1
|
Rocha PS, Silva AA, Queiroz-Junior CM, Braga AD, Moreira TP, Teixeira MM, Amaral FA. Trained immunity of synovial macrophages is associated with exacerbated joint inflammation and damage after Staphylococcus aureus infection. Inflamm Res 2024; 73:1995-2008. [PMID: 39340660 DOI: 10.1007/s00011-024-01946-w] [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: 05/09/2024] [Revised: 08/06/2024] [Accepted: 09/09/2024] [Indexed: 09/30/2024] Open
Abstract
OBJECTIVES Investigate whether and which synoviocytes would acquire trained immunity characteristics that could exacerbate joint inflammation following a secondary Staphylococcus aureus infection. METHODS Lipopolysaccharide (LPS) and S. aureus were separately or double injected (21 days of interval) into the tibiofemoral joint cavity of male C57BL/6 mice. At different time points after these stimulations, mechanical nociception was analyzed followed by the analysis of signs of inflammation and damage in the affected joints. The trained immunity markers, including the glycolytic and mTOR pathway, were analyzed in whole tissue or isolated synoviocytes. A group of mice was treated with Rapamycin, an mTOR inhibitor before LPS or S. aureus stimulation. RESULTS The double LPS - S. aureus hit promoted intense joint inflammation and damage compared to single joint stimulation, including markers in synoviocyte activation, production of proinflammatory cytokines, persistent nociception, and bone damage, despite not reducing the bacterial clearance. The double LPS - S. aureus hit joints increased the synovial macrophage population expressing CX3CR1 alongside triggering established epigenetic modifications associated with trained immunity events in these cells, such as the upregulation of the mTOR signaling pathway (p-mTOR and HIF1α) and the trimethylation of histone H3. Mice treated with Rapamycin presented reduced CX3CR1+ macrophage activation, joint inflammation, and bone damage. CONCLUSIONS There is a trained immunity phenotype in CX3CR1+ synovial macrophages that contributes to the exacerbation of joint inflammation and damage during septic arthritis caused by S. aureus.
Collapse
Affiliation(s)
- Peter Silva Rocha
- Department of Biochemistry and Immunology, Institute of Biological Sciences, Universidade Federal de Minas Gerais, Belo Horizonte, 31270-901, MG, Brazil
| | - Adryan Aparecido Silva
- Department of Biochemistry and Immunology, Institute of Biological Sciences, Universidade Federal de Minas Gerais, Belo Horizonte, 31270-901, MG, Brazil
| | - Celso Martins Queiroz-Junior
- Department of Morphology, Institute of Biological Sciences, Universidade Federal de Minas Gerais, Belo Horizonte, 31270-901, MG, Brazil
| | - Amanda Dias Braga
- Department of Morphology, Institute of Biological Sciences, Universidade Federal de Minas Gerais, Belo Horizonte, 31270-901, MG, Brazil
| | - Thaiane Pinto Moreira
- Department of Microbiology, Institute of Biological Sciences, Universidade Federal de Minas Gerais, Belo Horizonte, 31270-901, MG, Brazil
| | - Mauro Martins Teixeira
- Department of Biochemistry and Immunology, Institute of Biological Sciences, Universidade Federal de Minas Gerais, Belo Horizonte, 31270-901, MG, Brazil
| | - Flávio Almeida Amaral
- Department of Biochemistry and Immunology, Institute of Biological Sciences, Universidade Federal de Minas Gerais, Belo Horizonte, 31270-901, MG, Brazil.
| |
Collapse
|
2
|
Huerta MÁ, Molina-Álvarez M, García MM, Tejada MA, Goicoechea C, Ghasemlou N, Ruiz-Cantero MC, Cobos EJ. The role of neutrophils in pain: systematic review and meta-analysis of animal studies. Pain 2024:00006396-990000000-00754. [PMID: 39450928 DOI: 10.1097/j.pain.0000000000003450] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2024] [Accepted: 09/10/2024] [Indexed: 10/26/2024]
Abstract
ABSTRACT The peripheral inflammatory response is an attractive therapeutic target for pain treatment. Neutrophils are the first circulating inflammatory cells recruited to sites of injury, but their contribution to pain outcomes is unclear. We performed a systematic review and meta-analysis of original preclinical studies, which evaluated the effect of preemptive neutrophil depletion on pain outcomes (PROSPERO registration number: CRD42022364004). Literature search (PubMed, January 19, 2023) identified 49 articles, which were meta-analyzed using a random-effects model. The risk of bias was evaluated using SYRCLE's tool. The pooled effect considering all studies showed that neutrophil depletion induced a consistent pain reduction. Inflammatory, joint, neuropathic, and visceral pain showed significant pain alleviation by neutrophil depletion with medium-large effect sizes. However, muscle and postoperative pain were not significantly alleviated by neutrophil depletion. Further analysis showed a differential contribution of neutrophils to pain outcomes. Neutrophils had a higher impact on mechanical hyperalgesia, followed by nociceptive behaviors and mechanical allodynia, with a smaller contribution to thermal hyperalgesia. Interspecies (mice or rats) differences were not appreciated. Analyses regarding intervention unveiled a lower pain reduction for some commonly used methods for neutrophil depletion, such as injection of antineutrophil serum or an anti-Gr-1 antibody, than for other agents such as administration of an anti-Ly6G antibody, fucoidan, vinblastine, CXCR1/2 inhibitors, and etanercept. In conclusion, the contribution of neutrophils to pain depends on pain etiology (experimental model), pain outcome, and the neutrophil depletion strategy. Further research is needed to improve our understanding on the mechanisms of these differences.
Collapse
Affiliation(s)
- Miguel Á Huerta
- Department of Pharmacology, Faculty of Medicine, University of Granada, Granada, Spain
- Institute of Neuroscience, Biomedical Research Center, University of Granada, Granada, Spain
- Biosanitary Research Institute ibs.GRANADA, Granada, Spain
| | - Miguel Molina-Álvarez
- Area of Pharmacology, Nutrition and Bromatology, Department of Basic Health Sciences, Rey Juan Carlos University, Asociated Unit I+D+i Instituto de Química Médica (IQM) CSIC-URJC, Alcorcón, Spain
- High Performance Experimental Pharmacology Research Group, Rey Juan Carlos University (PHARMAKOM), Alcorcón, Spain
| | - Miguel M García
- Area of Pharmacology, Nutrition and Bromatology, Department of Basic Health Sciences, Rey Juan Carlos University, Asociated Unit I+D+i Instituto de Química Médica (IQM) CSIC-URJC, Alcorcón, Spain
- High Performance Experimental Pharmacology Research Group, Rey Juan Carlos University (PHARMAKOM), Alcorcón, Spain
| | - Miguel A Tejada
- Department of Pharmacology, Faculty of Medicine, University of Granada, Granada, Spain
- Institute of Neuroscience, Biomedical Research Center, University of Granada, Granada, Spain
- Biosanitary Research Institute ibs.GRANADA, Granada, Spain
| | - Carlos Goicoechea
- Area of Pharmacology, Nutrition and Bromatology, Department of Basic Health Sciences, Rey Juan Carlos University, Asociated Unit I+D+i Instituto de Química Médica (IQM) CSIC-URJC, Alcorcón, Spain
- High Performance Experimental Pharmacology Research Group, Rey Juan Carlos University (PHARMAKOM), Alcorcón, Spain
| | - Nader Ghasemlou
- Pain Chronobiology & Neuroimmunology Laboratory, Departments of Anesthesiology and Biomedical & Molecular Sciences, Queen's University, Kingston, ON, Canada
| | - M Carmen Ruiz-Cantero
- Laboratori de Química Farmacèutica (Unitat Associada al CSIC), Facultat de Farmàcia i Ciències de l'Alimentació, Universitat de Barcelona, Barcelona, Spain
| | - Enrique J Cobos
- Department of Pharmacology, Faculty of Medicine, University of Granada, Granada, Spain
- Institute of Neuroscience, Biomedical Research Center, University of Granada, Granada, Spain
- Biosanitary Research Institute ibs.GRANADA, Granada, Spain
- Teófilo Hernando Institute for Drug Discovery, Madrid, Spain
| |
Collapse
|
3
|
Huerta MÁ, Tejada MÁ, Nieto FR. Fucoidan as a Promising Drug for Pain Treatment: Systematic Review and Meta-Analysis. Mar Drugs 2024; 22:290. [PMID: 39057399 PMCID: PMC11277653 DOI: 10.3390/md22070290] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2024] [Revised: 06/15/2024] [Accepted: 06/18/2024] [Indexed: 07/28/2024] Open
Abstract
Fucoidan is a polymer of L-fucose and L-fucose-4-sulphate naturally found in marine sources that inhibits p-selectin, preventing neutrophil recruitment to the site of injury. Fucoidan is employed in many studies as a tool to investigate the contribution of neutrophils to pain, showing analgesic effects. We performed a systematic review and meta-analysis to quantify the analgesic effects of pretreatment with fucoidan reported in the available preclinical studies. In addition, we summarized the articles which have studied the therapeutic effects of fucoidan in pathological pain at preclinical and clinical levels. The results of this systematic review reveal that pretreatment with fucoidan is a powerful tool which reduces neutrophil infiltration by 70-90% at early time points. This meta-analysis showed that preventative treatment with fucoidan produced a significant pain reduction. In addition, several preclinical studies have observed that fucoidan treatment reduces the pain that is associated with various pathologies. Finally, fucoidan has also been tested in several clinical trials, with some degree of analgesic efficacy, but they were mostly small pilot studies. Considering all the above information, it can be concluded that fucoidan is not only a preclinical tool for studying the role of neutrophils in pain but also a promising therapeutic strategy for pain treatment.
Collapse
Affiliation(s)
- Miguel Á. Huerta
- Department of Pharmacology, University of Granada, 18016 Granada, Spain; (M.Á.H.); (M.Á.T.)
- Institute of Neuroscience, Biomedical Research Center, University of Granada, 18016 Granada, Spain
- Biosanitary Research Institute ibs.GRANADA, 18012 Granada, Spain
| | - Miguel Á. Tejada
- Department of Pharmacology, University of Granada, 18016 Granada, Spain; (M.Á.H.); (M.Á.T.)
- Institute of Neuroscience, Biomedical Research Center, University of Granada, 18016 Granada, Spain
- Biosanitary Research Institute ibs.GRANADA, 18012 Granada, Spain
| | - Francisco R. Nieto
- Department of Pharmacology, University of Granada, 18016 Granada, Spain; (M.Á.H.); (M.Á.T.)
- Institute of Neuroscience, Biomedical Research Center, University of Granada, 18016 Granada, Spain
- Biosanitary Research Institute ibs.GRANADA, 18012 Granada, Spain
| |
Collapse
|
4
|
Marín-Prida J, Rodríguez-Ulloa A, Besada V, Llopiz-Arzuaga A, Batista NV, Hernández-González I, Pavón-Fuentes N, Marciano Vieira ÉL, Falcón-Cama V, Acosta EF, Martínez-Donato G, Cervantes-Llanos M, Lingfeng D, González LJ, Fernández-Massó JR, Guillén-Nieto G, Pentón-Arias E, Amaral FA, Teixeira MM, Pentón-Rol G. The effects of Phycocyanobilin on experimental arthritis involve the reduction in nociception and synovial neutrophil infiltration, inhibition of cytokine production, and modulation of the neuronal proteome. Front Immunol 2023; 14:1227268. [PMID: 37936684 PMCID: PMC10627171 DOI: 10.3389/fimmu.2023.1227268] [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: 05/23/2023] [Accepted: 10/02/2023] [Indexed: 11/09/2023] Open
Abstract
Introduction The antinociceptive and pharmacological activities of C-Phycocyanin (C-PC) and Phycocyanobilin (PCB) in the context of inflammatory arthritis remain unexplored so far. In the present study, we aimed to assess the protective actions of these compounds in an experimental mice model that replicates key aspects of human rheumatoid arthritis. Methods Antigen-induced arthritis (AIA) was established by intradermal injection of methylated bovine serum albumin in C57BL/6 mice, and one hour before the antigen challenge, either C-PC (2, 4, or 8 mg/kg) or PCB (0.1 or 1 mg/kg) were administered intraperitoneally. Proteome profiling was also conducted on glutamate-exposed SH-SY5Y neuronal cells to evaluate the PCB impact on this key signaling pathway associated with nociceptive neuronal sensitization. Results and discussion C-PC and PCB notably ameliorated hypernociception, synovial neutrophil infiltration, myeloperoxidase activity, and the periarticular cytokine concentration of IFN-γ, TNF-α, IL-17A, and IL-4 dose-dependently in AIA mice. In addition, 1 mg/kg PCB downregulated the gene expression for T-bet, RORγ, and IFN-γ in the popliteal lymph nodes, accompanied by a significant reduction in the pathological arthritic index of AIA mice. Noteworthy, neuronal proteome analysis revealed that PCB modulated biological processes such as pain, inflammation, and glutamatergic transmission, all of which are involved in arthritic pathology. Conclusions These findings demonstrate the remarkable efficacy of PCB in alleviating the nociception and inflammation in the AIA mice model and shed new light on mechanisms underlying the PCB modulation of the neuronal proteome. This research work opens a new avenue to explore the translational potential of PCB in developing a therapeutic strategy for inflammation and pain in rheumatoid arthritis.
Collapse
Affiliation(s)
- Javier Marín-Prida
- Center for Research and Biological Evaluations, Institute of Pharmacy and Food, University of Havana, Havana, Cuba
| | - Arielis Rodríguez-Ulloa
- Division of Biomedical Research, Center for Genetic Engineering and Biotechnology, Havana, Cuba
| | - Vladimir Besada
- Division of Biomedical Research, Center for Genetic Engineering and Biotechnology, Havana, Cuba
- China-Cuba Biotechnology Joint Innovation Center (CCBJIC), Yongzhou Zhong Gu Biotechnology Co. Ltd, Yongzhou, China
| | - Alexey Llopiz-Arzuaga
- Division of Biomedical Research, Center for Genetic Engineering and Biotechnology, Havana, Cuba
- Department of Cellular Engineering and Biocatalysis , Institute of Biotechnology, National Autonomous University of Mexico (UNAM), Cuernavaca, Mexico
| | - Nathália Vieira Batista
- Laboratory of Immunopharmacology, Department of Biochemistry and Immunology, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| | | | - Nancy Pavón-Fuentes
- Immunochemical Department, International Center for Neurological Restoration (CIREN), Havana, Cuba
| | - Érica Leandro Marciano Vieira
- Translational Psychoneuroimmunology Group, School of Medicine, Federal University of Minas Gerais (UFMG), Belo Horizonte, Brazil
| | - Viviana Falcón-Cama
- Division of Biomedical Research, Center for Genetic Engineering and Biotechnology, Havana, Cuba
- Departments of Physiological or Morphological Sciences, Latin American School of Medicine (ELAM), Havana, Cuba
| | - Emilio F. Acosta
- Department of Characterization, Center for Advanced Studies of Cuba, Havana, Cuba
| | - Gillian Martínez-Donato
- Division of Biomedical Research, Center for Genetic Engineering and Biotechnology, Havana, Cuba
| | - Majel Cervantes-Llanos
- Division of Biomedical Research, Center for Genetic Engineering and Biotechnology, Havana, Cuba
| | - Dai Lingfeng
- China-Cuba Biotechnology Joint Innovation Center (CCBJIC), Yongzhou Zhong Gu Biotechnology Co. Ltd, Yongzhou, China
| | - Luis J. González
- Division of Biomedical Research, Center for Genetic Engineering and Biotechnology, Havana, Cuba
| | | | - Gerardo Guillén-Nieto
- Division of Biomedical Research, Center for Genetic Engineering and Biotechnology, Havana, Cuba
- Departments of Physiological or Morphological Sciences, Latin American School of Medicine (ELAM), Havana, Cuba
| | - Eduardo Pentón-Arias
- Division of Biomedical Research, Center for Genetic Engineering and Biotechnology, Havana, Cuba
- Departments of Physiological or Morphological Sciences, Latin American School of Medicine (ELAM), Havana, Cuba
| | - Flávio Almeida Amaral
- Laboratory of Immunopharmacology, Department of Biochemistry and Immunology, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| | - Mauro Martins Teixeira
- Laboratory of Immunopharmacology, Department of Biochemistry and Immunology, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| | - Giselle Pentón-Rol
- Division of Biomedical Research, Center for Genetic Engineering and Biotechnology, Havana, Cuba
- Departments of Physiological or Morphological Sciences, Latin American School of Medicine (ELAM), Havana, Cuba
| |
Collapse
|
5
|
McDonald PP, Leifer FG, Basso J, Lasala D, Li D, Chen KJ, Zhang J, Perkins WR, Cipolla DC. Brensocatib (an oral, reversible inhibitor of dipeptidyl peptidase-1) attenuates disease progression in two animal models of rheumatoid arthritis. Front Immunol 2023; 14:1231047. [PMID: 37638021 PMCID: PMC10451067 DOI: 10.3389/fimmu.2023.1231047] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2023] [Accepted: 07/28/2023] [Indexed: 08/29/2023] Open
Abstract
Rheumatoid arthritis (RA) is a painful and incurable disease characterized by chronic joint inflammation and a progressive destruction of cartilage and bone. Although current treatments have improved clinical outcomes for some patients, the high relapse rates and sizeable proportion of non-responders emphasize the need for further research. Arthritic joints are massively infiltrated by neutrophils, which influence inflammatory and immune processes by releasing cytokines, chemokines, eicosanoids, and neutrophil serine proteases (NSPs) - all of which are known to contribute to RA initiation and progression. Active NSPs are generated from zymogens at the promyelocytic stage of neutrophil differentiation under the action of dipeptidyl peptidase 1 (DPP-1) and DPP-1 knockout mice are resistant to the development of arthritis. Thus, DPP-1 inhibition represents a promising therapeutic approach in RA. In this study, we assessed the efficacy of a potent and highly selective DPP-1 inhibitor, brensocatib, in two well established RA models - rat collagen-induced arthritis (CIA) and mouse collagen antibody-induced arthritis (CAIA). In both models, brensocatib at 3 and 30 mg/kg/day significantly reduced bone marrow NSP levels, in keeping with prior pharmacodynamic studies in rodents. More importantly, brensocatib treatment significantly improved disease score at both dosages in both rodent models. In the mouse CAIA model, brensocatib even proved at least as potent as anti-TNF antibodies in diminishing both the histopathological score and neutrophil infiltration into arthritic joints. Together, these results show that brensocatib alters RA disease progression in rodents and supports the need for its further evaluation as a potential therapeutic option, or to complement existing RA treatments.
