1
|
Peng C, Vecchio EA, Nguyen ATN, De Seram M, Tang R, Keov P, Woodman OL, Chen YC, Baell J, May LT, Zhao P, Ritchie RH, Qin CX. Biased receptor signalling and intracellular trafficking profiles of structurally distinct formylpeptide receptor 2 agonists. Br J Pharmacol 2024; 181:4677-4692. [PMID: 39154373 DOI: 10.1111/bph.17310] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2023] [Revised: 05/24/2024] [Accepted: 06/13/2024] [Indexed: 08/20/2024] Open
Abstract
BACKGROUND There is increasing interest in developing FPR2 agonists (compound 43, ACT-389949 and BMS-986235) as potential pro-resolving therapeutics, with ACT-389949 and BMS-986235 having entered phase I clinical development. FPR2 activation leads to diverse downstream outputs. ACT-389949 was observed to cause rapid tachyphylaxis, while BMS-986235 and compound 43 induced cardioprotective effects in preclinical models. We aim to characterise the differences in ligand-receptor engagement and downstream signalling and trafficking bias profile. EXPERIMENTAL APPROACH Concentration-response curves to G protein dissociation, β-arrestin recruitment, receptor trafficking and second messenger signalling were generated using FPR2 ligands (BMS-986235, ACT-389949, compound 43 and WKYMVm), in HEK293A cells. Log(τ/KA) was obtained from the operational model for bias analysis using WKYMVm as a reference ligand. Docking of FPR2 ligands into the active FPR2 cryoEM structure (PDBID: 7T6S) was performed using ICM pro software. KEY RESULTS Bias analysis revealed that WKYMVm and ACT-389949 shared a very similar bias profile. In comparison, BMS-986235 and compound 43 displayed approximately 5- to 50-fold bias away from β-arrestin recruitment and trafficking pathways, while being 35- to 60-fold biased towards cAMP inhibition and pERK1/2. Molecular docking predicted key amino acid interactions at the FPR2 shared between WKYMVm and ACT-389949, but not with BMS-986235 and compound 43. CONCLUSION AND IMPLICATIONS In vitro characterisation demonstrated that WKYMVm and ACT-389949 differ from BMS-986235 and compound 43 in their signalling and protein coupling profile. This observation may be explained by differences in the ligand-receptor interactions. In vitro characterisation provided significant insights into identifying the desired bias profile for FPR2-based pharmacotherapy.
Collapse
Affiliation(s)
- Cheng Peng
- Drug Discovery Biology, Monash Institute of Pharmaceutical Sciences, Monash University, Melbourne, Victoria, Australia
| | - Elizabeth A Vecchio
- Drug Discovery Biology, Monash Institute of Pharmaceutical Sciences, Monash University, Melbourne, Victoria, Australia
| | - Anh T N Nguyen
- Drug Discovery Biology, Monash Institute of Pharmaceutical Sciences, Monash University, Melbourne, Victoria, Australia
| | - Mia De Seram
- Drug Discovery Biology, Monash Institute of Pharmaceutical Sciences, Monash University, Melbourne, Victoria, Australia
| | - Ruby Tang
- Drug Discovery Biology, Monash Institute of Pharmaceutical Sciences, Monash University, Melbourne, Victoria, Australia
| | - Peter Keov
- Drug Discovery Biology, Monash Institute of Pharmaceutical Sciences, Monash University, Melbourne, Victoria, Australia
| | - Owen L Woodman
- Drug Discovery Biology, Monash Institute of Pharmaceutical Sciences, Monash University, Melbourne, Victoria, Australia
| | - Yung-Chih Chen
- Monash Victorian Heart Institute, Blackburn Road Clayton, Monash University, Melbourne, Victoria, Australia
| | - Jonathan Baell
- Medicinal Chemistry, Monash Institute of Pharmaceutical Sciences, Monash University, Melbourne, Vitoria, Australia
| | - Lauren T May
- Drug Discovery Biology, Monash Institute of Pharmaceutical Sciences, Monash University, Melbourne, Victoria, Australia
| | - Peishen Zhao
- Drug Discovery Biology, Monash Institute of Pharmaceutical Sciences, Monash University, Melbourne, Victoria, Australia
| | - Rebecca H Ritchie
- Drug Discovery Biology, Monash Institute of Pharmaceutical Sciences, Monash University, Melbourne, Victoria, Australia
| | - Cheng Xue Qin
- Drug Discovery Biology, Monash Institute of Pharmaceutical Sciences, Monash University, Melbourne, Victoria, Australia
| |
Collapse
|
2
|
Yao Q, Min K, Zhao M, Chen X, Weng D, Zhou Y. Identification and validation of hub differential genes in pulmonary sarcoidosis. Front Immunol 2024; 15:1466029. [PMID: 39364409 PMCID: PMC11446782 DOI: 10.3389/fimmu.2024.1466029] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2024] [Accepted: 09/05/2024] [Indexed: 10/05/2024] Open
Abstract
A total of 138 cDEGs were screened from mediastinal lymph nodes and peripheral whole blood. Among them, 6 hub cDEGs including CTSS, CYBB, FPR2, MNDA, TLR1 and TLR8 with elevated degree and betweenness levels were illustrated in protein-protein interaction network. In comparison to healthy controls, CTSS (1.61 vs. 1.05), CYBB (1.68 vs. 1.07), FPR2 (2.77 vs. 0.96), MNDA (2.14 vs. 1.23), TLR1 (1.56 vs. 1.09), and TLR8 (2.14 vs. 0.98) displayed notably elevated expression levels within pulmonary sarcoidosis PBMC samples (P < 0.0001 for FPR2 and P < 0.05 for others), echoing with prior mRNA microarray findings. The most significant functional pathways were immune response, inflammatory response, plasma membrane and extracellular exosome, with 6 hub cDEGs distributing along these pathways. CTSS, CYBB, FPR2, MNDA, TLR1, and TLR8 could be conducive to improving the diagnostic process and understanding the underlying mechanisms of pulmonary sarcoidosis.
Collapse
Affiliation(s)
- Qian Yao
- Department of Respiratory Medicine, Shanghai Pulmonary Hospital, Tongji University School of Medicine, Shanghai, China
- Clinical Research Center, Shanghai Pulmonary Hospital, Tongji University School of Medicine, Shanghai, China
| | - Keting Min
- Clinical Research Center, Shanghai Pulmonary Hospital, Tongji University School of Medicine, Shanghai, China
| | - Mengmeng Zhao
- Department of Respiratory Medicine, Shanghai Pulmonary Hospital, Tongji University School of Medicine, Shanghai, China
| | - Xianqiu Chen
- Department of Respiratory Medicine, Shanghai Pulmonary Hospital, Tongji University School of Medicine, Shanghai, China
| | - Dong Weng
- Department of Respiratory Medicine, Shanghai Pulmonary Hospital, Tongji University School of Medicine, Shanghai, China
| | - Ying Zhou
- Department of Respiratory Medicine, Shanghai Pulmonary Hospital, Tongji University School of Medicine, Shanghai, China
- Clinical Research Center, Shanghai Pulmonary Hospital, Tongji University School of Medicine, Shanghai, China
| |
Collapse
|
3
|
AlOmar S, Mitchell JL, AlZahrani E. Lipoxin A4 analogue, BML-111, reduces platelet activation and protects from thrombosis. Thromb J 2024; 22:39. [PMID: 38654303 PMCID: PMC11036777 DOI: 10.1186/s12959-024-00606-7] [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: 12/24/2023] [Accepted: 04/10/2024] [Indexed: 04/25/2024] Open
Abstract
Formyl peptide receptors (FPRs) are members of seven transmembrane G protein-coupled receptors superfamily that exhibit different responses based on the nature of stimulating ligand type. FPRs have been shown to be present in platelets and regulate their function. However, the effect of formyl peptide receptor 2 (FPR2/ALX) lipid ligands on platelets has not yet been addressed. Hence, we sought to study the role of FPR2/ALX ligand and lipoxin A4 lipid analogue, BML-111, in the modulation of platelet function and thrombus formation. Immunofluorescence microscopy showed subcellular distribution and peripheral mobilisation of FPR2/ALX in stimulated platelets. This variation in distribution was further confirmed using flow cytometry. BML-111 inhibited a range of platelet activities in a dose-dependent manner in response to several platelet agonists. This included aggregation, fibrinogen binding to integrin αIIbβ3, α-granule secretion, dense granule secretion, Ca2 + mobilisation and integrin αIIbβ3-mediated outside-in signaling. The selectivity of BML-111 for FPR2/ALX was confirmed using FPR2/ALX deficient mice in flow cytometry assays. In vitro thrombus formation was also inhibited by various concentrations of BML-111. Moreover, the levels of vasodilator stimulated phosphorylation (VASP-S157) increased significantly after BML-111 treatment in resting and stimulated platelets via protein kinase A (PKA) independently of cyclic adenosine monophosphate (cAMP) signaling. Together, our findings demonstrate the significance of BML-111 as a modulator of platelet function via FPR2/ALX and unravel the thrombo-protective potentials of BML-111 induced signaling against thrombo-inflammatory diseases.
Collapse
Affiliation(s)
- Shatha AlOmar
- Department of Clinical Laboratory Sciences, King Saud University, Prince Turki Ibn Abdulaziz Al Awwal Rd, 12371, Riyadh, Saudi Arabia.
- School of Pharmacy, University of Reading, Reading, UK.
| | | | | |
Collapse
|
4
|
Chen K, Gong W, Huang J, Yoshimura T, Ming Wang J. Developmental and homeostatic signaling transmitted by the G-protein coupled receptor FPR2. Int Immunopharmacol 2023; 118:110052. [PMID: 37003185 PMCID: PMC10149111 DOI: 10.1016/j.intimp.2023.110052] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2022] [Revised: 03/02/2023] [Accepted: 03/15/2023] [Indexed: 04/03/2023]
Abstract
Formyl peptide receptor 2 (FPR2) and its mouse counterpart Fpr2 are the members of the G protein-coupled receptor (GPCR) family. FPR2 is the only member of the FPRs that interacts with ligands from different sources. FPR2 is expressed in myeloid cells as well as epithelial cells, endothelial cells, neurons, and hepatocytes. During the past years, some unusual properties of FPR2 have attracted intense attention because FPR2 appears to possess dual functions by activating or inhibiting intracellular signal pathways based on the nature, concentration of the ligands, and the temporal and spatial settings of the microenvironment in vivo, the cell types it interacts with. Therefore, FPR2 controls an abundant array of developmental and homeostatic signaling cascades, in addition to its "classical" capacity to mediate the migration of hematopoietic and non-hematopoietic cells including malignant cells. In this review, we summarize recent development in FPR2 research, particularly in its role in diseases, therefore helping to establish FPR2 as a potential target for therapeutic intervention.
Collapse
Affiliation(s)
- Keqiang Chen
- Laboratory of Cancer Innovation, Center for Cancer Research, National Cancer Institute at Frederick, Frederick, MD, USA.
| | - Wanghua Gong
- Basic Research Program, Leidos Biomedical Research, Inc., Frederick, MD, USA
| | - Jiaqiang Huang
- Laboratory of Cancer Innovation, Center for Cancer Research, National Cancer Institute at Frederick, Frederick, MD, USA; College of Life Sciences, Beijing Jiaotong University, Beijing, PR China
| | - Teizo Yoshimura
- Laboratory of Cancer Innovation, Center for Cancer Research, National Cancer Institute at Frederick, Frederick, MD, USA
| | - Ji Ming Wang
- Laboratory of Cancer Innovation, Center for Cancer Research, National Cancer Institute at Frederick, Frederick, MD, USA
| |
Collapse
|
5
|
Qin CX, Norling LV, Vecchio EA, Brennan EP, May LT, Wootten D, Godson C, Perretti M, Ritchie RH. Formylpeptide receptor 2: Nomenclature, structure, signalling and translational perspectives: IUPHAR review 35. Br J Pharmacol 2022; 179:4617-4639. [PMID: 35797341 PMCID: PMC9545948 DOI: 10.1111/bph.15919] [Citation(s) in RCA: 32] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2021] [Revised: 05/22/2022] [Accepted: 06/09/2022] [Indexed: 12/26/2022] Open
Abstract
We discuss the fascinating pharmacology of formylpeptide receptor 2 (FPR2; often referred to as FPR2/ALX since it binds lipoxin A4 ). Initially identified as a low-affinity 'relative' of FPR1, FPR2 presents complex and diverse biology. For instance, it is activated by several classes of agonists (from peptides to proteins and lipid mediators) and displays diverse expression patterns on myeloid cells as well as epithelial cells and endothelial cells, to name a few. Over the last decade, the pharmacology of FPR2 has progressed from being considered a weak chemotactic receptor to a master-regulator of the resolution of inflammation, the second phase of the acute inflammatory response. We propose that exploitation of the biology of FPR2 offers innovative ways to rectify chronic inflammatory states and represents a viable avenue to develop novel therapies. Recent elucidation of FPR2 structure will facilitate development of the anti-inflammatory and pro-resolving drugs of next decade.
