased actin polymerization, which is important for β2 integrin activation. Mn2+ -induced cell spreading is also inhibited after MFN2 knockdown. MFN2 deficiency limited the maturation of β2 integrin activation during the neutrophil-directed differentiation of HL60 cells, which is indicated by CD35 and CD87 markers. MFN2 knockdown in β2-integrin activation-matured cells (CD87high population) also inhibits integrin activation, indicating that MFN2 directly affects β2 integrin activation. Our study illustrates the function of MFN2 in leukocyte adhesion and may provide new insights into the development and treatment of MFN2 deficiency-related diseases.
Collapse
Affiliation(s)
- Wei Liu
- Department of Immunology, School of Medicine, UConn Health, Farmington, Connecticut, USA
| | - Alan Y Hsu
- Department of Biological Sciences, Purdue University, West Lafayette, Indiana, USA
| | - Yueyang Wang
- Department of Biological Sciences, Purdue University, West Lafayette, Indiana, USA
| | - Tao Lin
- Department of Immunology, School of Medicine, UConn Health, Farmington, Connecticut, USA
| | - Hao Sun
- Department of Medicine, University of California, San Diego, La Jolla, California, USA
| | - Joel S Pachter
- Department of Immunology, School of Medicine, UConn Health, Farmington, Connecticut, USA
| | - Alex Groisman
- Department of Physics, University of California San Diego, La Jolla, California, USA
| | | | | | - Liang Hu
- Cardiovascular Institute of Zhengzhou University, Department of Cardiology, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Penghua Wang
- Department of Immunology, School of Medicine, UConn Health, Farmington, Connecticut, USA
| | - Qing Deng
- Department of Biological Sciences, Purdue University, West Lafayette, Indiana, USA.,Purdue Institute for Inflammation, Immunology, & Infectious Disease, Purdue University, West Lafayette, Indiana, USA.,Purdue University Center for Cancer Research, Purdue University, West Lafayette, Indiana, USA
| | - Zhichao Fan
- Department of Immunology, School of Medicine, UConn Health, Farmington, Connecticut, USA
| |
Collapse
|
36
|
Park B, Lee M, Kim SD, Jeong YS, Kim JC, Yang S, Kim HY, Bae YS. Activation of formyl peptide receptor 1 elicits therapeutic effects against collagen-induced arthritis. J Cell Mol Med 2021; 25:8936-8946. [PMID: 34378309 PMCID: PMC8435430 DOI: 10.1111/jcmm.16854] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2021] [Revised: 07/27/2021] [Accepted: 08/02/2021] [Indexed: 02/02/2023] Open
Abstract
Rheumatoid arthritis (RA) is an autoimmune disorder which shows production of autoantibodies, inflammation, bone erosion, swelling and pain in joints. In this study, we examined the effects of an immune-modulating peptide, WKYMVm, that is an agonist for formyl peptide receptors (FPRs). Administration of WKYMVm into collagen-induced arthritis (CIA) mice, an animal model for RA, attenuated paw thickness, clinical scores, production of type II collagen-specific antibodies and inflammatory cytokines. WKYMVm treatment also decreased the numbers of TH 1 and TH 17 cells in the spleens of CIA mice. WKYMVm attenuated TH 1 and TH 17 differentiation in a dendritic cell (DC)-dependent manner. WKYMVm-induced beneficial effects against CIA and WKYMVm-attenuated TH 1 and TH 17 differentiation were reversed by cyclosporin H but not by WRW4, indicating a crucial role of FPR1. We also found that WKYMVm augmented IL-10 production from lipopolysaccharide-stimulated DCs and WKYMVm failed to suppress TH 1 and TH 17 differentiation in the presence of anti-IL-10 antibody. The therapeutic administration of WKYMVm also elicited beneficial outcome against CIA. Collectively, we demonstrate that WKYMVm stimulation of FPR1 in DCs suppresses the generation of TH 1 and TH 17 cells via IL-10 production, providing novel insight into the function of FPR1 in regulating CIA pathogenesis.
Collapse
Affiliation(s)
- Byunghyun Park
- Department of Biological Sciences, Sungkyunkwan University, Suwon, Korea
| | - Mingyu Lee
- Department of Health Sciences and Technology, Samsung Advanced Institute for Health Sciences and Technology, Sungkyunkwan University, Seoul, Korea
| | - Sang Doo Kim
- Department of Biological Sciences, Sungkyunkwan University, Suwon, Korea
| | - Yu Sun Jeong
- Department of Biological Sciences, Sungkyunkwan University, Suwon, Korea
| | - Ji Cheol Kim
- Department of Biological Sciences, Sungkyunkwan University, Suwon, Korea
| | - Siyoung Yang
- Department of Pharmacology, Ajou University School of Medicine, Suwon, Korea
| | - Hye Young Kim
- Laboratory of Mucosal Immunology, Department of Biomedical Sciences, Seoul National University College of Medicine, Seoul, Korea
| | - Yoe-Sik Bae
- Department of Biological Sciences, Sungkyunkwan University, Suwon, Korea.,Department of Health Sciences and Technology, Samsung Advanced Institute for Health Sciences and Technology, Sungkyunkwan University, Seoul, Korea
| |
Collapse
|
37
|
Iffah R, Gavins FNE. Thromboinflammation in coronavirus disease 2019: The clot thickens. Br J Pharmacol 2021; 179:2100-2107. [PMID: 34128218 PMCID: PMC8444860 DOI: 10.1111/bph.15594] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2021] [Revised: 05/20/2021] [Accepted: 05/27/2021] [Indexed: 12/15/2022] Open
Abstract
Since the start of the severe acute respiratory syndrome coronavirus 2 (SARS‐CoV‐2) pandemic, a disease that has become one of the world's greatest global health challenges, the role of the immune system has been at the forefront of scientific studies. The pathophysiology of coronavirus disease 2019 (COVID‐19) is complex, which is evident in those at higher risk for poor outcome. Multiple systems contribute to thrombosis and inflammation seen in COVID‐19 patients, including neutrophil and platelet activation, and endothelial dysfunction. Understanding how the immune system functions in different patient cohorts (particularly given recent emerging events with the Oxford/AstraZeneca vaccine) is vital to understanding the pathophysiology of this devastating disease and for the subsequent development of novel therapeutic targets and to facilitate possible drug repurposing strategies that could benefit society on a global scale.
