1
|
Shao G, Wang X, Zheng Y, Ma J, Wang L, Yan Z, Sun Z, Zhang S, Wu H, Lv Y, Huang H, Li J, Zhu T, Yang B, Wang N, Chen T, Guo X, Jin Y, Kang J, Wang H, Cao Y, Fu C. Identification of ANXA1 as a Novel Upstream Negative Regulator of Notch1 Function in AML. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024:e2409726. [PMID: 39447086 DOI: 10.1002/advs.202409726] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/15/2024] [Revised: 10/06/2024] [Indexed: 10/26/2024]
Abstract
Abnormal Notch1 expression has an important role in tumorigenesis. However, upstream control mechanisms for Notch1 are still insufficiently understood. Acute myeloid leukemia (AML) is one of the most common and lethal blood malignancies with limited possibilities for treatment. Thus, new therapeutic targets are urgently needed to improve current ineffective therapies. Herein, high Annexin A1 (ANXA1) expression is found correlated with hyperproliferation of AML cells, and then ANXA1 is identified as a novel negative regulator of Notch1 function in AML. Mechanistically, ANXA1 directly bound to the intracellular domain of Notch1 (NICD) to target this tumor suppressor for degradation. Furthermore, NICD executed its tumor suppressive function through activation of the p15 promoter. Thus, ablation of the Notch1-p15-mediated tumor suppression by ANXA1 provided a novel mechanism of AML proliferation. In human AML patients, a mutual exclusive relation is discovered between ANXA1 and Notch1/p15, corroborating mechanistic discovery. On the basis of these results, it is reasonably speculated that targeting ANXA1 would provide an effective approach for treatment of AML. In support of this new therapeutic paradigm, provided proof-of-concept data by antagonizing ANXA1 using NICD inhibitory peptides.
Collapse
Affiliation(s)
- Gang Shao
- Zhejiang Provincial Key Laboratory of Silkworm Bioreactor and Biomedicine, College of Life Sciences and Medicine, Zhejiang Sci-Tech University, Hangzhou, 310018, China
- Department of Oncology, No.903 Hospital of PLA Joint Logistic Support Force, Hangzhou, 310013, China
- College of Life Sciences, China Jiliang University, Hangzhou, 310018, China
| | - Xi Wang
- Department of Oncology, No.903 Hospital of PLA Joint Logistic Support Force, Hangzhou, 310013, China
| | - Yiting Zheng
- Zhejiang Provincial Key Laboratory of Silkworm Bioreactor and Biomedicine, College of Life Sciences and Medicine, Zhejiang Sci-Tech University, Hangzhou, 310018, China
| | - Junjie Ma
- Zhejiang Provincial Key Laboratory of Silkworm Bioreactor and Biomedicine, College of Life Sciences and Medicine, Zhejiang Sci-Tech University, Hangzhou, 310018, China
| | - Lei Wang
- Zhejiang Provincial Key Laboratory of Silkworm Bioreactor and Biomedicine, College of Life Sciences and Medicine, Zhejiang Sci-Tech University, Hangzhou, 310018, China
| | - Zhibin Yan
- Zhejiang Provincial Key Laboratory of Silkworm Bioreactor and Biomedicine, College of Life Sciences and Medicine, Zhejiang Sci-Tech University, Hangzhou, 310018, China
| | - Zeyu Sun
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310003, China
| | - Shuyuan Zhang
- Department of Neurosurgery, Zhejiang Cancer Hospital, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, 310022, China
| | - Hongzhang Wu
- Zhejiang Provincial Key Laboratory of Silkworm Bioreactor and Biomedicine, College of Life Sciences and Medicine, Zhejiang Sci-Tech University, Hangzhou, 310018, China
| | - Yudie Lv
- Zhejiang Provincial Key Laboratory of Silkworm Bioreactor and Biomedicine, College of Life Sciences and Medicine, Zhejiang Sci-Tech University, Hangzhou, 310018, China
- College of Life Sciences, China Jiliang University, Hangzhou, 310018, China
| | - Hemiao Huang
- Zhejiang Provincial Key Laboratory of Silkworm Bioreactor and Biomedicine, College of Life Sciences and Medicine, Zhejiang Sci-Tech University, Hangzhou, 310018, China
| | - Jianhu Li
- Department of Hematology, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310003, China
| | - Tianyi Zhu
- Zhejiang Provincial Key Laboratory for Cancer Molecular Cell Biology, Life Sciences Institute, Zhejiang University, Hangzhou, 310058, China
| | - Bing Yang
- Zhejiang Provincial Key Laboratory for Cancer Molecular Cell Biology, Life Sciences Institute, Zhejiang University, Hangzhou, 310058, China
| | - Nanxi Wang
- State Key Laboratory Cultivation Base for TCM Quality and Efficacy, School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, 210046, China
| | - Tao Chen
- Sartorius (Shanghai) Trading Co., Ltd., Shanghai, 200120, China
| | - Xuancheng Guo
- Hangzhou Acnovia Biotech Co., Ltd., Hangzhou, 310018, China
| | - Yuanting Jin
- College of Life Sciences, China Jiliang University, Hangzhou, 310018, China
| | - Jian Kang
- Oncogenic Signalling and Growth Control Program, Peter MacCallum Cancer Centre, 305 Grattan street, Melbourne, Victoria, 3000, Australia
- Sir Peter MacCallum Department of Oncology, University of Melbourne, Melbourne, Victoria, 3000, Australia
| | - Huafeng Wang
- Department of Hematology, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310003, China
| | - Yihai Cao
- Department of Microbiology, Tumor and Cell Biology, Karolinska Institute, Stockholm, 171 77, Sweden
| | - Caiyun Fu
- Zhejiang Provincial Key Laboratory of Silkworm Bioreactor and Biomedicine, College of Life Sciences and Medicine, Zhejiang Sci-Tech University, Hangzhou, 310018, China
- Key Laboratory of Preclinical Study for New Drugs of Gansu Province, Research Unit of Peptide Science, Chinese Academy of Medical Sciences, 2019RU066, Lanzhou University, Lanzhou, 730000, China
| |
Collapse
|
2
|
Scharf P, Sandri S, Rizzetto F, Xavier LF, Grosso D, Correia-Silva RD, Farsky PS, Gil CD, Farsky SHP. GPCRs overexpression and impaired fMLP-induced functions in neutrophils from chronic kidney disease patients. Front Immunol 2024; 15:1387566. [PMID: 39253088 PMCID: PMC11381270 DOI: 10.3389/fimmu.2024.1387566] [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: 02/18/2024] [Accepted: 08/02/2024] [Indexed: 09/11/2024] Open
Abstract
Introduction G-protein coupled receptors (GPCRs) expressed on neutrophils regulate their mobilization from the bone marrow into the blood, their half-live in the circulation, and their pro- and anti-inflammatory activities during inflammation. Chronic kidney disease (CKD) is associated with systemic inflammatory responses, and neutrophilia is a hallmark of CKD onset and progression. Nonetheless, the role of neutrophils in CKD is currently unclear. Methods Blood and renal tissue were collected from non-dialysis CKD (grade 3 - 5) patients to evaluate GPCR neutrophil expressions and functions in CKD development. Results CKD patients presented a higher blood neutrophil-to-lymphocyte ratio (NLR), which was inversely correlated with the glomerular filtration rate (eGFR). A higher frequency of neutrophils expressing the senescent GPCR receptor (CXCR4) and activation markers (CD18+CD11b+CD62L+) was detected in CKD patients. Moreover, CKD neutrophils expressed higher amounts of GPCR formyl peptide receptors (FPR) 1 and 2, known as neutrophil pro- and anti-inflammatory receptors, respectively. Cytoskeletal organization, migration, and production of reactive oxygen species (ROS) by CKD neutrophils were impaired in response to the FPR1 agonist (fMLP), despite the higher expression of FPR1. In addition, CKD neutrophils presented enhanced intracellular, but reduced membrane expression of the protein Annexin A1 (AnxA1), and an impaired ability to secrete it into the extracellular compartment. Secreted and phosphorylated AnxA1 is a recognized ligand of FPR2, pivotal in anti-inflammatory and efferocytosis effects. CKD renal tissue presented a low number of neutrophils, which were AnxA1+. Conclusion Together, these data highlight that CKD neutrophils overexpress GPCRs, which may contribute to an unbalanced aging process in the circulation, migration into inflamed tissues, and efferocytosis.
Collapse
Affiliation(s)
- Pablo Scharf
- Department of Clinical and Toxicological Analyses, School of Pharmaceutical Sciences, University of São Paulo, São Paulo, Brazil
| | - Silvana Sandri
- Department of Clinical and Toxicological Analyses, School of Pharmaceutical Sciences, University of São Paulo, São Paulo, Brazil
| | - Felipe Rizzetto
- Department of Clinical and Toxicological Analyses, School of Pharmaceutical Sciences, University of São Paulo, São Paulo, Brazil
- Lagoa Federal Hospital, Rio de Janeiro, Rio de Janeiro, Brazil
| | - Luana Filippi Xavier
- Department of Clinical and Toxicological Analyses, School of Pharmaceutical Sciences, University of São Paulo, São Paulo, Brazil
| | | | - Rebeca D Correia-Silva
- Department of Morphology and Genetics, Federal University of São Paulo, São Paulo, São Paulo, Brazil
| | - Pedro S Farsky
- Dante Pazzanese Institute of Cardiology of Sao Paulo, São Paulo, São Paulo, Brazil
| | - Cristiane D Gil
- Department of Morphology and Genetics, Federal University of São Paulo, São Paulo, São Paulo, Brazil
| | - Sandra Helena Poliselli Farsky
- Department of Clinical and Toxicological Analyses, School of Pharmaceutical Sciences, University of São Paulo, São Paulo, Brazil
| |
Collapse
|
3
|
Wang X, Yang B, Xiong T, Qiu Y, Qin Y, Liang X, Lu D, Yang X. Identification of potential biomarkers of gout through weighted gene correlation network analysis. Front Immunol 2024; 15:1367019. [PMID: 38686389 PMCID: PMC11056514 DOI: 10.3389/fimmu.2024.1367019] [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: 01/08/2024] [Accepted: 03/28/2024] [Indexed: 05/02/2024] Open
Abstract
Background Although hyperuricemia is not always associated with acute gouty arthritis, uric acid is a significant risk factor for gout. Therefore, we investigated the specific mechanism of uric acid activity. Methods Using the gout-associated transcriptome dataset GSE160170, we conducted differential expression analysis to identify differentially expressed genes (DEGs). Moreover, we discovered highly linked gene modules using weighted gene coexpression network analysis (WGCNA) and evaluated their intersection. Subsequently, we screened for relevant biomarkers using the cytoHubba and Mcode algorithms in the STRING database, investigated their connection to immune cells and constructed a competitive endogenous RNA (ceRNA) network to identify upstream miRNAs and lncRNAs. We also collected PBMCs from acute gouty arthritis patients and healthy individuals and constructed a THP-1 cell gout inflammatory model, RT-qPCR and western blotting (WB) were used to detect the expression of C-X-C motif ligand 8 (CXCL8), C-X-C motif ligand 2 (CXCL2), and C-X-C motif ligand 1 (CXCL1). Finally, we predicted relevant drug targets through hub genes, hoping to find better treatments. Results According to differential expression analysis, there were 76 upregulated and 28 downregulated mRNAs in GSE160170. Additionally, WGCNA showed that the turquoise module was most strongly correlated with primary gout; 86 hub genes were eventually obtained upon intersection. IL1β, IL6, CXCL8, CXCL1, and CXCL2 are the principal hub genes of the protein-protein interaction (PPI) network. Using RT-qPCR and WB, we found that there were significant differences in the expression levels of CXCL8, CXCL1, and CXCL2 between the gouty group and the healthy group, and we also predicted 10 chemicals related to these proteins. Conclusion In this study, we screened and validated essential genes using a variety of bioinformatics tools to generate novel ideas for the diagnosis and treatment of gout.
Collapse
Affiliation(s)
- Xinyi Wang
- Department of Endocrinology, First Affiliated Hospital, Guangxi Medical University, Nanning, China
| | - Bing Yang
- Department of Geriatric Endocrinology and Metabolism, First Affiliated Hospital, Guangxi Medical University, Nanning, China
| | - Tian Xiong
- Department of Geriatric Endocrinology and Metabolism, First Affiliated Hospital, Guangxi Medical University, Nanning, China
| | - Yu Qiu
- Department of Geriatric Endocrinology and Metabolism, First Affiliated Hospital, Guangxi Medical University, Nanning, China
| | - Yingfen Qin
- Department of Endocrinology, First Affiliated Hospital, Guangxi Medical University, Nanning, China
| | - Xinghuan Liang
- Department of Endocrinology, First Affiliated Hospital, Guangxi Medical University, Nanning, China
| | - Decheng Lu
- Department of Endocrinology, Wuming Hospital, Guangxi Medical University, Nanning, China
| | - Xi Yang
- Department of Geriatric Endocrinology and Metabolism, First Affiliated Hospital, Guangxi Medical University, Nanning, China
- Guangxi Key Laboratory of Precision Medicine in Cardio-cerebrovascular Diseases Control and Prevention, Nanning, China
- Guangxi Clinical Research Center for Cardio-cerebrovascular Diseases, Nanning, China
| |
Collapse
|
4
|
Broering MF, Oseliero Filho PL, Borges PP, da Silva LCC, Knirsch MC, Xavier LF, Scharf P, Sandri S, Stephano MA, de Oliveira FA, Sayed IM, Gamarra LF, Das S, Fantini MCA, Farsky SHP. Development of Ac2-26 Mesoporous Microparticle System as a Potential Therapeutic Agent for Inflammatory Bowel Diseases. Int J Nanomedicine 2024; 19:3537-3554. [PMID: 38638365 PMCID: PMC11024051 DOI: 10.2147/ijn.s451589] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2023] [Accepted: 03/29/2024] [Indexed: 04/20/2024] Open
Abstract
Introduction Inflammatory bowel diseases (IBDs) disrupt the intestinal epithelium, leading to severe chronic inflammation. Current therapies cause adverse effects and are expensive, invasive, and ineffective for most patients. Annexin A1 (AnxA1) is a pivotal endogenous anti-inflammatory and tissue repair protein in IBD. Nanostructured compounds loading AnxA1 or its active N-terminal mimetic peptides improve IBD symptomatology. Methods To further explore their potential as a therapeutic candidate, the AnxA1 N-terminal mimetic peptide Ac2-26 was incorporated into SBA-15 ordered mesoporous silica and covered with EL30D-55 to deliver it by oral treatment into the inflamed gut. Results The systems SBA-Ac2-26 developed measurements revealed self-assembled rod-shaped particles, likely on the external surface of SBA-15, and 88% of peptide incorporation. SBA-15 carried the peptide Ac2-26 into cultured Raw 264.7 macrophages and Caco-2 epithelial cells. Moreover, oral administration of Eudragit-SBA-15-Ac2-26 (200 μg; once a day; for 4 days) reduced colitis clinical symptoms, inflammation, and improved epithelium recovery in mice under dextran-sodium sulfate-induced colitis. Discussion The absorption of SBA-15 in gut epithelial cells is typically low; however, the permeable inflamed barrier can enable microparticles to cross, being phagocyted by macrophages. These findings suggest that Ac2-26 is successfully delivered and binds to its receptors in both epithelial and immune cells, aligning with the clinical results. Conclusion Our findings demonstrate a simple and cost-effective approach to delivering Ac2-26 orally into the inflamed gut, highlighting its potential as non-invasive IBD therapy.