Collapse
Affiliation(s)
- Cheng Xue Qin
- Drug Discovery Biology, Monash Institute of Pharmaceutical SciencesMonash UniversityMelbourneVictoriaAustralia
| | - Lucy V. Norling
- William Harvey Research Institute, Barts and the London School of MedicineQueen Mary University of LondonLondonUK
| | - Elizabeth A. Vecchio
- Drug Discovery Biology, Monash Institute of Pharmaceutical SciencesMonash UniversityMelbourneVictoriaAustralia
| | - Eoin P. Brennan
- Diabetes Complications Research Centre, Conway Institute and School of MedicineUniversity College DublinDublinIreland
| | - Lauren T. May
- Drug Discovery Biology, Monash Institute of Pharmaceutical SciencesMonash UniversityMelbourneVictoriaAustralia
| | - Denise Wootten
- Drug Discovery Biology, Monash Institute of Pharmaceutical SciencesMonash UniversityMelbourneVictoriaAustralia
| | - Catherine Godson
- Diabetes Complications Research Centre, Conway Institute and School of MedicineUniversity College DublinDublinIreland
| | - Mauro Perretti
- William Harvey Research Institute, Barts and the London School of MedicineQueen Mary University of LondonLondonUK
| | - Rebecca H. Ritchie
- Drug Discovery Biology, Monash Institute of Pharmaceutical SciencesMonash UniversityMelbourneVictoriaAustralia
| |
Collapse
|
6
|
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
|
7
|
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
|
8
|
Andrews D, Godson C. Lipoxins and synthetic lipoxin mimetics: Therapeutic potential in renal diseases. Biochim Biophys Acta Mol Cell Biol Lipids 2021; 1866:158940. [PMID: 33839296 DOI: 10.1016/j.bbalip.2021.158940] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2021] [Revised: 03/29/2021] [Accepted: 03/31/2021] [Indexed: 12/26/2022]
Abstract
Inflammation and its timely resolution are critical to ensuring effective host defence and appropriate tissue repair after injury. Unresolved inflammation typifies many renal pathologies. The key drivers of the inflammatory response are well defined and targeted by conventional anti-inflammatory therapeutics. However, these are associated with undesirable side effects including immune suppression. More recently, there is growing appreciation that specialized lipid mediators [SPMs] including lipoxins promote the resolution of inflammation and endogenous repair mechanisms without compromising host defence. We discuss the pro-resolving bioactions of lipoxins and recent work that aims to harness their therapeutic potential in the context of kidney disease.
Collapse
Affiliation(s)
- Darrell Andrews
- Diabetes Complications Research Centre, Conway Institute and School of Medicine, University College Dublin, Dublin, Ireland
| | - Catherine Godson
- Diabetes Complications Research Centre, Conway Institute and School of Medicine, University College Dublin, Dublin, Ireland.
| |
Collapse
|
9
|
Vergelli C, Khlebnikov AI, Crocetti L, Guerrini G, Cantini N, Kirpotina LN, Schepetkin IA, Cilibrizzi A, Quinn MT, Rossi P, Paoli P, Giovannoni MP. Synthesis, biological evaluation, molecular modeling, and structural analysis of new pyrazole and pyrazolone derivatives as N-formyl peptide receptors agonists. Chem Biol Drug Des 2021; 98:582-603. [PMID: 34148303 PMCID: PMC8446315 DOI: 10.1111/cbdd.13913] [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: 02/03/2021] [Revised: 04/30/2021] [Accepted: 06/06/2021] [Indexed: 11/27/2022]
Abstract
N‐formyl peptide receptors (FPR1, FPR2, and FPR3) play key roles in the regulation of inflammatory processes, and recently, it was demonstrated that FPR1 and FPR2 have a dual role in the progression/suppression of some cancers. Therefore, FPRs represent an important therapeutic target for the treatment of both cancer and inflammatory diseases. Previously, we identified selective or mixed FPR agonists with pyridazinone or pyridinone scaffolds showing a common 4‐(bromophenyl)acetamide fragment, which was essential for activity. We report here new pyrazole and pyrazolone derivatives as restricted analogues of the above 6‐membered compounds, all exhibiting the same 4‐bromophenylacetamide side chain. Most new products had low or absent FPR agonist activity, suggesting that the pyrazole nucleus was not appropriate for FPR agonists. This hypothesis was confirmed by molecular modeling studies, which highlighted that the five‐membered scaffold was responsible for a worse arrangement of the molecules in the receptor binding site.
Collapse
Affiliation(s)
- Claudia Vergelli
- Neurofarba, Pharmaceutical and Nutraceutical Section, University of Florence, Sesto Fiorentino, Italy
| | | | - Letizia Crocetti
- Neurofarba, Pharmaceutical and Nutraceutical Section, University of Florence, Sesto Fiorentino, Italy
| | - Gabriella Guerrini
- Neurofarba, Pharmaceutical and Nutraceutical Section, University of Florence, Sesto Fiorentino, Italy
| | - Niccolò Cantini
- Neurofarba, Pharmaceutical and Nutraceutical Section, University of Florence, Sesto Fiorentino, Italy
| | - Liliya N Kirpotina
- Department of Microbiology and Immunology, Montana State University, Bozeman, MT, USA
| | - Igor A Schepetkin
- Department of Microbiology and Immunology, Montana State University, Bozeman, MT, USA
| | | | - Mark T Quinn
- Department of Microbiology and Immunology, Montana State University, Bozeman, MT, USA
| | - Patrizia Rossi
- Department of Industrial Engineering, University of Florence, Florence, Italy
| | - Paola Paoli
- Department of Industrial Engineering, University of Florence, Florence, Italy
| | - Maria Paola Giovannoni
- Neurofarba, Pharmaceutical and Nutraceutical Section, University of Florence, Sesto Fiorentino, Italy
| |
Collapse
|
10
|
Formyl peptide receptor 2, as an important target for ligands triggering the inflammatory response regulation: a link to brain pathology. Pharmacol Rep 2021; 73:1004-1019. [PMID: 34105114 PMCID: PMC8413167 DOI: 10.1007/s43440-021-00271-x] [Citation(s) in RCA: 33] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2021] [Revised: 04/13/2021] [Accepted: 04/30/2021] [Indexed: 12/28/2022]
Abstract
Formyl peptide receptors (FPRs) belong to the family of seven-transmembrane G protein-coupled receptors. Among them, FPR2 is a low affinity receptor for N-formyl peptides and is considered the most promiscuous member of FPRs. FPR2 is able to recognize a broad variety of endogenous or exogenous ligands, ranging from lipid to proteins and peptides, including non-formylated peptides. Due to this property FPR2 has the ability to modulate both pro- and anti-inflammatory response, depending on the nature of the bound agonist and on the different recognition sites of the receptor. Thus, FPR2 takes part not only in the proinflammatory response but also in the resolution of inflammation (RoI) processes. Recent data have indicated that the malfunction of RoI may be the background for some central nervous system (CNS) disorders. Therefore, much interest is focused on endogenous molecules called specialized pro-resolving mediators (SPMs), as well as on new synthetic FPR2 agonists, which kick-start the resolution of inflammation (RoI) and modulate its course. Here, we shed some light on the general characteristics of the FPR family in humans and in the experimental animals. Moreover, we present a guide to understanding the "double faced" action of FPR2 activation in the context of immune-related diseases of the CNS.
Collapse
|
11
|
Klooster JPT, Bol-Schoenmakers M, van Summeren K, van Vliet ALW, de Haan CAM, van Kuppeveld FJM, Verkoeijen S, Pieters R. Enterocytes, fibroblasts and myeloid cells synergize in anti-bacterial and anti-viral pathways with IL22 as the central cytokine. Commun Biol 2021; 4:631. [PMID: 34045640 PMCID: PMC8160143 DOI: 10.1038/s42003-021-02176-0] [Citation(s) in RCA: 4] [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: 08/20/2020] [Accepted: 05/03/2021] [Indexed: 12/30/2022] Open
Abstract
IL22 is an important cytokine involved in the intestinal defense mechanisms against microbiome. By using ileum-derived organoids, we show that the expression of anti-microbial peptides (AMPs) and anti-viral peptides (AVPs) can be induced by IL22. In addition, we identified a bacterial and a viral route, both leading to IL22 production by T cells, but via different pathways. Bacterial products, such as LPS, induce enterocyte-secreted SAA1, which triggers the secretion of IL6 in fibroblasts, and subsequently IL22 in T cells. This IL22 induction can then be enhanced by macrophage-derived TNFα in two ways: by enhancing the responsiveness of T cells to IL6 and by increasing the expression of IL6 by fibroblasts. Viral infections of intestinal cells induce IFNβ1 and subsequently IL7. IFNβ1 can induce the expression of IL6 in fibroblasts and the combined activity of IL6 and IL7 can then induce IL22 expression in T cells. We also show that IL22 reduces the expression of viral entry receptors (e.g. ACE2, TMPRSS2, DPP4, CD46 and TNFRSF14), increases the expression of anti-viral proteins (e.g. RSAD2, AOS, ISG20 and Mx1) and, consequently, reduces the viral infection of neighboring cells. Overall, our data indicates that IL22 contributes to the innate responses against both bacteria and viruses.
Collapse
Affiliation(s)
- Jean Paul Ten Klooster
- Research Centre Healthy and Sustainable Living, Innovative Testing in Life Sciences and Chemistry, University of Applied Sciences Utrecht, Utrecht, The Netherlands.
| | - Marianne Bol-Schoenmakers
- Institute for Risk Assessment Sciences, Population Health Sciences Division, Utrecht University, Utrecht, The Netherlands
| | - Kitty van Summeren
- Research Centre Healthy and Sustainable Living, Innovative Testing in Life Sciences and Chemistry, University of Applied Sciences Utrecht, Utrecht, The Netherlands
| | - Arno L W van Vliet
- Virology Section, Infectious Disease and Immunology Division, Department of Biomolecular Health Sciences, Faculty of Veterinary Medicine, Utrecht University, Utrecht, The Netherlands
| | - Cornelis A M de Haan
- Virology Section, Infectious Disease and Immunology Division, Department of Biomolecular Health Sciences, Faculty of Veterinary Medicine, Utrecht University, Utrecht, The Netherlands
| | - Frank J M van Kuppeveld
- Virology Section, Infectious Disease and Immunology Division, Department of Biomolecular Health Sciences, Faculty of Veterinary Medicine, Utrecht University, Utrecht, The Netherlands
| | - Saertje Verkoeijen
- Research Centre Healthy and Sustainable Living, Innovative Testing in Life Sciences and Chemistry, University of Applied Sciences Utrecht, Utrecht, The Netherlands
| | - Raymond Pieters
- Research Centre Healthy and Sustainable Living, Innovative Testing in Life Sciences and Chemistry, University of Applied Sciences Utrecht, Utrecht, The Netherlands
- Institute for Risk Assessment Sciences, Population Health Sciences Division, Utrecht University, Utrecht, The Netherlands
| |
Collapse
|
12
|
Ali M, Yang F, Jansen JA, Walboomers XF. Lipoxin suppresses inflammation via the TLR4/MyD88/NF-κB pathway in periodontal ligament cells. Oral Dis 2019; 26:429-438. [PMID: 31814225 PMCID: PMC7074052 DOI: 10.1111/odi.13250] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2019] [Revised: 11/25/2019] [Accepted: 11/26/2019] [Indexed: 01/16/2023]
Abstract
Objective The objective of the present study was to evaluate the anti‐inflammatory effects of lipoxin A4 (LXA4) for the treatment of periodontitis in an in vitro model. Methods Human PDLCs were challenged with Escherichia coli (E. coli) lipopolysaccharide (LPS) to evoke an inflammatory response. This was done either in monoculture or in coculture with THP‐1, a monocytic cell line. Thereafter, cytokine expression was measured by ELISA, with or without LXA4. In addition, the effects of LXA4 were analyzed on the TLR‐MyD88‐NF‐κB (TMN)‐mediated intracellular signal pathway using immunocytochemistry. Results In response to LPS, the level of the pro‐inflammatory cytokine tumor necrosis factor alpha increased, whereas the anti‐inflammatory cytokine interleukin‐4 decreased significantly (p < .05). These effects were consistently reversed when LPS‐challenged PDLCs were also treated with LXA4. The results in the coculture system were comparable to the monoculture. Immunohistochemistry and quantitative assessment confirmed the importance of the TMN signal pathway in these processes. Conclusion These results corroborate earlier findings that PDLCs play an important role in inflammation. Moreover, LXA4 might offer new approaches for the therapeutic treatment of periodontal disease.