Collapse
|
6
|
Moreira TP, Sousa CDFD, Melo Costa VRD, Queiroz-Junior CM, Santos FM, Bonilha CS, Ésper LM, Nogueira ML, Cunha TM, Teixeira MM, Costa VV, de Souza DDG. Tumour necrosis factor plays a deleterious role in the pathogenesis of chikungunya virus infection. Immunology 2023; 168:444-458. [PMID: 36164989 DOI: 10.1111/imm.13583] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2021] [Accepted: 09/01/2022] [Indexed: 11/28/2022] Open
Abstract
Arthralgia is a hallmark of chikungunya virus (CHIKV) infection and can be very debilitating and associated with a robust local inflammatory response. Many pathophysiological aspects associated with the disease remain to be elucidated. Here, we describe a novel model of CHIKV infection in immunocompetent mice and evaluate the role of tumour necrosis factor in the pathogenesis of the disease. C57BL/6 wild type (WT) or TNF receptor 1 deficient (TNFR1-/- ) mice were inoculated with 1 × 106 PFU of CHIKV in the paw. Alternatively, etanercept was used to inhibit TNF in infected WT mice. Hypernociception, inflammatory and virological analysis were performed. Inoculation of CHIKV into WT mice induced persistent hypernociception. There was significant viral replication in target organs and local production of inflammatory mediators in early time-points after infection. CHIKV infection was associated with specific humoral IgM and IgG responses. In TNFR1-/- mice, there was a decrease in the hypernociception threshold, which was associated with a milder local inflammatory response in the paw but delayed viral clearance. Local or systemic treatment with etanercept reduced CHIKV-induced hypernociception. This is the first study to describe hypernociception, a clinical correlation of arthralgia, in immunocompetent mice infected with CHIKV. It also demonstrates the dual role of TNF in contributing to viral clearance but driving tissue damage and hypernociception. Inhibition of TNF may have therapeutic benefits but its role in viral clearance suggests that viral levels must be monitored in CHIKV-infected patients and that TNF inhibitors should ideally be used in combination with anti-viral drugs.
Collapse
Affiliation(s)
- Thaiane Pinto Moreira
- Institute of Biological Sciences, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| | | | | | | | - Franciele Martins Santos
- Institute of Biological Sciences, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| | - Caio Santos Bonilha
- Institute of Biological Sciences, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brazil.,Department of Pharmacology, Ribeirão Preto Medical School, University of São Paulo, São Paulo, Brazil
| | - Lísia Maria Ésper
- Institute of Biological Sciences, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| | - Mauricio Lacerda Nogueira
- Department of Dermatological, Infectious and Parasitic Diseases, Medical School of São José do Rio Preto, São Paulo, São José do Rio Preto, Brazil
| | - Thiago Mattar Cunha
- Department of Pharmacology, Ribeirão Preto Medical School, University of São Paulo, São Paulo, Brazil
| | - Mauro Martins Teixeira
- Institute of Biological Sciences, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| | - Vivian Vasconcelos Costa
- Institute of Biological Sciences, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| | - Daniele da Glória de Souza
- Institute of Biological Sciences, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| |
Collapse
|
7
|
Felix FB, Dias J, Vago JP, Martins DG, Beltrami VA, Fernandes DDO, Menezes Dos Santos ACP, Queiroz-Junior CM, de Sousa LP, Amaral FA, Soriani FM, Teixeira MM, Pinho V. Blocking the HGF-MET pathway induces resolution of neutrophilic inflammation by promoting neutrophil apoptosis and efferocytosis. Pharmacol Res 2023; 188:106640. [PMID: 36627004 DOI: 10.1016/j.phrs.2022.106640] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/29/2022] [Revised: 12/07/2022] [Accepted: 12/27/2022] [Indexed: 01/09/2023]
Abstract
Inflammation resolution is an active process that involves cellular events such as apoptosis and efferocytosis, which are key steps in the restoration of tissue homeostasis. Hepatocyte growth factor (HGF) is a growth factor mostly produced by mesenchymal-origin cells and has been described to act via MET receptor tyrosine kinase. The HGF/MET axis is essential for determining the progression and severity of inflammatory and immune-mediated disorders. Here, we investigated the effect of blocking the HGF/MET signalling pathway by PF-04217903 on the resolution of established models of neutrophilic inflammation. In a self-resolving model of gout induced by MSU crystals, HGF expression on periarticular tissue peaked at 12 h, the same time point that neutrophils reach their maximal accumulation in the joints. The HGF/MET axis was activated in this model, as demonstrated by increased levels of MET phosphorylation in neutrophils (Ly6G+ cells). In addition, the number of neutrophils was reduced in the knee exudate after PF-04217903 treatment, an effect accompanied by increased neutrophil apoptosis and efferocytosis and enhanced expression of Annexin A1, a key molecule for inflammation resolution. Reduced MPO activity, IL-1β and CXCL1 levels were also observed in periarticular tissue. Importantly, PF-04217903 reduced the histopathological score and hypernociceptive response. Similar findings were obtained in LPS-induced neutrophilic pleurisy. In human neutrophils, the combined use of LPS and HGF increased MET phosphorylation and provided a prosurvival signal, whereas blocking MET with PF-04217903 induced caspase-dependent neutrophil apoptosis. Taken together, these data demonstrate that blocking HGF/MET signalling may be a potential therapeutic strategy for inducing the resolution of neutrophilic inflammatory responses.
Collapse
Affiliation(s)
- Franciel Batista Felix
- Departamento de Morfologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Julia Dias
- Departamento de Morfologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Juliana Priscila Vago
- Experimental Rheumatology, Department of Rheumatology, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, Nijmegen, the Netherlands
| | - Débora Gonzaga Martins
- Departamento de Morfologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Vinícius Amorim Beltrami
- Departamento de Morfologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Débora de Oliveira Fernandes
- Departamento de Morfologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | | | - Celso Martins Queiroz-Junior
- Departamento de Morfologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Lirlândia Pires de Sousa
- Departamento de Análises Clínicas e Toxicológicas, Faculdade de Farmácia, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Flávio Almeida Amaral
- Departamento de Bioquímica e Imunologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Frederico Marianetti Soriani
- Departamento de Genética, Ecologia e Evolução, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Mauro Martins Teixeira
- Departamento de Bioquímica e Imunologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Vanessa Pinho
- Departamento de Morfologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil.
| |
Collapse
|
8
|
The Therapeutic Effect of Phosphopeptide P140 Attenuates Inflammation Induced by Uric Acid Crystals in Gout Arthritis Mouse Model. Cells 2022; 11:cells11233709. [PMID: 36496970 PMCID: PMC9740613 DOI: 10.3390/cells11233709] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2022] [Revised: 11/17/2022] [Accepted: 11/18/2022] [Indexed: 11/23/2022] Open
Abstract
Gout is a painful form of inflammatory arthritis characterized by the deposition of monosodium urate (MSU) crystals in the joints. The aim of this study was to investigate the effect of peptide P140 on the inflammatory responses in crystal-induced mouse models of gout and cell models including MSU-treated human cells. Injection of MSU crystals into the knee joint of mice induced neutrophil influx and inflammatory hypernociception. Injection of MSU crystals subcutaneously into the hind paw induced edema and increased pro-inflammatory cytokines levels. Treatment with P140 effectively reduced hypernociception, the neutrophil influx, and pro-inflammatory cytokine levels in these experimental models. Furthermore, P140 modulated neutrophils chemotaxis in vitro and increased apoptosis pathways through augmented caspase 3 activity and reduced NFκB phosphorylation. Moreover, P140 increased the production of the pro-resolving mediator annexin A1 and decreased the expression of the autophagy-related ATG5-ATG12 complex and HSPA8 chaperone protein. Overall, these findings suggest that P140 exerts a significant beneficial effect in a neutrophilic inflammation observed in the model of gout that can be of special interest in the design of new therapeutic strategies.
Collapse
|
9
|
de Oliveira DP, Garcia EDF, de Oliveira MA, Candido LCM, Coelho FM, Costa VV, Batista NV, Queiroz-Junior CM, Brito LF, Sousa LP, Souza DG, Amaral FA, de Pádua RM, Teixeira MM, Braga FC. cis-Aconitic Acid, a Constituent of Echinodorus grandiflorus Leaves, Inhibits Antigen-Induced Arthritis and Gout in Mice. PLANTA MEDICA 2022; 88:1123-1131. [PMID: 34763354 DOI: 10.1055/a-1676-4371] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/13/2023]
Abstract
cis-Aconitic acid is a constituent from the leaves of Echinodorus grandiflorus, a medicinal plant traditionally used in Brazil to treat inflammatory conditions, including arthritic diseases. The present study aimed to investigate the anti-arthritic effect of cis-aconitic acid in murine models of antigen-induced arthritis and monosodium urate-induced gout. The possible underlying mechanisms of action was evaluated in THP-1 macrophages. Oral treatment with cis-aconitic acid (10, 30, and 90 mg/kg) reduced leukocyte accumulation in the joint cavity and C-X-C motif chemokine ligand 1 and IL-1β levels in periarticular tissue. cis-Aconitic acid treatment reduced joint inflammation in tissue sections of antigen-induced arthritis mice and these effects were associated with decreased mechanical hypernociception. Administration of cis-aconitic acid (30 mg/kg p. o.) also reduced leukocyte accumulation in the joint cavity after the injection of monosodium urate crystals. cis-Aconitic acid reduced in vitro the release of TNF-α and phosphorylation of IκBα in lipopolysaccharide-stimulated THP-1 macrophages, suggesting that inhibition of nuclear factor kappa B activation was an underlying mechanism of cis-aconitic acid-induced anti-inflammatory effects. In conclusion, cis-aconitic acid has significant anti-inflammatory effects in antigen-induced arthritis and monosodium urate-induced arthritis in mice, suggesting its potential for the treatment of inflammatory diseases of the joint in humans. Additionally, our findings suggest that this compound may contribute to the anti-inflammatory effect previously reported for E. grandiflorus extracts.
Collapse
Affiliation(s)
- Diego Pinto de Oliveira
- Department of Pharmaceutical Products, Faculty of Pharmacy, Universidade Federal de Minas Gerais, Belo Horizonte, MG, Brazil
| | - Eliana de Faria Garcia
- Department of Pharmaceutical Products, Faculty of Pharmacy, Universidade Federal de Minas Gerais, Belo Horizonte, MG, Brazil
| | - Mariana Assíria de Oliveira
- Department of Pharmaceutical Products, Faculty of Pharmacy, Universidade Federal de Minas Gerais, Belo Horizonte, MG, Brazil
| | - Luiza C M Candido
- Department of Microbiology, Institute of Biological Sciences, Universidade Federal de Minas Gerais, Belo Horizonte, MG, Brazil
| | - Fernanda M Coelho
- Department of Microbiology, Institute of Biological Sciences, Universidade Federal de Minas Gerais, Belo Horizonte, MG, Brazil
| | - Vivian Vasconcelos Costa
- Department of Microbiology, Institute of Biological Sciences, Universidade Federal de Minas Gerais, Belo Horizonte, MG, Brazil
| | - Nathália Vieira Batista
- Department of Biochemistry and Immunology, Institute of Biological Sciences, Universidade Federal de Minas Gerais, Belo Horizonte, MG, Brazil
| | - Celso Martins Queiroz-Junior
- Department of Biochemistry and Immunology, Institute of Biological Sciences, Universidade Federal de Minas Gerais, Belo Horizonte, MG, Brazil
| | - Larissa Froede Brito
- Department of Clinical and Toxicological Analyses, Faculty of Pharmacy, Universidade Federal de Minas Gerais, Belo Horizonte, MG, Brazil
| | - Lirlândia Pires Sousa
- Department of Clinical and Toxicological Analyses, Faculty of Pharmacy, Universidade Federal de Minas Gerais, Belo Horizonte, MG, Brazil
| | - Daniele G Souza
- Department of Microbiology, Institute of Biological Sciences, Universidade Federal de Minas Gerais, Belo Horizonte, MG, Brazil
| | - Flávio Almeida Amaral
- Department of Biochemistry and Immunology, Institute of Biological Sciences, Universidade Federal de Minas Gerais, Belo Horizonte, MG, Brazil
| | - Rodrigo Maia de Pádua
- Department of Pharmaceutical Products, Faculty of Pharmacy, Universidade Federal de Minas Gerais, Belo Horizonte, MG, Brazil
| | - Mauro Martins Teixeira
- Department of Biochemistry and Immunology, Institute of Biological Sciences, Universidade Federal de Minas Gerais, Belo Horizonte, MG, Brazil
| | - Fernão Castro Braga
- Department of Pharmaceutical Products, Faculty of Pharmacy, Universidade Federal de Minas Gerais, Belo Horizonte, MG, Brazil
| |
Collapse
|
10
|
Correa LB, Pádua TA, Alabarse PVG, Saraiva EM, Garcia EB, Amendoeira FC, Ferraris FK, Fukada SY, Rosas EC, Henriques MG. Protective effect of methyl gallate on murine antigen-induced arthritis by inhibiting inflammatory process and bone erosion. Inflammopharmacology 2022; 30:251-266. [PMID: 35112275 DOI: 10.1007/s10787-021-00922-8] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2021] [Accepted: 12/28/2021] [Indexed: 12/15/2022]
Abstract
Methyl gallate (MG) is a plant-derived phenolic compound known to present remarkable anti-inflammatory effect in different experimental models, such as paw oedema, pleurisy, zymosan-induced arthritis and colitis. Herein we investigated the effect of MG in the mice model of antigen-induced arthritis (AIA), a model with complex inflammatory response, driven primally by immune process and that cause bone and cartilage erosion similarly found in rheumatoid arthritis. Arthritis was induced by intra-articular injection of albumin methylated from bovine serum (mBSA) in C57BL/6 male mice previously immunized. The dose-response analysis of MG (0.7-70 mg/kg; p.o) showed that maximum inhibition was reached with the dose of 7 mg/kg on paw oedema and cell infiltration induced by AIA at 7 h. Treatment with MG (7 mg/kg; p.o) or with the positive control, dexamethasone (Dexa, 10 mg/kg, ip) reduced AIA oedema formation, leukocyte infiltration, release of extracellular DNA and cytokine production 7 and 24 h (acute response). Mice treated daily with MG for 7 days showed no significant weight loss or liver and kidney toxicity contrary to dexamethasone that induced some degree of toxicity. Prolonged treatment with MG inhibited the late inflammatory response (28 days) reducing oedema formation, cell infiltration, synovial hyperplasia, pannus formation and cartilage degradation as observed in histopathological analyses. Ultimately, MG reduced bone resorption as evidenced by a decrease in tartrate-resistant acid phosphate (TRAP)-positive cells number in femur histology. Altogether, we demonstrate that MG ameliorates the inflammatory reaction driven primarily by the immune process, suggesting a potential therapeutic application in arthritis treatment.
Collapse
Affiliation(s)
- Luana Barbosa Correa
- Laboratory of Applied Pharmacology, Farmanguinhos, Oswaldo Cruz Foundation, Rio de Janeiro, Brazil.,National Institute for Science and Technology on Innovation on Diseases of Neglected Populations (INCT/IDPN), Oswaldo Cruz Foundation, Rio de Janeiro, Brazil
| | - Tatiana Almeida Pádua
- Laboratory of Applied Pharmacology, Farmanguinhos, Oswaldo Cruz Foundation, Rio de Janeiro, Brazil.,National Institute for Science and Technology on Innovation on Diseases of Neglected Populations (INCT/IDPN), Oswaldo Cruz Foundation, Rio de Janeiro, Brazil
| | - Paulo Vinicius Gil Alabarse
- Department of Biomolecular Sciences, School of Pharmaceutical Sciences of Ribeirão Preto, University of São Paulo, Ribeirão Preto, SP, Brazil
| | - Elvira Maria Saraiva
- Laboratory of Immunobiology of Leishmaniasis, Department of Immunology, Institute of Microbiology Paulo de Góes, Federal University of Rio de Janeiro, Rio de Janeiro, RJ, Brazil
| | - Esdras Barbosa Garcia
- Laboratory of Pharmacology, Department of Pharmacology and Toxicology, National Institute of Health Quality Control (INCQS), Oswaldo Cruz Foundation, Rio de Janeiro, Brazil
| | - Fabio Coelho Amendoeira
- Laboratory of Pharmacology, Department of Pharmacology and Toxicology, National Institute of Health Quality Control (INCQS), Oswaldo Cruz Foundation, Rio de Janeiro, Brazil
| | - Fausto Klabund Ferraris
- Laboratory of Pharmacology, Department of Pharmacology and Toxicology, National Institute of Health Quality Control (INCQS), Oswaldo Cruz Foundation, Rio de Janeiro, Brazil
| | - Sandra Yasuyo Fukada
- Department of Biomolecular Sciences, School of Pharmaceutical Sciences of Ribeirão Preto, University of São Paulo, Ribeirão Preto, SP, Brazil
| | - Elaine Cruz Rosas
- Laboratory of Applied Pharmacology, Farmanguinhos, Oswaldo Cruz Foundation, Rio de Janeiro, Brazil.,National Institute for Science and Technology on Innovation on Diseases of Neglected Populations (INCT/IDPN), Oswaldo Cruz Foundation, Rio de Janeiro, Brazil
| | - Maria G Henriques
- Laboratory of Applied Pharmacology, Farmanguinhos, Oswaldo Cruz Foundation, Rio de Janeiro, Brazil. .,National Institute for Science and Technology on Innovation on Diseases of Neglected Populations (INCT/IDPN), Oswaldo Cruz Foundation, Rio de Janeiro, Brazil.
| |
Collapse
|
11
|
Schneider AH, Machado CC, Veras FP, Maganin AGDM, de Souza FFL, Barroso LC, de Oliveira RDR, Alves-Filho JC, Cunha TM, Fukada SY, Louzada-Júnior P, da Silva TA, Cunha FQ. Neutrophil extracellular traps mediate joint hyperalgesia induced by immune inflammation. Rheumatology (Oxford) 2021; 60:3461-3473. [PMID: 33367912 DOI: 10.1093/rheumatology/keaa794] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2020] [Revised: 10/19/2020] [Indexed: 11/13/2022] Open
Abstract
OBJECTIVE To evaluate the role of neutrophil extracellular traps (NETs) in the genesis of joint hyperalgesia using an experimental model of arthritis and transpose the findings to clinical investigation. METHODS C57BL/6 mice were subjected to antigen-induced arthritis (AIA) and treated with Pulmozyme (PLZ) to degrade NETs or Cl-amidine to inhibit NET production. Oedema formation, the histopathological score and mechanical hyperalgesia were evaluated. NETs were injected intra-articularly in wild type (WT), Tlr4-/-, Tlr9-/-, Tnfr1-/- and Il1r-/- mice, and the levels of cytokines and Cox2 expression were quantified. NETs were also quantified from human neutrophils isolated from RA patients and individual controls. RESULTS AIA mice had increased NET concentration in joints, accompanied by increased Padi4 gene expression in the joint cells. Treatment of AIA mice with a peptidyl arginine deiminase 4 inhibitor or with PLZ inhibited the joint hyperalgesia. Moreover, the injection of NETs into joints of naïve animals generated a dose-dependent reduction of mechanical threshold, an increase of articular oedema, inflammatory cytokine production and cyclooxygenase-2 expression. In mice deficient for Tnfr1, Il1r, Tlr4 and Tlr9, joint hyperalgesia induced by NETs was prevented. Last, we found that neutrophils from RA patients were more likely to release NETs, and the increase in synovial fluid NET concentration correlated with an increase in joint pain. CONCLUSION The findings indicate that NETs cause hyperalgesia possibly through Toll-like receptor (TLR)-4 and TLR-9. These data support the idea that NETs contribute to articular pain, and this pathway can be an alternative target for the treatment of pain in RA.