Collapse
Affiliation(s)
- Milena Fronza Broering
- Department of Clinical and Toxicological Analyses, School of Pharmaceutical Sciences, University of São Paulo, São Paulo, SP, Brazil
- Department of Biomedical and Nutritional Sciences, University of Massachusetts, Lowell, MA, USA
| | - Pedro Leonidas Oseliero Filho
- Department of Applied Physics, Physics Institute, University of Sao Paulo, São Paulo, Brazil
- Materials Innovation Factory, University of Liverpool, Liverpool, MSY, UK
| | - Pâmela Pacassa Borges
- Department of Clinical and Toxicological Analyses, School of Pharmaceutical Sciences, University of São Paulo, São Paulo, SP, Brazil
| | | | - Marcos Camargo Knirsch
- Department of Biochemical and Pharmaceutical Technology, School of Pharmaceutical Sciences, University of São Paulo, São Paulo, SP, Brazil
| | - Luana Filippi Xavier
- Department of Clinical and Toxicological Analyses, School of Pharmaceutical Sciences, University of São Paulo, São Paulo, SP, Brazil
| | - Pablo Scharf
- Department of Clinical and Toxicological Analyses, School of Pharmaceutical Sciences, University of São Paulo, São Paulo, SP, Brazil
| | - Silvana Sandri
- Department of Clinical and Toxicological Analyses, School of Pharmaceutical Sciences, University of São Paulo, São Paulo, SP, Brazil
| | - Marco Antonio Stephano
- Department of Biochemical and Pharmaceutical Technology, School of Pharmaceutical Sciences, University of São Paulo, São Paulo, SP, Brazil
| | - Fernando Anselmo de Oliveira
- Instituto do Cérebro, Instituto Israelita de Ensino e Pesquisa, Sociedade Beneficente Israelita Brasileira Hospital Albert Einstein, São Paulo, SP, Brazil
| | - Ibrahim M Sayed
- Department of Biomedical and Nutritional Sciences, University of Massachusetts, Lowell, MA, USA
| | - Lionel Fernel Gamarra
- Instituto do Cérebro, Instituto Israelita de Ensino e Pesquisa, Sociedade Beneficente Israelita Brasileira Hospital Albert Einstein, São Paulo, SP, Brazil
| | - Soumita Das
- Department of Biomedical and Nutritional Sciences, University of Massachusetts, Lowell, MA, USA
| | - Márcia C A Fantini
- Department of Applied Physics, Physics Institute, University of Sao Paulo, São Paulo, Brazil
| | - Sandra H P Farsky
- Department of Clinical and Toxicological Analyses, School of Pharmaceutical Sciences, University of São Paulo, São Paulo, SP, Brazil
| |
Collapse
|
5
|
Ricotta TQN, Dos Santos LM, Oliveira LG, Souza-Testasicca MC, Nascimento FC, Vago JP, Carvalho AFS, Queiroz-Junior CM, Sousa LP, Fernandes AP. Annexin A1 improves immune responses and control of tissue parasitism during Leishmania amazonensis infection in BALB/c mice. Biomed Pharmacother 2024; 172:116254. [PMID: 38340398 DOI: 10.1016/j.biopha.2024.116254] [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: 12/14/2023] [Revised: 02/01/2024] [Accepted: 02/02/2024] [Indexed: 02/12/2024] Open
Abstract
Leishmaniases, a group of diseases caused by the species of the protozoan parasite Leishmania, remains a significant public health concern worldwide. Host immune responses play a crucial role in the outcome of Leishmania infections, and several mediators that regulate inflammatory responses are potential targets for therapeutic approaches. Annexin A1 (AnxA1), an endogenous protein endowed with anti-inflammatory and pro-resolving properties, has emerged as a potential player. We have shown that during L. braziliensis infection, deficiency of AnxA1 exacerbates inflammatory responses but does not affect parasite burden. Here, we have investigated the role of AnxA1 in L. amazonensis infection, given the non-healing and progressive lesions characteristic of this infectious model. Infection of AnxA1 KO BALB/c mice resulted in increased lesion size and tissue damage associated with higher parasite burdens and enhanced inflammatory response. Notably, therapeutic application of the AnxA1 peptidomimetic Ac2-26 improves control of parasite replication and increases IL-10 production in vivo and in vitro, in both WT and AnxA1 KO mice. Conversely, administration of WRW4, an inhibitor of FPR2/3, resulted in larger lesions and decreased production of IL-10, suggesting that the effects of AnxA1 during L. amazonensis infection are associated with the engagement of these receptors. Our study illuminates the role of AnxA1 in L. amazonensis infection, demonstrating its impact on the susceptibility phenotype of BALB/c mice. Furthermore, our results indicate that targeting the AnxA1 pathway by using the Ac2-26 peptide could represent a promising alternative for new treatments for leishmaniasis.
Collapse
Affiliation(s)
- Tiago Queiroga Nery Ricotta
- Departamento de Análises Clínicas e Toxicológicas, Faculdade de Farmácia, Universidade Federal de Minas Gerais, Brazil
| | - Liliane Martins Dos Santos
- Departamento de Análises Clínicas e Toxicológicas, Faculdade de Farmácia, Universidade Federal de Minas Gerais, Brazil
| | - Leandro Gonzaga Oliveira
- Departamento de Análises Clínicas e Toxicológicas, Faculdade de Farmácia, Universidade Federal de Minas Gerais, Brazil
| | | | - Frederico Crepaldi Nascimento
- Departamento de Análises Clínicas e Toxicológicas, Faculdade de Farmácia, Universidade Federal de Minas Gerais, Brazil
| | - Juliana P Vago
- Departamento de Análises Clínicas e Toxicológicas, Faculdade de Farmácia, Universidade Federal de Minas Gerais, Brazil
| | - Antônio Felipe S Carvalho
- Departamento de Análises Clínicas e Toxicológicas, Faculdade de Farmácia, Universidade Federal de Minas Gerais, Brazil
| | | | - Lirlândia P Sousa
- Departamento de Análises Clínicas e Toxicológicas, Faculdade de Farmácia, Universidade Federal de Minas Gerais, Brazil
| | - Ana Paula Fernandes
- Departamento de Análises Clínicas e Toxicológicas, Faculdade de Farmácia, Universidade Federal de Minas Gerais, Brazil.
| |
Collapse
|
6
|
Yu H, Xue W, Yu H, Song Y, Liu X, Qin L, Wang S, Bao H, Gu H, Chen G, Zhao D, Tu Y, Cheng J, Wang L, Ai Z, Hu D, Wang L, Peng A. Single-cell transcriptomics reveals variations in monocytes and Tregs between gout flare and remission. JCI Insight 2023; 8:e171417. [PMID: 38063198 PMCID: PMC10795830 DOI: 10.1172/jci.insight.171417] [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: 04/12/2023] [Accepted: 10/25/2023] [Indexed: 12/18/2023] Open
Abstract
Gout commonly manifests as a painful, self-limiting inflammatory arthritis. Nevertheless, the understanding of the inflammatory and immune responses underlying gout flares and remission remains ambiguous. Here, based on single-cell RNA-Seq and an independent validation cohort, we identified the potential mechanism of gout flare, which likely involves the upregulation of HLA-DQA1+ nonclassical monocytes and is related to antigen processing and presentation. Furthermore, Tregs also play an essential role in the suppressive capacity during gout remission. Cell communication analysis suggested the existence of altered crosstalk between monocytes and other T cell types, such as Tregs. Moreover, we observed the systemic upregulation of inflammatory and cytokine genes, primarily in classical monocytes, during gout flares. All monocyte subtypes showed increased arachidonic acid metabolic activity along with upregulation of prostaglandin-endoperoxide synthase 2 (PTGS2). We also detected a decrease in blood arachidonic acid and an increase in leukotriene B4 levels during gout flares. In summary, our study illustrates the distinctive immune cell responses and systemic inflammation patterns that characterize the transition from gout flares to remission, and it suggests that blood monocyte subtypes and Tregs are potential intervention targets for preventing recurrent gout attacks and progression.
Collapse
Affiliation(s)
- Hanjie Yu
- Center for Nephrology and Clinical Metabolomics and Division of Nephrology, Shanghai Tenth People’s Hospital, and
| | - Wen Xue
- Center for Nephrology and Clinical Metabolomics and Division of Nephrology, Shanghai Tenth People’s Hospital, and
| | - Hanqing Yu
- Center for Nephrology and Clinical Metabolomics and Division of Nephrology, Shanghai Tenth People’s Hospital, and
| | - Yaxiang Song
- Center for Nephrology and Clinical Metabolomics and Division of Nephrology, Shanghai Tenth People’s Hospital, and
| | - Xinying Liu
- Center for Nephrology and Clinical Metabolomics and Division of Nephrology, Shanghai Tenth People’s Hospital, and
| | - Ling Qin
- Center for Nephrology and Clinical Metabolomics and Division of Nephrology, Shanghai Tenth People’s Hospital, and
| | - Shu Wang
- Center for Nephrology and Clinical Metabolomics and Division of Nephrology, Shanghai Tenth People’s Hospital, and
| | - Hui Bao
- Center for Nephrology and Clinical Metabolomics and Division of Nephrology, Shanghai Tenth People’s Hospital, and
| | - Hongchen Gu
- Center for Nephrology and Clinical Metabolomics and Division of Nephrology, Shanghai Tenth People’s Hospital, and
| | - Guangqi Chen
- Center for Nephrology and Clinical Metabolomics and Division of Nephrology, Shanghai Tenth People’s Hospital, and
| | - Dake Zhao
- Center for Nephrology and Clinical Metabolomics and Division of Nephrology, Shanghai Tenth People’s Hospital, and
| | - Yang Tu
- Center for Nephrology and Clinical Metabolomics and Division of Nephrology, Shanghai Tenth People’s Hospital, and
| | - Jiafen Cheng
- Center for Nephrology and Clinical Metabolomics and Division of Nephrology, Shanghai Tenth People’s Hospital, and
| | - Liya Wang
- Center for Nephrology and Clinical Metabolomics and Division of Nephrology, Shanghai Tenth People’s Hospital, and
| | - Zisheng Ai
- Department of Medical Statistics, Tongji University School of Medicine, Shanghai, China
| | - Dayong Hu
- Center for Nephrology and Clinical Metabolomics and Division of Nephrology, Shanghai Tenth People’s Hospital, and
| | - Ling Wang
- Center for Nephrology and Clinical Metabolomics and Division of Nephrology, Shanghai Tenth People’s Hospital, and
| | - Ai Peng
- Center for Nephrology and Clinical Metabolomics and Division of Nephrology, Shanghai Tenth People’s Hospital, and
| |
Collapse
|
7
|
Zaninelli TH, Martelossi-Cebinelli G, Saraiva-Santos T, Borghi SM, Fattori V, Casagrande R, Verri WA. New drug targets for the treatment of gout arthritis: what's new? Expert Opin Ther Targets 2023; 27:679-703. [PMID: 37651647 DOI: 10.1080/14728222.2023.2247559] [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: 04/12/2023] [Revised: 06/14/2023] [Accepted: 08/09/2023] [Indexed: 09/02/2023]
Abstract
INTRODUCTION Gout arthritis (GA) is an intermittent inflammatory disease affecting approximately 10% of the worldwide population. Symptomatic phases (acute flares) are timely spaced by asymptomatic periods. During an acute attack, redness, joint swelling, limited movement, and excruciating pain are common symptoms. However, the current available therapies are not fully effective in reducing symptoms and offer numerous side effects. Therefore, unveiling new drug targets and effector molecules are required in developing novel GA therapeutics. AREAS COVERED This review discusses the pathophysiological mechanisms of GA and explores potential pharmacological targets to ameliorate disease outcome. In addition, we listed promising pre-clinical studies demonstrating effector molecules with therapeutical potential. Among those, we emphasized the importance of natural products, including traditional Chinese medicine formulas and their multitarget mechanisms of action. EXPERT OPINION In our search, we observed that there is a massive gap between pre-clinical and clinical knowledge. Only a minority (4.4%) of clinical trials aimed to intervene by applying natural products or current hot targets described herein. In this sense, we envisage four possibilities for GA therapeutics, which include the repurposing of existing therapies, ALX/FPR2 agonism for improvement in disease outcome, the use of multitarget drugs (e.g. natural products), and targeting the neuroinflammatory component of GA.
Collapse
Affiliation(s)
- Tiago H Zaninelli
- Laboratory of Pain, Inflammation, Neuropathy, and Cancer, Department of Pathology, Centre of Biological Sciences, Londrina State University, Londrina, Paraná, Brazil
| | - Geovana Martelossi-Cebinelli
- Laboratory of Pain, Inflammation, Neuropathy, and Cancer, Department of Pathology, Centre of Biological Sciences, Londrina State University, Londrina, Paraná, Brazil
| | - Telma Saraiva-Santos
- Laboratory of Pain, Inflammation, Neuropathy, and Cancer, Department of Pathology, Centre of Biological Sciences, Londrina State University, Londrina, Paraná, Brazil
| | - Sergio M Borghi
- Laboratory of Pain, Inflammation, Neuropathy, and Cancer, Department of Pathology, Centre of Biological Sciences, Londrina State University, Londrina, Paraná, Brazil
- Center for Research in Health Sciences, University of Northern Londrina, Londrina, Brazil
| | - Victor Fattori
- Laboratory of Pain, Inflammation, Neuropathy, and Cancer, Department of Pathology, Centre of Biological Sciences, Londrina State University, Londrina, Paraná, Brazil
- Vascular Biology Program, Department of Surgery, Boston Children's Hospital-Harvard Medical School, Karp Research Building, Boston, MA, USA
| | - Rubia Casagrande
- Laboratory of Antioxidants and Inflammation, Department of Pharmaceutical Sciences, Centre of Health Sciences, Londrina State University, Londrina, Brazil
| | - Waldiceu A Verri
- Laboratory of Pain, Inflammation, Neuropathy, and Cancer, Department of Pathology, Centre of Biological Sciences, Londrina State University, Londrina, Paraná, Brazil
| |
Collapse
|
8
|
Grossi LC, Zaidan I, Souza JAM, Carvalho AFS, Sanches RCO, Cardoso C, Lara ES, Montuori-Andrade ACM, Bruscoli S, Marchetti MC, Riccardi C, Teixeira MM, Tavares LP, Vago JP, Sousa LP. GILZ Modulates the Recruitment of Monocytes/Macrophages Endowed with a Resolving Phenotype and Favors Resolution of Escherichia coli Infection. Cells 2023; 12:1403. [PMID: 37408237 DOI: 10.3390/cells12101403] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2023] [Revised: 04/30/2023] [Accepted: 05/12/2023] [Indexed: 07/07/2023] Open
Abstract
Macrophages are important effectors of inflammation resolution that contribute to the elimination of pathogens and apoptotic cells and restoration of homeostasis. Pre-clinical studies have evidenced the anti-inflammatory and pro-resolving actions of GILZ (glucocorticoid-induced leucine zipper). Here, we evaluated the role of GILZ on the migration of mononuclear cells under nonphlogistic conditions and Escherichia coli-evoked peritonitis. TAT-GILZ (a cell-permeable GILZ-fusion protein) injection into the pleural cavity of mice induced monocyte/macrophage influx alongside increased CCL2, IL-10 and TGF-β levels. TAT-GILZ-recruited macrophages showed a regulatory phenotype, exhibiting increased expression of CD206 and YM1. During the resolving phase of E. coli-induced peritonitis, marked by an increased recruitment of mononuclear cells, lower numbers of these cells and CCL2 levels were found in the peritoneal cavity of GILZ-deficient mice (GILZ-/-) when compared to WT. In addition, GILZ-/- showed higher bacterial loads, lower apoptosis/efferocytosis counts and a lower number of macrophages with pro-resolving phenotypes. TAT-GILZ accelerated resolution of E. coli-evoked neutrophilic inflammation, which was associated with increased peritoneal numbers of monocytes/macrophages, enhanced apoptosis/efferocytosis counts and bacterial clearance through phagocytosis. Taken together, we provided evidence that GILZ modulates macrophage migration with a regulatory phenotype, inducing bacterial clearance and accelerating the resolution of peritonitis induced by E. coli.