Collapse
Affiliation(s)
- Muhanad Ali
- Department of Dentistry - Biomaterials, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Fang Yang
- Department of Dentistry - Biomaterials, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, Nijmegen, The Netherlands
| | - John A Jansen
- Department of Dentistry - Biomaterials, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, Nijmegen, The Netherlands
| | - X Frank Walboomers
- Department of Dentistry - Biomaterials, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, Nijmegen, The Netherlands
| |
Collapse
|
13
|
Raabe CA, Gröper J, Rescher U. Biased perspectives on formyl peptide receptors. BIOCHIMICA ET BIOPHYSICA ACTA-MOLECULAR CELL RESEARCH 2019; 1866:305-316. [DOI: 10.1016/j.bbamcr.2018.11.015] [Citation(s) in RCA: 34] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/18/2018] [Revised: 11/30/2018] [Accepted: 11/30/2018] [Indexed: 02/07/2023]
|
14
|
|
15
|
The ectoenzyme-side of matrix metalloproteinases (MMPs) makes inflammation by serum amyloid A (SAA) and chemokines go round. Immunol Lett 2018; 205:1-8. [PMID: 29870759 DOI: 10.1016/j.imlet.2018.06.001] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2018] [Revised: 05/16/2018] [Accepted: 06/01/2018] [Indexed: 12/18/2022]
Abstract
During an inflammatory response, a large number of distinct mediators appears in the affected tissues or in the blood circulation. These include acute phase proteins such as serum amyloid A (SAA), cytokines and chemokines and proteolytic enzymes. Although these molecules are generated within a cascade sequence in specific body compartments allowing for independent action, their co-appearance in space and time during acute or chronic inflammation points toward important mutual interactions. Pathogen-associated molecular patterns lead to fast induction of the pro-inflammatory endogenous pyrogens, which are evoking the acute phase response. Interleukin-1, tumor necrosis factor-α and interferons simultaneously trigger different cell types, including leukocytes, endothelial cells and fibroblasts for tissue-specific or systemic production of chemokines and matrix metalloproteinases (MMPs). In addition, SAA induces chemokines and both stimulate secretion of MMPs from multiple cell types. As a consequence, these mediators may cooperate to enhance the inflammatory response. Indeed, SAA synergizes with chemokines to increase chemoattraction of monocytes and granulocytes. On the other hand, MMPs post-translationally modify chemokines and SAA to reduce their activity. Indeed, MMPs internally cleave SAA with loss of its cytokine-inducing and direct chemotactic potential whilst retaining its capacity to synergize with chemokines in leukocyte migration. Finally, MMPs truncate chemokines at their NH2- or COOH-terminal end, resulting in reduced or enhanced chemotactic activity. Therefore, the complex interactions between chemokines, SAA and MMPs either maintain or dampen the inflammatory response.
Collapse
|
16
|
Sodin-Semrl S, Antico G, Mikus R, Lakota K, Varga J, Fiore S. Lipoxin A4 and Serum Amyloid a Differentially Modulate Phospholipase D in Human Fibroblast-Like Synoviocytes. EUR J INFLAMM 2017. [DOI: 10.1177/1721727x0900700102] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022] Open
Abstract
Lipoxin A4 (LXA4) and scrum amyloid A (SAA) are endogenous negative and positive modulators of inflammation, respectively. Both molecules bind the shared lipoxin A4 receptor (ALX) and elicit opposing effects on the production of inflammatory cytokines and matrix metalloproteinases. The aim of these studies is to examine the divergence of the intracellular signaling pathways triggered by lipid LXA4 (1 nM) and protein SAA (200 nM) ligands of ALX. Phospholipase D (PLD) is a phosphohydrolase enzyme that catalyzes the generation of phosphatidic acid (PA) from membrane phospholipids. Our results showed that in fibroblast-like synoviocytes, activation of PLD occurred only in response to LXA4, and not SAA. PA (30 μM) mimicked LXA4 and demonstrated inhibition of IL-8 production induced by SAA or interleukin-1β. In sharp contrast to LXA4, SAA confirmed the stimulation of IL-8 release as determined previously. Taken together, these findings suggest that two physiologic ligands sharing the common ALX receptor, LXA4 and SAA, differentially regulate the level of PLD activation and differentially modulate IL-8. These results may have important implications for understanding the regulation of inflammatory responses under physiologic and pathological conditions.
Collapse
Affiliation(s)
- S. Sodin-Semrl
- University Medical Centre, Department of Rheumatology, Ljubljana, Slovenia
| | - G. Antico
- University Medical Centre, Department of Rheumatology, Ljubljana, Slovenia
- Northwestern University Feinberg School of Medicine, Department of Pathology, Chicago, IL, USA
| | - R. Mikus
- University Medical Centre, Department of Rheumatology, Ljubljana, Slovenia
- University of Illinois at Chicago, Department of Medicine, Section of Rheumatology, IL, USA
| | - K. Lakota
- University Medical Centre, Department of Rheumatology, Ljubljana, Slovenia
| | - J. Varga
- University Medical Centre, Department of Rheumatology, Ljubljana, Slovenia
- Northwestern University Feinberg School of Medicine, Division of Rheumatology, Chicago, IL, USA; Present address
| | - S. Fiore
- University Medical Centre, Department of Rheumatology, Ljubljana, Slovenia
- Genentech Inc., Clinical Science Development ITGR, 1 DNA Way, M/S 211, S. San Francisco, CA, USA
| |
Collapse
|
17
|
Xie X, Yang M, Ding Y, Yu L, Chen J. Formyl peptide receptor 2 expression predicts poor prognosis and promotes invasion and metastasis in epithelial ovarian cancer. Oncol Rep 2017; 38:3297-3308. [PMID: 29039544 PMCID: PMC5783575 DOI: 10.3892/or.2017.6034] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2017] [Accepted: 08/31/2017] [Indexed: 12/29/2022] Open
Abstract
Formyl peptide receptor 2 (FPR2) has been identified as a member of the G protein-coupled chemoattractant receptor (GPCR) family and has been implicated as playing a role in both inflammation and cancer development. Epithelial ovarian cancer (EOC) has been suggested to be correlated with both infectious and non-infectious inflammation. To date, the role of FPR2 in EOC remains poorly understood and controversial. In the present study, we aimed to investigate the potential of FPR2 in regulating EOC. We performed immunohistochemistry and RT-qPCR to analyzed expression of FPR2 in EOC tissues and the correlation between FPR2 and EOC clinicopathological characteristics as well as prognosis were also analyzed. To test the role of FPR2 in EOC cell migration, we established FPR2-knockdown SKOV3 cells and performed wound-healing, Transwell and angiogenesis assays to detect the metastatic potential of these EOC cells. Our studies found that FPR2 was overexpressed in EOC tissues and was positively correlated with EOC clinicopathological characteristics including the International Federation of Gynecology and Obstetrics (FIGO) stage, histological grade and ovarian cancer type. Survival analyses suggested that FPR2 overexpression indicated the poorer prognosis of EOC patients and FPR2 may act as an independent risk factor for EOC prognosis. FPR2 knockdown decreased the migration potential of the ovarian cancer cells. Moreover, serum amyloid A (SAA) may stimulate the migration of SKOV3 cells through FPR2. The present study suggested that FPR2 promoted the invasion and metastasis of EOC and it could be a prognostic marker for EOC.
Collapse
Affiliation(s)
- Xiaohui Xie
- Department of Obstetrics and Gynecology, Second Xiangya Hospital of Central South University, Changsha, Hunan 410011, P.R. China
| | - Mengyuan Yang
- Department of Obstetrics and Gynecology, Second Xiangya Hospital of Central South University, Changsha, Hunan 410011, P.R. China
| | - Yiling Ding
- Department of Obstetrics and Gynecology, Second Xiangya Hospital of Central South University, Changsha, Hunan 410011, P.R. China
| | - Ling Yu
- Department of Obstetrics and Gynecology, Second Xiangya Hospital of Central South University, Changsha, Hunan 410011, P.R. China
| | - Jianlin Chen
- Department of Obstetrics and Gynecology, Second Xiangya Hospital of Central South University, Changsha, Hunan 410011, P.R. China
| |
Collapse
|
18
|
He HQ, Ye RD. The Formyl Peptide Receptors: Diversity of Ligands and Mechanism for Recognition. Molecules 2017; 22:E455. [PMID: 28335409 PMCID: PMC6155412 DOI: 10.3390/molecules22030455] [Citation(s) in RCA: 192] [Impact Index Per Article: 24.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2017] [Accepted: 03/09/2017] [Indexed: 12/21/2022] Open
Abstract
The formyl peptide receptors (FPRs) are G protein-coupled receptors that transduce chemotactic signals in phagocytes and mediate host-defense as well as inflammatory responses including cell adhesion, directed migration, granule release and superoxide production. In recent years, the cellular distribution and biological functions of FPRs have expanded to include additional roles in homeostasis of organ functions and modulation of inflammation. In a prototype, FPRs recognize peptides containing N-formylated methionine such as those produced in bacteria and mitochondria, thereby serving as pattern recognition receptors. The repertoire of FPR ligands, however, has expanded rapidly to include not only N-formyl peptides from microbes but also non-formyl peptides of microbial and host origins, synthetic small molecules and an eicosanoid. How these chemically diverse ligands are recognized by the three human FPRs (FPR1, FPR2 and FPR3) and their murine equivalents is largely unclear. In the absence of crystal structures for the FPRs, site-directed mutagenesis, computer-aided ligand docking and structural simulation have led to the identification of amino acids within FPR1 and FPR2 that interact with several formyl peptides. This review article summarizes the progress made in the understanding of FPR ligand diversity as well as ligand recognition mechanisms used by these receptors.
Collapse
Affiliation(s)
- Hui-Qiong He
- School of Pharmacy, Shanghai Jiao Tong University, Shanghai 200240, China.