Collapse
Affiliation(s)
- Raayma Iffah
- Department of Life Sciences, Centre for Inflammation Research and Translational Medicine (CIRTM), Brunel University London, London, UK
| | - Felicity N E Gavins
- Department of Life Sciences, Centre for Inflammation Research and Translational Medicine (CIRTM), Brunel University London, London, UK
| |
Collapse
|
38
|
Wee Y, Yang C, Chen S, Yen Y, Wang C. Inositol hexaphosphate modulates the behavior of macrophages through alteration of gene expression involved in pathways of pro- and anti-inflammatory responses, and resolution of inflammation pathways. Food Sci Nutr 2021; 9:3240-3249. [PMID: 34136188 PMCID: PMC8194914 DOI: 10.1002/fsn3.2286] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2021] [Revised: 03/13/2021] [Accepted: 03/25/2021] [Indexed: 12/19/2022] Open
Abstract
Inositol hexaphosphate (IP6) is a dietary compound commonly obtained from corn, rice, etc. Although we may consume significant amount of IP6 daily, it is unclear whether this diet will impact macrophages' fate and function. Therefore, we characterized the underlying relationship between IP6 and macrophage polarization in this study. We specifically examined the signature gene expression profiles associated with pro- and anti-inflammatory responses, and resolution of inflammation pathways in macrophages under the influence of IP6. Interestingly, our data suggested that IP6 polarizes bone marrow-derived macrophages (BMDM) into an M2a-like subtype. Our results also demonstrated that IP6 reduces lipopolysaccharide-induced apoptosis and pro-inflammatory responses in macrophages. In contrast, the expression levels of genes related to anti-inflammatory responses and resolution of inflammation pathways are upregulated. Our findings collectively demonstrated that IP6 has profound modulation effects on macrophages, which warrant further research on the therapeutic benefits of IP6 for inflammatory diseases.
Collapse
Affiliation(s)
- Yinshen Wee
- Department of PathologyUniversity of UtahSalt Lake CityUTUSA
| | | | - Shau‐Kwaun Chen
- Institute of NeuroscienceNational Chengchi UniversityTaipeiTaiwan
| | - Yu‐Chun Yen
- Biostatistics CenterOffice of Data ScienceTaipei Medical UniversityTaipeiTaiwan
| | - Ching‐Shuen Wang
- School of DentistryCollege of Oral MedicineTaipei Medical UniversityTaipeiTaiwan
| |
Collapse
|
39
|
Grewal T, Rentero C, Enrich C, Wahba M, Raabe CA, Rescher U. Annexin Animal Models-From Fundamental Principles to Translational Research. Int J Mol Sci 2021; 22:ijms22073439. [PMID: 33810523 PMCID: PMC8037771 DOI: 10.3390/ijms22073439] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2021] [Revised: 03/18/2021] [Accepted: 03/24/2021] [Indexed: 02/07/2023] Open
Abstract
Routine manipulation of the mouse genome has become a landmark in biomedical research. Traits that are only associated with advanced developmental stages can now be investigated within a living organism, and the in vivo analysis of corresponding phenotypes and functions advances the translation into the clinical setting. The annexins, a family of closely related calcium (Ca2+)- and lipid-binding proteins, are found at various intra- and extracellular locations, and interact with a broad range of membrane lipids and proteins. Their impacts on cellular functions has been extensively assessed in vitro, yet annexin-deficient mouse models generally develop normally and do not display obvious phenotypes. Only in recent years, studies examining genetically modified annexin mouse models which were exposed to stress conditions mimicking human disease often revealed striking phenotypes. This review is the first comprehensive overview of annexin-related research using animal models and their exciting future use for relevant issues in biology and experimental medicine.
Collapse
Affiliation(s)
- Thomas Grewal
- School of Pharmacy, Faculty of Medicine and Health, University of Sydney, Sydney, NSW 2006, Australia;
- Correspondence: (T.G.); (U.R.); Tel.: +61-(0)2-9351-8496 (T.G.); +49-(0)251-83-52121 (U.R.)
| | - Carles Rentero
- Departament de Biomedicina, Unitat de Biologia Cel·lular, Facultat de Medicina i Ciències de la Salut, Universitat de Barcelona, 08036 Barcelona, Spain; (C.R.); (C.E.)