Collapse
Affiliation(s)
- Laís C Grossi
- Signaling in Inflammation Lab., Departamento de Análises Clínicas e Toxicológicas, Faculdade de Farmácia, Universidade Federal de Minas Gerais, Belo Horizonte 31270-901, Brazil
- Programa de Pós-Graduação em Ciências Farmacêuticas, Faculdade de Farmácia, Universidade Federal de Minas Gerais, Belo Horizonte 31270-901, Brazil
| | - Isabella Zaidan
- Signaling in Inflammation Lab., Departamento de Análises Clínicas e Toxicológicas, Faculdade de Farmácia, Universidade Federal de Minas Gerais, Belo Horizonte 31270-901, Brazil
- Programa de Pós-Graduação em Ciências Farmacêuticas, Faculdade de Farmácia, Universidade Federal de Minas Gerais, Belo Horizonte 31270-901, Brazil
| | - Jéssica Amanda Marques Souza
- Signaling in Inflammation Lab., Departamento de Análises Clínicas e Toxicológicas, Faculdade de Farmácia, Universidade Federal de Minas Gerais, Belo Horizonte 31270-901, Brazil
- Programa de Pós-Graduação em Ciências Farmacêuticas, Faculdade de Farmácia, Universidade Federal de Minas Gerais, Belo Horizonte 31270-901, Brazil
| | - Antônio Felipe S Carvalho
- Signaling in Inflammation Lab., Departamento de Análises Clínicas e Toxicológicas, Faculdade de Farmácia, Universidade Federal de Minas Gerais, Belo Horizonte 31270-901, Brazil
- Programa de Pós-Graduação em Ciências Farmacêuticas, Faculdade de Farmácia, Universidade Federal de Minas Gerais, Belo Horizonte 31270-901, Brazil
- Hospital das Clínicas da Universidade Federal de Minas Gerais/Ebserh, Belo Horizonte 30130-100, Brazil
| | - Rodrigo C O Sanches
- Departamento de Bioquímica e Imunologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte 31270-901, Brazil
| | - Camila Cardoso
- Signaling in Inflammation Lab., Departamento de Análises Clínicas e Toxicológicas, Faculdade de Farmácia, Universidade Federal de Minas Gerais, Belo Horizonte 31270-901, Brazil
- Programa de Pós-Graduação em Ciências Farmacêuticas, Faculdade de Farmácia, Universidade Federal de Minas Gerais, Belo Horizonte 31270-901, Brazil
| | - Edvaldo S Lara
- Signaling in Inflammation Lab., Departamento de Análises Clínicas e Toxicológicas, Faculdade de Farmácia, Universidade Federal de Minas Gerais, Belo Horizonte 31270-901, Brazil
| | - Ana Clara M Montuori-Andrade
- Signaling in Inflammation Lab., Departamento de Análises Clínicas e Toxicológicas, Faculdade de Farmácia, Universidade Federal de Minas Gerais, Belo Horizonte 31270-901, Brazil
- Programa de Pós-Graduação em Ciências Farmacêuticas, Faculdade de Farmácia, Universidade Federal de Minas Gerais, Belo Horizonte 31270-901, Brazil
| | - Stefano Bruscoli
- Department of Medicine and Surgery, Section of Pharmacology, University of Perugia, 06132 Perugia, Italy
| | - Maria Cristina Marchetti
- Department of Medicine and Surgery, Section of Pharmacology, University of Perugia, 06132 Perugia, Italy
| | - Carlo Riccardi
- Department of Medicine and Surgery, Section of Pharmacology, University of Perugia, 06132 Perugia, Italy
| | - Mauro M Teixeira
- Centro de Pesquisa e Desenvolvimento de Fármacos, Departamento de Bioquímica e Imunologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte 31270-901, Brazil
| | - Luciana P Tavares
- Pulmonary and Critical Care Medicine Division, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA 02115, USA
| | - Juliana P Vago
- Experimental Rheumatology, Department of Rheumatology, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, 6525 GA Nijmegen, The Netherlands
| | - Lirlândia P Sousa
- Signaling in Inflammation Lab., Departamento de Análises Clínicas e Toxicológicas, Faculdade de Farmácia, Universidade Federal de Minas Gerais, Belo Horizonte 31270-901, Brazil
- Programa de Pós-Graduação em Ciências Farmacêuticas, Faculdade de Farmácia, Universidade Federal de Minas Gerais, Belo Horizonte 31270-901, Brazil
- Centro de Pesquisa e Desenvolvimento de Fármacos, Departamento de Bioquímica e Imunologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte 31270-901, Brazil
| |
Collapse
|
9
|
Resende F, de Araújo S, Tavares LP, Teixeira MM, Costa VV. The Multifaceted Role of Annexin A1 in Viral Infections. Cells 2023; 12:1131. [PMID: 37190040 PMCID: PMC10137178 DOI: 10.3390/cells12081131] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2023] [Revised: 04/04/2023] [Accepted: 04/06/2023] [Indexed: 05/17/2023] Open
Abstract
Dysregulated inflammatory responses are often correlated with disease severity during viral infections. Annexin A1 (AnxA1) is an endogenous pro-resolving protein that timely regulates inflammation by activating signaling pathways that culminate with the termination of response, clearance of pathogen and restoration of tissue homeostasis. Harnessing the pro-resolution actions of AnxA1 holds promise as a therapeutic strategy to control the severity of the clinical presentation of viral infections. In contrast, AnxA1 signaling might also be hijacked by viruses to promote pathogen survival and replication. Therefore, the role of AnxA1 during viral infections is complex and dynamic. In this review, we provide an in-depth view of the role of AnxA1 during viral infections, from pre-clinical to clinical studies. In addition, this review discusses the therapeutic potential for AnxA1 and AnxA1 mimetics in treating viral infections.
Collapse
Affiliation(s)
- Filipe Resende
- Post-Graduation Program of Cell Biology, Department of Morphology, Biological Sciences Institute, Federal University of Minas Gerais, Belo Horizonte 31270-901, Brazil
- Center for Research and Development of Drugs, Biological Sciences Institute, Federal University of Minas Gerais, Belo Horizonte 31270-901, Brazil
| | - Simone de Araújo
- Center for Research and Development of Drugs, Biological Sciences Institute, Federal University of Minas Gerais, Belo Horizonte 31270-901, Brazil
| | - Luciana Pádua Tavares
- Pulmonary and Critical Care Medicine Division, Department of Medicine, Brigham and Women’s Hospital and Harvard Medical School, Boston, MA 02115, USA;
| | - Mauro Martins Teixeira
- Center for Research and Development of Drugs, Biological Sciences Institute, Federal University of Minas Gerais, Belo Horizonte 31270-901, Brazil
- Department of Biochemistry and Immunology, Biological Sciences Institute, Federal University of Minas Gerais, Belo Horizonte 31270-901, Brazil
| | - Vivian Vasconcelos Costa
- Post-Graduation Program of Cell Biology, Department of Morphology, Biological Sciences Institute, Federal University of Minas Gerais, Belo Horizonte 31270-901, Brazil
- Center for Research and Development of Drugs, Biological Sciences Institute, Federal University of Minas Gerais, Belo Horizonte 31270-901, Brazil
| |
Collapse
|
10
|
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
|
11
|
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
|
12
|
Dadaya E, Koubala B, Ndjonka D, Zingué S, Laya A, Atsang G. Hydromethanolic Root Extract of Gnidia Kraussiana Demonstrates Anti-Inflammatory Effect Through Anti-Oxidant Activity Enhancement in a Rodent Model of Gout. Dose Response 2023; 21:15593258221148015. [PMID: 36743195 PMCID: PMC9893086 DOI: 10.1177/15593258221148015] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2021] [Accepted: 12/09/2022] [Indexed: 02/03/2023] Open
Abstract
Gout is a metabolic arthritis that originates from increased accumulation of monosodium urate (MSU) crystals in joints. This work aimed to evaluate the antioxidant and anti-inflammatory activities of the hydromethanolic extract of Gnidia kraussiana (HEGK) using model of Gouty arthritis on mice. The total polyphenol, flavonoid, tannin content and the antioxidant activity of HEGK were also evaluated. MSU-injected mice were treated daily for 3 days with HEGK (25, 50 and 100 mg/kg). Indomethacin and colchicin were used as reference drugs. Paw oedema and body temperature were measured at different time intervals post-injection. Malondialdehyde, acid phosphatase, β-Galactosidase, catalase, superoxide dismutase and glutathione levels were evaluated. HEGK is rich in polyphenol (129.93 mg/100 g), flavonoid (67.78 mg/100 g) and tannin conferring it a high antioxidant activity. Acute oral toxicity of HEGK resulted in LD50 greater than 2000 mg/kg. Oral administration of HEGK induced a significant decrease in the oedema of legs injected with urate crystals and reduced the release of acid phosphatase and β-Galactosidase. A model of oxidative damage was successfully established, revealing a significant increase in malondialdehyde and inhibition of antioxidants, including superoxide dismutase, catalase and glutathione activity. Thus, HEGK can actively inhibit the effect of inflammatory mediators in gouty arthritis.
Collapse
Affiliation(s)
- Elizé Dadaya
- Standard Institution, University of Maroua, Cameroon
| | - Benoit Koubala
- Standard Institution, University of Maroua, Cameroon,Benoit Koubala, Standard Institution, University of Maroua, Maroua 814, Cameroon.
| | | | | | - Alphonse Laya
- Standard Institution, University of Maroua, Cameroon
| | - Gisèle Atsang
- Standard Institution, University of Maroua, Cameroon
| |
Collapse
|
13
|
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
|
14
|
de Araújo S, de Melo Costa VR, Santos FM, de Sousa CDF, Moreira TP, Gonçalves MR, Félix FB, Queiroz-Junior CM, Campolina-Silva GH, Nogueira ML, Sugimoto MA, Bonilha CS, Perretti M, Souza DG, Costa VV, Teixeira MM. Annexin A1-FPR2/ALX Signaling Axis Regulates Acute Inflammation during Chikungunya Virus Infection. Cells 2022; 11:cells11172717. [PMID: 36078125 PMCID: PMC9454528 DOI: 10.3390/cells11172717] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2022] [Revised: 08/24/2022] [Accepted: 08/24/2022] [Indexed: 12/03/2022] Open
Abstract
Chikungunya (CHIKV) is an arthritogenic alphavirus that causes a self-limiting disease usually accompanied by joint pain and/or polyarthralgia with disabling characteristics. Immune responses developed during the acute phase of CHIKV infection determine the rate of disease progression and resolution. Annexin A1 (AnxA1) is involved in both initiating inflammation and preventing over-response, being essential for a balanced end of inflammation. In this study, we investigated the role of the AnxA1-FPR2/ALX pathway during CHIKV infection. Genetic deletion of AnxA1 or its receptor enhanced inflammatory responses driven by CHIKV. These knockout mice showed increased neutrophil accumulation and augmented tissue damage at the site of infection compared with control mice. Conversely, treatment of wild-type animals with the AnxA1 mimetic peptide (Ac2–26) reduced neutrophil accumulation, decreased local concentration of inflammatory mediators and diminished mechanical hypernociception and paw edema induced by CHIKV-infection. Alterations in viral load were mild both in genetic deletion or with treatment. Combined, our data suggest that the AnxA1-FPR2/ALX pathway is a potential therapeutic strategy to control CHIKV-induced acute inflammation and polyarthralgia.
Collapse
Affiliation(s)
- Simone de Araújo
- Graduate Program in Biological Sciences Physiology and Pharmacology, Department of Physiology and Biophysics, Institute of Biological Sciences, Federal University of Minas Gerais, Belo Horizonte 31270-901, Brazil
- Drug Research and Development Center, Institute of Biological Sciences, Federal University of Minas Gerais, Belo Horizonte 31270-901, Brazil
| | - Victor R. de Melo Costa
- Drug Research and Development Center, Institute of Biological Sciences, Federal University of Minas Gerais, Belo Horizonte 31270-901, Brazil
| | - Franciele M. Santos
- Drug Research and Development Center, Institute of Biological Sciences, Federal University of Minas Gerais, Belo Horizonte 31270-901, Brazil
| | - Carla D. Ferreira de Sousa
- Drug Research and Development Center, Institute of Biological Sciences, Federal University of Minas Gerais, Belo Horizonte 31270-901, Brazil
| | - Thaiane P. Moreira
- Drug Research and Development Center, Institute of Biological Sciences, Federal University of Minas Gerais, Belo Horizonte 31270-901, Brazil
| | - Matheus R. Gonçalves
- Drug Research and Development Center, Institute of Biological Sciences, Federal University of Minas Gerais, Belo Horizonte 31270-901, Brazil
| | - Franciel B. Félix
- Drug Research and Development Center, Institute of Biological Sciences, Federal University of Minas Gerais, Belo Horizonte 31270-901, Brazil
| | - Celso M. Queiroz-Junior
- Drug Research and Development Center, Institute of Biological Sciences, Federal University of Minas Gerais, Belo Horizonte 31270-901, Brazil
| | - Gabriel H. Campolina-Silva
- Drug Research and Development Center, Institute of Biological Sciences, Federal University of Minas Gerais, Belo Horizonte 31270-901, Brazil
- Department of Obstetrics, Gynecology and Reproduction, CHU de Quebec Research Center (CHUL), Université Laval, Quebec, QC G1V 0A6, Canada
| | - Maurício Lacerda Nogueira
- Department of Dermatological, Infections, and Parasitic Diseases, School of Medicine (FAMERP), São José do Rio Preto, São Paulo 15090-000, Brazil
| | - Michelle A. Sugimoto
- Barts and The London School of Medicine and Dentistry, William Harvey Research Institute, Queen Mary University of London, London E1 4NS, UK
| | - Caio S. Bonilha
- Drug Research and Development Center, Institute of Biological Sciences, Federal University of Minas Gerais, Belo Horizonte 31270-901, Brazil
- Center for Research on Inflammatory Diseases, University of São Paulo, São Paulo 05508-000, Brazil
- Institute of Infection, Immunity and Inflammation, University of Glasgow, Glasgow G12 8QQ, UK
| | - Mauro Perretti
- Barts and The London School of Medicine and Dentistry, William Harvey Research Institute, Queen Mary University of London, London E1 4NS, UK
- Centre for Inflammation and Therapeutic Innovation, Queen Mary University of London, London E1 4NS, UK
| | - Danielle G. Souza
- Graduate Program in Microbiology, Department of Microbiology, Institute of Biological Sciences, Federal University of Minas Gerais, Belo Horizonte 31270-901, Brazil
| | - Vivian V. Costa
- Drug Research and Development Center, Institute of Biological Sciences, Federal University of Minas Gerais, Belo Horizonte 31270-901, Brazil
- Graduate Program in Cell Biology, Department of Morphology, Federal University of Minas Gerais, Belo Horizonte 31270-901, Brazil
- Correspondence: (V.V.C.); (M.M.T.); Tel.: +55-31-3409-3082 (V.V.C.); +55-31-3409-2651 (M.M.T.)