- Institute of Chinese Medical Sciences, University of Macau, Macau SAR 999078, China.
| | - Richard D Ye
- Institute of Chinese Medical Sciences, University of Macau, Macau SAR 999078, China.
| |
Collapse
|
19
|
De Buck M, Gouwy M, Wang JM, Van Snick J, Opdenakker G, Struyf S, Van Damme J. Structure and Expression of Different Serum Amyloid A (SAA) Variants and their Concentration-Dependent Functions During Host Insults. Curr Med Chem 2017; 23:1725-55. [PMID: 27087246 PMCID: PMC5405626 DOI: 10.2174/0929867323666160418114600] [Citation(s) in RCA: 156] [Impact Index Per Article: 19.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2016] [Revised: 03/31/2016] [Accepted: 04/15/2016] [Indexed: 12/23/2022]
Abstract
Serum amyloid A (SAA) is, like C-reactive protein (CRP), an acute phase protein and can be used as a diagnostic, prognostic or therapy follow-up marker for many diseases. Increases in serum levels of SAA are triggered by physical insults to the host, including infection, trauma, inflammatory reactions and cancer. The order of magnitude of increase in SAA levels varies considerably, from a 10- to 100-fold during limited inflammatory events to a 1000-fold increase during severe bacterial infections and acute exacerbations of chronic inflammatory diseases. This broad response range is reflected by SAA gene duplications resulting in a cluster encoding several SAA variants and by multiple biological functions of SAA. SAA variants are single-domain proteins with simple structures and few post-translational modifications. SAA1 and SAA2 are inducible by inflammatory cytokines, whereas SAA4 is constitutively produced. We review here the regulated expression of SAA in normal and transformed cells and compare its serum levels in various disease states. At low concentrations (10-100 ng/ml), early in an inflammatory response, SAA induces chemokines or matrix degrading enzymes via Toll-like receptors and functions as an activator and chemoattractant through a G protein-coupled receptor. When an infectious or inflammatory stimulus persists, the liver continues to produce more SAA (> 1000 ng/ml) to become an antimicrobial agent by functioning as a direct opsonin of bacteria or by interference with virus infection of host cells. Thus, SAA regulates innate and adaptive immunity and this information may help to design better drugs to treat specific diseases.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Jo Van Damme
- University of Leuven, Department of Microbiology and Immunology, Rega Institute for Medical Research, Laboratory of Molecular Immunology, Minderbroedersstraat 10, 3000 Leuven, Belgium.
| |
Collapse
|
20
|
Tsai YF, Yang SC, Hwang TL. Formyl peptide receptor modulators: a patent review and potential applications for inflammatory diseases (2012-2015). Expert Opin Ther Pat 2016; 26:1139-1156. [PMID: 27454150 DOI: 10.1080/13543776.2016.1216546] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
INTRODUCTION The activation of leukocytes and the subsequent immune cascade play an essential role in sterile and infectious inflammation. Dysregulation of these immune responses or excess leukocyte activation can induce tissue damage, organ dysfunction and mortality. Formyl peptide receptors (FPRs) are functionally diverse pattern recognition receptors responsible for recognizing different endogenous damage-associated molecular patterns or exogenous pathogen-associated molecular patterns. FPRs mediate leukocyte activation during inflammation. FPR1 antagonists and FPR2 agonists have demonstrated significant anti-inflammatory effects based on in vitro and in vivo studies. An increasing number of synthesized compounds targeting FPRs, especially potential FPR1 antagonists and FPR2 agonists, have been disclosed in patents. Areas covered: This article summarizes the current pharmacology patents related to FPR family modulators and their therapeutic indications based on a review of patent applications disclosed between 2012 and 2015. Expert opinion: In this review, FPR1 modulators comprise β-1,3-glucan synthase inhibitors containing an FPR ligand moiety, template-fixed peptidomimetics, cyclosporin H, and dipeptide derivatives. FPR2 modulators include phenylurea, bridged spiro[2.4]heptane ester, naphthalene, aminotriazole, polycyclic pyrrolidine-2,5-dione, imidazolidine-2,4-dione, (2-ureidoacetamido)alkyl, amide, oxazolyl-methylether, oxazole, thiazole, and crystalline potassium salt derivatives. These compounds have potential applications for human conditions such as inflammatory lung diseases, ischemia-reperfusion injury, sepsis, inflammatory bowel disease, and wound healing. FPRs are emerging as important targets for treating leukocyte-dominant inflammation.
Collapse
Affiliation(s)
- Yung-Fong Tsai
- a Graduate Institute of Natural Products, School of Traditional Medicine, College of Medicine , Chang Gung University , Taoyuan , Taiwan.,b Graduate Institute of Clinical Medical Sciences, College of Medicine , Chang Gung University , Taoyuan , Taiwan.,c Department of Anesthesiology , Chang Gung Memorial Hospital , Taoyuan , Taiwan
| | - Shun-Chin Yang
- d Department of Anesthesiology , Taipei Veterans General Hospital and National Yang-Ming University , Taipei , Taiwan.,e Division of Natural Products, Graduate Institute of Biomedical Sciences, College of Medicine , Chang Gung University , Taoyuan , Taiwan
| | - Tsong-Long Hwang
- a Graduate Institute of Natural Products, School of Traditional Medicine, College of Medicine , Chang Gung University , Taoyuan , Taiwan.,c Department of Anesthesiology , Chang Gung Memorial Hospital , Taoyuan , Taiwan.,e Division of Natural Products, Graduate Institute of Biomedical Sciences, College of Medicine , Chang Gung University , Taoyuan , Taiwan.,f Chinese Herbal Medicine Research Team, Healthy Aging Research Centre , Chang Gung University , Taoyuan , Taiwan.,g Research Center for Industry of Human Ecology and Graduate Institute of Health Industry Technology , Chang Gung University of Science and Technology , Taoyuan , Taiwan
| |
Collapse
|
21
|
Connolly M, Rooney PR, McGarry T, Maratha AX, McCormick J, Miggin SM, Veale DJ, Fearon U. Acute serum amyloid A is an endogenous TLR2 ligand that mediates inflammatory and angiogenic mechanisms. Ann Rheum Dis 2016; 75:1392-8. [PMID: 26290589 DOI: 10.1136/annrheumdis-2015-207655] [Citation(s) in RCA: 47] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2015] [Accepted: 08/01/2015] [Indexed: 02/04/2023]
Abstract
INTRODUCTION Acute-phase serum amyloid A (A-SAA) has cytokine-like properties and is expressed at sites of inflammation. We examined whether A-SAA-induced pro-inflammatory mechanisms are mediated through Toll-like receptor 2 (TLR2) in rheumatoid arthritis (RA). METHODS The effect of A-SAA on human embryonic kidney (HEK), TLR2 or TLR4 cells was quantified by nuclear factor (NF)-κB luciferase reporter assays. A-SAA-induced RASFC and dHMVEC function were performed in the presence of a specific neutralising anti-TLR2 mAb (OPN301) (1 μg/mL) and matched IgG isotype control Ab (1 μg/mL). Cell surface expression of intracellular adhesion molecule (ICAM)-1, chemokine expression, cell migration, invasion and angiogenesis were assessed by flow cytometry, ELISA, Matrigel invasion chambers and tube formation assays. MyD88 expression was assessed by real-time PCR and western blot. RESULTS A-SAA induced TLR2 activation through induction of NF-κB (p<0.05), but failed to induce NF-κB in HEK-TLR4 cells, confirming specificity for TLR2. A-SAA-induced proliferation, invasion and migration were significantly inhibited in the presence of anti-TLR2 (all p<0.05), with no significant effect observed for tumour necrosis factor-α-induced events. Additionally, A-SAA-induced ICAM-1, interleukin-8, monocyte chemoattractant protein-1, RANTES and GRO-α expression were significantly reduced in the presence of anti-TLR2 (all p<0.05), as was A-SAA induced angiogenesis (p<0.05). Finally, A-SAA induced MyD88 signalling in RASFC and dHMVEC (p<0.05). CONCLUSIONS A-SAA is an endogenous ligand for TLR2, inducing pro-inflammatory effects in RA. Blocking the A-SAA/TLR2 interaction may be a potential therapeutic intervention in RA.
Collapse
Affiliation(s)
- Mary Connolly
- Centre for Arthritis and Rheumatic Diseases, Dublin Academic Medical Centre and Conway Institute of Biomolecular and Biomedical Research, Dublin 4, Ireland
| | - Peter R Rooney
- Centre for Arthritis and Rheumatic Diseases, Dublin Academic Medical Centre and Conway Institute of Biomolecular and Biomedical Research, Dublin 4, Ireland
| | - Trudy McGarry
- Centre for Arthritis and Rheumatic Diseases, Dublin Academic Medical Centre and Conway Institute of Biomolecular and Biomedical Research, Dublin 4, Ireland
| | - Ashwini X Maratha
- Department of Biology, Institute of Immunology, National University of Ireland Maynooth, Maynooth, County Kildare, Ireland
| | - Jennifer McCormick
- Centre for Arthritis and Rheumatic Diseases, Dublin Academic Medical Centre and Conway Institute of Biomolecular and Biomedical Research, Dublin 4, Ireland
| | - Sinead M Miggin
- Department of Biology, Institute of Immunology, National University of Ireland Maynooth, Maynooth, County Kildare, Ireland
| | - Douglas J Veale
- Centre for Arthritis and Rheumatic Diseases, Dublin Academic Medical Centre and Conway Institute of Biomolecular and Biomedical Research, Dublin 4, Ireland
| | - Ursula Fearon
- Centre for Arthritis and Rheumatic Diseases, Dublin Academic Medical Centre and Conway Institute of Biomolecular and Biomedical Research, Dublin 4, Ireland
| |
Collapse
|
22
|
Serum Amyloid A Activation of Inflammatory and Adhesion Molecules in Human Coronary Artery and Umbilical Vein Endothelial Cells. EUR J INFLAMM 2016. [DOI: 10.1177/1721727x0700500203] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022] Open
Abstract
Inflammation is considered to be the driving force leading to atherogenic and atherosclerotic mechanisms. Increased levels of SAA predict the risk of coronary artery disease and even mortality from cardiovascular disease in humans. Recent animal and human studies have indicated that SAA plays a causal role in atherogenesis, although it is largely unclear how this occurs. The objectives of this study are to understand the role of SAA in activating possible atherogenic inflammatory responses in human coronary artery endothelial cells (HCAEC) and to compare them with human umbilical vein endothelial cells (HUVEC). Our hypothesis is that vein and artery endothelial cells have different expression patterns and levels, leading to differential inflammatory responses. HUVEC and HCAEC were grown in order to analyze the effects of SAA on endothelial expression of pro-inflammatory cytokines, such as IL-6, chemokines, such as IL-8, and adhesion molecules (s-ICAM, s-VCAM, E-selectin) by reverse transcription-PCR and ELISAs. We compared the dose responses of SAA between HUVEC and HCAEC. SAA activated both HUVEC and HCAEC pro-inflammatory factors in a dose-dependent manner. In comparison however, HCAEC showed a strikingly greater sensitivity to SAA, with a higher level of expression of all pro-inflammatory markers at much lower concentrations of SAA, and their much greater stimulation at higher SAA concentrations. SAA also generated a dose-dependent positive feedback response on its own mRNA expression in HCAEC as compared to HUVEC. In summary, there are distinct significant differences in the levels of inflammatory markers and adhesion molecules between HUVEC and HCAEC SAA induced dose responses that could potentially account for HCAEC greater susceptibility to inflammation and atherogenesis.
Collapse
|
23
|
Frydas S, Papaioannou N, Papazahariadou M, Hatzistilianou M, Karagouni E, Trakatelli M, Brellou G, Petrarca C, Castellani ML, Conti P, Riccioni G, Patruno A, Grilli A. Inhibition of MCP-1 and MIP-2 Chemokines in Murine Trichinellosis: Effect of the Anti-Inflammatory Compound L-Mimosine. Int J Immunopathol Pharmacol 2016; 18:85-94. [PMID: 15698514 DOI: 10.1177/039463200501800110] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
Abstract
Mimosine, is a plant amino-acid which has been reported to block DNA replication in mammalian cells and to arrest cells reversibly towards the end of the G1 phase or at the beginning of the S phase. In this study, 42 mice were infected with T. spiralis, a nematode parasite, and treated with the anti-inflammatory compound L-mimosine, to determine if any alteration in the chronic inflammatory state occurred, by investigating the host's immunological response. MCP-1, a C-C chemokine and MIP-2, a C-X-C chemokine were tested and measured in the sera of infected animals, after 1, 10, 20, 30, 40, 50 and 60 days postinfection, by ELISA method. The diaphragm/muscle and the masseters of the infected mice, were tested for inflammatory response. We found that MCP-1 was partially inhibited by L-mimosine, while MIP-2 was totally inhibited. Moreover, in sections of the diaphragm and masseters, the infiltration of inflammatory cells such as macrophages, lymphocytes and eosinophils were more intense in untreated animals compared to those treated with L-mimosine. These findings show, that L-mimosine may have an inhibitory effect on MCP-1 and MIP-2 serum levels in Trichinellosis and may influence the recruitment of inflammatory cells and the intensity of the inflammatory reaction in this parasitic disease.
Collapse
Affiliation(s)
- S Frydas
- Parasitology Dept. Veterinary Faculty, Aristotelian University of Thessaloniki, Greece.