Collapse
Affiliation(s)
- Ayda Henriques Schneider
- Center of Research of Inflammatory Diseases, CRID.,Department of Pharmacology, Ribeirão Preto Medical School
| | - Caio Cavalcante Machado
- Center of Research of Inflammatory Diseases, CRID.,Department of Medicine, Clinical Immunology Division, Medicine Faculty of Ribeirão Preto
| | - Flávio Protásio Veras
- Center of Research of Inflammatory Diseases, CRID.,Department of Pharmacology, Ribeirão Preto Medical School
| | | | - Flávio Falcão Lima de Souza
- Center of Research of Inflammatory Diseases, CRID.,Department of Medicine, Clinical Immunology Division, Medicine Faculty of Ribeirão Preto
| | - Lívia Corrêa Barroso
- Center of Research of Inflammatory Diseases, CRID.,Department of Medicine, Clinical Immunology Division, Medicine Faculty of Ribeirão Preto
| | - Renê Donizeti Ribeiro de Oliveira
- Center of Research of Inflammatory Diseases, CRID.,Department of Medicine, Clinical Immunology Division, Medicine Faculty of Ribeirão Preto
| | - José Carlos Alves-Filho
- Center of Research of Inflammatory Diseases, CRID.,Department of Pharmacology, Ribeirão Preto Medical School
| | - Thiago Mattar Cunha
- Center of Research of Inflammatory Diseases, CRID.,Department of Pharmacology, Ribeirão Preto Medical School
| | - Sandra Yasuyo Fukada
- Center of Research of Inflammatory Diseases, CRID.,Department of Bio Molecular Sciences, School of Pharmaceutical Science, University of São Paulo, Ribeirão Preto, São Paulo
| | - Paulo Louzada-Júnior
- Center of Research of Inflammatory Diseases, CRID.,Department of Medicine, Clinical Immunology Division, Medicine Faculty of Ribeirão Preto
| | - Tarcília Aparecida da Silva
- Center of Research of Inflammatory Diseases, CRID.,Department of Oral Surgery and Pathology, Faculty of Dentistry, Federal University of Minas Gerais, Belo Horizonte, Brazil
| | - Fernando Queiroz Cunha
- Center of Research of Inflammatory Diseases, CRID.,Department of Pharmacology, Ribeirão Preto Medical School
| |
Collapse
|
12
|
Felix FB, Vago JP, Fernandes DDO, Martins DG, Moreira IZ, Gonçalves WA, Costa WC, Araújo JMD, Queiroz-Junior CM, Campolina-Silva GH, Soriani FM, Sousa LP, Grespan R, Teixeira MM, Pinho V. Biochanin A Regulates Key Steps of Inflammation Resolution in a Model of Antigen-Induced Arthritis via GPR30/PKA-Dependent Mechanism. Front Pharmacol 2021; 12:662308. [PMID: 33995086 PMCID: PMC8114065 DOI: 10.3389/fphar.2021.662308] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2021] [Accepted: 03/29/2021] [Indexed: 12/12/2022] Open
Abstract
Biochanin A (BCA) is a natural organic compound of the class of phytochemicals known as flavonoids and isoflavone subclass predominantly found in red clover (Trifolium pratense). It has anti-inflammatory activity and some pro-resolving actions, such as neutrophil apoptosis. However, the effect of BCA in the resolution of inflammation is still poorly understood. In this study, we investigated the effects of BCA on the neutrophilic inflammatory response and its resolution in a model of antigen-induced arthritis. Male wild-type BALB/c mice were treated with BCA at the peak of the inflammatory process (12 h). BCA decreased the accumulation of migrated neutrophils, and this effect was associated with reduction of myeloperoxidase activity, IL-1β and CXCL1 levels, and the histological score in periarticular tissues. Joint dysfunction, as seen by mechanical hypernociception, was improved by treatment with BCA. The resolution interval (Ri) was also quantified, defining profiles of acute inflammatory parameters that include the amplitude and duration of the inflammatory response monitored by the neutrophil infiltration. BCA treatment shortened Ri from ∼23 h observed in vehicle-treated mice to ∼5.5 h, associated with an increase in apoptotic events and efferocytosis, both key steps for the resolution of inflammation. These effects of BCA were prevented by H89, an inhibitor of protein kinase A (PKA) and G15, a selective G protein–coupled receptor 30 (GPR30) antagonist. In line with the in vivo data, BCA also increased the efferocytic ability of murine bone marrow–derived macrophages. Collectively, these data indicate for the first time that BCA resolves neutrophilic inflammation acting in key steps of the resolution of inflammation, requiring activation of GPR30 and via stimulation of cAMP-dependent signaling.
Collapse
Affiliation(s)
- Franciel Batista Felix
- Departamento de Morfologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Juliana Priscila Vago
- Departamento de Morfologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Débora de Oliveira Fernandes
- Departamento de Morfologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Débora Gonzaga Martins
- Departamento de Morfologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Isabella Zaidan Moreira
- Departamento de Análises Clínicas e Toxicológicas, Faculdade de Farmácia, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - William Antonio Gonçalves
- Departamento de Morfologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Walyson Coelho Costa
- Departamento de Morfologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | | | - Celso Martins Queiroz-Junior
- Departamento de Morfologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | | | - Frederico Marianetti Soriani
- Departamento de Genética, Ecologia e Evolução, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Lirlândia Pires Sousa
- Departamento de Análises Clínicas e Toxicológicas, Faculdade de Farmácia, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Renata Grespan
- Departamento de Fisiologia, Universidade Federal de Sergipe, São Cristovão, Brazil
| | - Mauro Martins Teixeira
- Departamento de Bioquímica e Imunologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Vanessa Pinho
- Departamento de Morfologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| |
Collapse
|
13
|
S. mansoni SmKI-1 Kunitz-domain: Leucine point mutation at P1 site generates enhanced neutrophil elastase inhibitory activity. PLoS Negl Trop Dis 2021; 15:e0009007. [PMID: 33465126 PMCID: PMC7846107 DOI: 10.1371/journal.pntd.0009007] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2020] [Revised: 01/29/2021] [Accepted: 11/24/2020] [Indexed: 12/14/2022] Open
Abstract
The Schistosoma mansoni SmKI-1 protein is composed of two domains: a Kunitz-type serine protease inhibitor motif (KD) and a C-terminus domain with no similarity outside the genera. Our previous work has demonstrated that KD plays an essential role in neutrophil elastase (NE) binding blockage, in neutrophil influx and as a potential anti-inflammatory molecule. In order to enhance NE blocking capacity, we analyzed the KD sequence from a structure-function point of view and designed specific point mutations in order to enhance NE affinity. We substituted the P1 site residue at the reactive site for a leucine (termed RL-KD), given its central role for KD's inhibition to NE. We have also substituted a glutamic acid that strongly interacts with the P1 residue for an alanine, to help KD to be buried on NE S1 site (termed EA-KD). KD and the mutant proteins were evaluated in silico by molecular docking to human NE, expressed in Escherichia coli and tested towards its NE inhibitory activity. Both mutated proteins presented enhanced NE inhibitory activity in vitro and RL-KD presented the best performance. We further tested RL-KD in vivo in an experimental model of monosodium urate (MSU)-induced acute arthritis. RL-KD showed reduced numbers of total cells and neutrophils in the mouse knee cavity when compared to KD. Nevertheless, both RL-KD and KD reduced mice hypernociception in a similar fashion. In summary, our results demonstrated that both mutated proteins showed enhanced NE inhibitory activity in vitro. However, RL-KD had a prominent effect in diminishing inflammatory parameters in vivo.
Collapse
|
14
|
Lopes F, Vicentini FA, Cluny NL, Mathews AJ, Lee BH, Almishri WA, Griffin L, Gonçalves W, Pinho V, McKay DM, Hirota SA, Swain MG, Pittman QJ, Sharkey KA. Brain TNF drives post-inflammation depression-like behavior and persistent pain in experimental arthritis. Brain Behav Immun 2020; 89:224-232. [PMID: 32592863 DOI: 10.1016/j.bbi.2020.06.023] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/18/2020] [Revised: 06/02/2020] [Accepted: 06/18/2020] [Indexed: 12/21/2022] Open
Abstract
Patients with rheumatoid arthritis experience chronic pain, depression and fatigue, even when inflammation of the joints is well controlled. To study the relationship between arthritis, depression, and sustained pain when articular inflammation is no longer observed, we tested the hypothesis that brain TNF drives post-inflammation depression-like behavior and persistent pain in experimental arthritis. The murine model of antigen-induced arthritis (AIA) was used to evaluate the effects of knee inflammation on sustained pain and depression-like behavior. We measured joint pain using an automated dynamic plantar algesiometer and depression-like behavior with the tail suspension test. Cytokines were measured by Luminex assay and ELISA. TNF in the brain was blocked by intracerebroventricular injection of anti-TNF antibodies. Histological damage and elevated levels of cytokines were observed in the knee 24 h after antigen treatment, but not at 13 days. Reduced pain thresholds were seen 24 h and 13 days after treatment. Depression-like behavior was observed on day 13. Treatment with the antidepressant imipramine reduced both depression-like behavior and persistent pain. However, blocking joint pain with the analgesic dipyrone did not alter depression-like behavior. Elevated levels of TNF, CCL2, and CXCL-1 were observed in the hippocampus 24 h after treatment, with TNF remaining elevated at day 13. Intracerebroventricular infusion of an anti-TNF antibody blocked depression-like behavior and reduced persistent pain. We have demonstrated that depression-like behavior and pain is sustained in AIA mice after the resolution of inflammation. These changes are associated with elevated levels of TNF in the hippocampus and are dependent upon brain TNF. The findings reveal an important mechanistic link between the expression of chronic pain and depression in experimental arthritis. Furthermore, they suggest treating depression in rheumatoid arthritis may positively impact other debilitating features of this condition.
Collapse
Affiliation(s)
- Fernando Lopes
- Institute of Parasitology, McGill University, Ste-Anne-de-Bellevue, QC, Canada; Department of Microbiology and Immunology, McGill University, Montreal, QC, Canada.
| | - Fernando A Vicentini
- Department of Physiology and Pharmacology, University of Calgary, Calgary, AB, Canada; Hotchkiss Brain Institute, University of Calgary, Calgary, AB, Canada; Calvin, Phoebe and Joan Snyder Institute for Chronic Diseases, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada
| | - Nina L Cluny
- Department of Physiology and Pharmacology, University of Calgary, Calgary, AB, Canada; Hotchkiss Brain Institute, University of Calgary, Calgary, AB, Canada; Calvin, Phoebe and Joan Snyder Institute for Chronic Diseases, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada
| | - Alexander J Mathews
- Department of Physiology and Pharmacology, University of Calgary, Calgary, AB, Canada; Hotchkiss Brain Institute, University of Calgary, Calgary, AB, Canada; Calvin, Phoebe and Joan Snyder Institute for Chronic Diseases, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada
| | - Benjamin H Lee
- Department of Physiology and Pharmacology, University of Calgary, Calgary, AB, Canada; Hotchkiss Brain Institute, University of Calgary, Calgary, AB, Canada; Calvin, Phoebe and Joan Snyder Institute for Chronic Diseases, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada
| | - Wagdi A Almishri
- Calvin, Phoebe and Joan Snyder Institute for Chronic Diseases, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada; Division of Gastroenterology and Hepatology, Department of Medicine, University of Calgary, Calgary, AB, Canada
| | - Lateece Griffin
- Department of Physiology and Pharmacology, University of Calgary, Calgary, AB, Canada; Hotchkiss Brain Institute, University of Calgary, Calgary, AB, Canada; Calvin, Phoebe and Joan Snyder Institute for Chronic Diseases, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada
| | - William Gonçalves
- Departamento de Morfologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, MG, Brazil
| | - Vanessa Pinho
- Departamento de Morfologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, MG, Brazil
| | - Derek M McKay
- Department of Physiology and Pharmacology, University of Calgary, Calgary, AB, Canada; Calvin, Phoebe and Joan Snyder Institute for Chronic Diseases, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada
| | - Simon A Hirota
- Department of Physiology and Pharmacology, University of Calgary, Calgary, AB, Canada; Calvin, Phoebe and Joan Snyder Institute for Chronic Diseases, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada
| | - Mark G Swain
- Calvin, Phoebe and Joan Snyder Institute for Chronic Diseases, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada; Division of Gastroenterology and Hepatology, Department of Medicine, University of Calgary, Calgary, AB, Canada
| | - Quentin J Pittman
- Department of Physiology and Pharmacology, University of Calgary, Calgary, AB, Canada; Hotchkiss Brain Institute, University of Calgary, Calgary, AB, Canada; Calvin, Phoebe and Joan Snyder Institute for Chronic Diseases, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada
| | - Keith A Sharkey
- Department of Physiology and Pharmacology, University of Calgary, Calgary, AB, Canada; Hotchkiss Brain Institute, University of Calgary, Calgary, AB, Canada; Calvin, Phoebe and Joan Snyder Institute for Chronic Diseases, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada.
| |
Collapse
|
15
|
Boff D, Oliveira VLS, Queiroz Junior CM, Galvão I, Batista NV, Gouwy M, Menezes GB, Cunha TM, Verri Junior WA, Proost P, Teixeira MM, Amaral FA. Lipoxin A 4 impairs effective bacterial control and potentiates joint inflammation and damage caused by Staphylococcus aureus infection. FASEB J 2020; 34:11498-11510. [PMID: 32741032 DOI: 10.1096/fj.201802830rr] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2018] [Revised: 05/12/2020] [Accepted: 05/15/2020] [Indexed: 02/07/2023]
Abstract
Staphylococcus aureus is the main cause of septic arthritis in humans, a disease associated with high morbidity and mortality. Inflammation triggered in response to infection is fundamental to control bacterial growth but may cause permanent tissue damage. Here, we evaluated the role of Lipoxin A4 (LXA4 ) in S aureus-induced arthritis in mice. Septic arthritis was induced by S aureus injection into tibiofemoral joints. At different time points, we evaluated cell recruitment and bacterial load in the joint, the production of pro-inflammatory molecules, and LXA4 in inflamed tissue and analyzed joint damage and dysfunction. LXA4 was investigated using genetically modified mice or by pharmacological blockade of its synthesis and receptor. CD11c+ cells were evaluated in lymph nodes by confocal microscopy and flow cytometry and dendritic cell chemotaxis using the Boyden chamber. Absence or pharmacological blockade of 5-lipoxygenase (5-LO) reduced joint inflammation and dysfunction and was associated with better control of infection at 4 to 7 days after the infection. There was an increase in LXA4 in joints of S aureus-infected mice and administration of LXA4 reversed the phenotype in 5-LO-/- mice. Blockade or absence of the LXA4 receptor FPR2 has a phenotype similar to 5-LO-/- mice. Mechanistically, LXA4 appeared to control migration and function of dendritic cells, cells shown to be crucial for adequate protective responses in the model. Thus, after the first days of infection when symptoms become evident therapies that inhibit LXA4 synthesis or action could be useful for treatment of S aureus-induced arthritis.