| | - Mauro M. Teixeira
- Drug Research and Development Center, Institute of Biological Sciences, Federal University of Minas Gerais, Belo Horizonte 31270-901, Brazil
- Department of Biochemistry and Immunology, Institute of Biological Sciences, Federal University of Minas Gerais, Belo Horizonte 31270-901, Brazil
- Correspondence: (V.V.C.); (M.M.T.); Tel.: +55-31-3409-3082 (V.V.C.); +55-31-3409-2651 (M.M.T.)
| |
Collapse
|
15
|
Zhao J, Wei K, Jiang P, Chang C, Xu L, Xu L, Shi Y, Guo S, Xue Y, He D. Inflammatory Response to Regulated Cell Death in Gout and Its Functional Implications. Front Immunol 2022; 13:888306. [PMID: 35464445 PMCID: PMC9020265 DOI: 10.3389/fimmu.2022.888306] [Citation(s) in RCA: 20] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2022] [Accepted: 03/17/2022] [Indexed: 02/03/2023] Open
Abstract
Gout, a chronic inflammatory arthritis disease, is characterized by hyperuricemia and caused by interactions between genetic, epigenetic, and metabolic factors. Acute gout symptoms are triggered by the inflammatory response to monosodium urate crystals, which is mediated by the innate immune system and immune cells (e.g., macrophages and neutrophils), the NACHT, LRR, and PYD domains-containing protein 3 (NLRP3) inflammasome activation, and pro-inflammatory cytokine (e.g., IL-1β) release. Recent studies have indicated that the multiple programmed cell death pathways involved in the inflammatory response include pyroptosis, NETosis, necroptosis, and apoptosis, which initiate inflammatory reactions. In this review, we explore the correlation and interactions among these factors and their roles in the pathogenesis of gout to provide future research directions and possibilities for identifying potential novel therapeutic targets and enhancing our understanding of gout pathogenesis.
Collapse
Affiliation(s)
- Jianan Zhao
- Guanghua Clinical Medical College, Shanghai University of Traditional Chinese Medicine, Shanghai, China.,Department of Rheumatology, Shanghai Guanghua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China.,Institute of Arthritis Research in Integrative Medicine, Shanghai Academy of Traditional Chinese Medicine, Shanghai, China
| | - Kai Wei
- Guanghua Clinical Medical College, Shanghai University of Traditional Chinese Medicine, Shanghai, China.,Department of Rheumatology, Shanghai Guanghua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China.,Institute of Arthritis Research in Integrative Medicine, Shanghai Academy of Traditional Chinese Medicine, Shanghai, China
| | - Ping Jiang
- Guanghua Clinical Medical College, Shanghai University of Traditional Chinese Medicine, Shanghai, China.,Department of Rheumatology, Shanghai Guanghua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China.,Institute of Arthritis Research in Integrative Medicine, Shanghai Academy of Traditional Chinese Medicine, Shanghai, China
| | - Cen Chang
- Guanghua Clinical Medical College, Shanghai University of Traditional Chinese Medicine, Shanghai, China.,Department of Rheumatology, Shanghai Guanghua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China.,Institute of Arthritis Research in Integrative Medicine, Shanghai Academy of Traditional Chinese Medicine, Shanghai, China
| | - Lingxia Xu
- Guanghua Clinical Medical College, Shanghai University of Traditional Chinese Medicine, Shanghai, China.,Department of Rheumatology, Shanghai Guanghua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China.,Institute of Arthritis Research in Integrative Medicine, Shanghai Academy of Traditional Chinese Medicine, Shanghai, China
| | - Linshuai Xu
- Guanghua Clinical Medical College, Shanghai University of Traditional Chinese Medicine, Shanghai, China.,Department of Rheumatology, Shanghai Guanghua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China.,Institute of Arthritis Research in Integrative Medicine, Shanghai Academy of Traditional Chinese Medicine, Shanghai, China
| | - Yiming Shi
- Guanghua Clinical Medical College, Shanghai University of Traditional Chinese Medicine, Shanghai, China.,Department of Rheumatology, Shanghai Guanghua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China.,Institute of Arthritis Research in Integrative Medicine, Shanghai Academy of Traditional Chinese Medicine, Shanghai, China
| | - Shicheng Guo
- Computation and Informatics in Biology and Medicine, University of Wisconsin-Madison, Madison, WI, United States.,Department of Medical Genetics, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, WI, United States
| | - Yu Xue
- Department of Rheumatology, Huashan Hospital, Institute of Rheumatology, Immunology and Allergy, Fudan University, Shanghai, China
| | - Dongyi He
- Guanghua Clinical Medical College, Shanghai University of Traditional Chinese Medicine, Shanghai, China.,Department of Rheumatology, Shanghai Guanghua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China.,Institute of Arthritis Research in Integrative Medicine, Shanghai Academy of Traditional Chinese Medicine, Shanghai, China.,Arthritis Institute of Integrated Traditional and Western Medicine, Shanghai Chinese Medicine Research Institute, Shanghai, China
| |
Collapse
|
16
|
Liu C, Zhou M, Jiang W, Ye S, Tian S, Jiang C, Hao K, Li H, Hu Q. GPR105-Targeted Therapy Promotes Gout Resolution as a Switch Between NETosis and Apoptosis of Neutrophils. Front Immunol 2022; 13:870183. [PMID: 35432308 PMCID: PMC9005804 DOI: 10.3389/fimmu.2022.870183] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2022] [Accepted: 03/02/2022] [Indexed: 12/24/2022] Open
Abstract
The fate of infiltrating neutrophils in inflamed joints determines the development of acute gouty arthritis (AGA). GPR105 highly expressed in human neutrophils is sensitive to monosodium urate crystals (MSU); nevertheless, the roles of GPR105 in AGA remain unclear. Here, we show that GPR105 is significantly upregulated in peripheral polymorphonuclear neutrophils of AGA patients. GPR105 knockout (GPR105−/−) prevented NETosis and induced apoptosis of neutrophils under MSU exposure, as well as attenuating inflammatory cascades in AGA. Mechanistically, GPR105 deletion activated cAMP-PKA signals, thereby disrupting Raf-Mek1/2-Erk1/2 pathway-mediated NADPH oxidase activation, contributing to inhibition of NETosis. Whereas, cAMP-PKA activation resulting in GPR105 deficiency modulated PI3K-Akt pathway to regulate apoptosis. More importantly, suppression of cAMP-PKA pathway by SQ22536 and H-89 restored NETosis instead of apoptosis in GPR105−/− neutrophils, promoting MSU-induced gout flares. Interestingly, lobetyolin was screened out as a potent GPR105 antagonist using molecular docking-based virtual screening and in vitro activity test, which efficiently attenuated MSU-induced inflammatory response interacting with GPR105. Taken together, our study implicated that modulating cell death patterns between NETosis and apoptosis through targeting GPR105 could be a potential therapeutic strategy for the treatment of AGA.
Collapse
Affiliation(s)
- Chunxiao Liu
- State Key Laboratory of Natural Medicines, Key Laboratory of Drug Metabolism and Pharmacokinetics, China Pharmaceutical University, Nanjing, China.,School of Pharmacy, China Pharmaceutical University, Nanjing, China
| | - Mengze Zhou
- State Key Laboratory of Natural Medicines, Key Laboratory of Drug Metabolism and Pharmacokinetics, China Pharmaceutical University, Nanjing, China
| | - Wenjiao Jiang
- State Key Laboratory of Natural Medicines, Key Laboratory of Drug Metabolism and Pharmacokinetics, China Pharmaceutical University, Nanjing, China
| | - Shumin Ye
- State Key Laboratory of Natural Medicines, Key Laboratory of Drug Metabolism and Pharmacokinetics, China Pharmaceutical University, Nanjing, China
| | - Sheng Tian
- College of Pharmaceutical Sciences, Soochow University, Suzhou, China
| | - Cheng Jiang
- School of Pharmacy, China Pharmaceutical University, Nanjing, China
| | - Kun Hao
- State Key Laboratory of Natural Medicines, Key Laboratory of Drug Metabolism and Pharmacokinetics, China Pharmaceutical University, Nanjing, China
| | - Huanqiu Li
- College of Pharmaceutical Sciences, Soochow University, Suzhou, China
| | - Qinghua Hu
- State Key Laboratory of Natural Medicines, Key Laboratory of Drug Metabolism and Pharmacokinetics, China Pharmaceutical University, Nanjing, China.,School of Pharmacy, China Pharmaceutical University, Nanjing, China
| |
Collapse
|
17
|
Costa VV, Sugimoto MA, Hubner J, Bonilha CS, Queiroz-Junior CM, Gonçalves-Pereira MH, Chen J, Gobbetti T, Libanio Rodrigues GO, Bambirra JL, Passos IB, Machado Lopes CE, Moreira TP, Bonjour K, Melo RCN, Oliveira MAP, Andrade MVM, Sousa LP, Souza DG, Santiago HDC, Perretti M, Teixeira MM. Targeting the Annexin A1-FPR2/ALX pathway for host-directed therapy in dengue disease. eLife 2022; 11:73853. [PMID: 35293862 PMCID: PMC8959599 DOI: 10.7554/elife.73853] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2021] [Accepted: 03/15/2022] [Indexed: 11/13/2022] Open
Abstract
Host immune responses contribute to dengue's pathogenesis and severity, yet the possibility that failure in endogenous inflammation resolution pathways could characterise the disease has not been contemplated. The pro-resolving protein Annexin A1 (AnxA1) is known to counterbalance overexuberant inflammation and mast cell (MC) activation. We hypothesised that inadequate AnxA1 engagement underlies the cytokine storm and vascular pathologies associated with dengue disease. Levels of AnxA1 were examined in the plasma of dengue patients and infected mice. Immunocompetent, interferon (alpha and beta) receptor one knockout (KO), AnxA1 KO, and formyl peptide receptor 2 (FPR2) KO mice were infected with dengue virus (DENV) and treated with the AnxA1 mimetic peptide Ac2-26 for analysis. In addition, the effect of Ac2-26 on DENV-induced MC degranulation was assessed in vitro and in vivo. We observed that circulating levels of AnxA1 were reduced in dengue patients and DENV-infected mice. Whilst the absence of AnxA1 or its receptor FPR2 aggravated illness in infected mice, treatment with AnxA1 agonistic peptide attenuated disease manifestationsatteanuated the symptoms of the disease. Both clinical outcomes were attributed to modulation of DENV-mediated viral load-independent MC degranulation. We have thereby identified that altered levels of the pro-resolving mediator AnxA1 are of pathological relevance in DENV infection, suggesting FPR2/ALX agonists as a therapeutic target for dengue disease.
Collapse
Affiliation(s)
- Vivian Vasconcelos Costa
- Department of Morphology, Institute of Biological Sciences, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Michelle A Sugimoto
- Department of Biochemistry and Immunology, Institute of Biological Sciences, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil.,School of Medicine, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil.,William Harvey Research Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London, United Kingdom
| | - Josy Hubner
- Department of Morphology, Institute of Biological Sciences, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Caio S Bonilha
- Department of Biochemistry and Immunology, Institute of Biological Sciences, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil.,Institute of Infection, Immunity and Inflammation, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow, United Kingdom
| | - Celso Martins Queiroz-Junior
- Department of Morphology, Institute of Biological Sciences, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Marcela Helena Gonçalves-Pereira
- Department of Biochemistry and Immunology, Institute of Biological Sciences, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Jianmin Chen
- William Harvey Research Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London, United Kingdom
| | - Thomas Gobbetti
- William Harvey Research Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London, United Kingdom
| | - Gisele Olinto Libanio Rodrigues
- Department of Biochemistry and Immunology, Institute of Biological Sciences, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Jordana L Bambirra
- Department of Microbiology, Institute of Biological Sciences, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Ingredy B Passos
- Department of Morphology, Institute of Biological Sciences, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Carla Elizabeth Machado Lopes
- Department of Morphology, Institute of Biological Sciences, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Thaiane P Moreira
- Department of Microbiology, Institute of Biological Sciences, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Kennedy Bonjour
- Department of Biology, Institute of Biological Sciences, Federal University of Juiz de Fora, Juiz de Fora, Brazil
| | - Rossana C N Melo
- Department of Biology, Institute of Biological Sciences, Federal University of Juiz de Fora, Juiz de Fora, Brazil
| | - Milton A P Oliveira
- Tropical Pathology and Public Health Institute, Universidade Federal de Goiás, Goiânia, Brazil
| | | | - Lirlândia Pires Sousa
- Department of Clinical and Toxicological Analyses, School of Pharmacy, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Danielle Gloria Souza
- Department of Microbiology, Institute of Biological Sciences, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Helton da Costa Santiago
- Department of Biochemistry and Immunology, Institute of Biological Sciences, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Mauro Perretti
- William Harvey Research Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London, United Kingdom.,Centre for Inflammation and Therapeutic Innovation, Queen Mary University of London, London, United Kingdom
| | - Mauro Martins Teixeira
- Department of Biochemistry and Immunology, Institute of Biological Sciences, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| |
Collapse
|
18
|
Chavez-Dominguez R, Perez-Medina M, Aguilar-Cazares D, Galicia-Velasco M, Meneses-Flores M, Islas-Vazquez L, Camarena A, Lopez-Gonzalez JS. Old and New Players of Inflammation and Their Relationship With Cancer Development. Front Oncol 2021; 11:722999. [PMID: 34881173 PMCID: PMC8645998 DOI: 10.3389/fonc.2021.722999] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2021] [Accepted: 11/04/2021] [Indexed: 12/18/2022] Open
Abstract
Pathogens or genotoxic agents continuously affect the human body. Acute inflammatory reaction induced by a non-sterile or sterile environment is triggered for the efficient elimination of insults that caused the damage. According to the insult, pathogen-associated molecular patterns, damage-associated molecular patterns, and homeostasis-altering molecular processes are released to facilitate the arrival of tissue resident and circulating cells to the injured zone to promote harmful agent elimination and tissue regeneration. However, when inflammation is maintained, a chronic phenomenon is induced, in which phagocytic cells release toxic molecules damaging the harmful agent and the surrounding healthy tissues, thereby inducing DNA lesions. In this regard, chronic inflammation has been recognized as a risk factor of cancer development by increasing the genomic instability of transformed cells and by creating an environment containing proliferation signals. Based on the cancer immunoediting concept, a rigorous and regulated inflammation process triggers participation of innate and adaptive immune responses for efficient elimination of transformed cells. When immune response does not eliminate all transformed cells, an equilibrium phase is induced. Therefore, excessive inflammation amplifies local damage caused by the continuous arrival of inflammatory/immune cells. To regulate the overstimulation of inflammatory/immune cells, a network of mechanisms that inhibit or block the cell overactivity must be activated. Transformed cells may take advantage of this process to proliferate and gradually grow until they become preponderant over the immune cells, preserving, increasing, or creating a microenvironment to evade the host immune response. In this microenvironment, tumor cells resist the attack of the effector immune cells or instruct them to sustain tumor growth and development until its clinical consequences. With tumor development, evolving, complex, and overlapping microenvironments are arising. Therefore, a deeper knowledge of cytokine, immune, and tumor cell interactions and their role in the intricated process will impact the combination of current or forthcoming therapies.