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
24
|
Domizio S, Barbante E, Puglielli C, Clementini E, Domizio R, Sabatino GMD, Albanese A, Colosimo C, Sabatino G. Excessively High Magnetic Resonance Signal in Preterm Infants and Neuropsychobehavioural Follow-up at 2 Years. Int J Immunopathol Pharmacol 2016; 18:365-75. [PMID: 15888258 DOI: 10.1177/039463200501800218] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022] Open
Abstract
The diffuse excessive high-signal intensity (DEHSI) findings in the T2 weighted scans of white matter (WM), besides the corresponding low signal in the T1 weighted images, are usually more evident around the periventricular regions. It is not clear whether the DEHSI should be considered as a diffuse WM injury rather than a sign of delayed maturation of the WM. Eighty nine preterm infants at the full-term equivalent age (FEA) were studied using conventional Magnetic Resonance (MR) imaging of the brain. Based on the MR findings, the infants studied were divided into three groups: the control group presenting normal WM, the DEHSI group and the group with other WM lesions. Ten newborns were not included in the statistical analysis because they presented evidence of precedent germinal matrix hemorrhage (GMH-IVH) which cannot be considered as WM lesions. Seventy nine infants were enrolled in a program of neuropsychobehavioural study follow-up until 24 months of age. Each infant was evaluated for those variables which mostly affect the occurrence of neuropsychomotor disability. In the DEHSI infant group, significantly lower mean pH and mean base excess (BE) values were found in comparison to controls, while the mean birth weight (BW) was significantly higher. No significant difference was observed between the mean 1st minute Apgar Score, mean birth gestational age (GA) and assisted ventilation mean duration of controls and DEHSI groups. Finally, no significant difference between the parameters studied was found by comparing the WM lesion infants group to the DEHSI infants one. Our observations, together with follow-up studies, even up to school age, confirm that DEHSI has a clinical significance and cannot be considered as a simple indicator of delayed WM maturation.
Collapse
Affiliation(s)
- S Domizio
- Neonatal Intensive Care Unit, University G. D'Annunzio, Chieti, Italy
| | | | | | | | | | | | | | | | | |
Collapse
|
25
|
Di Giannantonio M, Frydas S, Kempuraj D, Karagouni E, Hatzistilianou M, Conti CM, Boucher W, Papadopoulou N, Donelan J, Cao J, Madhappan B, Boscolo P, Petrarca C, Castellani L, Quartesan L, Doyle R, Ferro FM. Cytokines in Stress. Int J Immunopathol Pharmacol 2016; 18:1-5. [PMID: 15698505 DOI: 10.1177/039463200501800101] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2023] Open
|
26
|
Cavalieri S, Rotoli M, Feliciani C, Amerio P. Expression of the High-Affinity Laminin Receptor (67 kDa) in Normal Human Skin and Appendages. Int J Immunopathol Pharmacol 2016; 18:223-31. [PMID: 15888241 DOI: 10.1177/039463200501800205] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
Abstract
The interaction of cells with extracellular matrix components plays a significant role in the regulation of cell biology. Laminin is a large glycoprotein involved in fundamental interactions between cells and the basement membrane. Several cell surface receptors are responsible for cell-matrix interactions. The 67 kDa high affinity laminin receptor, 67LR, is involved in the adhesion of normal cells to the laminin network and is also associated with the metastatic phenotype of some tumoral cells. We have investigated the expression of laminin and of the 67LR in normal human skin using immunoperoxidase staining. Twenty samples of skin were analyzed. Antibody against laminin reacted in a continuous linear band at the dermal-epidermal junction, as well as basement membranes of hair follicles, sebaceous and eccrine sweat glands, and dermal blood vessels. The epidermis and the follicular epithelium were negative for laminin. The 67LR seemed not to be expressed on the basal surface of basal keratinocytes. The major expression of this receptor may be detected in the upper half of the spinous layer and in the granular layer. The cells of the outer root sheath in hair follicle showed the same immunohistochemical pattern described for epidermis. In sebaceous glands and in eccrine sweat glands the secreting epithelium was positive. Endothelial cells of dermal blood vessels were routinely positive for 67LR. We observed that the expression of the 67LR in normal human skin is mostly located in epidermal areas in which the keratinizing process was particularly advanced.
Collapse
Affiliation(s)
- S Cavalieri
- Department of Dermatology, Catholic University of the Sacred Heart, Rome, Italy
| | | | | | | |
Collapse
|
27
|
De Buck M, Gouwy M, Wang JM, Van Snick J, Proost P, Struyf S, Van Damme J. The cytokine-serum amyloid A-chemokine network. Cytokine Growth Factor Rev 2015; 30:55-69. [PMID: 26794452 DOI: 10.1016/j.cytogfr.2015.12.010] [Citation(s) in RCA: 99] [Impact Index Per Article: 9.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2015] [Revised: 12/22/2015] [Accepted: 12/22/2015] [Indexed: 12/14/2022]
Abstract
Levels of serum amyloid A (SAA), a major acute phase protein in humans, are increased up to 1000-fold upon infection, trauma, cancer or other inflammatory events. However, the exact role of SAA in host defense is yet not fully understood. Several pro- and anti-inflammatory properties have been ascribed to SAA. Here, the regulated production of SAA by cytokines and glucocorticoids is discussed first. Secondly, the cytokine and chemokine inducing capacity of SAA and its receptor usage are reviewed. Thirdly, the direct (via FPR2) and indirect (via TLR2) chemotactic effects of SAA and its synergy with chemokines are unraveled. Altogether, a complex cytokine-SAA-chemokine network is established, in which SAA plays a key role in regulating the inflammatory response.
Collapse
Affiliation(s)
- Mieke De Buck
- KU Leuven, University of Leuven, Department of Microbiology and Immunology, Rega Institute for Medical Research, Laboratory of Molecular Immunology, Minderbroedersstraat 10, 3000 Leuven, Belgium.
| | - Mieke Gouwy
- KU Leuven, University of Leuven, Department of Microbiology and Immunology, Rega Institute for Medical Research, Laboratory of Molecular Immunology, Minderbroedersstraat 10, 3000 Leuven, Belgium.
| | - Ji Ming Wang
- Cancer and Inflammation Program, Center for Cancer Research, National Cancer Institute at Frederick, Frederick, MD 21702, USA.
| | - Jacques Van Snick
- Ludwig Cancer Research, Brussels Branch, Brussels, Belgium; e Duve Institute, Université Catholique de Louvain, Brussels, Belgium.
| | - Paul Proost
- KU Leuven, University of Leuven, Department of Microbiology and Immunology, Rega Institute for Medical Research, Laboratory of Molecular Immunology, Minderbroedersstraat 10, 3000 Leuven, Belgium.
| | - Sofie Struyf
- KU Leuven, University of Leuven, Department of Microbiology and Immunology, Rega Institute for Medical Research, Laboratory of Molecular Immunology, Minderbroedersstraat 10, 3000 Leuven, Belgium.
| | - Jo Van Damme
- KU Leuven, University of Leuven, Department of Microbiology and Immunology, Rega Institute for Medical Research, Laboratory of Molecular Immunology, Minderbroedersstraat 10, 3000 Leuven, Belgium.
| |
Collapse
|
28
|
O'Neill L, Rooney P, Molloy D, Connolly M, McCormick J, McCarthy G, Veale DJ, Murphy CC, Fearon U, Molloy E. Regulation of Inflammation and Angiogenesis in Giant Cell Arteritis by Acute-Phase Serum Amyloid A. Arthritis Rheumatol 2015; 67:2447-56. [DOI: 10.1002/art.39217] [Citation(s) in RCA: 50] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2014] [Accepted: 05/21/2015] [Indexed: 12/11/2022]
Affiliation(s)
- Lorraine O'Neill
- St. Vincent's University Hospital and Dublin Academic Medical Centre; Dublin Ireland
| | - Peadar Rooney
- St. Vincent's University Hospital and Dublin Academic Medical Centre; Dublin Ireland
| | - Danielle Molloy
- St. Vincent's University Hospital and Dublin Academic Medical Centre; Dublin Ireland
| | - Mary Connolly
- St. Vincent's University Hospital and Dublin Academic Medical Centre; Dublin Ireland
| | - Jennifer McCormick
- St. Vincent's University Hospital and Dublin Academic Medical Centre; Dublin Ireland
| | - Geraldine McCarthy
- Mater Misericordiae University Hospital and Dublin Academic Medical Centre; Dublin Ireland
| | - Douglas J. Veale
- St. Vincent's University Hospital and Dublin Academic Medical Centre; Dublin Ireland
| | - Conor C. Murphy
- Royal College of Surgeons of Ireland and Royal Victoria Eye and Ear Hospital; Dublin Ireland
| | - Ursula Fearon
- St. Vincent's University Hospital and Dublin Academic Medical Centre; Dublin Ireland
| | - Eamonn Molloy
- St. Vincent's University Hospital and Dublin Academic Medical Centre; Dublin Ireland
| |
Collapse
|
29
|
Ye RD, Sun L. Emerging functions of serum amyloid A in inflammation. J Leukoc Biol 2015; 98:923-9. [PMID: 26130702 DOI: 10.1189/jlb.3vmr0315-080r] [Citation(s) in RCA: 215] [Impact Index Per Article: 21.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2015] [Accepted: 06/02/2015] [Indexed: 12/12/2022] Open
Abstract
SAA is a major acute-phase protein produced in large quantity during APR. The rise of SAA concentration in blood circulation during APR has been a clinical marker for active inflammation. In the past decade, research has been conducted to determine whether SAA plays an active role during inflammation and if so, how it influences the course of inflammation. These efforts have led to the discovery of cytokine-like activities of rhSAA, which is commercially available and widely used in most of the published studies. SAA activates multiple receptors, including the FPR2, the TLRs TLR2 and TLR4, the scavenger receptor SR-BI, and the ATP receptor P2X7. More recent studies have shown that SAA not only activates transcription factors, such as NF-κB, but also plays a role in epigenetic regulation through a MyD88-IRF4-Jmjd3 pathway. It is postulated that the activation of these pathways leads to induced expression of proinflammatory factors and a subset of proteins expressed by the M2 macrophages. These functional properties set SAA apart from well-characterized inflammatory factors, such as LPS and TNF-α, suggesting that it may play a homeostatic role during the course of inflammation. Ongoing and future studies are directed to addressing unresolved issues, including the difference between rSAA and native SAA isoforms and the exact functions of SAA in physiologic and pathologic settings.
Collapse
Affiliation(s)
- Richard D Ye
- *School of Pharmacy, Shanghai Jiao Tong University, Shanghai, China; and Department of Pharmacology, University of Illinois at Chicago, Illinois, USA
| | - Lei Sun
- *School of Pharmacy, Shanghai Jiao Tong University, Shanghai, China; and Department of Pharmacology, University of Illinois at Chicago, Illinois, USA
| |
Collapse
|
30
|
Lakota K, Carns M, Podlusky S, Mrak-Poljsak K, Hinchcliff M, Lee J, Tomsic M, Sodin-Semrl S, Varga J. Serum amyloid A is a marker for pulmonary involvement in systemic sclerosis. PLoS One 2015; 10:e0110820. [PMID: 25629975 PMCID: PMC4321755 DOI: 10.1371/journal.pone.0110820] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2014] [Accepted: 09/21/2014] [Indexed: 01/19/2023] Open
Abstract
Inflammation in systemic sclerosis (SSc) is a prominent, but incompletely characterized feature in early stages of the disease. The goal of these studies was to determine the circulating levels, clinical correlates and biological effects of the acute phase protein serum amyloid A (SAA), a marker of inflammation, in patients with SSc. Circulating levels of SAA were determined by multiplex assays in serum from 129 SSc patients and 98 healthy controls. Correlations between SAA levels and clinical and laboratory features of disease were analyzed. The effects of SAA on human pulmonary fibroblasts were studied ex vivo. Elevated levels of SAA were found in 25% of SSc patients, with the highest levels in those with early-stage disease and diffuse cutaneous involvement. Significant negative correlations of SAA were found with forced vital capacity and diffusion capacity for carbon monoxide. Patients with elevated SAA had greater dyspnea and more frequent interstitial lung disease, and had worse scores on patient-reported outcome measures. Incubation with recombinant SAA induced dose-dependent stimulation of IL-6 and IL-8 in normal lung fibroblasts in culture. Serum levels of the inflammatory marker SAA are elevated in patients with early diffuse cutaneous SSc, and correlate with pulmonary involvement. In lung fibroblasts, SAA acts as a direct stimulus for increased cytokine production. These findings suggest that systemic inflammation in SSc may be linked to lung involvement and SAA could serve as a potential biomarker for this complication.