Collapse
Affiliation(s)
- Daiane Boff
- Immunopharmacology, Department of Biochemistry and Immunology, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil.,Laboratory of Molecular Immunology, Department of Microbiology and Immunology, Rega Institute for Medical Research, KU Leuven, Leuven, Belgium
| | - Vivian Louise Soares Oliveira
- Immunopharmacology, Department of Biochemistry and Immunology, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Celso M Queiroz Junior
- Department of Morphology, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Izabela Galvão
- Immunopharmacology, Department of Biochemistry and Immunology, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Nathalia Vieira Batista
- Immunopharmacology, Department of Biochemistry and Immunology, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Mieke Gouwy
- Laboratory of Molecular Immunology, Department of Microbiology and Immunology, Rega Institute for Medical Research, KU Leuven, Leuven, Belgium
| | - Gustavo Batista Menezes
- Departamento de Morfologia, Centro de Biologia Gastrointestinal, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Thiago Mattar Cunha
- Department of Pharmacology, Faculdade de Medicina de Ribeirao Preto, Center for Research in Inflammatory Diseases, Universidade de São Paulo, São Paulo, Brazil
| | | | - Paul Proost
- Laboratory of Molecular Immunology, Department of Microbiology and Immunology, Rega Institute for Medical Research, KU Leuven, Leuven, Belgium
| | - Mauro Martins Teixeira
- Immunopharmacology, Department of Biochemistry and Immunology, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Flávio Almeida Amaral
- Immunopharmacology, Department of Biochemistry and Immunology, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| |
Collapse
|
16
|
Mechanism of aspirin-induced inhibition on the secondary hyperalgesia in osteoarthritis model rats. Heliyon 2020; 6:e03963. [PMID: 32478188 PMCID: PMC7248669 DOI: 10.1016/j.heliyon.2020.e03963] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2019] [Revised: 04/24/2020] [Accepted: 05/06/2020] [Indexed: 01/23/2023] Open
Abstract
Aims The daily activity of osteoarthritis (OA) patients is limited by chronic pain and central sensitization. Although non-steroidal anti-inflammatory drugs (NSAIDs) and acetaminophen are the first-line drugs for the treatment of OA-related pain, their efficacy on central sensitization remains unclear. In the present study, we evaluated the effect of acetylsalicylic acid (ASA, Aspirin) using an OA model induced by monosodium iodoacetate (MIA), which has a similar disease progression to human OA. Main methods Secondary hyperalgesia was assessed at the plantar surface of the hind paw by Von Frey test. We evaluated the expression of acid-sensing ion channel 3 (ASIC3) in dorsal root ganglia and that of tumor necrosis factor-α (TNF-α) and interleukin-1β (IL-1β) in the spinal cord, which may cause secondary hyperalgesia in OA, by immunohistochemical analysis and real-time qPCR. Key findings The administration of ASA attenuated secondary hyperalgesia at 1–3 weeks after MIA, while celecoxib, a selective cyclooxygenase (COX)-2 inhibitor, failed to attenuate secondary hyperalgesia at week 2 after MIA injection, suggesting that ASA exerts its analgesic effect through a COX-2-independent pathway. Immunohistochemical analysis of the dorsal root ganglia indicated that ASA reduced the expression of ASIC3 during OA progression. Expression of TNF-α mRNA, but not IL-1β mRNA, in the spinal cord following MIA injection was suppressed by ASA administration. Significance These findings suggest that ASA may have the ability to attenuate secondary hyperalgesia through suppression of ASIC3 and/or TNF-α expression. ASA is therefore a clinically useful analgesic drug for treatment of secondary hyperalgesia in OA.
Collapse
|
17
|
In-depth characterization of a novel live-attenuated Mayaro virus vaccine candidate using an immunocompetent mouse model of Mayaro disease. Sci Rep 2020; 10:5306. [PMID: 32210270 PMCID: PMC7093544 DOI: 10.1038/s41598-020-62084-x] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2019] [Accepted: 02/24/2020] [Indexed: 02/06/2023] Open
Abstract
Mayaro virus (MAYV) is endemic in South American countries where it is responsible for sporadic outbreaks of acute febrile illness. The hallmark of MAYV infection is a highly debilitating and chronic arthralgia. Although MAYV emergence is a potential threat, there are no specific therapies or licensed vaccine. In this study, we developed a murine model of MAYV infection that emulates many of the most relevant clinical features of the infection in humans and tested a live-attenuated MAYV vaccine candidate (MAYV/IRES). Intraplantar inoculation of a WT strain of MAYV into immunocompetent mice induced persistent hypernociception, transient viral replication in target organs, systemic production of inflammatory cytokines, chemokines and specific humoral IgM and IgG responses. Inoculation of MAYV/IRES in BALB/c mice induced strong specific cellular and humoral responses. Moreover, MAYV/IRES vaccination of immunocompetent and interferon receptor-defective mice resulted in protection from disease induced by the virulent wt MAYV strain. Thus, this study describes a novel model of MAYV infection in immunocompetent mice and highlights the potential role of a live-attenuated MAYV vaccine candidate in host's protection from disease induced by a virulent MAYV strain.
Collapse
|
18
|
Gonçalves WA, Rezende BM, de Oliveira MPE, Ribeiro LS, Fattori V, da Silva WN, Prazeres PHDM, Queiroz-Junior CM, Santana KTDO, Costa WC, Beltrami VA, Costa VV, Birbrair A, Verri WA, Lopes F, Cunha TM, Teixeira MM, Amaral FA, Pinho V. Sensory Ganglia-Specific TNF Expression Is Associated With Persistent Nociception After Resolution of Inflammation. Front Immunol 2020; 10:3120. [PMID: 32038637 PMCID: PMC6984351 DOI: 10.3389/fimmu.2019.03120] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2019] [Accepted: 12/20/2019] [Indexed: 12/26/2022] Open
Abstract
Joint pain is a distressing symptom of arthritis, and it is frequently persistent even after treatments which reduce local inflammation. Continuous production of algogenic factors activate/sensitize nociceptors in the joint structures and contribute to persistent pain, a challenging and difficult condition to treat. TNF is a crucial cytokine for the pathogenesis of several rheumatic diseases, and its inhibition is a mainstay of treatment to control joint symptoms, including pain. Here, we sought to investigate the inflammatory changes and the role of TNF in dorsal root ganglia (DRG) during persistent hypernociception after the resolution of acute joint inflammation. Using a model of antigen-induced arthritis, the peak of joint inflammation occurred 12–24 h after local antigen injection and was characterized by an intense influx of neutrophils, pro-inflammatory cytokine production, and joint damage. We found that inflammatory parameters in the joint returned to basal levels between 6 and 8 days after antigen-challenge, characterizing the resolving phase of joint inflammation. Mechanical hyperalgesia was persistent up to 14 days after joint insult. The persistent nociception was associated with the inflammatory status of DRG after cessation of acute joint inflammation. The late state of neuroinflammation in the ipsilateral side was evidenced by gene expression of TNF, TNFR2, IL-6, IL-1β, CXCL2, COX2, and iNOS in lumbar DRG (L3-L5) and leukocyte adhesion in the lumbar intumescent vessels between days 6 and 8. Moreover, there were signs of resident macrophage activation in DRG, as evidenced by an increase in Iba1-positive cells. Intrathecal or systemic injection of etanercept, an agent clinically utilized for TNF neutralization, at day 7 post arthritis induction, alleviated the persistent joint hyperalgesia by specific action in DRG. Our data suggest that neuroinflammation in DRG after the resolution of acute joint inflammation drives continuous neural sensitization resulting in persistent joint nociception in a TNF-dependent mechanism.
Collapse
Affiliation(s)
- William Antonio Gonçalves
- Departamento de Morfologia, Instituto de Ciências Biológicas (ICB), Universidade Federal de Minas Gerais (UFMG), Belo Horizonte, Brazil
| | - Barbara Maximino Rezende
- Departamento de Enfermagem Básica, Escola de Enfermagem da Universidade Federal de Minas Gerais (UFMG), Belo Horizonte, Brazil
| | - Marcos Paulo Esteves de Oliveira
- Departamento de Morfologia, Instituto de Ciências Biológicas (ICB), Universidade Federal de Minas Gerais (UFMG), Belo Horizonte, Brazil
| | - Lucas Secchim Ribeiro
- Biomediziniches Zentrum (BMZ), Institut für Angeborene Immunität, Rheinische Friedrich-Wilhelms-Universität Bonn, Venusberg, Germany
| | - Victor Fattori
- Departamento de Patologia, Center of Biological Sciences, Londrina State University, Londrina, Brazil
| | - Walison Nunes da Silva
- Departamento de Patologia, Instituto de Ciências Biológicas (ICB), Universidade Federal de Minas Gerais (UFMG), Belo Horizonte, Brazil
| | | | - Celso Martins Queiroz-Junior
- Departamento de Morfologia, Instituto de Ciências Biológicas (ICB), Universidade Federal de Minas Gerais (UFMG), Belo Horizonte, Brazil
| | - Karina Talita de Oliveira Santana
- Departamento de Genética, Ecologia e Evolução, Instituto de Ciências Biológicas (ICB), Universidade Federal de Minas Gerais (UFMG), Belo Horizonte, Brazil
| | - Walyson Coelho Costa
- Departamento de Morfologia, Instituto de Ciências Biológicas (ICB), Universidade Federal de Minas Gerais (UFMG), Belo Horizonte, Brazil
| | - Vinícius Amorim Beltrami
- Departamento de Morfologia, Instituto de Ciências Biológicas (ICB), Universidade Federal de Minas Gerais (UFMG), Belo Horizonte, Brazil
| | - Vivian Vasconcelos Costa
- Departamento de Morfologia, Instituto de Ciências Biológicas (ICB), Universidade Federal de Minas Gerais (UFMG), Belo Horizonte, Brazil
| | - Alexander Birbrair
- Departamento de Patologia, Instituto de Ciências Biológicas (ICB), Universidade Federal de Minas Gerais (UFMG), Belo Horizonte, Brazil
| | - Waldiceu A Verri
- Departamento de Patologia, Center of Biological Sciences, Londrina State University, Londrina, Brazil
| | - Fernando Lopes
- Institute of Parasitology and Department of Microbiology and Immunology, McGill University, Montreal, QC, Canada
| | - Thiago Mattar Cunha
- Departamento de Farmacologia, Faculdade de Medicina de Ribeirão Preto, Universidade de São Paulo (USP), Ribeirão Preto, Brazil
| | - Mauro Martins Teixeira
- Departamento de Bioquímica e Imunologia, Instituto de Ciências Biológicas (ICB), Universidade Federal de Minas Gerais (UFMG), Belo Horizonte, Brazil
| | - Flávio Almeida Amaral
- Departamento de Bioquímica e Imunologia, Instituto de Ciências Biológicas (ICB), Universidade Federal de Minas Gerais (UFMG), Belo Horizonte, Brazil
| | - Vanessa Pinho
- Departamento de Morfologia, Instituto de Ciências Biológicas (ICB), Universidade Federal de Minas Gerais (UFMG), Belo Horizonte, Brazil
| |
Collapse
|
19
|
Tavares LD, Galvão I, Costa VV, Batista NV, Rossi LCR, Brito CB, Reis AC, Queiroz-Junior CM, Braga AD, Coelho FM, Dias AC, Zamboni DS, Pinho V, Teixeira MM, Amaral FA, Souza DG. Phosphoinositide-3 kinase gamma regulates caspase-1 activation and leukocyte recruitment in acute murine gout. J Leukoc Biol 2019; 106:619-629. [PMID: 31392775 DOI: 10.1002/jlb.ma1118-470rr] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2019] [Revised: 07/09/2019] [Accepted: 07/14/2019] [Indexed: 12/20/2022] Open
Abstract
This study investigates the participation of PI3Kγ in the development of joint inflammation and dysfunction in an experimental model of acute gout in mice. Acute gout was induced by injection of monosodium urate (MSU) crystals into the tibiofemoral joint of mice. The involvement of PI3Kγ was evaluated using a selective inhibitor and mice deficient for PI3Kγ (PI3Kγ-/- ) or with loss of kinase activity. Neutrophils recovered from the inflamed joint were quantified and stained for phosphorylated Akt (pAkt) and production of reactive oxygen species (ROS). The adherence of leukocytes to the joint microvasculature was assessed by intravital microscopy and cleaved caspase-1 by Western blot. Injection of MSU crystals induced massive accumulation of neutrophils expressing phosphorylated Akt. In the absence of PI3Kγ, there was reduction of pAkt expression, chemokine production, and neutrophil recruitment. Genetic or pharmacological inhibition of PI3Kγ reduced the adherence of leukocytes to the joint microvasculature, even in joints with established inflammation. Neutrophils from PI3Kγ-/- mice produced less ROS than wild-type neutrophils. There was decreased joint damage and dysfunction in the absence of PI3Kγ. In addition, in the absence of PI3Kγ activity, there was reduction of cleaved caspase-1 and IL-1β production in synovial tissue after injection of MSU crystals and leukotriene B4 . Our studies suggest that PI3Kγ is crucial for MSU crystal-induced acute joint inflammation. It is necessary for regulating caspase-1 activation and for mediating neutrophil migration and activation. Drugs that impair PI3Kγ function may be useful to control acute gout inflammation.
Collapse
Affiliation(s)
- Lívia D Tavares
- Departamento de Microbiologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| | - Izabela Galvão
- Departamento de Morfologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| | - Vivian V Costa
- Departamento de Morfologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| | - Nathalia V Batista
- Departamento de Bioquímica e Imunologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| | - Lívia C R Rossi
- Departamento de Microbiologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| | - Camila B Brito
- Departamento de Microbiologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| | - Alesandra C Reis
- Departamento de Morfologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| | - Celso M Queiroz-Junior
- Departamento de Morfologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| | - Amanda D Braga
- Departamento de Morfologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| | - Fernanda M Coelho
- Vollum Institute, Oregon Health and Science University, Portland, Oregon, USA
| | - Ana C Dias
- Departamento de Bioquímica e Imunologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| | - Dario S Zamboni
- Department of Cell Biology, Medical School of Ribeirão Preto, University of São Paulo FMRP/USP, Ribeirão Preto, São Paulo, Brazil
| | - Vanessa Pinho
- Departamento de Morfologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| | - Mauro M Teixeira
- Departamento de Bioquímica e Imunologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| | - Flávio A Amaral
- Departamento de Bioquímica e Imunologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| | - Daniele G Souza
- Departamento de Microbiologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| |
Collapse
|
20
|
|
21
|
Valiate BVS, Alvarez RU, Karra L, Queiroz‐Júnior CM, Amaral FA, Levi‐Schaffer F, Teixeira MM. The immunoreceptor CD300a controls the intensity of inflammation and dysfunction in a model of Ag‐induced arthritis in mice. J Leukoc Biol 2019; 106:957-966. [DOI: 10.1002/jlb.3a1018-389r] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2018] [Revised: 03/21/2019] [Accepted: 05/10/2019] [Indexed: 12/12/2022] Open
Affiliation(s)
- Bruno V. S. Valiate
- Departamento de Bioquímica e ImunologiaInstituto de Ciências Biológicas, Universidade Federal de Minas Gerais Belo Horizonte Brazil
| | - Rodrigo U. Alvarez
- Departamento de Bioquímica e ImunologiaInstituto de Ciências Biológicas, Universidade Federal de Minas Gerais Belo Horizonte Brazil
| | - Laila Karra
- Pharmacology and Experimental Therapeutics UnitInstitute for Drug Research, School of Pharmacy, Faculty of Medicine, The Hebrew University of Jerusalem Jerusalem Israel
| | | | - Flavio A. Amaral
- Departamento de Bioquímica e ImunologiaInstituto de Ciências Biológicas, Universidade Federal de Minas Gerais Belo Horizonte Brazil
| | - Francesca Levi‐Schaffer
- Pharmacology and Experimental Therapeutics UnitInstitute for Drug Research, School of Pharmacy, Faculty of Medicine, The Hebrew University of Jerusalem Jerusalem Israel
| | - Mauro M. Teixeira
- Departamento de Bioquímica e ImunologiaInstituto de Ciências Biológicas, Universidade Federal de Minas Gerais Belo Horizonte Brazil
| |
Collapse
|
22
|
Reis PVM, Boff D, Verly RM, Melo-Braga MN, Cortés ME, Santos DM, Pimenta AMDC, Amaral FA, Resende JM, de Lima ME. LyeTxI-b, a Synthetic Peptide Derived From Lycosa erythrognatha Spider Venom, Shows Potent Antibiotic Activity in Vitro and in Vivo. Front Microbiol 2018; 9:667. [PMID: 29681894 PMCID: PMC5897548 DOI: 10.3389/fmicb.2018.00667] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2017] [Accepted: 03/21/2018] [Indexed: 01/07/2023] Open
Abstract
The antimicrobial peptide LyeTxI isolated from the venom of the spider Lycosa erythrognatha is a potential model to develop new antibiotics against bacteria and fungi. In this work, we studied a peptide derived from LyeTxI, named LyeTxI-b, and characterized its structural profile and its in vitro and in vivo antimicrobial activities. Compared to LyeTxI, LyeTxI-b has an acetylated N-terminal and a deletion of a His residue, as structural modifications. The secondary structure of LyeTxI-b is a well-defined helical segment, from the second amino acid to the amidated C-terminal, with no clear partition between hydrophobic and hydrophilic faces. Moreover, LyeTxI-b shows a potent antimicrobial activity against Gram-positive and Gram-negative planktonic bacteria, being 10-fold more active than the native peptide against Escherichia coli. LyeTxI-b was also active in an in vivo model of septic arthritis, reducing the number of bacteria load, the migration of immune cells, the level of IL-1β cytokine and CXCL1 chemokine, as well as preventing cartilage damage. Our results show that LyeTxI-b is a potential therapeutic model for the development of new antibiotics against Gram-positive and Gram-negative bacteria.