- Centre de Recerca Biomèdica CELLEX, Institut d’Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), 08036 Barcelona, Spain
| | - Carlos Enrich
- Departament de Biomedicina, Unitat de Biologia Cel·lular, Facultat de Medicina i Ciències de la Salut, Universitat de Barcelona, 08036 Barcelona, Spain; (C.R.); (C.E.)
- Centre de Recerca Biomèdica CELLEX, Institut d’Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), 08036 Barcelona, Spain
| | - Mohamed Wahba
- School of Pharmacy, Faculty of Medicine and Health, University of Sydney, Sydney, NSW 2006, Australia;
| | - Carsten A. Raabe
- Research Group Regulatory Mechanisms of Inflammation, Center for Molecular Biology of Inflammation (ZMBE) and Cells in Motion Interfaculty Center (CiM), Institute of Medical Biochemistry, University of Muenster, 48149 Muenster, Germany;
| | - Ursula Rescher
- Research Group Regulatory Mechanisms of Inflammation, Center for Molecular Biology of Inflammation (ZMBE) and Cells in Motion Interfaculty Center (CiM), Institute of Medical Biochemistry, University of Muenster, 48149 Muenster, Germany;
- Correspondence: (T.G.); (U.R.); Tel.: +61-(0)2-9351-8496 (T.G.); +49-(0)251-83-52121 (U.R.)
| |
Collapse
|
40
|
Ansari J, Gavins FNE. The impact of thrombo-inflammation on the cerebral microcirculation. Microcirculation 2021; 28:e12689. [PMID: 33638262 DOI: 10.1111/micc.12689] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2021] [Revised: 02/12/2021] [Accepted: 02/22/2021] [Indexed: 12/16/2022]
Abstract
The intertwined processes of thrombosis and inflammation (termed "thrombo-inflammation") are significant drivers of cerebrovascular diseases, and as such, they represent prime targets for drug discovery programs focusing on treatment and management of cerebrovascular diseases. Most cerebrovascular events result from chronic systemic microcirculatory dysfunction due to underlying conditions, for example, hypertension, diabetes mellitus, coronary artery disease, dyslipidemia, and sickle cell disease. Immune cells especially neutrophils play a critical role in the onset and maintenance of neuroinflammatory responses in the microcirculation. Neutrophils have the ability to drive both inflammatory and anti-inflammatory/pro-resolution effects depending on the underlying vascular state (physiological vs. pathological). In this article, we highlight the pathophysiological role of neutrophils in stroke and discuss ongoing pharmacotherapeutic strategies that are focused on identifying potential therapeutic targets for enhancing neuroprotection, mitigating inflammatory pathways, and enabling resolution.
Collapse
Affiliation(s)
- Junaid Ansari
- Department of Neurology, Louisiana State University Health Sciences Center-Shreveport, Shreveport, LA, USA
| | - Felicity N E Gavins
- Department of Life Sciences, The Centre for Inflammation Research and Translational Medicine (CIRTM), Brunel University London, Uxbridge, Middlesex, UK
| |
Collapse
|
41
|
Ansari J, Senchenkova EY, Vital SA, Al-Yafeai Z, Kaur G, Sparkenbaugh EM, Orr AW, Pawlinski R, Hebbel RP, Granger DN, Kubes P, Gavins FNE. Targeting the AnxA1/Fpr2/ALX pathway regulates neutrophil function, promoting thromboinflammation resolution in sickle cell disease. Blood 2021; 137:1538-1549. [PMID: 33512489 PMCID: PMC7976506 DOI: 10.1182/blood.2020009166] [Citation(s) in RCA: 37] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2020] [Accepted: 12/28/2020] [Indexed: 12/19/2022] Open
Abstract
Neutrophils play a crucial role in the intertwined processes of thrombosis and inflammation. An altered neutrophil phenotype may contribute to inadequate resolution, which is known to be a major pathophysiological contributor of thromboinflammatory conditions such as sickle cell disease (SCD). The endogenous protein annexin A1 (AnxA1) facilitates inflammation resolution via formyl peptide receptors (FPRs). We sought to comprehensively elucidate the functional significance of targeting the neutrophil-dependent AnxA1/FPR2/ALX pathway in SCD. Administration of AnxA1 mimetic peptide AnxA1Ac2-26 ameliorated cerebral thrombotic responses in Sickle transgenic mice via regulation of the FPR2/ALX (a fundamental receptor involved in resolution) pathway. We found direct evidence that neutrophils with SCD phenotype play a key role in contributing to thromboinflammation. In addition, AnxA1Ac2-26 regulated activated SCD neutrophils through protein kinase B (Akt) and extracellular signal-regulated kinases (ERK1/2) to enable resolution. We present compelling conceptual evidence that targeting the AnxA1/FPR2/ALX pathway may provide new therapeutic possibilities against thromboinflammatory conditions such as SCD.