Collapse
Affiliation(s)
- Katja Lakota
- Department of Rheumatology, University Medical Centre Ljubljana, Ljubljana,
Slovenia
- Division of Rheumatology, Feinberg School of Medicine, Northwestern
University, Chicago, United States of America
| | - Mary Carns
- Division of Rheumatology, Feinberg School of Medicine, Northwestern
University, Chicago, United States of America
| | - Sofia Podlusky
- Division of Rheumatology, Feinberg School of Medicine, Northwestern
University, Chicago, United States of America
| | - Katjusa Mrak-Poljsak
- Department of Rheumatology, University Medical Centre Ljubljana, Ljubljana,
Slovenia
| | - Monique Hinchcliff
- Division of Rheumatology, Feinberg School of Medicine, Northwestern
University, Chicago, United States of America
| | - Jungwha Lee
- Department of Preventive Medicine, Feinberg School of Medicine, Northwestern
University, Chicago, United States of America
| | - Matija Tomsic
- Department of Rheumatology, University Medical Centre Ljubljana, Ljubljana,
Slovenia
| | - Snezna Sodin-Semrl
- Department of Rheumatology, University Medical Centre Ljubljana, Ljubljana,
Slovenia
- University of Primorska, Faculty of Mathematics, Natural Sciences and
Information Technology, Koper, Slovenia
| | - John Varga
- Division of Rheumatology, Feinberg School of Medicine, Northwestern
University, Chicago, United States of America
| |
Collapse
|
31
|
Chen M, Zhou H, Cheng N, Qian F, Ye RD. Serum amyloid A1 isoforms display different efficacy at Toll-like receptor 2 and formyl peptide receptor 2. Immunobiology 2014; 219:916-23. [PMID: 25154907 PMCID: PMC4252704 DOI: 10.1016/j.imbio.2014.08.002] [Citation(s) in RCA: 43] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2014] [Revised: 07/22/2014] [Accepted: 08/03/2014] [Indexed: 01/13/2023]
Abstract
Serum amyloid A (SAA) is a major acute-phase protein and a precursor of amyloid A, the deposit of which leads to amyloidosis. Different alleles exist in SAA1, a predominant form of the human SAA gene family. Emerging evidence has shown correlations between these alleles and diseases including familiar Mediterranean fever and amyloidosis. However, it remains unclear how the proteins encoded by these SAA1 alleles act differently. Here we report the characterization of proteins encoded by SAA1.1, SAA1.3, and SAA1.5, in comparison to that encoded by SAA2.2, for their preference of the SAA receptors including formyl peptide receptor 2 (FPR2) and Toll-like receptor 2 (TLR2). SAA1.1 was more efficacious than SAA1.3 and SAA1.5 but equally efficacious to SAA2.2 in calcium mobilization and chemotaxis assays, which measure the activation of the G protein-coupled FPR2. In agreement with this, SAA1.1 and SAA2.2 induced more robust phosphorylation of ERK than SAA1.3 and SAA1.5. Only small differences were observed between the SAA1 proteins tested and SAA2.2 in TLR2-dependent NF-κB luciferase reporter assay. In comparison, SAA1.3 was most effective in stimulating ERK and p38 MAPK phosphorylation. Using bone marrow-derived macrophages from C57BL/10ScN (Tlr4lps-del) mice, we examined the SAA isoforms for their induction of selected pro- and anti-inflammatory cytokines. SAA1.3 was most potent in the induction of TNFα and IL-1rn, whereas SAA1.5 induced robust IL-10 expression. These results show differences of the SAA1 isoforms in their selectivity for the SAA receptors, which may affect their roles in modulating inflammation and immunity.
Collapse
Affiliation(s)
- Mingjie Chen
- School of Pharmacy, Shanghai Jiao Tong University, Shanghai, PR China
| | - Huibing Zhou
- School of Pharmacy, Shanghai Jiao Tong University, Shanghai, PR China
| | - Ni Cheng
- Department of Pharmacology, University of Illinois College of Medicine, Chicago, Illinois 60612, USA
| | - Feng Qian
- School of Pharmacy, Shanghai Jiao Tong University, Shanghai, PR China
| | - Richard D Ye
- School of Pharmacy, Shanghai Jiao Tong University, Shanghai, PR China; Department of Pharmacology, University of Illinois College of Medicine, Chicago, Illinois 60612, USA.
| |
Collapse
|
32
|
Rooney P, Connolly M, Gao W, McCormick J, Biniecka M, Sullivan O, Kirby B, Sweeney C, Molloy E, Markham T, Fearon U, Veale DJ. Notch-1 mediates endothelial cell activation and invasion in psoriasis. Exp Dermatol 2014; 23:113-8. [PMID: 24330353 DOI: 10.1111/exd.12306] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/11/2013] [Indexed: 01/20/2023]
Abstract
Notch receptor-ligand interactions are critical for cell proliferation, differentiation and survival; however, the role of Notch signalling in psoriasis remains to be elucidated. Serum amyloid A (A-SAA) is an acute-phase protein with cytokine-like properties, regulates cell survival pathways and is implicated in many inflammatory conditions. To examine the role of Notch-1 signalling in the pathogenesis of psoriasis, Notch-1, DLL-4, Jagged-1, Hrt-1/Hrt-2, A-SAA, Factor VIII and vascular endothelial growth factor (VEGF) mRNA and/or protein expression in psoriasis skin biopsies, serum and dHMVEC were assessed by immunohistology, dual-immunofluorescence, real-time PCR, ELISA and Western blotting. A-SAA-induced angiogenesis and invasion in the presence of Notch-1 siRNA was assessed by matrigel tube formation assays and Transwell invasion assay. Increased Notch-1, its ligand DLL-4 and Hrt-1 expression were demonstrated in lesional skin compared with non-lesional skin, with greatest expression observed in the dermal vasculature (P < 0.05). Dual-immunofluorescent staining demonstrated co-localization of Notch-1 to endothelial cell marker Factor VIII. A significant increase in A-SAA levels was demonstrated in psoriasis serum compared with healthy control serum (P < 0.05), and A-SAA expression was higher in lesional skin compared with non-lesional. In dHMVEC, A-SAA significantly induced Jagged-1, Hrt-1 and VEGF mRNA expression (P < 0.05) and activated Notch-1 IC indicative of transcriptional regulation. In contrast, A-SAA significantly inhibited DLL-4 mRNA expression (P < 0.05). Finally A-SAA-induced angiogenesis and invasion were inhibited by Notch-1 siRNA (P < 0.05). Notch receptor-ligand interactions mediate vascular dysfunction in psoriasis and may represent a potential therapeutic target.
Collapse
Affiliation(s)
- Peadar Rooney
- Department of Rheumatology, Dublin Academic Medical Centre and the Conway Institute of Biomolecular and Biomedical Research, UCD, Dublin 4, Ireland
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
33
|
Martini AC, Forner S, Bento AF, Rae GA. Neuroprotective effects of lipoxin A4 in central nervous system pathologies. BIOMED RESEARCH INTERNATIONAL 2014; 2014:316204. [PMID: 25276776 PMCID: PMC4174961 DOI: 10.1155/2014/316204] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/19/2014] [Accepted: 08/12/2014] [Indexed: 02/07/2023]
Abstract
Many diseases of the central nervous system are characterized and sometimes worsened by an intense inflammatory response in the affected tissue. It is now accepted that resolution of inflammation is an active process mediated by a group of mediators that can act in synchrony to switch the phenotype of cells, from a proinflammatory one to another that favors the return to homeostasis. This new genus of proresolving mediators includes resolvins, protectins, maresins, and lipoxins, the first to be discovered. In this short review we provide an overview of current knowledge into the cellular and molecular interactions of lipoxins in diseases of the central nervous system in which they appear to facilitate the resolution of inflammation, thus exerting a neuroprotective action.
Collapse
Affiliation(s)
- Alessandra Cadete Martini
- Departmento de Farmacologia, Centro de Ciências Biológicas, Universidade Federal de Santa Catarina (UFSC), Campus Universitário, Trindade, 88049-900 Florianópolis, SC, Brazil
| | - Stefânia Forner
- Departmento de Farmacologia, Centro de Ciências Biológicas, Universidade Federal de Santa Catarina (UFSC), Campus Universitário, Trindade, 88049-900 Florianópolis, SC, Brazil
| | - Allisson Freire Bento
- Centro de Inovação e Ensaios Pré-Clínicos (CIEnP), Av. Luiz Boiteux Piazza, 1302-Canasvieiras, 88056-000 Florianópolis, SC, Brazil
| | - Giles Alexander Rae
- Departmento de Farmacologia, Centro de Ciências Biológicas, Universidade Federal de Santa Catarina (UFSC), Campus Universitário, Trindade, 88049-900 Florianópolis, SC, Brazil
| |
Collapse
|
34
|
Uteroglobin, a possible ligand of the lipoxin receptor inhibits serum amyloid A-driven inflammation. Mediators Inflamm 2014; 2014:876395. [PMID: 24782597 PMCID: PMC3981015 DOI: 10.1155/2014/876395] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2013] [Revised: 01/31/2014] [Accepted: 02/07/2014] [Indexed: 11/18/2022] Open
Abstract
Serum amyloid A (SAA) production is increased by inflamed arthritic synovial tissue, where it acts as a cytokine/chemoattractant for inflammatory and immune cells and as an inducer of matrix degrading enzymes. SAA has been shown to bind lipoxin A4 receptor, a member of the formyl-peptide related 2 G-protein coupled receptor family (ALX) and elicit proinflammatory activities in human primary fibroblast-like synoviocytes (FLS). We report on the identification of uteroglobin, a small globular protein with potent anti-inflammatory activities, as a possible ligand of ALX. Uteroglobin-specific association with ALX was demonstrated by an enzyme immunoassay experiment employing a cell line engineered to express the human ALX receptor. Uteroglobin's interaction with ALX resulted in the inhibition of SAA responses, such as attenuation of phospholipase A2 activation and cellular chemotaxis. In FLS, uteroglobin showed an antagonism against SAA-induced interleukin-8 release and decreased cell migration. These novel roles described for uteroglobin via ALX may help elucidate genetic and clinical observations indicating that a polymorphism in the uteroglobin promoter is linked to disease outcome, specifically prediction of bone erosion in patients with rheumatoid arthritis or severity of IgA glomerulonephritis and sarcoidosis.
Collapse
|
35
|
Kojima F, Kapoor M, Kawai S, Crofford LJ. New insights into eicosanoid biosynthetic pathways: implications for arthritis. Expert Rev Clin Immunol 2014; 2:277-91. [DOI: 10.1586/1744666x.2.2.277] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023]
|
36
|
Alexander SPH, Benson HE, Faccenda E, Pawson AJ, Sharman JL, Spedding M, Peters JA, Harmar AJ. The Concise Guide to PHARMACOLOGY 2013/14: G protein-coupled receptors. Br J Pharmacol 2013; 170:1459-581. [PMID: 24517644 PMCID: PMC3892287 DOI: 10.1111/bph.12445] [Citation(s) in RCA: 509] [Impact Index Per Article: 42.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
The Concise Guide to PHARMACOLOGY 2013/14 provides concise overviews of the key properties of over 2000 human drug targets with their pharmacology, plus links to an open access knowledgebase of drug targets and their ligands (www.guidetopharmacology.org), which provides more detailed views of target and ligand properties. The full contents can be found at http://onlinelibrary.wiley.com/doi/10.1111/bph.12444/full. G protein-coupled receptors are one of the seven major pharmacological targets into which the Guide is divided, with the others being G protein-coupled receptors, ligand-gated ion channels, ion channels, catalytic receptors, nuclear hormone receptors, transporters and enzymes. These are presented with nomenclature guidance and summary information on the best available pharmacological tools, alongside key references and suggestions for further reading. A new landscape format has easy to use tables comparing related targets. It is a condensed version of material contemporary to late 2013, which is presented in greater detail and constantly updated on the website www.guidetopharmacology.org, superseding data presented in previous Guides to Receptors and Channels. It is produced in conjunction with NC-IUPHAR and provides the official IUPHAR classification and nomenclature for human drug targets, where appropriate. It consolidates information previously curated and displayed separately in IUPHAR-DB and the Guide to Receptors and Channels, providing a permanent, citable, point-in-time record that will survive database updates.