Collapse
Affiliation(s)
- Pablo V M Reis
- Departamento de Bioquímica e Imunologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Daiane Boff
- Departamento de Bioquímica e Imunologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Rodrigo M Verly
- Departamento de Química, Faculdade de Ciências Exatas, Universidade Federal dos Vales do Jequitinhonha e Mucuri, Diamantina, Brazil
| | - Marcella N Melo-Braga
- Departamento de Bioquímica e Imunologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - María E Cortés
- Departamento de Odontologia Restauradora, Faculdade de Odontologia, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Daniel M Santos
- Serviço de Proteômica e Aracnídeos - Diretoria de Pesquisa e Desenvolvimento, Fundação Ezequiel Dias, Belo Horizonte, Brazil
| | - Adriano M de C Pimenta
- Departamento de Bioquímica e Imunologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Flávio A Amaral
- Departamento de Bioquímica e Imunologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Jarbas M Resende
- Departamento de Química, Instituto de Ciências Exatas, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Maria E de Lima
- Departamento de Bioquímica e Imunologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| |
Collapse
|
23
|
Boff D, Crijns H, Teixeira MM, Amaral FA, Proost P. Neutrophils: Beneficial and Harmful Cells in Septic Arthritis. Int J Mol Sci 2018; 19:E468. [PMID: 29401737 PMCID: PMC5855690 DOI: 10.3390/ijms19020468] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2017] [Revised: 01/30/2018] [Accepted: 02/01/2018] [Indexed: 12/17/2022] Open
Abstract
Septic arthritis is an inflammatory joint disease that is induced by pathogens such as Staphylococcus aureus. Infection of the joint triggers an acute inflammatory response directed by inflammatory mediators including microbial danger signals and cytokines and is accompanied by an influx of leukocytes. The recruitment of these inflammatory cells depends on gradients of chemoattractants including formylated peptides from the infectious agent or dying cells, host-derived leukotrienes, complement proteins and chemokines. Neutrophils are of major importance and play a dual role in the pathogenesis of septic arthritis. On the one hand, these leukocytes are indispensable in the first-line defense to kill invading pathogens in the early stage of disease. However, on the other hand, neutrophils act as mediators of tissue destruction. Since the elimination of inflammatory neutrophils from the site of inflammation is a prerequisite for resolution of the acute inflammatory response, the prolonged stay of these leukocytes at the inflammatory site can lead to irreversible damage to the infected joint, which is known as an important complication in septic arthritis patients. Thus, timely reduction of the recruitment of inflammatory neutrophils to infected joints may be an efficient therapy to reduce tissue damage in septic arthritis.
Collapse
Affiliation(s)
- Daiane Boff
- Imunofarmacologia, Department of Biochemistry and Immunology, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte 31270-901, Brazil.
- Laboratory of Molecular Immunology, Department of Microbiology and Immunology, Rega Institute for Medical Research, KU Leuven, B-3000 Leuven, Belgium.
| | - Helena Crijns
- Imunofarmacologia, Department of Biochemistry and Immunology, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte 31270-901, Brazil.
- Laboratory of Molecular Immunology, Department of Microbiology and Immunology, Rega Institute for Medical Research, KU Leuven, B-3000 Leuven, Belgium.
| | - Mauro M Teixeira
- Imunofarmacologia, Department of Biochemistry and Immunology, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte 31270-901, Brazil.
| | - Flavio A Amaral
- Imunofarmacologia, Department of Biochemistry and Immunology, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte 31270-901, Brazil.
| | - Paul Proost
- Laboratory of Molecular Immunology, Department of Microbiology and Immunology, Rega Institute for Medical Research, KU Leuven, B-3000 Leuven, Belgium.
| |
Collapse
|
24
|
Boff D, Oliveira VLS, Queiroz Junior CM, Silva TA, Allegretti M, Verri WA, Proost P, Teixeira MM, Amaral FA. CXCR2 is critical for bacterial control and development of joint damage and pain in Staphylococcus aureus-induced septic arthritis in mouse. Eur J Immunol 2018; 48:454-463. [PMID: 29168180 DOI: 10.1002/eji.201747198] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2017] [Revised: 10/23/2017] [Accepted: 11/15/2017] [Indexed: 02/06/2023]
Abstract
Staphylococcus aureus is the main pathogen associated with septic arthritis. Upon infection, neutrophils are quickly recruited to the joint by different chemoattractants, especially CXCR1/2 binding chemokines. Although their excessive accumulation is associated with intense pain and permanent articular damage, neutrophils have an important function in controlling bacterial burden. This work aimed to study the role of CXCR2 in the control of infection, hypernociception and tissue damage in S. aureus-induced septic arthritis in mice. The kinetics of neutrophil recruitment correlated with the bacterial load recovered from inflamed joint after intra-articular injection of S. aureus. Treatment of mice from the start of infection with the non-competitive antagonist of CXCR1/2, DF2156A, reduced neutrophil accumulation, cytokine production in the tissue, joint hypernociception and articular damage. However, early DF2156A treatment increased the bacterial load locally. CXCR2 was important for neutrophil activation and clearance of bacteria in vitro and in vivo. Start of treatment with DF2156A 3 days after infection prevented increase in bacterial load and reduced the hypernociception in the following days, but did not improve tissue damage. In conclusion, treatment with DF2156A seems be effective in controlling tissue inflammation and dysfunction but its effects are highly dependent on the timing of the treatment start.
Collapse
Affiliation(s)
- Daiane Boff
- Imunofarmacologia, Department of Biochemistry and Immunology, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais Brazil.,Laboratory of Molecular Immunology, Department of Microbiology and Immunology, Rega Institute for Medical Research, KU Leuven, Leuven, Belgium
| | - Vivian L S Oliveira
- Imunofarmacologia, Department of Biochemistry and Immunology, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais Brazil
| | - Celso M Queiroz Junior
- Department of Morphology, Institute of Biological Sciences, Universidade Federal de Minas Gerais, Brazil
| | - Tarcília A Silva
- Department of Oral Surgery and Pathology, School of Dentistry, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| | | | - Waldiceu A Verri
- Department of Pathological Sciences, Centro de Ciências Biológicas, Universidade Estadual de Londrina, Brazil
| | - Paul Proost
- Laboratory of Molecular Immunology, Department of Microbiology and Immunology, Rega Institute for Medical Research, KU Leuven, Leuven, Belgium
| | - Mauro M Teixeira
- Imunofarmacologia, Department of Biochemistry and Immunology, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais Brazil
| | - Flavio A Amaral
- Imunofarmacologia, Department of Biochemistry and Immunology, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais Brazil
| |
Collapse
|
25
|
Lora VRMM, Clemente-Napimoga JT, Abdalla HB, Macedo CG, Canales GDLT, Barbosa CMR. Botulinum toxin type A reduces inflammatory hypernociception induced by arthritis in the temporomadibular joint of rats. Toxicon 2017; 129:52-57. [PMID: 28209481 DOI: 10.1016/j.toxicon.2017.02.010] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2016] [Revised: 02/07/2017] [Accepted: 02/11/2017] [Indexed: 02/02/2023]
Abstract
OBJECTIVE This study aimed to investigate the antinociceptive effects of Botulinum toxin type A (BoNT-A) on persistent inflammatory hypernociception induced by arthritis in the temporomandibular joint (TMJ) of rats. MATERIAL AND METHODS Wistar rats were induced to persistent inflammatory hypernociception in the left TMJ. Then, animals were treated with intra-TMJ injections of BoNT-A, using doses of 3.5, 7 and 14 U/kg. Saline was used as control group. Behavioral tests were applied to evaluated the effect of BoNT-A in the inflammatory hypernociception. After that, animals were euthanized and samples from peri-articular tissues and trigeminal ganglia were obtained for further analyses. RESULTS BoNT-A reduced the persistent inflammatory hypernociception induced by arthritis in the TMJ of rats. BoNT-A significantly reduced the peripheral release of the neurotransmitters Substance P and Calcitonin gene related peptide; and the pro-inflammatory cytokine IL-1β. Otherwise, BoNT-A had no effect in the peripheral release of glutamate and the cytokine TNF-α. CONCLUSION These results demonstrate that intra-articular injection of BoNT-A reduces the albumin-induced arthritis persistent hypernociception in TMJ of rats by peripheral inhibition of neuropeptides release.
Collapse
Affiliation(s)
- Victor Ricardo Manuel Muñoz Lora
- Department of Prosthodontics and Periodontology, Piracicaba Dental School, University of Campinas (UNICAMP), Av Limeira, 901, Piracicaba, São Paulo, zip code 13414-903, Brazil
| | - Juliana Trindade Clemente-Napimoga
- São Leopoldo Mandic Institute and Research Center, Rua Jose Rocha Junqueira, 13 - Swift, Campinas, São Paulo, zip code 13045-755, Brazil; Laboratory of Orofacial Pain, Department of Physiological Sciences, Piracicaba Dental School, University of Campinas (UNICAMP), Av Limeira, 901, Piracicaba, São Paulo, zip code 13414-903, Brazil.
| | - Henrique Ballassini Abdalla
- Laboratory of Orofacial Pain, Department of Physiological Sciences, Piracicaba Dental School, University of Campinas (UNICAMP), Av Limeira, 901, Piracicaba, São Paulo, zip code 13414-903, Brazil
| | - Cristina Gomes Macedo
- São Leopoldo Mandic Institute and Research Center, Rua Jose Rocha Junqueira, 13 - Swift, Campinas, São Paulo, zip code 13045-755, Brazil; Laboratory of Orofacial Pain, Department of Physiological Sciences, Piracicaba Dental School, University of Campinas (UNICAMP), Av Limeira, 901, Piracicaba, São Paulo, zip code 13414-903, Brazil
| | - Giancarlo de la Torre Canales
- Department of Prosthodontics and Periodontology, Piracicaba Dental School, University of Campinas (UNICAMP), Av Limeira, 901, Piracicaba, São Paulo, zip code 13414-903, Brazil
| | - Celia Marisa Rizzatti Barbosa
- Department of Prosthodontics and Periodontology, Piracicaba Dental School, University of Campinas (UNICAMP), Av Limeira, 901, Piracicaba, São Paulo, zip code 13414-903, Brazil
| |
Collapse
|
26
|
Godin AM, Araújo DP, Menezes RR, de Brito AMS, Melo ISF, Coura GME, Bastos LFS, Amaral FA, Teixeira MM, de Fátima Â, Coelho MM, Machado RR. 2-Phthalimidethanol and 2-phthalimidethyl nitrate inhibit mechanical allodynia, neutrophil recruitment and cytokine and chemokine production in a murine model of articular inflammation. Pharmacol Rep 2017; 69:691-695. [PMID: 28550800 DOI: 10.1016/j.pharep.2017.01.033] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2016] [Revised: 01/16/2017] [Accepted: 01/30/2017] [Indexed: 12/31/2022]
Abstract
BACKGROUND Phthalimide analogs have been shown to exhibit anti-inflammatory, analgesic and immunomodulatory activities in different preclinical assays. This study aimed to investigate the potential role of 2-phthalimidethanol (PTD-OH) and 2-phthalimidethyl nitrate (PTD-NO) in a murine model of antigen-induced articular inflammation. METHODS Articular inflammation was induced by intra-articular injection of methylated bovine serum albumin (mBSA) in the knee joint of immunized male C57BL/6J mice. The animals were pre-treated with PTD-OH or PTD-NO (500mg/kg, per os, - 1h). Nociceptive threshold was measured using an electronic von Frey apparatus. The total number of leukocytes in the synovial cavity was determined. Concentrations of tumor necrosis factor (TNF)-α and CXCL-1 and myeloperoxidase (MPO) activity were determined in periarticular tissue. RESULTS Both PTD-OH and PTD-NO inhibited at similar extent the mechanical allodynia, neutrophil recruitment to the synovial cavity and periarticular tissue and TNF-α and CXCL-1 production induced by intra-articular challenge with mBSA in immunized mice. CONCLUSIONS PTD-OH and PTD-NO exhibit a marked activity in a murine model of antigen-induced articular inflammation in immunized animals. These results reinforce the interest in the investigation of phthalimide analogs devoid of the glutarimide ring as candidates to analgesic and anti-inflammatory drugs.
Collapse
Affiliation(s)
- Adriana M Godin
- Departamento de Produtos Farmacêuticos, Faculdade de Farmácia, Universidade Federal de Minas Gerais, Belo Horizonte, MG, Brazil
| | - Débora P Araújo
- Departamento de Química, Instituto de Ciências Exatas, Universidade Federal de Minas Gerais, Belo Horizonte, MG, Brazil
| | - Raquel R Menezes
- Departamento de Produtos Farmacêuticos, Faculdade de Farmácia, Universidade Federal de Minas Gerais, Belo Horizonte, MG, Brazil
| | - Ana M S de Brito
- Departamento de Produtos Farmacêuticos, Faculdade de Farmácia, Universidade Federal de Minas Gerais, Belo Horizonte, MG, Brazil
| | - Ivo S F Melo
- Departamento de Produtos Farmacêuticos, Faculdade de Farmácia, Universidade Federal de Minas Gerais, Belo Horizonte, MG, Brazil
| | - Giovanna M E Coura
- Departamento de Produtos Farmacêuticos, Faculdade de Farmácia, Universidade Federal de Minas Gerais, Belo Horizonte, MG, Brazil
| | - Leandro F S Bastos
- Departamento de Produtos Farmacêuticos, Faculdade de Farmácia, Universidade Federal de Minas Gerais, Belo Horizonte, MG, Brazil
| | - Flávio A Amaral
- Departamento de Bioquímica e Imunologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, MG, Brazil
| | - Mauro M Teixeira
- Departamento de Bioquímica e Imunologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, MG, Brazil
| | - Ângelo de Fátima
- Departamento de Química, Instituto de Ciências Exatas, Universidade Federal de Minas Gerais, Belo Horizonte, MG, Brazil
| | - Márcio M Coelho
- Departamento de Produtos Farmacêuticos, Faculdade de Farmácia, Universidade Federal de Minas Gerais, Belo Horizonte, MG, Brazil
| | - Renes R Machado
- Departamento de Produtos Farmacêuticos, Faculdade de Farmácia, Universidade Federal de Minas Gerais, Belo Horizonte, MG, Brazil.
| |
Collapse
|
27
|
Galvão I, Vago JP, Barroso LC, Tavares LP, Queiroz-Junior CM, Costa VV, Carneiro FS, Ferreira TP, Silva PMR, Amaral FA, Sousa LP, Teixeira MM. Annexin A1 promotes timely resolution of inflammation in murine gout. Eur J Immunol 2017; 47:585-596. [PMID: 27995621 DOI: 10.1002/eji.201646551] [Citation(s) in RCA: 48] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2016] [Revised: 10/27/2016] [Accepted: 12/14/2016] [Indexed: 12/31/2022]
Abstract
Gout is a self-limited inflammatory disease caused by deposition of monosodium urate (MSU) crystals in the joints. Resolution of inflammation is an active process leading to restoration of tissue homeostasis. Here, we studied the role of Annexin A1 (AnxA1), a glucocorticoid-regulated protein that has anti-inflammatory and proresolving actions, in resolution of acute gouty inflammation. Injection of MSU crystals in the knee joint of mice induced inflammation that was associated with expression of AnxA1 during the resolving phase of inflammation. Neutralization of AnxA1 with antiserum or blockade of its receptor with BOC-1 (nonselective) or WRW4 (selective) prevented the spontaneous resolution of gout. There was greater neutrophil infiltration after challenge with MSU crystals in AnxA1 knockout mice (AnxA1-/- ) and delayed resolution associated to decreased neutrophil apoptosis and efferocytosis. Pretreatment of mice with AnxA1-active N-terminal peptide (Ac2-26 ) decreased neutrophil influx, IL-1β, and CXCL1 production in periarticular joint. Posttreatment with Ac2-26 decreased neutrophil accumulation, IL-1β, and hypernociception, and improved the articular histopathological score. Importantly, the therapeutic effects of Ac2-26 were associated with increased neutrophils apoptosis and shortened resolution intervals. In conclusion, AnxA1 plays a crucial role in the context of acute gouty inflammation by promoting timely resolution of inflammation.