Collapse
Affiliation(s)
- Junaid Ansari
- Department of Molecular and Cellular Physiology
- Department of Neurology, and
| | | | | | - Zaki Al-Yafeai
- Department of Pathology and Translational Pathobiology, Louisiana State University Health Sciences Center-Shreveport, Shreveport, LA
| | | | - Erica M Sparkenbaugh
- UNC Blood Research Center, Department of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC
| | - A Wayne Orr
- Department of Molecular and Cellular Physiology
- Department of Pathology and Translational Pathobiology, Louisiana State University Health Sciences Center-Shreveport, Shreveport, LA
- Department of Cellular Biology and Anatomy, Louisiana State University Health Sciences Center-Shreveport, Shreveport, LA
| | - Rafal Pawlinski
- UNC Blood Research Center, Department of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC
| | - Robert P Hebbel
- Department of Medicine, University of Minnesota Medical School, Minneapolis, MN
| | | | - Paul Kubes
- Departments of Physiology and Pharmacology, Microbiology and Immunology and Critical Care Medicine, Snyder Institute for Chronic Disease, University of Calgary, Calgary, AB, Canada; and
| | - Felicity N E Gavins
- Department of Molecular and Cellular Physiology
- Department of Neurology, and
- Department of Life Sciences, Brunel University London, United Kingdom
| |
Collapse
|
42
|
Liao WI, Wu SY, Tsai SH, Pao HP, Huang KL, Chu SJ. 2-Methoxyestradiol Protects Against Lung Ischemia/Reperfusion Injury by Upregulating Annexin A1 Protein Expression. Front Immunol 2021; 12:596376. [PMID: 33796096 PMCID: PMC8007881 DOI: 10.3389/fimmu.2021.596376] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2020] [Accepted: 02/22/2021] [Indexed: 11/13/2022] Open
Abstract
Background: 2-Methoxyestradiol (2ME), a natural 17-β estradiol metabolite, is a potent anti-inflammatory agent, but its effect on ischemia/reperfusion (IR)-induced acute lung inflammation remains unknown. Annexin A1 (AnxA1), a glucocorticoid-regulated protein, is effective at inhibiting neutrophil transendothelial migration by binding the formyl peptide receptors (FPRs). We aimed to investigate whether 2ME upregulates the expression of AnxA1 and protects against IR-induced lung damage. Methods: IR-mediated acute lung inflammation was induced by ischemia for 40 min followed by reperfusion for 60 min in an isolated, perfused rat lung model. The rat lungs were randomly treated with vehicle or 2ME, and the functional relevance of AnxA1 was determined using an anti-AnxA1 antibody or BOC2 (a pan-receptor antagonist of the FPR). In vitro, human primary alveolar epithelial cells (HPAECs) and rat neutrophils were pretreated with 2ME and an AnxA1 siRNA or anti-AnxA1 antibody and subjected to hypoxia-reoxygenation (HR). Results: 2ME significantly decreased all lung edema parameters, neutrophil infiltration, oxidative stress, proinflammatory cytokine production, lung cell apoptosis, tight junction protein disruption, and lung tissue injury in the IR-induced acute lung inflammation model. 2ME also increased the expression of the AnxA1 mRNA and protein and suppressed the activation of nuclear factor-κB (NF-κB). In vitro, 2ME attenuated HR-triggered NF-κB activation and interleukin-8 production in HPAECs, decreased transendothelial migration, tumor necrosis factor-α production, and increased apoptosis in neutrophils exposed to HR. These protective effects of 2ME were significantly abrogated by BOC2, the anti-AnxA1 antibody, or AnxA1 siRNA. Conclusions: 2ME ameliorates IR-induced acute lung inflammation by increasing AnxA1 expression. Based on these results, 2ME may be a promising agent for attenuating IR-induced lung injury.
Collapse
Affiliation(s)
- Wen-I Liao
- The Graduate Institute of Medical Sciences, National Defense Medical Center, Taipei, Taiwan
- Department of Emergency Medicine, Tri-Service General Hospital, National Defense Medical Center, Taipei, Taiwan
| | - Shu-Yu Wu
- Institute of Aerospace and Undersea Medicine, National Defense Medical Center, Taipei, Taiwan
| | - Shih-Hung Tsai
- Department of Emergency Medicine, Tri-Service General Hospital, National Defense Medical Center, Taipei, Taiwan
- Department of Physiology and Biophysics, Graduate Institute of Physiology, National Defense Medical Center, Taipei, Taiwan
| | - Hsin-Ping Pao
- The Graduate Institute of Medical Sciences, National Defense Medical Center, Taipei, Taiwan
| | - Kun-Lun Huang
- The Graduate Institute of Medical Sciences, National Defense Medical Center, Taipei, Taiwan
- Institute of Aerospace and Undersea Medicine, National Defense Medical Center, Taipei, Taiwan
| | - Shi-Jye Chu
- Department of Internal Medicine, Tri-Service General Hospital, National Defense Medical Center, Taipei, Taiwan
| |
Collapse
|
43
|
Regulation of Inflammation and Oxidative Stress by Formyl Peptide Receptors in Cardiovascular Disease Progression. Life (Basel) 2021; 11:life11030243. [PMID: 33804219 PMCID: PMC7998928 DOI: 10.3390/life11030243] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2021] [Revised: 03/08/2021] [Accepted: 03/14/2021] [Indexed: 12/23/2022] Open
Abstract
G protein-coupled receptors (GPCRs) are the most important regulators of cardiac function and are commonly targeted for medical therapeutics. Formyl-Peptide Receptors (FPRs) are members of the GPCR superfamily and play an emerging role in cardiovascular pathologies. FPRs can modulate oxidative stress through nicotinamide adenine dinucleotide phosphate (NADPH) oxidase-dependent reactive oxygen species (ROS) production whose dysregulation has been observed in different cardiovascular diseases. Therefore, many studies are focused on identifying molecular mechanisms of the regulation of ROS production. FPR1, FPR2 and FPR3 belong to the FPRs family and their stimulation triggers phosphorylation of intracellular signaling molecules and nonsignaling proteins that are required for NADPH oxidase activation. Some FPR agonists trigger inflammatory processes, while other ligands activate proresolving or anti-inflammatory pathways, depending on the nature of the ligands. In general, bacterial and mitochondrial formylated peptides activate a proinflammatory cell response through FPR1, while Annexin A1 and Lipoxin A4 are anti-inflammatory FPR2 ligands. FPR2 can also trigger a proinflammatory pathway and the switch between FPR2-mediated pro- and anti-inflammatory cell responses depends on conformational changes of the receptor upon ligand binding. Here we describe the detrimental or beneficial effects of the main FPR agonists and their potential role as new therapeutic and diagnostic targets in the progression of cardiovascular diseases.