Collapse
Affiliation(s)
- Stephen PH Alexander
- School of Life Sciences, University of Nottingham Medical SchoolNottingham, NG7 2UH, UK
| | - Helen E Benson
- The University/BHF Centre for Cardiovascular Science, University of EdinburghEdinburgh, EH16 4TJ, UK
| | - Elena Faccenda
- The University/BHF Centre for Cardiovascular Science, University of EdinburghEdinburgh, EH16 4TJ, UK
| | - Adam J Pawson
- The University/BHF Centre for Cardiovascular Science, University of EdinburghEdinburgh, EH16 4TJ, UK
| | - Joanna L Sharman
- The University/BHF Centre for Cardiovascular Science, University of EdinburghEdinburgh, EH16 4TJ, UK
| | | | - John A Peters
- Neuroscience Division, Medical Education Institute, Ninewells Hospital and Medical School, University of DundeeDundee, DD1 9SY, UK
| | - Anthony J Harmar
- The University/BHF Centre for Cardiovascular Science, University of EdinburghEdinburgh, EH16 4TJ, UK
| |
Collapse
|
37
|
Hawkins KE, DeMars KM, Singh J, Yang C, Cho HS, Frankowski JC, Doré S, Candelario-Jalil E. Neurovascular protection by post-ischemic intravenous injections of the lipoxin A4 receptor agonist, BML-111, in a rat model of ischemic stroke. J Neurochem 2013; 129:130-42. [PMID: 24225006 DOI: 10.1111/jnc.12607] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2013] [Revised: 10/23/2013] [Accepted: 11/08/2013] [Indexed: 02/03/2023]
Abstract
Resolution of inflammation is an emerging new strategy to reduce damage following ischemic stroke. Lipoxin A4 (LXA4 ) is an anti-inflammatory, pro-resolution lipid mediator with high affinity binding to ALX, the lipoxin A4 receptor. Since LXA4 is rapidly inactivated, potent analogs have been created, including the ALX agonist BML-111. We hypothesized that post-ischemic intravenous administration of BML-111 would provide protection to the neurovascular unit and reduce neuroinflammation in a rat stroke model. Animals were subjected to 90 min of middle cerebral artery occlusion (MCAO) and BML-111 was injected 100 min and 24 h after stroke onset and animals euthanized at 48 h. Post-ischemic treatment with BML-111 significantly reduced infarct size, decreased vasogenic edema, protected against blood-brain barrier disruption, and reduced hemorrhagic transformation. Matrix metalloproteinase-9 and matrix metalloproteinase-3 were significantly reduced following BML-111 treatment. Administration of BML-111 dramatically decreased microglial activation, as seen with CD68, and neutrophil infiltration and recruitment, as assessed by levels of myeloperoxidase and intracellular adhesion molecule-1. The tight junction protein zona occludens-1 was protected from degradation following treatment with BML-111. These results indicate that post-ischemic activation of ALX has pro-resolution effects that limit the inflammatory damage in the cerebral cortex and helps maintain blood-brain barrier integrity after ischemic stroke.
Collapse
Affiliation(s)
- Kimberly E Hawkins
- Department of Neuroscience, McKnight Brain Institute, University of Florida, Gainesville, Florida, USA
| | | | | | | | | | | | | | | |
Collapse
|
38
|
Ishii W, Liepnieks JJ, Yamada T, Benson MD, Kluve-Beckerman B. Human SAA1-derived amyloid deposition in cell culture: a consistent model utilizing human peripheral blood mononuclear cells and serum-free medium. Amyloid 2013; 20:61-71. [PMID: 23461622 DOI: 10.3109/13506129.2013.775941] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
Amyloid A (AA) amyloidosis is a fatal disease caused by extracellular deposition of fibrils derived from serum AA (SAA). AA amyloid fibril formation has previously been modeled in macrophage cultures using highly amyloidogenic mouse SAA1.1, but attempts to do the same with human SAA invariably failed. Our objective was to define conditions that support human SAA-derived amyloid formation in peripheral blood mononuclear cell (PBMC) cultures. Two conditions were found to be critical - omission of fetal calf serum and use of StemPro34, a lipid-enriched medium formulated for hematopoietic progenitor cells. Cultures maintained in serum-free StemPro34 and provided with recombinant human SAA1 in the complete absence of amyloid-enhancing factor exhibited amyloid deposition within 7 d. Amyloid co-localized with cell clusters that characteristically included cells of fibrocytic/dendritic morphology as well as macrophages. These cells formed networks that appeared to serve as scaffolding within and upon which amyloid accumulated. Cells in amyloid-forming cultures demonstrated increased adherence, survival and expression of extracellular matrix components. Of the three human SAA1 isoforms, SAA1.3 showed the most extensive amyloid deposition, consistent with it being the most prevalent isoform in Japanese patients with AA amyloidosis. Attesting to the reproducibility and general applicability of this model, amyloid formation has been documented in cultures established from eight PBMC donors.
Collapse
Affiliation(s)
- Wataru Ishii
- Department of Medicine (Neurology and Rheumatology), Shinshu Unviersity School of Medicine, Matsumoto, Japan
| | | | | | | | | |
Collapse
|
39
|
Distinct signaling cascades elicited by different formyl peptide receptor 2 (FPR2) agonists. Int J Mol Sci 2013; 14:7193-230. [PMID: 23549262 PMCID: PMC3645683 DOI: 10.3390/ijms14047193] [Citation(s) in RCA: 141] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2013] [Revised: 03/13/2013] [Accepted: 03/15/2013] [Indexed: 12/22/2022] Open
Abstract
The formyl peptide receptor 2 (FPR2) is a remarkably versatile transmembrane protein belonging to the G-protein coupled receptor (GPCR) family. FPR2 is activated by an array of ligands, which include structurally unrelated lipids and peptide/proteins agonists, resulting in different intracellular responses in a ligand-specific fashion. In addition to the anti-inflammatory lipid, lipoxin A4, several other endogenous agonists also bind FPR2, including serum amyloid A, glucocorticoid-induced annexin 1, urokinase and its receptor, suggesting that the activation of FPR2 may result in potent pro- or anti-inflammatory responses. Other endogenous ligands, also present in biological samples, include resolvins, amyloidogenic proteins, such as beta amyloid (Aβ)-42 and prion protein (Prp)106–126, the neuroprotective peptide, humanin, antibacterial peptides, annexin 1-derived peptides, chemokine variants, the neuropeptides, vasoactive intestinal peptide (VIP) and pituitary adenylate cyclase activating polypeptide (PACAP)-27, and mitochondrial peptides. Upon activation, intracellular domains of FPR2 mediate signaling to G-proteins, which trigger several agonist-dependent signal transduction pathways, including activation of phospholipase C (PLC), protein kinase C (PKC) isoforms, the phosphoinositide 3-kinase (PI3K)/protein kinase B (Akt) pathway, the mitogen-activated protein kinase (MAPK) pathway, p38MAPK, as well as the phosphorylation of cytosolic tyrosine kinases, tyrosine kinase receptor transactivation, phosphorylation and nuclear translocation of regulatory transcriptional factors, release of calcium and production of oxidants. FPR2 is an attractive therapeutic target, because of its involvement in a range of normal physiological processes and pathological diseases. Here, we review and discuss the most significant findings on the intracellular pathways and on the cross-communication between FPR2 and tyrosine kinase receptors triggered by different FPR2 agonists.
Collapse
|
40
|
Gori I, Rodriguez Y, Pellegrini C, Achtari C, Hornung D, Chardonnens E, Wunder D, Fiche M, Canny GO. Augmented epithelial multidrug resistance-associated protein 4 expression in peritoneal endometriosis: regulation by lipoxin A(4). Fertil Steril 2013; 99:1965-73.e2. [PMID: 23472950 DOI: 10.1016/j.fertnstert.2013.01.146] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2012] [Revised: 01/28/2013] [Accepted: 01/29/2013] [Indexed: 12/22/2022]
Abstract
OBJECTIVE To compare the expression of the prostaglandin (PG) E(2) transporter multidrug resistance-associated protein 4 (MRP4) in eutopic and ectopic endometrial tissue from endometriosis patients with that of control subjects and to examine whether MRP4 is regulated by the antiinflammatory lipid lipoxin A(4) (LXA(4)) in endometriotic epithelial cells. DESIGN Molecular analysis in human samples and a cell line. SETTING Two university hospitals and a private clinic. PATIENT(S) A total of 59 endometriosis patients and 32 age- and body mass index-matched control subjects undergoing laparoscopy or hysterectomy. INTERVENTION(S) Normal, eutopic, and ectopic endometrial biopsies as well as peritoneal fluid were obtained during surgery performed during the proliferative phase of the menstrual cycle. 12Z endometriotic epithelial cells were used for in vitro mechanistic studies. MAIN OUTCOME MEASURE(S) Tissue MRP4 mRNA levels were quantified by quantitative reverse-transcription polymerase chain reaction (qRT-PCR), and localization was analyzed with the use of immunohistochemistry. Cellular MRP4 mRNA and protein were quantified by qRT-PCR and Western blot, respectively. PGE(2) was measured in peritoneal fluid and cell supernatants using an enzyme immunoassay (EIA). RESULT(S) MRP4 was expressed in eutopic and ectopic endometrium, where it was overexpressed in peritoneal lesions and localized in the cytoplasm of glandular epithelial cells. LXA(4) attenuated MRP4 mRNA and protein levels in endometriotic epithelial cells in a dose-dependent manner, while not affecting the expression of enzymes involved in PGE(2) metabolism. Investigations employing receptor antagonists and small interfering RNA revealed that this occurred through estrogen receptor α. Accordingly, LXA(4) treatment inhibited extracellular PGE(2) release. CONCLUSION(S) We report for the first time that MRP4 is expressed in human endometrium, elevated in peritoneal endometriosis, and modulated by LXA(4) in endometriotic epithelial cells.
Collapse
Affiliation(s)
- Ilaria Gori
- Department of Gynecology, Obstetrics, and Medical Genetics, Lausanne University Hospital, Lausanne, Switzerland
| | | | | | | | | | | | | | | | | |
Collapse
|
41
|
Jin H, Li YH, Xu JS, Guo GQ, Chen DL, Bo Y. Lipoxin A4 analog attenuates morphine antinociceptive tolerance, withdrawal-induced hyperalgesia, and glial reaction and cytokine expression in the spinal cord of rat. Neuroscience 2012; 208:1-10. [DOI: 10.1016/j.neuroscience.2012.02.009] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2012] [Revised: 01/31/2012] [Accepted: 02/05/2012] [Indexed: 01/03/2023]
|
42
|
Kennedy A, Fearon U, Veale DJ, Godson C. Macrophages in synovial inflammation. Front Immunol 2011; 2:52. [PMID: 22566842 PMCID: PMC3342259 DOI: 10.3389/fimmu.2011.00052] [Citation(s) in RCA: 129] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2011] [Accepted: 09/19/2011] [Indexed: 01/09/2023] Open
Abstract
Synovial macrophages are one of the resident cell types in synovial tissue and while they remain relatively quiescent in the healthy joint, they become activated in the inflamed joint and, along with infiltrating monocytes/macrophages, regulate secretion of pro-inflammatory cytokines and enzymes involved in driving the inflammatory response and joint destruction. Synovial macrophages are positioned throughout the sub-lining layer and lining layer at the cartilage–pannus junction and mediate articular destruction. Sub-lining macrophages are now also considered as the most reliable biomarker for disease severity and response to therapy in rheumatoid arthritis (RA). There is a growing understanding of the molecular drivers of inflammation and an appreciation that the resolution of inflammation is an active process rather than a passive return to homeostasis, and this has implications for our understanding of the role of macrophages in inflammation. Macrophage phenotype determines the cytokine secretion profile and tissue destruction capabilities of these cells. Whereas inflammatory synovial macrophages have not yet been classified into one phenotype or another it is widely known that TNFα and IL-l, characteristically released by M1 macrophages, are abundant in RA while IL-10 activity, characteristic of M2 macrophages, is somewhat diminished. Here we will briefly review our current understanding of macrophages and macrophage polarization in RA as well as the elements implicated in controlling polarization, such as cytokines and transcription factors like NFκB, IRFs and NR4A, and pro-resolving factors, such as LXA4 and other lipid mediators which may promote a non-inflammatory, pro-resolving phenotype, and may represent a novel therapeutic paradigm.