Collapse
Affiliation(s)
- Rodolfo Chavez-Dominguez
- Departamento de Enfermedades Cronico-Degenerativas, Instituto Nacional de Enfermedades Respiratorias "Ismael Cosio Villegas", Mexico City, Mexico.,Posgrado en Ciencias Biologicas, Universidad Nacional Autonoma de Mexico, Mexico City, Mexico
| | - Mario Perez-Medina
- Departamento de Enfermedades Cronico-Degenerativas, Instituto Nacional de Enfermedades Respiratorias "Ismael Cosio Villegas", Mexico City, Mexico.,Laboratorio de Quimioterapia Experimental, Departamento de Bioquímica, Escuela Nacional de Ciencias Biológicas, Instituto Politécnico Nacional, Mexico City, Mexico
| | - Dolores Aguilar-Cazares
- Departamento de Enfermedades Cronico-Degenerativas, Instituto Nacional de Enfermedades Respiratorias "Ismael Cosio Villegas", Mexico City, Mexico
| | - Miriam Galicia-Velasco
- Departamento de Enfermedades Cronico-Degenerativas, Instituto Nacional de Enfermedades Respiratorias "Ismael Cosio Villegas", Mexico City, Mexico
| | - Manuel Meneses-Flores
- Departamento de Patología, Instituto Nacional de Enfermedades Respiratorias "Ismael Cosio Villegas", Mexico City, Mexico
| | - Lorenzo Islas-Vazquez
- Departamento de Enfermedades Cronico-Degenerativas, Instituto Nacional de Enfermedades Respiratorias "Ismael Cosio Villegas", Mexico City, Mexico
| | - Angel Camarena
- Laboratorio de Human Leukocyte Antigen (HLA), Instituto Nacional de Enfermedades Respiratorias "Ismael Cosio Villegas", Mexico City, Mexico
| | - Jose S Lopez-Gonzalez
- Departamento de Enfermedades Cronico-Degenerativas, Instituto Nacional de Enfermedades Respiratorias "Ismael Cosio Villegas", Mexico City, Mexico
| |
Collapse
|
19
|
Valiate BVS, Queiroz-Junior CM, Levi-Schaffer F, Galvão I, Teixeira MM. CD300a contributes to the resolution of articular inflammation triggered by MSU crystals by controlling neutrophil apoptosis. Immunology 2021; 164:305-317. [PMID: 34002852 DOI: 10.1111/imm.13371] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2021] [Revised: 04/26/2021] [Accepted: 04/29/2021] [Indexed: 12/27/2022] Open
Abstract
Gout is an inflammatory disease triggered by deposition of monosodium urate (MSU) crystals in the joints, resulting in high neutrophil influx and pain. Here, we studied the role of the inhibitory receptor CD300a in the resolution process in a murine model of gout. We found increased CD300a expression on neutrophils emigrated to the joint. When compared to WT mice, CD300a-/- mice had persistent neutrophil influx till 24 hr after MSU injection. This was associated with increased concentration of IL-1β and greater tissue damage in the joints of CD300a-/- mice. There was an increase in the percentage of apoptotic neutrophils in the synovial lavage of WT mice, as compared to CD300a-/- mice. This difference was reflected in the decline of efferocytic events in the synovial cavity of CD300a-/- mice 24 hr after MSU injection. A CD300a agonistic antibody was shown, for the first time, to increase apoptosis of human neutrophils, and this was associated with cleavage of caspase-8. In conclusion, our results reveal an important role of CD300a in the control of leucocyte infiltration, IL-1β production and caspase-8 cleavage in neutrophils, contributing to the resolution of inflammation triggered by MSU injection.
Collapse
Affiliation(s)
- Bruno V S Valiate
- Departamento de Bioquímica e Imunologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Celso M Queiroz-Junior
- Departamento de Morfologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Francesca Levi-Schaffer
- Pharmacology and Experimental Therapeutics Unit, Institute for Drug Research, School of Pharmacy, Faculty of Medicine, The Hebrew University of Jerusalem, Jerusalem, Israel
| | - Izabela Galvão
- Departamento de Morfologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Mauro M Teixeira
- Departamento de Bioquímica e Imunologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| |
Collapse
|
20
|
Autoinflammatory Features in Gouty Arthritis. J Clin Med 2021; 10:jcm10091880. [PMID: 33926105 PMCID: PMC8123608 DOI: 10.3390/jcm10091880] [Citation(s) in RCA: 47] [Impact Index Per Article: 15.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2021] [Revised: 04/16/2021] [Accepted: 04/22/2021] [Indexed: 12/16/2022] Open
Abstract
In the panorama of inflammatory arthritis, gout is the most common and studied disease. It is known that hyperuricemia and monosodium urate (MSU) crystal-induced inflammation provoke crystal deposits in joints. However, since hyperuricemia alone is not sufficient to develop gout, molecular-genetic contributions are necessary to better clinically frame the disease. Herein, we review the autoinflammatory features of gout, from clinical challenges and differential diagnosis, to the autoinflammatory mechanisms, providing also emerging therapeutic options available for targeting the main inflammatory pathways involved in gout pathogenesis. This has important implication as treating the autoinflammatory aspects and not only the dysmetabolic side of gout may provide an effective and safer alternative for patients even in the prevention of possible gouty attacks.
Collapse
|
21
|
Grewal T, Rentero C, Enrich C, Wahba M, Raabe CA, Rescher U. Annexin Animal Models-From Fundamental Principles to Translational Research. Int J Mol Sci 2021; 22:ijms22073439. [PMID: 33810523 PMCID: PMC8037771 DOI: 10.3390/ijms22073439] [Citation(s) in RCA: 27] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2021] [Revised: 03/18/2021] [Accepted: 03/24/2021] [Indexed: 02/07/2023] Open
Abstract
Routine manipulation of the mouse genome has become a landmark in biomedical research. Traits that are only associated with advanced developmental stages can now be investigated within a living organism, and the in vivo analysis of corresponding phenotypes and functions advances the translation into the clinical setting. The annexins, a family of closely related calcium (Ca2+)- and lipid-binding proteins, are found at various intra- and extracellular locations, and interact with a broad range of membrane lipids and proteins. Their impacts on cellular functions has been extensively assessed in vitro, yet annexin-deficient mouse models generally develop normally and do not display obvious phenotypes. Only in recent years, studies examining genetically modified annexin mouse models which were exposed to stress conditions mimicking human disease often revealed striking phenotypes. This review is the first comprehensive overview of annexin-related research using animal models and their exciting future use for relevant issues in biology and experimental medicine.
Collapse
Affiliation(s)
- Thomas Grewal
- School of Pharmacy, Faculty of Medicine and Health, University of Sydney, Sydney, NSW 2006, Australia;
- Correspondence: (T.G.); (U.R.); Tel.: +61-(0)2-9351-8496 (T.G.); +49-(0)251-83-52121 (U.R.)
| | - Carles Rentero
- Departament de Biomedicina, Unitat de Biologia Cel·lular, Facultat de Medicina i Ciències de la Salut, Universitat de Barcelona, 08036 Barcelona, Spain; (C.R.); (C.E.)
- Centre de Recerca Biomèdica CELLEX, Institut d’Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), 08036 Barcelona, Spain
| | - Carlos Enrich
- Departament de Biomedicina, Unitat de Biologia Cel·lular, Facultat de Medicina i Ciències de la Salut, Universitat de Barcelona, 08036 Barcelona, Spain; (C.R.); (C.E.)
- Centre de Recerca Biomèdica CELLEX, Institut d’Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), 08036 Barcelona, Spain
| | - Mohamed Wahba
- School of Pharmacy, Faculty of Medicine and Health, University of Sydney, Sydney, NSW 2006, Australia;
| | - Carsten A. Raabe
- Research Group Regulatory Mechanisms of Inflammation, Center for Molecular Biology of Inflammation (ZMBE) and Cells in Motion Interfaculty Center (CiM), Institute of Medical Biochemistry, University of Muenster, 48149 Muenster, Germany;
| | - Ursula Rescher
- Research Group Regulatory Mechanisms of Inflammation, Center for Molecular Biology of Inflammation (ZMBE) and Cells in Motion Interfaculty Center (CiM), Institute of Medical Biochemistry, University of Muenster, 48149 Muenster, Germany;
- Correspondence: (T.G.); (U.R.); Tel.: +61-(0)2-9351-8496 (T.G.); +49-(0)251-83-52121 (U.R.)
| |
Collapse
|
22
|
Camba-Gómez M, Gualillo O, Conde-Aranda J. New Perspectives in the Study of Intestinal Inflammation: Focus on the Resolution of Inflammation. Int J Mol Sci 2021; 22:ijms22052605. [PMID: 33807591 PMCID: PMC7962019 DOI: 10.3390/ijms22052605] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2021] [Revised: 02/25/2021] [Accepted: 03/02/2021] [Indexed: 12/14/2022] Open
Abstract
Inflammation is an essential physiological process that is directed to the protection of the organism against invading pathogens or tissue trauma. Most of the existing knowledge related to inflammation is focused on the factors and mechanisms that drive the induction phase of this process. However, since the recognition that the resolution of the inflammation is an active and tightly regulated process, increasing evidence has shown the relevance of this process for the development of chronic inflammatory diseases, such as inflammatory bowel disease. For that reason, with this review, we aimed to summarize the most recent and interesting information related to the resolution process in the context of intestinal inflammation. We discussed the advances in the understanding of the pro-resolution at intestine level, as well as the new mediators with pro-resolutive actions that could be interesting from a therapeutic point of view.
Collapse
Affiliation(s)
- Miguel Camba-Gómez
- Molecular and Cellular Gastroenterology, Health Research Institute of Santiago de Compostela (IDIS), 15706 Santiago de Compostela, Spain;
| | - Oreste Gualillo
- SERGAS (Servizo Galego de Saúde) and IDIS (Instituto de Investigación Sanitaria de Santiago), The NEIRID Lab (Neuroendocrine Interactions in Rheumatology and Inflammatory Diseases), Research Laboratory 9, Santiago University Clinical Hospital, 15706 Santiago de Compostela, Spain;
| | - Javier Conde-Aranda
- Molecular and Cellular Gastroenterology, Health Research Institute of Santiago de Compostela (IDIS), 15706 Santiago de Compostela, Spain;
- Correspondence: ; Tel.: +34-981-955-091
| |
Collapse
|
23
|
Galvão I, Melo EM, de Oliveira VLS, Vago JP, Queiroz-Junior C, de Gaetano M, Brennan E, Gahan K, Guiry PJ, Godson C, Teixeira MM. Therapeutic potential of the FPR2/ALX agonist AT-01-KG in the resolution of articular inflammation. Pharmacol Res 2021; 165:105445. [PMID: 33493655 DOI: 10.1016/j.phrs.2021.105445] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/25/2020] [Revised: 01/12/2021] [Accepted: 01/14/2021] [Indexed: 12/16/2022]
Abstract
The resolution of inflammation is a dynamic process, characterized by the biosynthesis of pro-resolving mediators, including the lipid Lipoxin A4 (LXA4). LXA4 acts on the N-formyl peptide receptor 2 (FPR2/ALX) to mediate anti-inflammatory and pro-resolving effects. In order to exploit the therapeutic potential of endogenous LXA4 in the context of inflammation we have recently developed synthetic LXA4 mimetics (sLXms) including a dimethyl-imidazole-containing FPR2/ALX agonist designated AT-01-KG. Here, we have investigated the effect of treatment with AT-01-KG in established models of articular inflammation. In a model of gout, mice were injected with MSU crystals and treated with AT-01-KG at the peak of inflammatory response. The treatment decreased the number of neutrophils in the knee exudate, an effect which was accompanied by low levels of myeloperoxidase, CXCL1 and IL-1β in periarticular tissue. AT-01-KG treatment led to reduced tissue damage and hypernociception. The effects of AT-01-KG on neutrophil accumulation were not observed in MSU treated FPR2/3-/-mice. Importantly, AT-01-KG induced resolution of articular inflammation by increasing neutrophil apoptosis and subsequent efficient efferocytosis. In a model of antigen-induced arthritis, AT-01-KG treatment also attenuated inflammatory responses. These data suggest that AT-01-KG may be a potential new therapy for neutrophilic inflammation of the joints.
Collapse
Affiliation(s)
- Izabela Galvão
- Departamento de Morfologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Eliza M Melo
- Departamento de Bioquímica e Imunologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Vivian L S de Oliveira
- Departamento de Bioquímica e Imunologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Juliana P Vago
- Departamento de Morfologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Celso Queiroz-Junior
- Departamento de Morfologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Monica de Gaetano
- UCD Diabetes Complications Research Centre, UCD Conway Institute School of Medicine, University College Dublin, Dublin, Ireland
| | - Eoin Brennan
- UCD Diabetes Complications Research Centre, UCD Conway Institute School of Medicine, University College Dublin, Dublin, Ireland
| | - Kevin Gahan
- Centre for Synthesis and Chemical Biology, School of Chemistry, University College Dublin, Dublin, Ireland
| | - Patrick J Guiry
- Centre for Synthesis and Chemical Biology, School of Chemistry, University College Dublin, Dublin, Ireland
| | - Catherine Godson
- UCD Diabetes Complications Research Centre, UCD Conway Institute School of Medicine, University College Dublin, Dublin, Ireland
| | - Mauro M Teixeira
- Departamento de Bioquímica e Imunologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil.
| |
Collapse
|
24
|
Sanches JM, Correia-Silva RD, Duarte GHB, Fernandes AMAP, Sánchez-Vinces S, Carvalho PO, Oliani SM, Bortoluci KR, Moreira V, Gil CD. Role of Annexin A1 in NLRP3 Inflammasome Activation in Murine Neutrophils. Cells 2021; 10:121. [PMID: 33440601 PMCID: PMC7827236 DOI: 10.3390/cells10010121] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2020] [Revised: 01/05/2021] [Accepted: 01/06/2021] [Indexed: 02/07/2023] Open
Abstract
This study evaluated the role of endogenous and exogenous annexin A1 (AnxA1) in the activation of the NLRP3 inflammasome in isolated peritoneal neutrophils. C57BL/6 wild-type (WT) and AnxA1 knockout mice (AnxA1-/-) received 0.3% carrageenan intraperitoneally and, after 3 h, the peritoneal exudate was collected. WT and AnxA1-/- neutrophils were then stimulated with lipopolysaccharide, followed by the NLRP3 agonists nigericin or ATP. To determine the exogenous effect of AnxA1, the neutrophils were pretreated with the AnxA1-derived peptide Ac2-26 followed by the NLRP3 agonists. Ac2-26 administration reduced NLRP3-derived IL-1β production by WT neutrophils after nigericin and ATP stimulation. However, IL-1β release was impaired in AnxA1-/- neutrophils stimulated by both agonists, and there was no further impairment in IL-1β release with Ac2-26 treatment before stimulation. Despite this, ATP- and nigericin-stimulated AnxA1-/- neutrophils had increased levels of cleaved caspase-1. The lipidomics of supernatants from nigericin-stimulated WT and AnxA1-/- neutrophils showed potential lipid biomarkers of cell stress and activation, including specific sphingolipids and glycerophospholipids. AnxA1 peptidomimetic treatment also increased the concentration of phosphatidylserines and oxidized phosphocholines, which are lipid biomarkers related to the inflammatory resolution pathway. Together, our results indicate that exogenous AnxA1 negatively regulates NLRP3-derived IL-1β production by neutrophils, while endogenous AnxA1 is required for the activation of the NLRP3 machinery.