Collapse
|
44
|
Thromboinflammation Model-on-A-Chip by Whole Blood Microfluidics on Fixed Human Endothelium. Diagnostics (Basel) 2021; 11:diagnostics11020203. [PMID: 33573079 PMCID: PMC7911484 DOI: 10.3390/diagnostics11020203] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2020] [Revised: 12/23/2020] [Accepted: 01/26/2021] [Indexed: 01/15/2023] Open
Abstract
Microfluidic devices have an established role in the study of platelets and coagulation factors in thrombosis, with potential diagnostic applications. However, few microfluidic devices have assessed the contribution of neutrophils to thrombus formation, despite increasing knowledge of neutrophils’ importance in cardiovascular thrombosis. We describe a thromboinflammation model which uses straight channels, lined with fixed human umbilical vein endothelial cells, after treatment with tumour necrosis factor-alpha. Re-calcified whole blood is perfused over the endothelium at venous and arterial shear rate. Neutrophil adhesion, platelet and fibrin thrombus formation, is measured over time by the addition of fluorescent antibodies to a whole blood sample. Fixed endothelium retains surface expression of adhesion molecules ICAM-1 and E-Selectin. Neutrophils adhere preferentially to platelet thrombi on the endothelium. Inhibitors of neutrophil adhesion and anti-inflammatory agents, such as isoquercetin, decrease neutrophil adhesion. Our model offers the advantage of the use of (1) fixed endothelium, (2) whole blood, instead of isolated neutrophils, and (3) a small amount of blood (1 mL). The characteristics of this thromboinflammation model provide the potential for further development for drug screening and point-of-care applications.
Collapse
|
45
|
Vallance TM, Sheard JJ, Meng Y, Torre EC, Patel K, Widera D, Vaiyapuri S. Development and characterization of a novel, megakaryocyte NF-κB reporter cell line for investigating inflammatory responses. J Thromb Haemost 2021; 19:107-120. [PMID: 33037735 DOI: 10.1111/jth.15118] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2020] [Revised: 08/27/2020] [Accepted: 09/24/2020] [Indexed: 11/27/2022]
Abstract
Essentials An easily detectable readout in megakaryocyte cell lines will enhance inflammatory research in these cells. Here, we report the development and characterization of a novel megakaryocyte NF-κB-reporter cell line (Meg-01R). Multiple inflammatory molecules modulate NF-κB activity in Meg-01R cells. Meg-01R cells respond to small molecule inhibitors such as IMD0354 and C87 that are known to inhibit NF-κB activity upon stimulation with TNFα. ABSTRACT: Background Because of the difficulties in acquiring large numbers of megakaryocytes, the impact of inflammatory responses on these cells and their ability to produce fully functional platelets under various pathological conditions has not been investigated in detail. Objectives The primary objective of this study is to develop and functionally characterize a novel megakaryocyte nuclear factor κB (NF-κB) reporter cell line to determine the effects of various inflammatory molecules on megakaryocytes and their signalling pathways. Methods A Meg-01-NF-κB-GFP-Luc (Meg-01R) cell line was developed by inserting a reporter NF-κB-GFP-Luc cassette into normal Meg-01 cells to produce luciferase following activation of NF-κB to enable easy detection of pro-inflammatory and reparative signalling. Results and conclusions Meg-01 and Meg-01R cells have comparable characteristics, including the expression of both GPIbα and integrin β3 . Meg-01R cells responded to various inflammatory molecules as measured by NF-κB-dependent bioluminescence. For example, inflammatory molecules such as tumor necrosis factor-α and Pam3CSK4 increased NF-κB activity, whereas an antimicrobial peptide, LL37, reduced its activity. Meg-01R cells were also found to be sensitive to inhibitors (IMD0354 and C87) of inflammatory pathways. Notably, Meg-01R cells were able to respond to lipopolysaccharide (LPS; non-ultrapure), although it was not able to react to ultrapure LPS because of the lack of sufficient TLR4 molecules on their surface. For the first time, we report the development and characterization of a novel megakaryocyte NF-κB reporter cell line (Meg-01R) as a robust tool to study the inflammatory responses/signalling of megakaryocytes upon stimulation with a broad range of inflammatory molecules that can affect NF-κB activity.