Collapse
Affiliation(s)
- Aisling Kennedy
- School of Medicine and Medical Sciences, University College Dublin Conway Institute Dublin, Ireland
| | | | | | | |
Collapse
|
43
|
Wu Y, Wang YP, Guo P, Ye XH, Wang J, Yuan SY, Yao SL, Shang Y. A lipoxin A4 analog ameliorates blood-brain barrier dysfunction and reduces MMP-9 expression in a rat model of focal cerebral ischemia-reperfusion injury. J Mol Neurosci 2011; 46:483-91. [PMID: 21845429 DOI: 10.1007/s12031-011-9620-5] [Citation(s) in RCA: 52] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2011] [Accepted: 08/04/2011] [Indexed: 12/15/2022]
Abstract
LXA(4) methyl ester (LXA(4)ME), a lipoxin A(4) analog, reduces ischemic insult in the rat models of transient or permanent cerebral ischemic injury. We investigated whether LXA(4)ME could ameliorate blood-brain barrier (BBB) dysfunction after stroke by reducing matrix metalloproteinase (MMP)-9 expression. Adult male rats were subjected to 2-h middle cerebral artery occlusion (MCAO) followed by 24-h reperfusion. Brain infarctions were detected by triphenyltetrazolium chloride (TTC) staining. BBB dysfunction was determined by examining brain edema and Evans Blue extravasation. Temporal expression of MMP-9 was determined by zymography and Western blot. The presence of tissue inhibitors of metalloproteinase-1 (TIMP-1) was also determined by Western blot in tissue protein sample. Brain edema and Evans Blue leakage were significantly reduced after stroke in the LXA(4)ME group and were associated with reduced brain infarct volumes. MMP-9 activity and expression were inhibited by LXA(4)ME after stroke. In addition, LXA(4)ME significantly increased TIMP-1 protein levels. Our results indicate that LXA(4)ME reduces brain injury by improving BBB function in a rat model of MCAO, and that a relationship exists between BBB permeability and MMP-9 expression following ischemic insult. Furthermore, these results suggest that LXA(4)ME-mediated reduction of MMP-9 following stroke are attributed to increased TIMP-1 expression.
Collapse
Affiliation(s)
- Yan Wu
- Department of Neurology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, No. 1277, Jiefang Avenue, Wuhan, 430022, China.
| | | | | | | | | | | | | | | |
Collapse
|
44
|
Hutchinson JL, Rajagopal SP, Sales KJ, Jabbour HN. Molecular regulators of resolution of inflammation: potential therapeutic targets in the reproductive system. Reproduction 2011; 142:15-28. [DOI: 10.1530/rep-11-0069] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
Inflammatory processes are central to reproductive events including ovulation, menstruation, implantation and labour, while inflammatory dysregulation is a feature of numerous reproductive pathologies. In recent years, there has been much research into the endogenous mechanisms by which inflammatory reactions are terminated and tissue homoeostasis is restored, a process termed resolution. The identification and characterisation of naturally occurring pro-resolution mediators including lipoxins and annexin A1 has prompted a shift in the field of anti-inflammation whereby resolution is now observed as an active process, triggered as part of a normal inflammatory response. This review will address the process of resolution, discuss available evidence for expression of pro-resolution factors in the reproductive tract and explore possible roles for resolution in physiological reproductive processes and associated pathologies.
Collapse
|
45
|
Macdonald LJ, Boddy SC, Denison FC, Sales KJ, Jabbour HN. A role for lipoxin A₄ as an anti-inflammatory mediator in the human endometrium. Reproduction 2011; 142:345-52. [PMID: 21555360 PMCID: PMC3139491 DOI: 10.1530/rep-11-0021] [Citation(s) in RCA: 50] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Lipoxin A(4) is a lipid mediator that elicits anti-inflammatory and pro-resolution actions via its receptor, formyl peptide receptor 2 (FPR2/ALX). In this study, we aimed to investigate the expression and potential role of lipoxin A(4) and FPR2/ALX in the regulation of inflammation associated with cyclical remodeling of the human endometrium across the menstrual cycle and during early pregnancy. Using quantitative RT-PCR analysis, we found that FPR2/ALX expression is upregulated during the menstrual phase of the cycle and in decidua tissue from the first trimester of pregnancy. We localized the site of expression of FPR2/ALX in menstrual phase endometrium and first-trimester decidua tissue to glandular epithelial cells and cells within the stromal compartment, including cells lining the blood vessels and immune cells. Measurement of serum lipoxin A(4) by ELISA revealed no difference in its levels across the menstrual cycle but an elevation in early pregnancy (P<0.001). We found that lipoxin A(4) was regulated by human chorionic gonadotrophin (hCG) during early pregnancy, because treatment of human decidua tissue with hCG increased lipoxin A(4) release (P<0.01). Finally, we have shown that lipoxin A(4) can suppress phorbol myristate acetate-induced expression of the inflammatory cytokines interleukin 6 and 8 in human endometrium and decidua tissue. These results demonstrate for the first time that lipoxin A(4) and its receptor FPR2/ALX can regulate inflammatory events in the human endometrium and decidua of early pregnancy.
Collapse
Affiliation(s)
- Linsay J Macdonald
- MRC Human Reproductive Sciences Unit, The Queen's Medical Research Institute, 47 Little France Crescent, Edinburgh, EH16 4TJ, UK
| | | | | | | | | |
Collapse
|
46
|
Connolly M, Marrelli A, Blades M, McCormick J, Maderna P, Godson C, Mullan R, FitzGerald O, Bresnihan B, Pitzalis C, Veale DJ, Fearon U. Acute serum amyloid A induces migration, angiogenesis, and inflammation in synovial cells in vitro and in a human rheumatoid arthritis/SCID mouse chimera model. THE JOURNAL OF IMMUNOLOGY 2010; 184:6427-37. [PMID: 20435930 DOI: 10.4049/jimmunol.0902941] [Citation(s) in RCA: 82] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Serum amyloid A (A-SAA), an acute-phase protein with cytokine-like properties, is expressed at sites of inflammation. This study investigated the effects of A-SAA on chemokine-regulated migration and angiogenesis using rheumatoid arthritis (RA) cells and whole-tissue explants in vitro, ex vivo, and in vivo. A-SAA levels were measured by real-time PCR and ELISA. IL-8 and MCP-1 expression was examined in RA synovial fibroblasts, human microvascular endothelial cells, and RA synovial explants by ELISA. Neutrophil transendothelial cell migration, cell adhesion, invasion, and migration were examined using transwell leukocyte/monocyte migration assays, invasion assays, and adhesion assays with or without anti-MCP-1/anti-IL-8. NF-kappaB was examined using a specific inhibitor and Western blotting. An RA synovial/SCID mouse chimera model was used to examine the effects of A-SAA on cell migration, proliferation, and angiogenesis in vivo. High expression of A-SAA was demonstrated in RA patients (p < 0.05). A-SAA induced chemokine expression in a time- and dose-dependent manner (p < 0.05). Blockade with anti-scavenger receptor class B member 1 and lipoxin A4 (A-SAA receptors) significantly reduced chemokine expression in RA synovial tissue explants (p < 0.05). A-SAA induced cell invasion, neutrophil-transendothelial cell migration, monocyte migration, and adhesion (all p < 0.05), effects that were blocked by anti-IL-8 or anti-MCP-1. A-SAA-induced chemokine expression was mediated through NF-kappaB in RA explants (p < 0.05). Finally, in the RA synovial/SCID mouse chimera model, we demonstrated for the first time in vivo that A-SAA directly induces monocyte migration from the murine circulation into RA synovial grafts, synovial cell proliferation, and angiogenesis (p < 0.05). A-SAA promotes cell migrational mechanisms and angiogenesis critical to RA pathogenesis.
Collapse
Affiliation(s)
- Mary Connolly
- Dublin Academic Medical Centre, University College Dublin, Dublin, Ireland
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
47
|
Neuroprotective effect of lipoxin A4 methyl ester in a rat model of permanent focal cerebral ischemia. J Mol Neurosci 2010; 42:226-34. [PMID: 20401639 DOI: 10.1007/s12031-010-9355-8] [Citation(s) in RCA: 39] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2009] [Accepted: 03/19/2010] [Indexed: 10/19/2022]
Abstract
Neuroprotective effect of lipoxin A(4) methyl ester (LXA(4) ME) was tested in a rat model of permanent middle cerebral artery occlusion. LXA(4) ME was administrated through intracerebroventricular injection immediately after middle cerebral artery was occluded. Administration of LXA(4) ME ameliorated neurological deficit, reduced infarct volume, attenuated histological damage, and decreased number of apoptotic neuron induced by ischemic insult. These neuroprotective effects of LXA(4) ME were associated with inhibition of neutrophil infiltration, lipid peroxidation, and astrocyte activation. In addition, LXA(4) ME also attenuated proinflammatory cytokines (TNF-alpha and IL-1beta) production. These data suggest that LXA(4) ME protects neuron against permanent cerebral ischemia by inhibiting inflammatory responses.
Collapse
|
48
|
Ye XH, Wu Y, Guo PP, Wang J, Yuan SY, Shang Y, Yao SL. Lipoxin A4 analogue protects brain and reduces inflammation in a rat model of focal cerebral ischemia reperfusion. Brain Res 2010; 1323:174-83. [DOI: 10.1016/j.brainres.2010.01.079] [Citation(s) in RCA: 67] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2009] [Revised: 01/26/2010] [Accepted: 01/28/2010] [Indexed: 12/30/2022]
|
49
|
Mullan RH, McCormick J, Connolly M, Bresnihan B, Veale DJ, Fearon U. A role for the high-density lipoprotein receptor SR-B1 in synovial inflammation via serum amyloid-A. THE AMERICAN JOURNAL OF PATHOLOGY 2010; 176:1999-2008. [PMID: 20304957 DOI: 10.2353/ajpath.2010.090014] [Citation(s) in RCA: 42] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Acute phase apoprotein Serum Amyloid A (A-SAA), which is strongly expressed in rheumatoid arthritis synovial membrane (RA SM), induces angiogenesis, adhesion molecule expression, and matrix metalloproteinase production through the G-coupled receptor FPRL-1. Here we report alternative signaling through the high-density lipoprotein receptor scavenger receptor-class B type 1 (SR-B1). Quantitative expression/localization of SR-B1 in RA SM, RA fibroblast-like cells (FLCs), and microvascular endothelial cells (ECs) was assessed by Western blotting and immunohistology/fluorescence. A-SAA-mediated effects were examined using a specific antibody against SR-B1 or amphipathic alpha-Helical Peptides (the SR-B1 antagonists L-37pA and D-37pA), in RA FLCs and ECs. Adhesion molecule expression and cytokine production were quantified using flow cytometry and ELISA. SR-B1 was strongly expressed in the RA SM lining layer and endothelial/perivascular regions compared with osteoarthritis SM or normal control synovium. Differential SR-B1 expression in RA FLC lines (n = 5) and ECs correlated closely with A-SAA, but not tumor necrosis factor alpha-induced intercellular adhesion molecule-1 upregulation. A-SAA-induced interleukin-6 and -8 production was inhibited in the presence of anti-SR-B1 in human microvascular endothelial cells and RA FLCs. Moreover, D-37pA and L-37pA inhibited A-SAA-induced vascular cell adhesion molecule-1 and intercellular adhesion molecule expression from ECs in a dose-dependent manner. As SR-B1 is expressed in RA synovial tissue and mediates A-SAA-induced pro-inflammatory pathways, a better understanding of A-SAA-mediated inflammatory pathways may lead to novel treatment strategies for RA.
Collapse
Affiliation(s)
- Ronan Hugh Mullan
- Education and Research Centre, St. Vincent's University Hospital, Elm Park, Dublin 4, Ireland
| | | | | | | | | | | |
Collapse
|
50
|
|