Collapse
Affiliation(s)
- José Marcos Sanches
- Programa de Pós-Graduação em Biologia Estrutural e Funcional, Universidade Federal de São Paulo, São Paulo 04023-900, Brazil; (J.M.S.); (R.D.C.-S.); (S.M.O.)
| | - Rebeca D. Correia-Silva
- Programa de Pós-Graduação em Biologia Estrutural e Funcional, Universidade Federal de São Paulo, São Paulo 04023-900, Brazil; (J.M.S.); (R.D.C.-S.); (S.M.O.)
| | - Gustavo H. B. Duarte
- Instituto de Química, Universidade Estadual de Campinas, Campinas 13083-862, São Paulo, Brazil;
| | - Anna Maria A. P. Fernandes
- Laboratório de Pesquisa Multidisciplinar, Universidade São Francisco, Bragança Paulista 12916-900, São Paulo, Brazil; (A.M.A.P.F.); (S.S.-V.); (P.O.C.)
| | - Salvador Sánchez-Vinces
- Laboratório de Pesquisa Multidisciplinar, Universidade São Francisco, Bragança Paulista 12916-900, São Paulo, Brazil; (A.M.A.P.F.); (S.S.-V.); (P.O.C.)
| | - Patrícia O. Carvalho
- Laboratório de Pesquisa Multidisciplinar, Universidade São Francisco, Bragança Paulista 12916-900, São Paulo, Brazil; (A.M.A.P.F.); (S.S.-V.); (P.O.C.)
| | - Sonia M. Oliani
- Programa de Pós-Graduação em Biologia Estrutural e Funcional, Universidade Federal de São Paulo, São Paulo 04023-900, Brazil; (J.M.S.); (R.D.C.-S.); (S.M.O.)
- Programa de Pós-Graduação em Biociências, Universidade Estadual Paulista (UNESP), Instituto de Biociências Letras e Ciências Exatas, São José do Rio Preto 15054-000, São Paulo, Brazil
| | - Karina R. Bortoluci
- Departamento de Ciências Biológicas e Centro de Terapia Celular e Molecular, Universidade Federal de São Paulo, São Paulo 04044-010, Brazil;
| | - Vanessa Moreira
- Departamento de Farmacologia, Universidade Federal de São Paulo, São Paulo 04044-020, Brazil;
| | - Cristiane D. Gil
- Programa de Pós-Graduação em Biologia Estrutural e Funcional, Universidade Federal de São Paulo, São Paulo 04023-900, Brazil; (J.M.S.); (R.D.C.-S.); (S.M.O.)
- Programa de Pós-Graduação em Biociências, Universidade Estadual Paulista (UNESP), Instituto de Biociências Letras e Ciências Exatas, São José do Rio Preto 15054-000, São Paulo, Brazil
| |
Collapse
|
25
|
Melo EM, Del Sarto J, Vago JP, Tavares LP, Rago F, Gonçalves APF, Machado MG, Aranda-Pardos I, Valiate BVS, Cassali GD, Pinho V, Sousa LP, A-Gonzalez N, Campagnole-Santos MJ, Bader M, Santos RAS, Machado AV, Ludwig S, Teixeira MM. Relevance of angiotensin-(1-7) and its receptor Mas in pneumonia caused by influenza virus and post-influenza pneumococcal infection. Pharmacol Res 2021; 163:105292. [PMID: 33171305 DOI: 10.1016/j.phrs.2020.105292] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/03/2020] [Revised: 10/22/2020] [Accepted: 10/22/2020] [Indexed: 12/12/2022]
Abstract
Resolution failure of exacerbated inflammation triggered by Influenza A virus (IAV) prevents return of pulmonary homeostasis and survival, especially when associated with secondary pneumococcal infection. Therapeutic strategies based on pro-resolving molecules have great potential against acute inflammatory diseases. Angiotensin-(1-7) [Ang-(1-7)] is a pro-resolving mediator that acts on its Mas receptor (MasR) to promote resolution of inflammation. We investigated the effects of Ang-(1-7) and the role of MasR in the context of primary IAV infection and secondary pneumococcal infection and evaluated pulmonary inflammation, virus titers and bacteria counts, and pulmonary damage. Therapeutic treatment with Ang-(1-7) decreased neutrophil recruitment, lung injury, viral load and morbidity after a primary IAV infection. Ang-(1-7) induced apoptosis of neutrophils and efferocytosis of these cells by alveolar macrophages, but had no direct effect on IAV replication in vitro. MasR-deficient (MasR-/-) mice were highly susceptible to IAV infection, displaying uncontrolled inflammation, increased viral load and greater lethality rate, as compared to WT animals. Ang-(1-7) was not protective in MasR-/- mice. Interestingly, Ang-(1-7) given during a sublethal dose of IAV infection greatly reduced morbidity associated with a subsequent S. pneumoniae infection, as seen by decrease in the magnitude of neutrophil influx, number of bacteria in the blood leading to a lower lethality. Altogether, these results show that Ang-(1-7) is highly protective against severe primary IAV infection and protects against secondary bacterial infection of the lung. These effects are MasR-dependent. Mediators of resolution of inflammation, such as Ang-(1-7), should be considered for the treatment of pulmonary viral infections.
Collapse
Affiliation(s)
- Eliza M Melo
- Laboratório de Imunofarmacologia, Departamento de Bioquímica e Imunologia, ICB, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| | - Juliana Del Sarto
- Laboratório de Imunofarmacologia, Departamento de Bioquímica e Imunologia, ICB, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brazil; Institute of Virology Muenster (IVM), Westfaelische Wilhelms-University Muenster, Von-Esmarch-Str. 56, D-48149 Muenster, Germany
| | - Juliana P Vago
- Laboratório de Imunofarmacologia, Departamento de Bioquímica e Imunologia, ICB, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| | - Luciana P Tavares
- Laboratório de Imunofarmacologia, Departamento de Bioquímica e Imunologia, ICB, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brazil; Pulmonary and Critical Care Medicine Division, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA 02115, USA
| | - Flávia Rago
- Laboratório de Imunofarmacologia, Departamento de Bioquímica e Imunologia, ICB, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| | - Ana Paula F Gonçalves
- Laboratório de Imunofarmacologia, Departamento de Bioquímica e Imunologia, ICB, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brazil; Imunologia de Doenças Virais, Centro de Pesquisa René Rachou, Fundação Oswaldo Cruz (FIOCRUZ-Minas), Belo Horizonte, Minas Gerais, Brazil
| | - Marina G Machado
- Laboratório de Imunofarmacologia, Departamento de Bioquímica e Imunologia, ICB, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brazil; Centre d'Infection et d'Immunité de Lille, INSERM U1019, CNRS UMR 8204, University of Lille, CHU Lille, Institut Pasteur de Lille, Lille, France
| | - Irene Aranda-Pardos
- Institute of Immunology, Westfaelische Wilhelms-University muenster, Röntgenstraße 21, D-48149 Muenster, Germany
| | - Bruno V S Valiate
- Laboratório de Imunofarmacologia, Departamento de Bioquímica e Imunologia, ICB, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| | - Geovanni D Cassali
- Laboratório de Patologia Comparada, Departamento de Patologia, ICB, Universidade Federal de Minas gerais, Belo Horizonte, Minas Gerais, Brazil
| | - Vanessa Pinho
- Laboratório de Imunofarmacologia, Departamento de Bioquímica e Imunologia, ICB, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| | - Lirlândia P Sousa
- Laboratório de sinalização da inflamação, Departamento de Análises Clínicase Toxicológicas, Faculdade de Farmácia, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Noelia A-Gonzalez
- Institute of Immunology, Westfaelische Wilhelms-University muenster, Röntgenstraße 21, D-48149 Muenster, Germany
| | - Maria José Campagnole-Santos
- Instituto Nacional de Ciência e Tecnologia em Nanobiofarmacêutica, Departamento de Fisiologia e Biofísica, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Michael Bader
- Max-Delbrück-Center for Molecular Medicine (MDC), Berlin, Germany
| | - Robson A S Santos
- Instituto Nacional de Ciência e Tecnologia em Nanobiofarmacêutica, Departamento de Fisiologia e Biofísica, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Alexandre V Machado
- Imunologia de Doenças Virais, Centro de Pesquisa René Rachou, Fundação Oswaldo Cruz (FIOCRUZ-Minas), Belo Horizonte, Minas Gerais, Brazil
| | - Stephan Ludwig
- Institute of Virology Muenster (IVM), Westfaelische Wilhelms-University Muenster, Von-Esmarch-Str. 56, D-48149 Muenster, Germany
| | - Mauro M Teixeira
- Laboratório de Imunofarmacologia, Departamento de Bioquímica e Imunologia, ICB, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brazil.
| |
Collapse
|
26
|
Sanches JM, Rossato L, Lice I, Alves de Piloto Fernandes AM, Bueno Duarte GH, Rosini Silva AA, de Melo Porcari A, de Oliveira Carvalho P, Gil CD. The role of annexin A1 in Candida albicans and Candida auris infections in murine neutrophils. Microb Pathog 2020; 150:104689. [PMID: 33307121 DOI: 10.1016/j.micpath.2020.104689] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2020] [Revised: 11/29/2020] [Accepted: 11/30/2020] [Indexed: 12/18/2022]
Abstract
Annexin A1 (AnxA1) is an anti-inflammatory protein expressed in various cell types, especially macrophages and neutrophils. Because neutrophils play important roles in infections and inflammatory processes and the relationship between AnxA1 and Candida spp. infections is not well-understood, our study examined whether AnxA1 can serve as a target protein for the regulation of the immune response during fungal infections. C57BL/6 wild-type (WT) and AnxA1 knockout (AnxA1-/-) peritoneal neutrophils were coinfected with Candida albicans or Candida auris for 4 h. AnxA1-/- neutrophils exhibited a marked increase in cyclooxygenase 2 (COX-2), phosphorylated extracellular signal-related kinase (ERK), p-38, and c-Jun N-terminal kinase (JNK) levels after coinfection with both Candida spp. A lipidomics approach showed that AnxA1 deficiency produced marked differences in the supernatant lipid profiles of both control neutrophils and neutrophils coinfected with Candida spp. compared with WT cells, especially the levels of glycerophospholipids and glycerolipids. Our results showed that endogenous AnxA1 regulates the neutrophil response under fungal infection conditions, altering lipid membrane organization and metabolism.
Collapse
Affiliation(s)
- José Marcos Sanches
- Departamento de Morfologia e Genética, Universidade Federal de São Paulo - UNIFESP, São Paulo, 04023-900, Brazil
| | - Luana Rossato
- Laboratório Especial de Micologia, Departamento de Medicina, UNIFESP, São Paulo, 04038-032, Brazil
| | - Izabella Lice
- Departamento de Morfologia e Genética, Universidade Federal de São Paulo - UNIFESP, São Paulo, 04023-900, Brazil
| | | | | | - Alex Aparecido Rosini Silva
- Laboratório de Pesquisa Multidisciplinar, Universidade São Francisco, Bragança Paulista, 12916-900, São Paulo, Brazil
| | - Andreia de Melo Porcari
- Laboratório de Pesquisa Multidisciplinar, Universidade São Francisco, Bragança Paulista, 12916-900, São Paulo, Brazil
| | - Patrícia de Oliveira Carvalho
- Laboratório de Pesquisa Multidisciplinar, Universidade São Francisco, Bragança Paulista, 12916-900, São Paulo, Brazil
| | - Cristiane Damas Gil
- Departamento de Morfologia e Genética, Universidade Federal de São Paulo - UNIFESP, São Paulo, 04023-900, Brazil.
| |
Collapse
|
27
|
Bonavita AG. Ac2-26 mimetic peptide of annexin A1 to treat severe COVID-19: A hypothesis. Med Hypotheses 2020; 145:110352. [PMID: 33129009 PMCID: PMC7577270 DOI: 10.1016/j.mehy.2020.110352] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2020] [Accepted: 10/17/2020] [Indexed: 02/07/2023]
Abstract
The Coronavirus Diseases-2019 (COVID-19) pandemic leads many researchers around the world to study the SARS-CoV-s2 infection and pathology to find a treatment for it. This generates a massive production of papers including pre-clinical, clinical and revisions but till now no specific treatment were identified. Meanwhile, like other coronavirus infections, COVID-19 leads to the cytokine storm syndrome resulting in hyperinflammation, exacerbated immune response and multiple organ dysfunctions indicating that drugs that modulate this response, as glucocorticoids could be a treatment option. However glucocorticoids have several side effects or usage limitations. In this sense a drug with anti-inflammatory effects and capable to reduce inflammation but with less after-effects could be a powerful tool to combat COVID-19. Thus the Ac2-26 Mimetic Peptide of Annexin A1 emerges as a possible therapy. The peptide has many anti-inflammatory effects described including the reduction of interleukin (IL)-6, one of the main mediators of cytokine storm syndrome. Therefore the hypothesis to use the Ac2-26 peptide to treat severe COVID-19 will be highlighted in this paper.