Collapse
Affiliation(s)
| | | | - Yiming Meng
- School of Pharmacy, University of Reading, Reading, UK
| | | | - Ketan Patel
- School of Biological Sciences, University of Reading, Reading, UK
| | - Darius Widera
- School of Pharmacy, University of Reading, Reading, UK
| | | |
Collapse
|
46
|
Jaén RI, Sánchez-García S, Fernández-Velasco M, Boscá L, Prieto P. Resolution-Based Therapies: The Potential of Lipoxins to Treat Human Diseases. Front Immunol 2021; 12:658840. [PMID: 33968061 PMCID: PMC8102821 DOI: 10.3389/fimmu.2021.658840] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2021] [Accepted: 04/07/2021] [Indexed: 02/05/2023] Open
Abstract
Inflammation is an a physiological response instead an essential response of the organism to injury and its adequate resolution is essential to restore homeostasis. However, defective resolution can be the precursor of severe forms of chronic inflammation and fibrosis. Nowadays, it is known that an excessive inflammatory response underlies the most prevalent human pathologies worldwide. Therefore, great biomedical research efforts have been driven toward discovering new strategies to promote the resolution of inflammation with fewer side-effects and more specificity than the available anti-inflammatory treatments. In this line, the use of endogenous specialized pro-resolving mediators (SPMs) has gained a prominent interest. Among the different SPMs described, lipoxins stand out as one of the most studied and their deficiency has been widely associated with a wide range of pathologies. In this review, we examined the current knowledge on the therapeutic potential of lipoxins to treat diseases characterized by a severe inflammatory background affecting main physiological systems, paying special attention to the signaling pathways involved. Altogether, we provide an updated overview of the evidence suggesting that increasing endogenously generated lipoxins may emerge as a new therapeutic approach to prevent and treat many of the most prevalent diseases underpinned by an increased inflammatory response.
Collapse
Affiliation(s)
- Rafael I. Jaén
- Instituto de Investigaciones Biomédicas Alberto Sols, CSIC-UAM, Madrid, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Cardiovasculares (CIBER-CV), Instituto de Salud Carlos III, Madrid, Spain
| | | | - María Fernández-Velasco
- Centro de Investigación Biomédica en Red de Enfermedades Cardiovasculares (CIBER-CV), Instituto de Salud Carlos III, Madrid, Spain
- Instituto de investigación del Hospital la Paz, IdiPaz, Madrid, Spain
| | - Lisardo Boscá
- Instituto de Investigaciones Biomédicas Alberto Sols, CSIC-UAM, Madrid, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Cardiovasculares (CIBER-CV), Instituto de Salud Carlos III, Madrid, Spain
- *Correspondence: Lisardo Boscá, ; Patricia Prieto,
| | - Patricia Prieto
- Instituto de Investigaciones Biomédicas Alberto Sols, CSIC-UAM, Madrid, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Cardiovasculares (CIBER-CV), Instituto de Salud Carlos III, Madrid, Spain
- Departamento de Farmacología, Farmacognosia y Botánica, Facultad de Farmacia, Universidad Complutense de Madrid, Madrid, Spain
- *Correspondence: Lisardo Boscá, ; Patricia Prieto,
| |
Collapse
|
47
|
Terstappen F, Calis JJA, Paauw ND, Joles JA, van Rijn BB, Mokry M, Plösch T, Lely AT. Developmental programming in human umbilical cord vein endothelial cells following fetal growth restriction. Clin Epigenetics 2020; 12:185. [PMID: 33256815 PMCID: PMC7708922 DOI: 10.1186/s13148-020-00980-9] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2020] [Accepted: 11/17/2020] [Indexed: 12/14/2022] Open
Abstract
BACKGROUND Fetal growth restriction (FGR) is associated with an increased susceptibility for various noncommunicable diseases in adulthood, including cardiovascular and renal disease. During FGR, reduced uteroplacental blood flow, oxygen and nutrient supply to the fetus are hypothesized to detrimentally influence cardiovascular and renal programming. This study examined whether developmental programming profiles, especially related to the cardiovascular and renal system, differ in human umbilical vein endothelial cells (HUVECs) collected from pregnancies complicated by placental insufficiency-induced FGR compared to normal growth pregnancies. Our approach, involving transcriptomic profiling by RNA-sequencing and gene set enrichment analysis focused on cardiovascular and renal gene sets and targeted DNA methylation assays, contributes to the identification of targets underlying long-term cardiovascular and renal diseases. RESULTS Gene set enrichment analysis showed several downregulated gene sets, most of them involved in immune or inflammatory pathways or cell cycle pathways. seven of the 22 significantly upregulated gene sets related to kidney development and four gene sets involved with cardiovascular health and function were downregulated in FGR (n = 11) versus control (n = 8). Transcriptomic profiling by RNA-sequencing revealed downregulated expression of LGALS1, FPR3 and NRM and upregulation of lincRNA RP5-855F14.1 in FGR compared to controls. DNA methylation was similar for LGALS1 between study groups, but relative hypomethylation of FPR3 and hypermethylation of NRM were present in FGR, especially in male offspring. Absolute differences in methylation were, however, small. CONCLUSION This study showed upregulation of gene sets related to renal development in HUVECs collected from pregnancies complicated by FGR compared to control donors. The differentially expressed gene sets related to cardiovascular function and health might be in line with the downregulated expression of NRM and upregulated expression of lincRNA RP5-855F14.1 in FGR samples; NRM is involved in cardiac remodeling, and lincRNAs are correlated with cardiovascular diseases. Future studies should elucidate whether the downregulated LGALS1 and FPR3 expressions in FGR are angiogenesis-modulating regulators leading to placental insufficiency-induced FGR or whether the expression of these genes can be used as a biomarker for increased cardiovascular risk. Altered DNA methylation might partly underlie FPR3 and NRM differential gene expression differences in a sex-dependent manner.