Collapse
Affiliation(s)
- Izabela Galvão
- Imunofarmacologia, Departamento de Bioquímica e Imunologia, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Juliana P Vago
- Imunofarmacologia, Departamento de Bioquímica e Imunologia, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil.,Departamento de Morfologia, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Livia C Barroso
- Imunofarmacologia, Departamento de Bioquímica e Imunologia, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Luciana P Tavares
- Imunofarmacologia, Departamento de Bioquímica e Imunologia, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Celso M Queiroz-Junior
- Departamento de Morfologia, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Vivian V Costa
- Imunofarmacologia, Departamento de Bioquímica e Imunologia, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Fernanda S Carneiro
- Departamento de Análises Clínicas e Toxicológicas, Faculdade de Farmácia, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Tatiana P Ferreira
- Laboratório de Inflamação, Instituto Oswaldo Cruz/FIOCRUZ, Rio de Janeiro, Brazil
| | - Patricia M R Silva
- Laboratório de Inflamação, Instituto Oswaldo Cruz/FIOCRUZ, Rio de Janeiro, Brazil
| | - Flávio A Amaral
- Imunofarmacologia, Departamento de Bioquímica e Imunologia, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Lirlândia P Sousa
- Imunofarmacologia, Departamento de Bioquímica e Imunologia, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil.,Departamento de Análises Clínicas e Toxicológicas, Faculdade de Farmácia, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Mauro M Teixeira
- Imunofarmacologia, Departamento de Bioquímica e Imunologia, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| |
Collapse
|
28
|
Cook AD, Louis C, Robinson MJ, Saleh R, Sleeman MA, Hamilton JA. Granulocyte macrophage colony-stimulating factor receptor α expression and its targeting in antigen-induced arthritis and inflammation. Arthritis Res Ther 2016; 18:287. [PMID: 27908288 PMCID: PMC5134062 DOI: 10.1186/s13075-016-1185-9] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2016] [Accepted: 11/16/2016] [Indexed: 12/23/2022] Open
Abstract
Background Blockade of granulocyte macrophage colony-stimulating factor (GM-CSF) and its receptor (GM-CSFRα) is being successfully tested in trials in rheumatoid arthritis (RA) with clinical results equivalent to those found with neutralization of the current therapeutic targets, TNF and IL-6. To explore further the role of GM-CSF as a pro-inflammatory cytokine, we examined the effect of anti-GM-CSFRα neutralization on myeloid cell populations in antigen-driven arthritis and inflammation models and also compared its effect with that of anti-TNF and anti-IL-6. Methods Cell population changes upon neutralization by monoclonal antibodies (mAbs) in the antigen-induced arthritis (AIA) and antigen-induced peritonitis (AIP) models were monitored by flow cytometry and microarray. Adoptive transfer of monocytes into the AIP cavity was used to assess the GM-CSF dependence of the development of macrophages and monocyte-derived dendritic cells (Mo-DCs) at a site of inflammation. Results Therapeutic administration of a neutralizing anti-GM-CSF mAb, but not of an anti-colony-stimulating factor (anti-CSF)-1 or an anti-CSF-1R mAb, ameliorated AIA disease. Using the anti-GM-CSFRα mAb, the relative surface expression of different inflammatory myeloid populations was found to be similar in the inflamed tissues in both the AIA and AIP models; however, the GM-CSFRα mAb, but not neutralizing anti-TNF and anti-IL-6 mAbs, preferentially depleted Mo-DCs from these sites. In addition, we were able to show that locally acting GM-CSF upregulated macrophage/Mo-DC numbers via GM-CSFR signalling in donor monocytes. Conclusions Our findings suggest that GM-CSF blockade modulates inflammatory responses differently to TNF and IL-6 blockade and may provide additional insight into how targeting the GM-CSF/GM-CSFRα system is providing efficacy in RA. Electronic supplementary material The online version of this article (doi:10.1186/s13075-016-1185-9) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Andrew D Cook
- Department of Medicine, Royal Melbourne Hospital, University of Melbourne, Parkville, Victoria, 3050, Australia.
| | - Cynthia Louis
- Department of Medicine, Royal Melbourne Hospital, University of Melbourne, Parkville, Victoria, 3050, Australia
| | - Matthew J Robinson
- Department of Respiratory, Inflammation and Autoimmunity, MedImmune Ltd, Granta Park, Cambridge, CB21 6GH, UK
| | - Reem Saleh
- Department of Medicine, Royal Melbourne Hospital, University of Melbourne, Parkville, Victoria, 3050, Australia
| | - Matthew A Sleeman
- Department of Respiratory, Inflammation and Autoimmunity, MedImmune Ltd, Granta Park, Cambridge, CB21 6GH, UK.,, Present Address: Regeneron, 777 Old Saw Mill River Rd, Tarrytown, NY, USA
| | - John A Hamilton
- Department of Medicine, Royal Melbourne Hospital, University of Melbourne, Parkville, Victoria, 3050, Australia
| |
Collapse
|
29
|
Correa LB, Pádua TA, Seito LN, Costa TEMM, Silva MA, Candéa ALP, Rosas EC, Henriques MG. Anti-inflammatory Effect of Methyl Gallate on Experimental Arthritis: Inhibition of Neutrophil Recruitment, Production of Inflammatory Mediators, and Activation of Macrophages. JOURNAL OF NATURAL PRODUCTS 2016; 79:1554-1566. [PMID: 27227459 DOI: 10.1021/acs.jnatprod.5b01115] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/05/2023]
Abstract
Methyl gallate (MG) is a prevalent phenolic acid in the plant kingdom, and its presence in herbal medicines might be related to its remarkable biological effects, such as its antioxidant, antitumor, and antimicrobial activities. Although some indirect evidence suggests anti-inflammatory activity for MG, there are no studies demonstrating this effect in animal models. Herein, we demonstrated that MG (0.7-70 mg/kg) inhibited zymosan-induced experimental arthritis in a dose-dependent manner. The oral administration of MG (7 mg/kg) attenuates arthritis induced by zymosan, affecting edema formation, leukocyte migration, and the production of inflammatory mediators (IL-1β, IL-6, TNF-α, CXCL-1, LTB4, and PGE2). Pretreatment with MG inhibited in vitro neutrophil chemotaxis elicited by CXCL-1, as well as the adhesion of these cells to TNF-α-primed endothelial cells. MG also impaired zymosan-stimulated macrophages by inhibiting IL-6 and NO production, COX-2 and iNOS expression, and intracellular calcium mobilization. Thus, MG is likely to present an anti-inflammatory effect by targeting multiple cellular events such as the production of various inflammatory mediators, as well as leukocyte activation and migration.
Collapse
Affiliation(s)
- Luana Barbosa Correa
- Laboratory of Applied Pharmacology, Farmanguinhos, and ‡National Institute for Science and Technology on Innovation on Neglected Diseases (INCT/IDN), Center for Technological Development in Health (CDTS), Oswaldo Cruz Foundation (Fiocruz) , Rio de Janeiro, RJ, Brazil
| | - Tatiana Almeida Pádua
- Laboratory of Applied Pharmacology, Farmanguinhos, and ‡National Institute for Science and Technology on Innovation on Neglected Diseases (INCT/IDN), Center for Technological Development in Health (CDTS), Oswaldo Cruz Foundation (Fiocruz) , Rio de Janeiro, RJ, Brazil
| | - Leonardo Noboru Seito
- Laboratory of Applied Pharmacology, Farmanguinhos, and ‡National Institute for Science and Technology on Innovation on Neglected Diseases (INCT/IDN), Center for Technological Development in Health (CDTS), Oswaldo Cruz Foundation (Fiocruz) , Rio de Janeiro, RJ, Brazil
| | - Thadeu Estevam Moreira Maramaldo Costa
- Laboratory of Applied Pharmacology, Farmanguinhos, and ‡National Institute for Science and Technology on Innovation on Neglected Diseases (INCT/IDN), Center for Technological Development in Health (CDTS), Oswaldo Cruz Foundation (Fiocruz) , Rio de Janeiro, RJ, Brazil
| | - Magaiver Andrade Silva
- Laboratory of Applied Pharmacology, Farmanguinhos, and ‡National Institute for Science and Technology on Innovation on Neglected Diseases (INCT/IDN), Center for Technological Development in Health (CDTS), Oswaldo Cruz Foundation (Fiocruz) , Rio de Janeiro, RJ, Brazil
| | - André Luis Peixoto Candéa
- Laboratory of Applied Pharmacology, Farmanguinhos, and ‡National Institute for Science and Technology on Innovation on Neglected Diseases (INCT/IDN), Center for Technological Development in Health (CDTS), Oswaldo Cruz Foundation (Fiocruz) , Rio de Janeiro, RJ, Brazil
| | - Elaine Cruz Rosas
- Laboratory of Applied Pharmacology, Farmanguinhos, and ‡National Institute for Science and Technology on Innovation on Neglected Diseases (INCT/IDN), Center for Technological Development in Health (CDTS), Oswaldo Cruz Foundation (Fiocruz) , Rio de Janeiro, RJ, Brazil
| | - Maria G Henriques
- Laboratory of Applied Pharmacology, Farmanguinhos, and ‡National Institute for Science and Technology on Innovation on Neglected Diseases (INCT/IDN), Center for Technological Development in Health (CDTS), Oswaldo Cruz Foundation (Fiocruz) , Rio de Janeiro, RJ, Brazil
| |
Collapse
|
30
|
Bas DB, Su J, Wigerblad G, Svensson CI. Pain in rheumatoid arthritis: models and mechanisms. Pain Manag 2016; 6:265-84. [PMID: 27086843 DOI: 10.2217/pmt.16.4] [Citation(s) in RCA: 49] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
Pain is one of the most challenging symptoms for patients with rheumatoid arthritis (RA). RA-related pain is frequently considered to be solely a consequence of inflammation in the joints; however, recent studies show that multiple mechanisms are involved. Indeed, RA pain may start even before the disease manifests, and frequently does not correlate with the degree of inflammation or pharmacological management. In this aspect, animal studies have the potential to provide new insights into the pathology that initiate and maintain pain in RA. The focus of this review is to describe the most commonly used animal models for studies of RA pathology, which have also been utilized in pain research, and to summarize findings providing potential clues to the mechanisms involved in the regulation of RA-induced pain.
Collapse
Affiliation(s)
- Duygu B Bas
- Department of Physiology & Pharmacology, Karolinska Institutet, Stockholm 171 77, Sweden
| | - Jie Su
- Department of Physiology & Pharmacology, Karolinska Institutet, Stockholm 171 77, Sweden
| | - Gustaf Wigerblad
- Department of Physiology & Pharmacology, Karolinska Institutet, Stockholm 171 77, Sweden
| | - Camilla I Svensson
- Department of Physiology & Pharmacology, Karolinska Institutet, Stockholm 171 77, Sweden
| |
Collapse
|
31
|
Galvão I, Dias ACF, Tavares LD, Rodrigues IPS, Queiroz-Junior CM, Costa VV, Reis AC, Ribeiro Oliveira RD, Louzada-Junior P, Souza DG, Leng L, Bucala R, Sousa LP, Bozza MT, Teixeira MM, Amaral FA. Macrophage migration inhibitory factor drives neutrophil accumulation by facilitating IL-1β production in a murine model of acute gout. J Leukoc Biol 2016; 99:1035-43. [PMID: 26868525 DOI: 10.1189/jlb.3ma0915-418r] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2015] [Accepted: 01/25/2016] [Indexed: 12/25/2022] Open
Abstract
This study evaluated the role of macrophage migration inhibitory factor in inflammation caused by monosodium urate crystals. The concentration of macrophage migration inhibitory factor was increased in synovial fluid of patients with acute gout, and there was a positive correlation between intra-articular macrophage migration inhibitory factor and IL-1β concentrations. In mice, the injection of monosodium urate crystals into the knee joint increased the levels of macrophage migration inhibitory factor in macrophages and in inflamed tissue. The injection of recombinant macrophage migration inhibitory factor into the joint of mice reproduced the inflammatory response observed in acute gout, including histologic changes, the recruitment of neutrophils, and increased levels of IL-1β and CXCL1. Importantly, the accumulation of neutrophils and the amount IL-1β in the joints were reduced in macrophage migration inhibitory factor-deficient mice when injected with monosodium urate crystals. We observed a similar effect when we blocked macrophage migration inhibitory factor with (S,R)-3-(4-hydroxyphenyl)-4,5-dihydro-5-isoxazole acetic acid or anti-macrophage migration inhibitory factor. In addition, the blockade of IL-1R and CXCR2 reduced recombinant macrophage migration inhibitory factor-induced neutrophil recruitment. Mechanistically, recombinant macrophage migration inhibitory factor is important for the synthesis of il1β mRNA in vivo and in isolated macrophages. Altogether, macrophage migration inhibitory factor promotes neutrophil accumulation and is important for IL-1β production, which are 2 crucial events contributing to the pathogenesis of acute gout.
Collapse
Affiliation(s)
- Izabela Galvão
- Department of Biochemistry and Immunology, Institute of Biological Sciences, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| | - Ana Carolina Fialho Dias
- Department of Biochemistry and Immunology, Institute of Biological Sciences, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| | - Livia Duarte Tavares
- Department of Microbiology, Institute of Biological Sciences, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| | - Irla Paula Stopa Rodrigues
- Department of Biochemistry and Immunology, Institute of Biological Sciences, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| | - Celso Martins Queiroz-Junior
- Department of Morphology, Institute of Biological Sciences, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| | - Vivian Vasconcelos Costa
- Department of Microbiology, Institute of Biological Sciences, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| | - Alesandra Corte Reis
- Department of Morphology, Institute of Biological Sciences, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| | | | - Paulo Louzada-Junior
- Division of Rheumatology, School of Medicine of Ribeirao Preto, Universidade de São Paulo, Brazil
| | - Daniele Glória Souza
- Department of Microbiology, Institute of Biological Sciences, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| | - Lin Leng
- Department of Medicine/Rheumatology, Yale University School of Medicine, The Anlyan Center, New Haven, Connecticut, USA
| | - Richard Bucala
- Department of Medicine/Rheumatology, Yale University School of Medicine, The Anlyan Center, New Haven, Connecticut, USA
| | - Lirlândia Pires Sousa
- Department of Clinical Analysis and Toxicological, Faculty of Pharmacy, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| | - Marcelo Torres Bozza
- Laboratory of Inflammation and Immunity, Department of Immunology, Institute of Microbiology, Universidade Federal do Rio de Janeiro, Brazil
| | - Mauro Martins Teixeira
- Department of Biochemistry and Immunology, Institute of Biological Sciences, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| | - Flávio Almeida Amaral
- Department of Biochemistry and Immunology, Institute of Biological Sciences, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| |
Collapse
|
32
|
Oliveira MC, Tavares LP, Vago JP, Batista NV, Queiroz-Junior CM, Vieira AT, Menezes GB, Sousa LP, van de Loo FAJ, Teixeira MM, Amaral FA, Ferreira AVM. Tumor Necrosis Factor, but Not Neutrophils, Alters the Metabolic Profile in Acute Experimental Arthritis. PLoS One 2016; 11:e0146403. [PMID: 26742100 PMCID: PMC4712146 DOI: 10.1371/journal.pone.0146403] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2015] [Accepted: 12/15/2015] [Indexed: 12/18/2022] Open
Abstract
Metabolic alterations are associated with arthritis apart from obesity. However, it is still unclear which is the underlying process behind these metabolic changes. Here, we investigate the role of tumor necrosis factor (TNF) in this process in an acute model of antigen-induced arthritis (AIA). Immunized male BALB/c mice received an intra-articular injection of PBS (control) or methylated bovine serum albumin (mBSA) into their knees, and were also pre-treated with different drugs: Etanercept, an anti-TNF drug, DF2156A, a CXCR1/2 receptor antagonist, or a monoclonal antibody RB6-8C5 to deplete neutrophils. Local challenge with mBSA evoked an acute neutrophil influx into the knee joint, and enhanced the joint nociception, along with a transient systemic metabolic alteration (higher levels of glucose and lipids, and altered adipocytokines). Pre-treatment with the conventional biological Etanercept, an inhibitor of TNF action, ameliorated the nociception and the acute joint inflammation dominated by neutrophils, and markedly improved many of the altered systemic metabolites (glucose and lipids), adipocytokines and PTX3. However, the lessening of metabolic changes was not due to diminished accumulation of neutrophils in the joint by Etanercept. Reduction of neutrophil recruitment by pre-treating AIA mice with DF2156A, or even the depletion of these cells by using RB6-8C5 reduced all of the inflammatory parameters and hypernociception developed after AIA challenge, but could not prevent the metabolic changes. Therefore, the induction of joint inflammation provoked acute metabolic alterations which were involved with TNF. We suggest that the role of TNF in arthritis-associated metabolic changes is not due to local neutrophils, which are the major cells present in this model, but rather due to cytokines.
Collapse
MESH Headings
- Adipokines/genetics
- Adipokines/metabolism
- Animals
- Anti-Inflammatory Agents, Non-Steroidal/pharmacology
- Antibodies, Monoclonal/pharmacology
- Arthritis, Experimental/chemically induced
- Arthritis, Experimental/drug therapy
- Arthritis, Experimental/metabolism
- Arthritis, Experimental/pathology
- C-Reactive Protein/genetics
- C-Reactive Protein/metabolism
- Cartilage, Articular/drug effects
- Cartilage, Articular/metabolism
- Cartilage, Articular/pathology
- Cattle
- Etanercept/pharmacology
- Gene Expression
- Glucose/metabolism
- Injections, Intra-Articular
- Lipid Metabolism/drug effects
- Male
- Mice
- Mice, Inbred BALB C
- Nerve Tissue Proteins/genetics
- Nerve Tissue Proteins/metabolism
- Neutrophil Infiltration/drug effects
- Neutrophils/drug effects
- Neutrophils/metabolism
- Neutrophils/pathology
- Receptors, Interleukin-8A/antagonists & inhibitors
- Receptors, Interleukin-8A/genetics
- Receptors, Interleukin-8A/metabolism
- Receptors, Interleukin-8B/antagonists & inhibitors
- Receptors, Interleukin-8B/genetics
- Receptors, Interleukin-8B/metabolism
- Serum Albumin, Bovine
- Sulfonamides/pharmacology
- Tumor Necrosis Factor-alpha/antagonists & inhibitors
- Tumor Necrosis Factor-alpha/genetics
- Tumor Necrosis Factor-alpha/metabolism
Collapse
Affiliation(s)
- Marina C. Oliveira
- Department of Nutrition, Nursing School, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
- Immunopharmacology, Department of Biochemistry and Immunology, Institute of Biological Sciences, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| | - Luciana P. Tavares
- Immunopharmacology, Department of Biochemistry and Immunology, Institute of Biological Sciences, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| | - Juliana P. Vago
- Immunopharmacology, Department of Biochemistry and Immunology, Institute of Biological Sciences, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
- Department of Clinical and Toxicological Analyses, Faculty of Pharmacy, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| | - Nathália V. Batista
- Immunopharmacology, Department of Biochemistry and Immunology, Institute of Biological Sciences, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| | - Celso M. Queiroz-Junior
- Department of Clinic, Pathology and Odontological Surgery, Faculty of Odontology, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| | - Angelica T. Vieira
- Immunopharmacology, Department of Biochemistry and Immunology, Institute of Biological Sciences, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| | - Gustavo B. Menezes
- Immunopharmacology, Department of Biochemistry and Immunology, Institute of Biological Sciences, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
- Department of Morphology, Institute of Biological Sciences, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| | - Lirlândia P. Sousa
- Department of Clinical and Toxicological Analyses, Faculty of Pharmacy, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| | - Fons A. J. van de Loo
- Experimental Rheumatology, Radboud university medical center, Nijmegen, The Netherlands
| | - Mauro M. Teixeira
- Immunopharmacology, Department of Biochemistry and Immunology, Institute of Biological Sciences, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| | - Flávio A. Amaral
- Immunopharmacology, Department of Biochemistry and Immunology, Institute of Biological Sciences, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| | - Adaliene V. M. Ferreira
- Department of Nutrition, Nursing School, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
- Immunopharmacology, Department of Biochemistry and Immunology, Institute of Biological Sciences, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
- * E-mail:
| |
Collapse
|
33
|
Abstract
Chemokines are essential mediators of leukocyte movement in vivo. In vitro assays of leukocyte migration cannot mimic the complex interactions with other cell types and matrix needed for cells to extravasate and migrate into tissues. Therefore, in vivo strategies to study the effects and potential relevance of chemokines for the migration of particular leukocyte subsets are necessary. Here, we describe methods to study the effects and endogenous role of chemokine in mice. Advantages and pitfalls of particular models are discussed and we focus on description in model's joint and pleural cavity inflammation and the effects and relevance of CXCR2 and CCR2 ligands on cell migration.