Collapse
Affiliation(s)
- Andre Gustavo Bonavita
- Grupo de Pesquisa em Farmacologia de Produtos Bioativos, Campus UFRJ-Macaé Professor Aloizio Teixeira Macaé, Universidade Federal do Rio de Janeiro, Rua Aloísio da Silva Gomes, 50, Macaé, RJ, Brazil.
| |
Collapse
|
28
|
Liu N, Meng B, Zeng L, Yin S, Hu Y, Li S, Fu Y, Zhang X, Xie C, Shu L, Yang M, Wang Y, Yang X. Discovery of a novel rice-derived peptide with significant anti-gout potency. Food Funct 2020; 11:10542-10553. [PMID: 33185232 DOI: 10.1039/d0fo01774d] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023]
Abstract
As a metabolic disease, gout, which seriously affects the normal life of patients, has become increasingly common in modern society. However, the existing medicines cannot completely meet the clinical needs. In the current study, a novel short peptide (named rice-derived-peptide-2 (RDP2), AAAAGAMPK-NH2, 785.97 Da) was isolated and identified from water extract of shelled Oryza sativa fruits, without toxic side effects but excellent stability. Our results indicated that RDP2 (the minimum effective concentration is 5 μg kg-1) induced a significant reduction in serum uric acid levels in hyperuricemic mice via suppressing xanthine oxidase activity and urate transporter 1 expression, as well as alleviated renal damage through inhibiting the activation of NLRP3 inflammasome. In addition, RDP2 can also alleviate paw swelling and inflammatory reactions in mice after subcutaneous injection of monosodium urate crystals. As mentioned above, we obtained a novel peptide which could work through all stages of gout, not only reducing uric acid levels and renal damage in hyperuricemic mice, but also alleviating inflammatory responses associated with acute gout attack, and thus provided a new peptide molecular template for the development of anti-gout drugs.
Collapse
Affiliation(s)
- Naixin Liu
- Department of Anatomy and Histology & Embryology, Faculty of Basic Medical Science, Kunming Medical University, Kunming, Yunnan 650500, China.
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
29
|
Rüger M, Kipp E, Schubert N, Schröder N, Pufe T, Stope MB, Kipp M, Blume C, Tauber SC, Brandenburg LO. The formyl peptide receptor agonist Ac2-26 alleviates neuroinflammation in a mouse model of pneumococcal meningitis. J Neuroinflammation 2020; 17:325. [PMID: 33121515 PMCID: PMC7596991 DOI: 10.1186/s12974-020-02006-w] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2020] [Accepted: 10/21/2020] [Indexed: 01/20/2023] Open
Abstract
Background Bacterial meningitis is still a cause of severe neurological disability. The brain is protected from penetrating pathogens by the blood-brain barrier and the innate immune system. The invading pathogens are recognized by pattern recognition receptors including the G-protein-coupled formyl peptide receptors (FPRs), which are expressed by immune cells of the central nervous system. FPRs show a broad spectrum of ligands, including pro- and anti-inflammatory ones. Here, we investigated the effects of the annexin A1 mimetic peptide Ac2-26 in a mouse model of pneumococcal meningitis. Methods Wildtype (WT) and Fpr1- and Fpr2-deficient mice were intrathecally infected with Streptococcus pneumoniae D39 (type 2). Subsequently, the different mice groups were treated by intraperitoneal injections of Ac2-26 (1 mg/kg body weight) 2, 8, and 24 h post-infection. The extent of inflammation was analyzed in various brain regions by means of immunohistochemistry and real-time reverse transcription polymerase chain reaction (RT-PCR) 30 h post-infection. Results Ac2-26-treated WT mice showed less severe neutrophil infiltration, paralleled by a reduced induction of pro-inflammatory glial cell responses in the hippocampal formation and cortex. While meningitis was ameliorated in Ac2-26-treated Fpr1-deficient mice, this protective effect was not observed in Fpr2-deficient mice. Irrespective of Ac2-26 treatment, inflammation was more severe in Fpr2-deficient compared to Fpr1-deficient mice. Conclusions In summary, this study demonstrates anti-inflammatory properties of Ac2-26 in a model of bacterial meningitis, which are mediated via FPR2, but not FPR1. Ac2-26 and other FPR2 modulators might be promising targets for the development of novel therapies for Streptococcus pneumoniae-induced meningitis.
Collapse
Affiliation(s)
- Marvin Rüger
- Institute of Anatomy, Rostock University Medical Center, Gertrudenstrasse 9, 18057, Rostock, Germany.,Department of Anatomy and Cell Biology, RWTH Aachen University, Aachen, Germany
| | - Eugenia Kipp
- Institute of Anatomy, Rostock University Medical Center, Gertrudenstrasse 9, 18057, Rostock, Germany.,Department of Anatomy and Cell Biology, RWTH Aachen University, Aachen, Germany
| | - Nadine Schubert
- Institute of Anatomy, Rostock University Medical Center, Gertrudenstrasse 9, 18057, Rostock, Germany.,Department of Anatomy and Cell Biology, RWTH Aachen University, Aachen, Germany
| | - Nicole Schröder
- Institute of Anatomy, Rostock University Medical Center, Gertrudenstrasse 9, 18057, Rostock, Germany.,Department of Anatomy and Cell Biology, RWTH Aachen University, Aachen, Germany
| | - Thomas Pufe
- Department of Anatomy and Cell Biology, RWTH Aachen University, Aachen, Germany
| | - Matthias B Stope
- Department of Urology, University Medicine Greifswald, Greifswald, Germany.,Department of Gynecology and Obstetrics, University Hospital Bonn, Bonn, Germany
| | - Markus Kipp
- Institute of Anatomy, Rostock University Medical Center, Gertrudenstrasse 9, 18057, Rostock, Germany.,Center for Transdisciplinary Neurosciences Rostock (CTNR), Rostock University Medical Center, Gelsheimer Strasse 20, 18147, Rostock, Germany
| | - Christian Blume
- Department of Neurosurgery, RWTH Aachen University, Pauwelsstrasse 30, 52074, Aachen, Germany
| | - Simone C Tauber
- Department of Neurology, RWTH University Hospital Aachen, Aachen, Germany
| | - Lars-Ove Brandenburg
- Institute of Anatomy, Rostock University Medical Center, Gertrudenstrasse 9, 18057, Rostock, Germany. .,Department of Anatomy and Cell Biology, RWTH Aachen University, Aachen, Germany. .,Center for Transdisciplinary Neurosciences Rostock (CTNR), Rostock University Medical Center, Gelsheimer Strasse 20, 18147, Rostock, Germany.
| |
Collapse
|
30
|
Fattori V, Staurengo-Ferrari L, Zaninelli TH, Casagrande R, Oliveira RD, Louzada-Junior P, Cunha TM, Alves-Filho JC, Teixeira MM, Cunha FQ, Amaral FA, Verri WA. IL-33 enhances macrophage release of IL-1β and promotes pain and inflammation in gouty arthritis. Inflamm Res 2020; 69:1271-1282. [PMID: 32886146 DOI: 10.1007/s00011-020-01399-x] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2020] [Revised: 07/29/2020] [Accepted: 08/30/2020] [Indexed: 12/17/2022] Open
Abstract
OBJECTIVE To investigate the role of IL-33 in gouty arthritis. MATERIAL 174 Balb/c (wild-type) and 54 ST2-/- mice were used in this study. In vitro experiments were conducted in bone marrow-derived macrophages (BMDMs). Synovial fluid samples from gouty arthritis (n = 7) and osteoarthritis (n = 8) hospital patients were used to measure IL-33 and sST2 levels. METHODS Gout was induced by injection of monosodium urate (MSU) crystals in the knee joint of mice. Pain was determined using the electronic von Frey and static weight bearing. Neutrophil recruitment was determined by H&E staining, Rosenfeld staining slides, and MPO activity. ELISA was used for cytokine and sST2 measurement. The priming effect of IL-33 was determined in BMDM. RESULTS Synovial fluid of gout patients showed higher IL-33 levels and neutrophil counts than osteoarthritis patients. In mice, the absence of ST2 prevented mechanical pain, knee joint edema, neutrophil recruitment to the knee joint, and lowered IL-1β and superoxide anion levels. In macrophages, IL-33 enhanced the release of IL-1β and TNF-α, and BMDMs from ST2-/- showed reduced levels of these cytokines after stimulus with MSU crystals. CONCLUSION IL-33 mediates gout pain and inflammation by boosting macrophages production of cytokines upon MSU crystals stimulus.
Collapse
Affiliation(s)
- Victor Fattori
- Laboratory of Pain, Inflammation, Neuropathy, and Cancer, Department of Pathology, Londrina State University, Londrina, Brazil
| | - Larissa Staurengo-Ferrari
- Laboratory of Pain, Inflammation, Neuropathy, and Cancer, Department of Pathology, Londrina State University, Londrina, Brazil
| | - Tiago H Zaninelli
- Laboratory of Pain, Inflammation, Neuropathy, and Cancer, Department of Pathology, Londrina State University, Londrina, Brazil
| | - Rubia Casagrande
- Department of Pharmaceutical Science, Londrina State University, Londrina, Brazil
| | - Rene D Oliveira
- Division of Clinical Immunology, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, Brazil
| | - Paulo Louzada-Junior
- Division of Clinical Immunology, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, Brazil
| | - Thiago M Cunha
- Department of Pharmacology, Center for Research in Inflammatory Diseases, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, Brazil
| | - Jose C Alves-Filho
- Department of Pharmacology, Center for Research in Inflammatory Diseases, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, Brazil
| | - Mauro M Teixeira
- Departamento de Bioquímica e Imunologia, Instituto de Ciências Biológicas (ICB), Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Fernando Q Cunha
- Department of Pharmacology, Center for Research in Inflammatory Diseases, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, Brazil
| | - Flavio A Amaral
- Departamento de Bioquímica e Imunologia, Instituto de Ciências Biológicas (ICB), Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Waldiceu A Verri
- Laboratory of Pain, Inflammation, Neuropathy, and Cancer, Department of Pathology, Londrina State University, Londrina, Brazil.
- Departamento de Ciências Patológicas, Centro de Ciências Biológicas, Universidade Estadual de Londrina, Rodovia Celso Garcia Cid, PR 445, KM 380, PO Box 10.011, Londrina, Parana, 86057-970, Brazil.
| |
Collapse
|
31
|
Tavares LP, Negreiros-Lima GL, Lima KM, E Silva PMR, Pinho V, Teixeira MM, Sousa LP. Blame the signaling: Role of cAMP for the resolution of inflammation. Pharmacol Res 2020; 159:105030. [PMID: 32562817 DOI: 10.1016/j.phrs.2020.105030] [Citation(s) in RCA: 72] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/08/2020] [Revised: 06/06/2020] [Accepted: 06/12/2020] [Indexed: 12/14/2022]
Abstract
A complex intracellular signaling governs different cellular responses in inflammation. Extracellular stimuli are sensed, amplified, and transduced through a dynamic cellular network of messengers converting the first signal into a proper response: production of specific mediators, cell activation, survival, or death. Several overlapping pathways are coordinated to ensure specific and timely induction of inflammation to neutralize potential harms to the tissue. Ideally, the inflammatory response must be controlled and self-limited. Resolution of inflammation is an active process that culminates with termination of inflammation and restoration of tissue homeostasis. Comparably to the onset of inflammation, resolution responses are triggered by coordinated intracellular signaling pathways that transduce the message to the nucleus. However, the key messengers and pathways involved in signaling transduction for resolution are still poorly understood in comparison to the inflammatory network. cAMP has long been recognized as an inducer of anti-inflammatory responses and cAMP-dependent pathways have been extensively exploited pharmacologically to treat inflammatory diseases. Recently, cAMP has been pointed out as coordinator of key steps of resolution of inflammation. Here, we summarize the evidence for the role of cAMP at inducing important features of resolution of inflammation.
Collapse
Affiliation(s)
- Luciana P Tavares
- Immunopharmacology Laboratory, Department of Biochemistry and Immunology, Instituto de Ciências Biológicas, UFMG, Belo Horizonte, Brazil; Signaling in Inflammation Laboratory, Department of Clinical and Toxicological Analysis, Faculdade de Farmácia, UFMG, Belo Horizonte, Brazil; Pulmonary and Critical Care Medicine Division, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA 02115, USA..
| | - Graziele L Negreiros-Lima
- Immunopharmacology Laboratory, Department of Biochemistry and Immunology, Instituto de Ciências Biológicas, UFMG, Belo Horizonte, Brazil; Signaling in Inflammation Laboratory, Department of Clinical and Toxicological Analysis, Faculdade de Farmácia, UFMG, Belo Horizonte, Brazil.
| | - Kátia M Lima
- Immunopharmacology Laboratory, Department of Biochemistry and Immunology, Instituto de Ciências Biológicas, UFMG, Belo Horizonte, Brazil; Signaling in Inflammation Laboratory, Department of Clinical and Toxicological Analysis, Faculdade de Farmácia, UFMG, Belo Horizonte, Brazil; Post-Graduation Program in Pharmaceutical Sciences, Faculdade de Farmácia, UFMG, Belo Horizonte, Brazil.
| | - Patrícia M R E Silva
- Inflammation Laboratory, Instituto Oswaldo Cruz, FIOCRUZ, Rio de Janeiro, Brazil.
| | - Vanessa Pinho
- Immunopharmacology Laboratory, Department of Biochemistry and Immunology, Instituto de Ciências Biológicas, UFMG, Belo Horizonte, Brazil; Department of Morphology, Instituto de Ciências Biológicas, UFMG, Belo Horizonte, Brazil.
| | - Mauro M Teixeira
- Immunopharmacology Laboratory, Department of Biochemistry and Immunology, Instituto de Ciências Biológicas, UFMG, Belo Horizonte, Brazil.
| | - Lirlândia P Sousa
- Immunopharmacology Laboratory, Department of Biochemistry and Immunology, Instituto de Ciências Biológicas, UFMG, Belo Horizonte, Brazil; Signaling in Inflammation Laboratory, Department of Clinical and Toxicological Analysis, Faculdade de Farmácia, UFMG, Belo Horizonte, Brazil; Post-Graduation Program in Pharmaceutical Sciences, Faculdade de Farmácia, UFMG, Belo Horizonte, Brazil.
| |
Collapse
|
32
|
Schröder N, Schaffrath A, Welter JA, Putzka T, Griep A, Ziegler P, Brandt E, Samer S, Heneka MT, Kaddatz H, Zhan J, Kipp E, Pufe T, Tauber SC, Kipp M, Brandenburg LO. Inhibition of formyl peptide receptors improves the outcome in a mouse model of Alzheimer disease. J Neuroinflammation 2020; 17:131. [PMID: 32331524 PMCID: PMC7181500 DOI: 10.1186/s12974-020-01816-2] [Citation(s) in RCA: 31] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2019] [Accepted: 04/16/2020] [Indexed: 12/11/2022] Open
Abstract
Background An important hallmark of Alzheimer’s disease (AD) is the increase of Aβ1-42 burden and its accumulation to senile plaques, leading the reactive gliosis and neurodegeneration. The modulation of glia cell function represents an attractive therapeutic strategy, but is currently limited by an incomplete understanding of its relevance for AD. The chemotactic G-protein coupled formyl peptide receptor (FPR), which is known to modulate Aβ1-42 uptake and signal transduction, might be one candidate molecule regulating glia function in AD. Here, we investigate whether the modulation of FPR exerts beneficial effects in an AD preclinical model. Methods To address this question, APP/PS1 double-transgenic AD mice were treated for 20 weeks with either the pro-inflammatory FPR agonist fMLF, the FPR1/2 antagonist Boc2 or the anti-inflammatory FPR2 agonist Ac2-26. Spatial learning and memory were evaluated using a Morris water maze test. Immunohistological staining, gene expression studies, and flow cytometry analyses were performed to study neuronal loss, gliosis, and Aß-load in the hippocampus and cortex, respectively. Results FPR antagonism by Boc2-treatment significantly improved spatial memory performance, reduced neuronal pathology, induced the expression of homeostatic growth factors, and ameliorated microglia, but not astrocyte, reactivity. Furthermore, the elevated levels of amyloid plaques in the hippocampus were reduced by Boc2-treatment, presumably by an induction of amyloid degradation. Conclusions We suggest that the modulation of FPR signaling cascades might be considered as a promising therapeutic approach for alleviating the cognitive deficits associated with early AD. Additional studies are now needed to address the downstream effectors as well as the safety profile of Boc2.