Collapse
Affiliation(s)
- Fieke Terstappen
- Division Woman and Baby, Department of Obstetrics, Wilhelmina Children's Hospital, University Medical Center Utrecht, Postbus 85090, 3508 AB, Utrecht, The Netherlands.
- Department for Developmental Origins of Disease, Wilhelmina Children's Hospital, University Medical Center Utrecht, Utrecht, The Netherlands.
| | - Jorg J A Calis
- Department of Cardiology, University Medical Center Utrecht, Utrecht, The Netherlands
- Center for Translational Immunology, University Medical Centre Utrecht, Utrecht, The Netherlands
| | - Nina D Paauw
- Division Woman and Baby, Department of Obstetrics, Wilhelmina Children's Hospital, University Medical Center Utrecht, Postbus 85090, 3508 AB, Utrecht, The Netherlands
| | - Jaap A Joles
- Department of Nephrology and Hypertension, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Bas B van Rijn
- Department of Obstetrics and Fetal Medicine, Erasmus MC University Medical Center Rotterdam, Rotterdam, The Netherlands
| | - Michal Mokry
- Department of Cardiology, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Torsten Plösch
- Department of Obstetrics and Gynaecology, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | - A Titia Lely
- Division Woman and Baby, Department of Obstetrics, Wilhelmina Children's Hospital, University Medical Center Utrecht, Postbus 85090, 3508 AB, Utrecht, The Netherlands
| |
Collapse
|
48
|
Vital SA, Senchenkova EY, Ansari J, Gavins FNE. Targeting AnxA1/Formyl Peptide Receptor 2 Pathway Affords Protection against Pathological Thrombo-Inflammation. Cells 2020; 9:cells9112473. [PMID: 33202930 PMCID: PMC7697101 DOI: 10.3390/cells9112473] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2020] [Revised: 10/30/2020] [Accepted: 11/04/2020] [Indexed: 12/14/2022] Open
Abstract
Stroke is a leading cause of death and disability globally and is associated with a number of co-morbidities including sepsis and sickle cell disease (SCD). Despite thrombo-inflammation underlying these co-morbidities, its pathogenesis remains complicated and drug discovery programs aimed at reducing and resolving the detrimental effects remain a major therapeutic challenge. The objective of this study was to assess whether the anti-inflammatory pro-resolving protein Annexin A1 (AnxA1) was able to reduce inflammation-induced thrombosis and suppress platelet activation and thrombus formation in the cerebral microvasculature. Using two distinct models of pathological thrombo-inflammation (lipopolysaccharide (LPS) and sickle transgenic mice (STM)), thrombosis was induced in the murine brain using photoactivation (light/dye) coupled with intravital microscopy. The heightened inflammation-induced microvascular thrombosis present in these two distinct thrombo-inflammatory models was inhibited significantly by the administration of AnxA1 mimetic peptide AnxA1Ac2-26 (an effect more pronounced in the SCD model vs. the endotoxin model) and mediated by the key resolution receptor, Fpr2/ALX. Furthermore, AnxA1Ac2-26 treatment was able to hamper platelet aggregation by reducing platelet stimulation and aggregation (by moderating αIIbβ3 and P-selectin). These findings suggest that targeting the AnxA1/Fpr2/ALX pathway represents an attractive novel treatment strategy for resolving thrombo-inflammation, counteracting e.g., stroke in high-risk patient cohorts.
Collapse
Affiliation(s)
- Shantel A. Vital
- Department of Molecular and Cellular Physiology, Louisiana State University Health Sciences Center-Shreveport, Shreveport, LA 71130, USA; (S.A.V.); (E.Y.S.); (J.A.)
| | - Elena Y. Senchenkova
- Department of Molecular and Cellular Physiology, Louisiana State University Health Sciences Center-Shreveport, Shreveport, LA 71130, USA; (S.A.V.); (E.Y.S.); (J.A.)
| | - Junaid Ansari
- Department of Molecular and Cellular Physiology, Louisiana State University Health Sciences Center-Shreveport, Shreveport, LA 71130, USA; (S.A.V.); (E.Y.S.); (J.A.)
- Department of Neurology, Louisiana State University Health Sciences Center-Shreveport, Shreveport, LA 71130, USA
| | - Felicity N. E. Gavins
- Department of Molecular and Cellular Physiology, Louisiana State University Health Sciences Center-Shreveport, Shreveport, LA 71130, USA; (S.A.V.); (E.Y.S.); (J.A.)