Collapse
Affiliation(s)
- F A Amaral
- Immunopharmacology, Department of Biochemistry and Immunology, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - D Boff
- Immunopharmacology, Department of Biochemistry and Immunology, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - M M Teixeira
- Immunopharmacology, Department of Biochemistry and Immunology, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil.
| |
Collapse
|
34
|
Vieira AT, Galvão I, Amaral FA, Teixeira MM, Nicoli JR, Martins FS. Oral treatment with Bifidobacterium longum 51A reduced inflammation in a murine experimental model of gout. Benef Microbes 2015; 6:799-806. [PMID: 26322542 DOI: 10.3920/bm2015.0015] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Gout is an acute inflammatory disease characterised by the presence of uric acid crystals in the joint. This event promotes neutrophil infiltration and activation that leads to tissue damage. We investigated here whether the oral administration of the probiotic strain Bifidobacterium longum 5(1A) (BL) could ameliorate monosodium urate crystal (MSU)-induced inflammation in a murine model of gout. Mice received oral administration of BL or saline daily for 7 days and then were injected with MSU in the knee cavity. Treatment with BL significantly alleviated the inflammatory parameters, as seen by reduced hypernociception, reduced neutrophil accumulation in the joint and myeloperoxidase activity in periarticular tissue. There was inhibition of the production of CXCL1 and interleukin(IL)-1β in joints. Levels of the anti-inflammatory cytokine IL-10 were significantly higher in the knee tissue of mice treated with than control mice injected with MSU. In conclusion, oral BL treatment reduced the inflammatory response in an experimental murine model of gout, suggesting it may be useful as an adjuvant treatment in patients with gout.
Collapse
Affiliation(s)
- A T Vieira
- 1 Department of Microbiology, Institute of Biological Sciences, Federal University of Minas Gerais, Avenida Presidente Antônio Carlos 6627, C.P. 486, Pampulha-Campus UFMG, 31270-901, Belo Horizonte, MG, Brazil.,2 Department of Biochemistry and Immunology, Institute of Biological Sciences, Federal University of Minas Gerais, Belo Horizonte, MG, Brazil
| | - I Galvão
- 2 Department of Biochemistry and Immunology, Institute of Biological Sciences, Federal University of Minas Gerais, Belo Horizonte, MG, Brazil
| | - F A Amaral
- 2 Department of Biochemistry and Immunology, Institute of Biological Sciences, Federal University of Minas Gerais, Belo Horizonte, MG, Brazil
| | - M M Teixeira
- 2 Department of Biochemistry and Immunology, Institute of Biological Sciences, Federal University of Minas Gerais, Belo Horizonte, MG, Brazil
| | - J R Nicoli
- 1 Department of Microbiology, Institute of Biological Sciences, Federal University of Minas Gerais, Avenida Presidente Antônio Carlos 6627, C.P. 486, Pampulha-Campus UFMG, 31270-901, Belo Horizonte, MG, Brazil
| | - F S Martins
- 1 Department of Microbiology, Institute of Biological Sciences, Federal University of Minas Gerais, Avenida Presidente Antônio Carlos 6627, C.P. 486, Pampulha-Campus UFMG, 31270-901, Belo Horizonte, MG, Brazil
| |
Collapse
|
35
|
Vieira AT, Macia L, Galvão I, Martins FS, Canesso MCC, Amaral FA, Garcia CC, Maslowski KM, De Leon E, Shim D, Nicoli JR, Harper JL, Teixeira MM, Mackay CR. A Role for Gut Microbiota and the Metabolite-Sensing Receptor GPR43 in a Murine Model of Gout. Arthritis Rheumatol 2015; 67:1646-56. [PMID: 25914377 DOI: 10.1002/art.39107] [Citation(s) in RCA: 180] [Impact Index Per Article: 20.0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2014] [Accepted: 03/03/2015] [Indexed: 12/12/2022]
Abstract
OBJECTIVE Host-microbial interactions are central in health and disease. Monosodium urate monohydrate (MSU) crystals cause gout by activating the NLRP3 inflammasome, leading to interleukin-1β (IL-1β) production and neutrophil recruitment. This study was undertaken to investigate the relevance of gut microbiota, acetate, and the metabolite-sensing receptor GPR43 in regulating inflammation in a murine model of gout. METHODS Gout was induced by the injection of MSU crystals into the knee joints of mice. Macrophages from the various animals were stimulated to determine inflammasome activation and production of reactive oxygen species (ROS). RESULTS Injection of MSU crystals caused joint inflammation, as seen by neutrophil influx, hypernociception, and production of IL-1β and CXCL1. These parameters were greatly decreased in germ-free mice, mice treated with antibiotics, and GPR-43-deficient mice. Recolonization or administration of acetate to germ-free mice restored inflammation in response to injection of MSU crystals. In vitro, macrophages produced ROS and assembled the inflammasome when stimulated with MSU. Macrophages from germ-free animals produced little ROS, and there was little inflammasome assembly. Similar results were observed in macrophages from GPR-43-deficient mice. Treatment of germ-free mice with acetate restored in vitro responsiveness of macrophages to MSU crystals. CONCLUSION In the absence of microbiota, there is decreased production of short-chain fatty acids that are necessary for adequate inflammasome assembly and IL-1β production in a manner that is at least partially dependent on GPR43. These results clearly show that the commensal microbiota shapes the host's ability to respond to an inflammasome-dependent acute inflammatory stimulus outside the gut.
Collapse
Affiliation(s)
- Angélica T Vieira
- Universidade Federal de Minas Gerais, Belo Horizonte, Brazil, and Monash University, Clayton, Victoria, Australia
| | | | - Izabela Galvão
- Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | | | | | - Flávio A Amaral
- Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | | | - Kendle M Maslowski
- Garvan Institute of Medical Research, Darlinghurst, New South Wales, Australia
| | - Ellen De Leon
- Garvan Institute of Medical Research, Darlinghurst, New South Wales, Australia
| | - Doris Shim
- Garvan Institute of Medical Research, Darlinghurst, New South Wales, Australia
| | | | | | | | | |
Collapse
|
36
|
Costa VV, Amaral FA, Coelho FM, Queiroz-Junior CM, Malagoli BG, Gomes JHS, Lopes F, Silveira KD, Sachs D, Fagundes CT, Tavares LD, Pinho V, Silva TA, Teixeira MM, Braga FC, Souza DG. Lithothamnion muelleri treatment ameliorates inflammatory and hypernociceptive responses in antigen-induced arthritis in mice. PLoS One 2015; 10:e0118356. [PMID: 25793994 PMCID: PMC4368517 DOI: 10.1371/journal.pone.0118356] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2014] [Accepted: 01/14/2015] [Indexed: 12/29/2022] Open
Abstract
Rheumatoid Arthritis (RA) is a chronic disease characterized by persistent inflammation and pain. Alternative therapies to reduce these symptoms are needed. Marine algae are valuable sources of diverse bioactive compounds. Lithothamnion muelleri (Hapalidiaceae) is a marine algae with anti-inflammatory, antitumor, and immunomodulatory properties. Here, we investigated the potential anti-inflammatory and analgesic activities of L. muelleri in a murine model of antigen-induced arthritis (AIA) in mice. Our results demonstrate that treatment with L. muelleri prevented inflammation and hypernociception in arthritic mice. Mechanistically, the crude extract and the polysaccharide-rich fractions of L. muelleri may act impairing the production of the chemokines CXCL1 and CXCL2, and consequently inhibit neutrophil influx to the knee joint by dampening the adhesion step of leukocyte recruitment in the knee microvessels. Altogether our results suggest that treatment with L.muelleri has a potential therapeutic application in arthritis treatment.
Collapse
Affiliation(s)
- Vivian V. Costa
- Laboratório de Interação Microrganismo-Hospedeiro, Departamento de Microbiologia, ICB, UFMG, Belo Horizonte, MG, Brazil
- Imunofarmacologia, Departamento de Bioquímica e Imunologia, ICB, UFMG, Belo Horizonte, MG, Brazil
- Interdisciplinary Research Group in Infectious Diseases, Singapore-MIT Alliance for Research and Technology, Singapore, Singapore
| | - Flavio A. Amaral
- Imunofarmacologia, Departamento de Bioquímica e Imunologia, ICB, UFMG, Belo Horizonte, MG, Brazil
| | - Fernanda M. Coelho
- Imunofarmacologia, Departamento de Bioquímica e Imunologia, ICB, UFMG, Belo Horizonte, MG, Brazil
| | - Celso M. Queiroz-Junior
- Laboratório de Interação Microrganismo-Hospedeiro, Departamento de Microbiologia, ICB, UFMG, Belo Horizonte, MG, Brazil
- Departamento de Clínica, Patologia e Cirurgia Odontológicas, Faculdade de Odontologia, UFMG, Belo Horizonte, MG, Brazil
| | - Bruna G. Malagoli
- Departamento de Produtos Farmacêuticos, Faculdade de Farmácia, UFMG, Belo Horizonte, MG, Brazil
| | - Jose Hugo S. Gomes
- Departamento de Produtos Farmacêuticos, Faculdade de Farmácia, UFMG, Belo Horizonte, MG, Brazil
| | - Fernando Lopes
- Imunofarmacologia, Departamento de Bioquímica e Imunologia, ICB, UFMG, Belo Horizonte, MG, Brazil
- Departamento de Morfologia, ICB, UFMG, Belo Horizonte, MG, Brazil
| | - Kátia D. Silveira
- Imunofarmacologia, Departamento de Bioquímica e Imunologia, ICB, UFMG, Belo Horizonte, MG, Brazil
| | - Daniela Sachs
- Departamento de Física e Química, Instituto de Ciências Exatas da Universidade Federal de Itajubá (UNIFEI), Itajubá, MG, Brazil
| | - Caio T. Fagundes
- Laboratório de Interação Microrganismo-Hospedeiro, Departamento de Microbiologia, ICB, UFMG, Belo Horizonte, MG, Brazil
- Imunofarmacologia, Departamento de Bioquímica e Imunologia, ICB, UFMG, Belo Horizonte, MG, Brazil
| | - Lívia D. Tavares
- Laboratório de Interação Microrganismo-Hospedeiro, Departamento de Microbiologia, ICB, UFMG, Belo Horizonte, MG, Brazil
- Imunofarmacologia, Departamento de Bioquímica e Imunologia, ICB, UFMG, Belo Horizonte, MG, Brazil
| | - Vanessa Pinho
- Imunofarmacologia, Departamento de Bioquímica e Imunologia, ICB, UFMG, Belo Horizonte, MG, Brazil
- Departamento de Morfologia, ICB, UFMG, Belo Horizonte, MG, Brazil
| | - Tarcilia A. Silva
- Imunofarmacologia, Departamento de Bioquímica e Imunologia, ICB, UFMG, Belo Horizonte, MG, Brazil
- Departamento de Clínica, Patologia e Cirurgia Odontológicas, Faculdade de Odontologia, UFMG, Belo Horizonte, MG, Brazil
| | - Mauro M. Teixeira
- Imunofarmacologia, Departamento de Bioquímica e Imunologia, ICB, UFMG, Belo Horizonte, MG, Brazil
| | - Fernão C. Braga
- Departamento de Produtos Farmacêuticos, Faculdade de Farmácia, UFMG, Belo Horizonte, MG, Brazil
| | - Danielle G. Souza
- Laboratório de Interação Microrganismo-Hospedeiro, Departamento de Microbiologia, ICB, UFMG, Belo Horizonte, MG, Brazil
- Imunofarmacologia, Departamento de Bioquímica e Imunologia, ICB, UFMG, Belo Horizonte, MG, Brazil
- * E-mail:
| |
Collapse
|
37
|
Camargo LL, Denadai-Souza A, Yshii LM, Mesquita FPN, Soares AG, Lima C, Schenka A, Grant A, Fernandes E, Muscará MN, Costa SKP. Peripheral neurokinin-1 receptors contribute to kaolin-induced acute monoarthritis in rats. Neuroimmunomodulation 2015; 22:373-84. [PMID: 26088412 DOI: 10.1159/000381549] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/20/2014] [Accepted: 03/06/2015] [Indexed: 11/19/2022] Open
Abstract
OBJECTIVE intra-articular co-injection of kaolin with carrageenan (CGN) in rodents is widely used as an experimental model of arthritis. However, the ability of kaolin to cause arthritis and related immune responses when administered alone is unclear. We evaluated the contribution of prostanoids and sensory C-fibres (and their neuropeptide substance P) to kaolin-induced inflammation in the rat knee. METHODS Wistar rats, 8-10 weeks old, received an intra-articular injection of kaolin (1-10 μg/joint) or saline into the knee joint. Knee inflammation, proinflammatory cytokines, pain behaviour and secondary tactile allodynia were assessed over 5 h, when synovial leukocyte counts, histopathological changes and proinflammatory cytokine levels were evaluated. RESULTS The intra-articular injection of kaolin caused a dose- and time-dependent knee swelling and impairment of motion that were associated with secondary tactile allodynia, elevated concentrations of IL-1β, IL-6 and TNFα, leukocyte infiltration, and histopathological changes in the ipsilateral hindpaw. The neurokinin-1 (NK1) receptor antagonist SR140333 or neonatal treatment with capsaicin markedly reduced the inflammatory parameters, cytokines and allodynia but failed to significantly inhibit the impaired motion. The cyclo-oxygenase inhibitor indomethacin partially inhibited knee oedema and allodynia but did not affect the leukocyte influx, myeloperoxidase activity or impaired motion in the kaolin-injected rat. CONCLUSIONS We show the first evidence that intra-articular injection of kaolin without CGN produced severe acute monoarthritis. This was highly dependent on substance P (released from C-fibres) and NK1 receptor activation, which stimulated local production of proinflammatory cytokines. This model may be of critical importance for mechanistic studies and screening new anti-inflammatory/analgesic drugs.
Collapse
Affiliation(s)
- Livia L Camargo
- Department of Pharmacology, Institute of Biomedical Sciences, University of Sx00E3;o Paulo (USP), Sx00E3;o Paulo, Brazil
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
38
|
Silva Quinteiro M, Henrique Napimoga M, Gomes Macedo C, Furtado Freitas F, Balassini Abdalla H, Bonfante R, Trindade Clemente-Napimoga J. 15-deoxy-Δ12,14-prostaglandin J2 reduces albumin-induced arthritis in temporomandibular joint of rats. Eur J Pharmacol 2014; 740:58-65. [PMID: 25016088 DOI: 10.1016/j.ejphar.2014.07.002] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2014] [Revised: 06/27/2014] [Accepted: 07/02/2014] [Indexed: 02/01/2023]
Abstract
The aim of this study was to evaluate the peripheral effect of 15-deoxy-Δ12,14-prostaglandin J2 (15d-PGJ2) in albumin-induced arthritis in temporomandibular joint (TMJ) of rats. Antigen-induced arthritis (AIA) was generated in rats with methylated bovine serum albumin (mBSA) diluted in complete Freund׳s adjuvant. Pretreatment with an intra-articular injection of 15d-PGJ2 (100 ng/TMJ) before mBSA intra-articular injection (10 µg/TMJ) (challenge) in immunized rats significantly reduced the albumin-induced arthritis inflammation. The results demonstrated that 15d-PGJ2 was able to inhibit plasma extravasation, leukocyte migration and the release of inflammatory cytokines IL-6, IL-12, IL-18 and the chemokine CINC-1 in the TMJ tissues. In addition, 15d-PGJ2 was able to increase the expression of the anti-adhesive molecule CD55 and the anti-inflammatory cytokine IL-10. Taken together, it is possible to suggest that 15d-PGJ2 inhibit leukocyte infiltration and subsequently inflammatory process, through a shift in the balance of the pro- and anti-adhesive properties. Thus, 15d-PGJ2 might be used as a potential anti-inflammatory drug to treat arthritis-induced inflammation of the temporomandibular joint.
Collapse
Affiliation(s)
- Mariana Silva Quinteiro
- Laboratory of Orofacial Pain, Department of Physiological Sciences, Piracicaba Dental School, University of Campinas, Brazil
| | - Marcelo Henrique Napimoga
- Laboratory of Immunology and Molecular Biology, São Leopoldo Mandic Institute and Research Center, Campinas, Brazil
| | - Cristina Gomes Macedo
- Laboratory of Orofacial Pain, Department of Physiological Sciences, Piracicaba Dental School, University of Campinas, Brazil
| | - Fabiana Furtado Freitas
- Laboratory of Orofacial Pain, Department of Physiological Sciences, Piracicaba Dental School, University of Campinas, Brazil
| | - Henrique Balassini Abdalla
- Laboratory of Orofacial Pain, Department of Physiological Sciences, Piracicaba Dental School, University of Campinas, Brazil
| | - Ricardo Bonfante
- Laboratory of Orofacial Pain, Department of Physiological Sciences, Piracicaba Dental School, University of Campinas, Brazil
| | | |
Collapse
|
39
|
Russo RC, Garcia CC, Teixeira MM, Amaral FA. The CXCL8/IL-8 chemokine family and its receptors in inflammatory diseases. Expert Rev Clin Immunol 2014; 10:593-619. [DOI: 10.1586/1744666x.2014.894886] [Citation(s) in RCA: 317] [Impact Index Per Article: 31.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
|
40
|
Garin A, Johnson Z, Hermant A, Beltran F, Ratinaud Y, Michel A, Krohn S, Gaudet M, Carboni S, Ji H, Missotten M, Leger O, Power C, Proudfoot A. Chemokine receptor antagonist development. Methods Mol Biol 2013; 1013:67-92. [PMID: 23625494 DOI: 10.1007/978-1-62703-426-5_6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/10/2023]
Abstract
This chapter describes assays that focus on the characterization of compounds identified in high--throughput screening campaigns, and the subsequent medicinal chemistry programs. They cover methods to determine potency in buffer, the effect of whole blood on the compounds' activity and finally the pharmacokinetic (PK)/pharmacodynamic (PD) -relationship of the compounds in a rodent species.