- Department of Neurology, Louisiana State University Health Sciences Center-Shreveport, Shreveport, LA 71130, USA
- Department of Life Sciences, Centre for Inflammation Research and Translational Medicine (CIRTM), Brunel University London, Uxbridge, Middlesex UB8 3PH, UK
- Correspondence: ; Tel.: +44-(0)-1895-267151
| |
Collapse
|
49
|
Trojan E, Bryniarska N, Leśkiewicz M, Regulska M, Chamera K, Szuster-Głuszczak M, Leopoldo M, Lacivita E, Basta-Kaim A. The Contribution of Formyl Peptide Receptor Dysfunction to the Course of Neuroinflammation: A Potential Role in the Brain Pathology. Curr Neuropharmacol 2020; 18:229-249. [PMID: 31629396 PMCID: PMC7327951 DOI: 10.2174/1570159x17666191019170244] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2019] [Revised: 08/01/2019] [Accepted: 10/15/2019] [Indexed: 12/27/2022] Open
Abstract
Chronic inflammatory processes within the central nervous system (CNS) are in part responsible for the development of neurodegenerative and psychiatric diseases. These processes are associated with, among other things, the increased and disturbed activation of microglia and the elevated production of proinflammatory factors. Recent studies indicated that the disruption of the process of resolution of inflammation (RoI) may be the cause of CNS disorders. It is shown that the RoI is regulated by endogenous molecules called specialized pro-resolving mediators (SPMs), which interact with specific membrane receptors. Some SPMs activate formyl peptide receptors (FPRs), which belong to the family of seven-transmembrane G protein-coupled receptors. These receptors take part not only in the proinflammatory response but also in the resolution of the inflammation process. Therefore, the activation of FPRs might have complex consequences. This review discusses the potential role of FPRs, and in particular the role of FPR2 subtype, in the brain under physiological and pathological conditions and their involvement in processes underlying neurodegenerative and psychiatric disorders as well as ischemia, the pathogenesis of which involves the dysfunction of inflammatory processes.
Collapse
Affiliation(s)
- Ewa Trojan
- Laboratory of Immunoendocrinology, Department of Experimental Neuroendocrinology, Maj Institute of Pharmacology, Polish Academy of Sciences, 12 Smetna St. 31-343 Krakow, Poland
| | - Natalia Bryniarska
- Laboratory of Immunoendocrinology, Department of Experimental Neuroendocrinology, Maj Institute of Pharmacology, Polish Academy of Sciences, 12 Smetna St. 31-343 Krakow, Poland
| | - Monika Leśkiewicz
- Laboratory of Immunoendocrinology, Department of Experimental Neuroendocrinology, Maj Institute of Pharmacology, Polish Academy of Sciences, 12 Smetna St. 31-343 Krakow, Poland
| | - Magdalena Regulska
- Laboratory of Immunoendocrinology, Department of Experimental Neuroendocrinology, Maj Institute of Pharmacology, Polish Academy of Sciences, 12 Smetna St. 31-343 Krakow, Poland
| | - Katarzyna Chamera
- Laboratory of Immunoendocrinology, Department of Experimental Neuroendocrinology, Maj Institute of Pharmacology, Polish Academy of Sciences, 12 Smetna St. 31-343 Krakow, Poland
| | - Magdalena Szuster-Głuszczak
- Laboratory of Immunoendocrinology, Department of Experimental Neuroendocrinology, Maj Institute of Pharmacology, Polish Academy of Sciences, 12 Smetna St. 31-343 Krakow, Poland
| | - Marcello Leopoldo
- Department of Pharmacy - Drug Sciences, University of Bari, via Orabona 4, 70125 Bari, Italy
| | - Enza Lacivita
- Department of Pharmacy - Drug Sciences, University of Bari, via Orabona 4, 70125 Bari, Italy
| | - Agnieszka Basta-Kaim
- Laboratory of Immunoendocrinology, Department of Experimental Neuroendocrinology, Maj Institute of Pharmacology, Polish Academy of Sciences, 12 Smetna St. 31-343 Krakow, Poland
| |
Collapse
|
50
|
Kim AS, Conte MS. Specialized pro-resolving lipid mediators in cardiovascular disease, diagnosis, and therapy. Adv Drug Deliv Rev 2020; 159:170-179. [PMID: 32697951 PMCID: PMC10980506 DOI: 10.1016/j.addr.2020.07.011] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2020] [Revised: 07/06/2020] [Accepted: 07/16/2020] [Indexed: 12/12/2022]
Abstract
Persistent inflammation is the key aggravator in many cardiovascular diseases, including atherosclerosis, aneurysm, injury/reperfusion, thrombosis, and neointimal hyperplasia following surgical or percutaneous interventions. Resolution is an active process orchestrated by specialized pro-resolving lipid mediators (SPMs) which tamp down acute inflammatory signals, promote healing and facilitate a return to homeostasis. SPMs are endogenously derived from poly-unsaturated fatty acids, and their biologic activity is mediated via specific G-protein coupled receptor binding. The potency of SPM in regulating the inflammatory response has encouraged investigation into their therapeutic and diagnostic use in cardiovascular pathologies. Herein we describe the translational groundwork which has established the synthesis and interactions of SPM in cardiovascular and hematologic cells, the therapeutic effects of SPM in animal models of cardiovascular disease, and some early technologies that harness and attempt to optimize SPM delivery and "resolution pharmacology". Further studies are required to precisely determine the mechanisms of resolution in the cardiovascular system and to determine the clinical settings in which SPM can be utilized to optimize patient outcomes.
Collapse
Affiliation(s)
- Alexander S Kim
- Division of Vascular and Endovascular Surgery, Cardiovascular Research Institute, University of California, San Francisco, USA
| | - Michael S Conte
- Division of Vascular and Endovascular Surgery, Cardiovascular Research Institute, University of California, San Francisco, USA.
| |
Collapse
|