Collapse
Affiliation(s)
- Alexandre Garin
- Geneva Research Centre, Division of Autoimmune and Inflammatory Diseases, Merck Serono, Geneva, Switzerland
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
41
|
Silveira KD, Coelho FM, Vieira AT, Barroso LC, Queiroz-Junior CM, Costa VV, Sousa LFC, Oliveira ML, Bader M, Silva TA, Santos RAS, Silva ACSE, Teixeira MM. Mechanisms of the anti-inflammatory actions of the angiotensin type 1 receptor antagonist losartan in experimental models of arthritis. Peptides 2013; 46:53-63. [PMID: 23727291 DOI: 10.1016/j.peptides.2013.05.012] [Citation(s) in RCA: 56] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/10/2013] [Revised: 05/14/2013] [Accepted: 05/14/2013] [Indexed: 12/28/2022]
Abstract
Angiotensin (Ang) II and its AT1 receptors have been implicated in the pathogenesis of rheumatoid arthritis. Activation of the counter-regulatory Ang-(1-7)-Mas receptor axis may contribute to some of the effects of AT₁ receptor blockers (ARBs). In this study, we have used losartan, an ARB, to investigate the role of and the mechanisms by which AT₁ receptors participated in two experimental models of arthritis: antigen-induced arthritis (AIA) in mice and adjuvant-induced arthritis (AdIA) in rats. Treatment with losartan decreased neutrophil recruitment, hypernociception and the production of TNF-α, IL-1β and chemokine (C-X-C motif) ligand 1 in mice subjected to AIA. Histopathological analysis showed significant reduction of tissue injury and inflammation and decreased proteoglycan loss. In addition to decreasing cytokine production, losartan directly reduced leukocyte rolling and adhesion. Anti-inflammatory effects of losartan were not associated to Mas receptor activation and/or Ang-(1-7) production. Anti-inflammatory effects were reproduced in rats subjected to AdIA. This study shows that ARBs have potent anti-inflammatory effects in animal models of arthritis. Mechanistically, reduction of leukocyte accumulation and of joint damage was associated with local inhibition of cytokine production and direct inhibition of leukocyte-endothelium interactions. The anti-inflammatory actions of losartan were accompanied by functional improvement of the joint, as seen by reduced joint hypernociception. These findings support the use of ARBs for the treatment of human arthritis and provide potential mechanisms for the anti-inflammatory actions of these compounds.
Collapse
MESH Headings
- Angiotensin I/biosynthesis
- Angiotensin II Type 1 Receptor Blockers/pharmacology
- Animals
- Anti-Inflammatory Agents, Non-Steroidal/pharmacology
- Arthritis, Experimental/drug therapy
- Arthritis, Rheumatoid/drug therapy
- Cell Adhesion/drug effects
- Chemokine CXCL1/biosynthesis
- Disease Models, Animal
- Female
- Hyperalgesia/drug therapy
- Inflammation/drug therapy
- Interleukin-1beta/biosynthesis
- Leukocyte Rolling/drug effects
- Losartan/pharmacology
- Male
- Mice
- Mice, Inbred C57BL
- Neutrophil Infiltration/drug effects
- Peptide Fragments/biosynthesis
- Proto-Oncogene Mas
- Proto-Oncogene Proteins/metabolism
- Rats
- Rats, Sprague-Dawley
- Receptor, Angiotensin, Type 1/drug effects
- Receptor, Angiotensin, Type 1/metabolism
- Receptors, G-Protein-Coupled/metabolism
- Tumor Necrosis Factor-alpha/biosynthesis
Collapse
Affiliation(s)
- Kátia D Silveira
- Immunopharmacology, Department of Biochemistry and Immunology, Institute of Biological Sciences, Brazil
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
42
|
Doufas AG, Tian L, Padrez KA, Suwanprathes P, Cardell JA, Maecker HT, Panousis P. Experimental pain and opioid analgesia in volunteers at high risk for obstructive sleep apnea. PLoS One 2013; 8:e54807. [PMID: 23382975 PMCID: PMC3558510 DOI: 10.1371/journal.pone.0054807] [Citation(s) in RCA: 81] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2012] [Accepted: 12/17/2012] [Indexed: 01/20/2023] Open
Abstract
Background Obstructive sleep apnea (OSA) is characterized by recurrent nocturnal hypoxia and sleep disruption. Sleep fragmentation caused hyperalgesia in volunteers, while nocturnal hypoxemia enhanced morphine analgesic potency in children with OSA. This evidence directly relates to surgical OSA patients who are at risk for airway compromise due to postoperative use of opioids. Using accepted experimental pain models, we characterized pain processing and opioid analgesia in male volunteers recruited based on their risk for OSA. Methods After approval from the Intitutional Review Board and informed consent, we assessed heat and cold pain thresholds and tolerances in volunteers after overnight polysomnography (PSG). Three pro-inflammatory and 3 hypoxia markers were determined in the serum. Pain tests were performed at baseline, placebo, and two effect site concentrations of remifentanil (1 and 2 µg/ml), an μ-opioid agonist. Linear mixed effects regression models were employed to evaluate the association of 3 PSG descriptors [wake after sleep onset, number of sleep stage shifts, and lowest oxyhemoglobin saturation (SaO2) during sleep] and all serum markers with pain thresholds and tolerances at baseline, as well as their changes under remifentanil. Results Forty-three volunteers (12 normal and 31 with a PSG-based diagnosis of OSA) were included in the analysis. The lower nadir SaO2 and higher insulin growth factor binding protein-1 (IGFBP-1) were associated with higher analgesic sensitivity to remifentanil (SaO2, P = 0.0440; IGFBP-1, P = 0.0013). Other pro-inflammatory mediators like interleukin-1β and tumor necrosis factor-α (TNF-α) were associated with an enhanced sensitivity to the opioid analgesic effect (IL-1β, P = 0.0218; TNF-α, P = 0.0276). Conclusions Nocturnal hypoxemia in subjects at high risk for OSA was associated with an increased potency of opioid analgesia. A serum hypoxia marker (IGFBP-1) was associated with hypoalgesia and increased potency to opioid analgesia; other pro-inflammatory mediators also predicted an enhanced opioid potency. Trial Registration: ClinicalTrials.gov NCT00672737.
Collapse
Affiliation(s)
- Anthony G Doufas
- Department of Anesthesiology, Perioperative and Pain Medicine, Stanford University School of Medicine, Stanford, California, USA.
| | | | | | | | | | | | | |
Collapse
|
43
|
Severin IC, Soares A, Hantson J, Teixeira M, Sachs D, Valognes D, Scheer A, Schwarz MK, Wells TNC, Proudfoot AEI, Shaw J. Glycosaminoglycan analogs as a novel anti-inflammatory strategy. Front Immunol 2012; 3:293. [PMID: 23087686 PMCID: PMC3472544 DOI: 10.3389/fimmu.2012.00293] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2012] [Accepted: 09/03/2012] [Indexed: 01/07/2023] Open
Abstract
Heparin, a glycosaminoglycan (GAG), has both anti-inflammatory and anti-coagulant properties. The clinical use of heparin against inflammation, however, has been limited by concerns about increased bleeding. While the anti-coagulant activity of heparin is well understood, its anti-inflammatory properties are less so. Heparin is known to bind to certain cytokines, including chemokines, small proteins which mediate inflammation through their control of leukocyte migration and activation. Molecules which can interrupt the chemokine-GAG interaction without inhibiting coagulation could therefore, represent a new class of anti-inflammatory agents. In the present study, two approaches were undertaken, both focusing on the heparin-chemokine relationship. In the first, a structure based strategy was used: after an initial screening of potential small molecule binders using protein NMR on a target chemokine, binding molecules were optimized through structure-based design. In the second approach, commercially available short oligosaccharides were polysulfated. In vitro, these molecules prevented chemokine-GAG binding and chemokine receptor activation without disrupting coagulation. However, in vivo, these compounds caused variable results in a murine peritoneal recruitment assay, with a general increase of cell recruitment. In more disease specific models, such as antigen-induced arthritis and delayed-type hypersensitivity, an overall decrease in inflammation was noted, suggesting that the primary anti-inflammatory effect may also involve factors beyond the chemokine system.
Collapse
Affiliation(s)
| | | | | | - Mauro Teixeira
- Departmento de Bioquimica e Imunologia, Instituto de Ciencias Biologicas, Universidade Federal de Minas GeraisBelo Horizonte, Brazil
| | - Daniela Sachs
- Departmento de Bioquimica e Imunologia, Instituto de Ciencias Biologicas, Universidade Federal de Minas GeraisBelo Horizonte, Brazil
| | | | | | | | | | - Amanda E. I. Proudfoot
- Merck Serono Geneva Research CentreGeneva, Switzerland,*Correspondence: Amanda E. I. Proudfoot and Jeffrey Shaw, Merck Serono Geneva Research Centre, 9, Chemin des Mines, 1202 Geneva, Switzerland. e-mail: ;
| | - Jeffrey Shaw
- Merck Serono Geneva Research CentreGeneva, Switzerland,*Correspondence: Amanda E. I. Proudfoot and Jeffrey Shaw, Merck Serono Geneva Research Centre, 9, Chemin des Mines, 1202 Geneva, Switzerland. e-mail: ;
| |
Collapse
|
44
|
Amaral FA, Costa VV, Tavares LD, Sachs D, Coelho FM, Fagundes CT, Soriani FM, Silveira TN, Cunha LD, Zamboni DS, Quesniaux V, Peres RS, Cunha TM, Cunha FQ, Ryffel B, Souza DG, Teixeira MM. NLRP3 inflammasome-mediated neutrophil recruitment and hypernociception depend on leukotriene B(4) in a murine model of gout. ACTA ACUST UNITED AC 2012; 64:474-84. [PMID: 21952942 DOI: 10.1002/art.33355] [Citation(s) in RCA: 197] [Impact Index Per Article: 16.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
Abstract
OBJECTIVE Deposition of monosodium urate monohydrate (MSU) crystals in the joints promotes an intense inflammatory response and joint dysfunction. This study evaluated the role of the NLRP3 inflammasome and 5-lipoxygenase (5-LOX)-derived leukotriene B(4) (LTB(4) ) in driving tissue inflammation and hypernociception in a murine model of gout. METHODS Gout was induced by injecting MSU crystals into the joints of mice. Wild-type mice and mice deficient in NLRP3, ASC, caspase 1, interleukin-1β (IL-1β), IL-1 receptor type I (IL-1RI), IL-18R, myeloid differentiation factor 88 (MyD88), or 5-LOX were used. Evaluations were performed to assess neutrophil influx, LTB(4) activity, cytokine (IL-1β, CXCL1) production (by enzyme-linked immunosorbent assay), synovial microvasculature cell adhesion (by intravital microscopy), and hypernociception. Cleaved caspase 1 and production of reactive oxygen species (ROS) were analyzed in macrophages by Western blotting and fluorometric assay, respectively. RESULTS Injection of MSU crystals into the knee joints of mice induced neutrophil influx and neutrophil-dependent hypernociception. MSU crystal-induced neutrophil influx was CXCR2-dependent and relied on the induction of CXCL1 in an NLRP3/ASC/caspase 1/IL-1β/MyD88-dependent manner. LTB(4) was produced rapidly after injection of MSU crystals, and this was necessary for caspase 1-dependent IL-1β production and consequent release of CXCR2-acting chemokines in vivo. In vitro, macrophages produced LTB(4) after MSU crystal injection, and LTB(4) was relevant in the MSU crystal-induced maturation of IL-1β. Mechanistically, LTB(4) drove MSU crystal-induced production of ROS and ROS-dependent activation of the NLRP3 inflammasome. CONCLUSION These results reveal the role of the NLRP3 inflammasome in mediating MSU crystal-induced inflammation and dysfunction of the joints, and highlight a previously unrecognized role of LTB(4) in driving NLRP3 inflammasome activation in response to MSU crystals, both in vitro and in vivo.
Collapse
|
45
|
A model of DENV-3 infection that recapitulates severe disease and highlights the importance of IFN-γ in host resistance to infection. PLoS Negl Trop Dis 2012; 6:e1663. [PMID: 22666512 PMCID: PMC3362616 DOI: 10.1371/journal.pntd.0001663] [Citation(s) in RCA: 52] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2012] [Accepted: 04/11/2012] [Indexed: 12/19/2022] Open
Abstract
There are few animal models of dengue infection, especially in immunocompetent mice. Here, we describe alterations found in adult immunocompetent mice inoculated with an adapted Dengue virus (DENV-3) strain. Infection of mice with the adapted DENV-3 caused inoculum-dependent lethality that was preceded by several hematological and biochemical changes and increased virus dissemination, features consistent with severe disease manifestation in humans. IFN-γ expression increased after DENV-3 infection of WT mice and this was preceded by increase in expression of IL-12 and IL-18. In DENV-3-inoculated IFN-γ(-/-) mice, there was enhanced lethality, which was preceded by severe disease manifestation and virus replication. Lack of IFN-γ production was associated with diminished NO-synthase 2 (NOS2) expression and higher susceptibility of NOS2(-/-) mice to DENV-3 infection. Therefore, mechanisms of protection to DENV-3 infection rely on IFN-γ-NOS2-NO-dependent control of viral replication and of disease severity, a pathway showed to be relevant for resistance to DENV infection in other experimental and clinical settings. Thus, the model of DENV-3 infection in immunocompetent mice described here represents a significant advance in animal models of severe dengue disease and may provide an important tool to the elucidation of immunopathogenesis of disease and of protective mechanisms associated with infection.
Collapse
|
46
|
Queiroz-Junior CM, Madeira MFM, Coelho FM, Oliveira CR, Cândido LCM, Garlet GP, Teixeira MM, Souza DDG, Silva TAD. Experimental arthritis exacerbates Aggregatibacter actinomycetemcomitans-induced periodontitis in mice. J Clin Periodontol 2012; 39:608-16. [DOI: 10.1111/j.1600-051x.2012.01886.x] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/24/2012] [Indexed: 01/19/2023]
Affiliation(s)
| | | | - Fernanda Matos Coelho
- Department of Biochemistry and Immunology, Instituto de Ciências Biológicas; Universidade Federal de Minas Gerais; Minas Gerais; Brazil
| | | | - Luíza Castro Menezes Cândido
- Department of Biochemistry and Immunology, Instituto de Ciências Biológicas; Universidade Federal de Minas Gerais; Minas Gerais; Brazil
| | - Gustavo Pompermaier Garlet
- Department of Biological Sciences; School of Dentistry of Bauru, Universidade de São Paulo; São Paulo; Brazil
| | - Mauro Martins Teixeira
- Department of Biochemistry and Immunology, Instituto de Ciências Biológicas; Universidade Federal de Minas Gerais; Minas Gerais; Brazil
| | - Daniele da Glória Souza
- Department of Microbiology, Instituto de Ciências Biológicas; Universidade Federal de Minas Gerais; Minas Gerais; Brazil
| | | |
Collapse
|
47
|
Quinteiro M, Napimoga M, Mesquita K, Clemente-Napimoga J. The indirect antinociceptive mechanism of 15d-PGJ2 on rheumatoid arthritis-induced TMJ inflammatory pain in rats. Eur J Pain 2012; 16:1106-15. [DOI: 10.1002/j.1532-2149.2012.00114.x] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023]
Affiliation(s)
- M.S. Quinteiro
- Laboratory of Biopathology and Molecular Biology; University of Uberaba; Brazil
| | | | - K.P. Mesquita
- Laboratory of Biopathology and Molecular Biology; University of Uberaba; Brazil
| | | |
Collapse
|
48
|
Queiroz-Junior CM, Madeira MFM, Coelho FM, Costa VV, Bessoni RLC, Sousa LFDC, Garlet GP, Souza DDGD, Teixeira MM, Silva TAD. Experimental arthritis triggers periodontal disease in mice: involvement of TNF-α and the oral Microbiota. THE JOURNAL OF IMMUNOLOGY 2011; 187:3821-30. [PMID: 21890656 DOI: 10.4049/jimmunol.1101195] [Citation(s) in RCA: 72] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/29/2023]
Abstract
Rheumatoid arthritis (RA) and periodontal disease (PD) are prevalent chronic inflammatory disorders that affect bone structures. Individuals with RA are more likely to experience PD, but how disease in joints could induce PD remains unknown. This study aimed to experimentally mimic clinical parameters of RA-induced PD and to provide mechanistic findings to explain this association. Chronic Ag-induced arthritis (AIA) was triggered by injection of methylated BSA in the knee joint of immunized mice. Anti-TNF-α was used to assess the role of this cytokine. Intra-articular challenge induced infiltration of cells, synovial hyperplasia, bone resorption, proteoglycan loss, and increased expression of cytokines exclusively in challenged joints. Simultaneously, AIA resulted in severe alveolar bone loss, migration of osteoclasts, and release of proinflammatory cytokines in maxillae. Anti-TNF-α therapy prevented the development of both AIA and PD. AIA did not modify bacterial counts in the oral cavity. PD, but not AIA, induced by injection of Ag in immunized mice was decreased by local treatment with antiseptic, which decreased the oral microbiota. AIA was associated with an increase in serum C-reactive protein levels and the expression of the transcription factors RORγ and Foxp3 in cervical lymph nodes. There were higher titers of anti-collagen I IgG, and splenocytes were more responsive to collagen I in AIA mice. In conclusion, AIA-induced PD was dependent on TNF-α and the oral microbiota. Moreover, PD was associated with changes in expression of lymphocyte transcription factors, presence of anti-collagen Abs, and increased reactivity to autoantigens.
Collapse
Affiliation(s)
- Celso Martins Queiroz-Junior
- Departamento de Clínica, Patologia e Cirurgia Odontológicas, Faculdade de Odontologia, Universidade Federal de Minas Gerais, Minas Gerais, Brazil
| | | | | | | | | | | | | | | | | | | |
Collapse
|