Collapse
Affiliation(s)
- Nicole Schröder
- Institute of Anatomy, Rostock University Medical Center, Gertrudenstrasse 9, D-18057, Rostock, Germany.,Department of Anatomy and Cell Biology, RWTH Aachen University, Aachen, Germany
| | - Anja Schaffrath
- Institute of Anatomy, Rostock University Medical Center, Gertrudenstrasse 9, D-18057, Rostock, Germany.,Department of Anatomy and Cell Biology, RWTH Aachen University, Aachen, Germany
| | - Josua A Welter
- Institute of Anatomy, Rostock University Medical Center, Gertrudenstrasse 9, D-18057, Rostock, Germany.,Department of Anatomy and Cell Biology, RWTH Aachen University, Aachen, Germany
| | - Tim Putzka
- Institute of Anatomy, Rostock University Medical Center, Gertrudenstrasse 9, D-18057, Rostock, Germany.,Department of Anatomy and Cell Biology, RWTH Aachen University, Aachen, Germany
| | - Angelika Griep
- Department of Neurodegenerative Diseases and Gerontopsychiatry, University of Bonn, Bonn, Germany
| | - Patrick Ziegler
- Institute for Occupational and Social Medicine, RWTH Aachen University, Aachen, Germany
| | - Elisa Brandt
- Department of Anatomy and Cell Biology, RWTH Aachen University, Aachen, Germany
| | - Sebastian Samer
- Institute of Anatomy, Rostock University Medical Center, Gertrudenstrasse 9, D-18057, Rostock, Germany.,Department of Anatomy and Cell Biology, RWTH Aachen University, Aachen, Germany
| | - Michael T Heneka
- Department of Neurodegenerative Diseases and Gerontopsychiatry, University of Bonn, Bonn, Germany.,German Center for Neurodegenerative Diseases (DZNE), Bonn, Germany
| | - Hannes Kaddatz
- Institute of Anatomy, Rostock University Medical Center, Gertrudenstrasse 9, D-18057, Rostock, Germany
| | - Jiangshan Zhan
- Institute of Anatomy, Rostock University Medical Center, Gertrudenstrasse 9, D-18057, Rostock, Germany
| | - Eugenia Kipp
- Institute of Anatomy, Rostock University Medical Center, Gertrudenstrasse 9, D-18057, Rostock, Germany.,Department of Anatomy and Cell Biology, RWTH Aachen University, Aachen, Germany
| | - Thomas Pufe
- Department of Anatomy and Cell Biology, RWTH Aachen University, Aachen, Germany
| | - Simone C Tauber
- Department of Neurology, RWTH University Hospital Aachen, Aachen, Germany
| | - Markus Kipp
- Institute of Anatomy, Rostock University Medical Center, Gertrudenstrasse 9, D-18057, Rostock, Germany
| | - Lars-Ove Brandenburg
- Institute of Anatomy, Rostock University Medical Center, Gertrudenstrasse 9, D-18057, Rostock, Germany. .,Department of Anatomy and Cell Biology, RWTH Aachen University, Aachen, Germany.
| |
Collapse
|
33
|
Sanches JM, Branco LM, Duarte GHB, Oliani SM, Bortoluci KR, Moreira V, Gil CD. Annexin A1 Regulates NLRP3 Inflammasome Activation and Modifies Lipid Release Profile in Isolated Peritoneal Macrophages. Cells 2020; 9:cells9040926. [PMID: 32283822 PMCID: PMC7226734 DOI: 10.3390/cells9040926] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2020] [Revised: 04/05/2020] [Accepted: 04/06/2020] [Indexed: 02/07/2023] Open
Abstract
Annexin A1 (AnxA1) is a potent anti-inflammatory protein that downregulates proinflammatory cytokine release. This study evaluated the role of AnxA1 in the regulation of NLRP3 inflammasome activation and lipid release by starch-elicited murine peritoneal macrophages. C57bl/6 wild-type (WT) and AnxA1-null (AnxA1-/-) mice received an intraperitoneal injection of 1.5% starch solution for macrophage recruitment. NLRP3 was activated by priming cells with lipopolysaccharide for 3 h, followed by nigericin (1 h) or ATP (30 min) incubation. As expected, nigericin and ATP administration decreased elicited peritoneal macrophage viability and induced IL-1β release, more pronounced in the AnxA1-/- cells than in the control peritoneal macrophages. In addition, nigericin-activated AnxA1-/- macrophages showed increased levels of NLRP3, while points of co-localization of the AnxA1 protein and NLRP3 inflammasome were detected in WT cells, as demonstrated by ultrastructural analysis. The lipidomic analysis showed a pronounced release of prostaglandins in nigericin-stimulated WT peritoneal macrophages, while ceramides were detected in AnxA1-/- cell supernatants. Different eicosanoid profiles were detected for both genotypes, and our results suggest that endogenous AnxA1 regulates the NLRP3-derived IL-1β and lipid mediator release in macrophages.
Collapse
Affiliation(s)
- José Marcos Sanches
- Departamento de Morfologia e Genética, Universidade Federal de São Paulo, São Paulo 04023-900, Brazil;
- Faculdade de Medicina, Universidade do Oeste Paulista, Guarujá, São Paulo 11410-980, Brazil
| | - Laura Migliari Branco
- Departamento de Ciências Biológicas e Centro de Terapia Celular e Molecular, Universidade Federal de São Paulo, São Paulo 04044-010, Brazil; (L.M.B.); (K.R.B.)
| | | | - Sonia Maria Oliani
- Programa de Pós-Graduação em Biociências, Instituto de Biociências, Letras e Ciências Exatas (IBILCE), Universidade Estadual Paulista, São José do Rio Preto, São Paulo 15054-000, Brazil;
| | - Karina Ramalho Bortoluci
- Departamento de Ciências Biológicas e Centro de Terapia Celular e Molecular, Universidade Federal de São Paulo, São Paulo 04044-010, Brazil; (L.M.B.); (K.R.B.)
| | - Vanessa Moreira
- Departamento de Farmacologia, Universidade Federal de São Paulo, São Paulo 04044-020, Brazil;
| | - Cristiane Damas Gil
- Departamento de Morfologia e Genética, Universidade Federal de São Paulo, São Paulo 04023-900, Brazil;
- Programa de Pós-Graduação em Biociências, Instituto de Biociências, Letras e Ciências Exatas (IBILCE), Universidade Estadual Paulista, São José do Rio Preto, São Paulo 15054-000, Brazil;
- Correspondence: ; Tel.: +55-011-5576-4268
| |
Collapse
|
34
|
Kourtzelis I, Hajishengallis G, Chavakis T. Phagocytosis of Apoptotic Cells in Resolution of Inflammation. Front Immunol 2020; 11:553. [PMID: 32296442 PMCID: PMC7137555 DOI: 10.3389/fimmu.2020.00553] [Citation(s) in RCA: 160] [Impact Index Per Article: 40.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2020] [Accepted: 03/11/2020] [Indexed: 01/04/2023] Open
Abstract
Efficient inflammation resolution is important not only for the termination of the inflammatory response but also for the restoration of tissue integrity. An integral process to resolution of inflammation is the phagocytosis of dying cells by macrophages, known as efferocytosis. This function is mediated by a complex and well-orchestrated network of interactions amongst specialized phagocytic receptors, bridging molecules, as well as “find-me” and “eat-me” signals. Efferocytosis serves not only as a waste disposal mechanism (clearance of the apoptotic cells) but also promotes a pro-resolving phenotype in efferocytic macrophages and thereby termination of inflammation. Alterations in cellular metabolism are critical for shaping the phenotype and function of efferocytic macrophages, thus, representing an important determinant of macrophage plasticity. Impaired efferocytosis can result in inflammation-associated pathologies or autoimmunity. The present mini review summarizes current knowledge regarding the mechanisms regulating macrophage efferocytosis during clearance of inflammation.
Collapse
Affiliation(s)
- Ioannis Kourtzelis
- Institute for Clinical Chemistry and Laboratory Medicine, University Hospital and Faculty of Medicine, Technische Universität Dresden, Dresden, Germany.,Hull York Medical School, York Biomedical Research Institute, University of York, York, United Kingdom
| | - George Hajishengallis
- Laboratory of Innate Immunity and Inflammation, Penn Dental Medicine, Department of Basic and Translational Sciences, University of Pennsylvania, Philadelphia, PA, United States
| | - Triantafyllos Chavakis
- Institute for Clinical Chemistry and Laboratory Medicine, University Hospital and Faculty of Medicine, Technische Universität Dresden, Dresden, Germany.,Centre for Cardiovascular Science, Queen's Medical Research Institute, University of Edinburgh, Edinburgh, United Kingdom
| |
Collapse
|
35
|
Machado MG, Tavares LP, Souza GVS, Queiroz-Junior CM, Ascenção FR, Lopes ME, Garcia CC, Menezes GB, Perretti M, Russo RC, Teixeira MM, Sousa LP. The Annexin A1/FPR2 pathway controls the inflammatory response and bacterial dissemination in experimental pneumococcal pneumonia. FASEB J 2019; 34:2749-2764. [PMID: 31908042 DOI: 10.1096/fj.201902172r] [Citation(s) in RCA: 48] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2019] [Revised: 11/29/2019] [Accepted: 12/09/2019] [Indexed: 01/10/2023]
Abstract
Streptococcus pneumoniae is a major cause of community-acquired pneumonia leading to high mortality rates. Inflammation triggered by pneumococcal infection is necessary for bacterial clearance but must be spatially and temporally regulated to prevent further tissue damage and bacterial dissemination. Annexin A1 (AnxA1) mainly acts through Formyl Peptide Receptor 2 (FPR2) inducing the resolution of inflammation. Here, we have evaluated the role of AnxA1 and FPR2 during pneumococcal pneumonia in mice. For that, AnxA1, Fpr2/3 knockout (KO) mice and wild-type (WT) controls were infected intranasally with S pneumoniae. AnxA1 and Fpr2/3 KO mice were highly susceptible to infection, displaying uncontrolled inflammation, increased bacterial dissemination, and pulmonary dysfunction compared to WT animals. Mechanistically, the absence of AnxA1 resulted in the loss of lung barrier integrity and increased neutrophil activation upon S pneumoniae stimulation. Importantly, treatment of WT or AnxA1 KO-infected mice with Ac2-26 decreased inflammation, lung damage, and bacterial burden in the airways by increasing macrophage phagocytosis. Conversely, Ac2-26 peptide was ineffective to afford protection in Fpr2/3 KO mice during infection. Altogether, these findings show that AnxA1, via FPR2, controls inflammation and bacterial dissemination during pneumococcal pneumonia by promoting host defenses, suggesting AnxA1-based peptides as a novel therapeutic strategy to control pneumococcal pneumonia.
Collapse
Affiliation(s)
- Marina Gomes Machado
- Laboratório de sinalização na inflamação, Departamento de Análises Clínicas e Toxicológicas, Faculdade de Farmácia, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil.,Laboratório de Imunofarmacologia, Departamento de Bioquímica e Imunologia, ICB, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Luciana Pádua Tavares
- Laboratório de Imunofarmacologia, Departamento de Bioquímica e Imunologia, ICB, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Geovanna V Santos Souza
- Laboratório de sinalização na inflamação, Departamento de Análises Clínicas e Toxicológicas, Faculdade de Farmácia, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil.,Laboratório de Imunofarmacologia, Departamento de Bioquímica e Imunologia, ICB, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Celso M Queiroz-Junior
- Departamento de Morfologia, ICB, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Fernando Roque Ascenção
- Laboratório de Imunofarmacologia, Departamento de Bioquímica e Imunologia, ICB, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Mateus Eustáquio Lopes
- Departamento de Morfologia, ICB, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Cristiana Couto Garcia
- Laboratório de Vírus Respiratórios e do Sarampo, Instituto Oswaldo Cruz (Fiocruz), Rio de Janeiro, Brazil
| | - Gustavo Batista Menezes
- Departamento de Morfologia, ICB, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Mauro Perretti
- William Harvey Research Institute, Barts and The London School of Medicine, Queen Mary University of London, London, United Kingdom
| | - Remo Castro Russo
- Laboratório de Imunologia e Mecânica Pulmonar, Departamento de Fisiologia e Biofísica, ICB, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Mauro Martins Teixeira
- Laboratório de Imunofarmacologia, Departamento de Bioquímica e Imunologia, ICB, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Lirlândia Pires Sousa
- Laboratório de sinalização na inflamação, Departamento de Análises Clínicas e Toxicológicas, Faculdade de Farmácia, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil.,Laboratório de Imunofarmacologia, Departamento de Bioquímica e Imunologia, ICB, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| |
Collapse
|
36
|
Investigating MIF in Mouse Model of Gout. Methods Mol Biol 2019. [PMID: 31745884 DOI: 10.1007/978-1-4939-9936-1_19] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register]
Abstract
Mice are widely used to assess the pathogenesis of diseases. An experimental model of gout consists of the injection of uric acid crystals into joints of mice, which reproduce inflammation and functional changes of the human disease. Uric acid crystals activate synoviocytes culminating in the release of IL-1β and neutrophil recruitment, key inflammatory elements in gouty arthritis. Since MIF plays an important role in orchestrating gout inflammation, we detail valuable procedures to investigate uric acid crystal-induced joint inflammation in mice and give options for further understanding the functions of MIF in gouty arthritis in vivo and in vitro.
Collapse
|
37
|
Galvão I, Athayde RM, Perez DA, Reis AC, Rezende L, de Oliveira VLS, Rezende BM, Gonçalves WA, Sousa LP, Teixeira MM, Pinho V. ROCK Inhibition Drives Resolution of Acute Inflammation by Enhancing Neutrophil Apoptosis. Cells 2019; 8:E964. [PMID: 31450835 PMCID: PMC6769994 DOI: 10.3390/cells8090964] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2019] [Revised: 08/19/2019] [Accepted: 08/22/2019] [Indexed: 02/07/2023] Open
Abstract
Uncontrolled inflammation leads to tissue damage and it is central for the development of chronic inflammatory diseases and autoimmunity. An acute inflammatory response is finely regulated by the action of anti-inflammatory and pro-resolutive mediators, culminating in the resolution of inflammation and restoration of homeostasis. There are few studies investigating intracellular signaling pathways associated with the resolution of inflammation. Here, we investigate the role of Rho-associated kinase (ROCK), a serine/threonine kinase, in a model of self-resolving neutrophilic inflammatory. We show that ROCK activity, evaluated by P-MYPT-1 kinetics, was higher during the peak of lipopolysaccharide-induced neutrophil influx in the pleural cavity of mice. ROCK inhibition by treatment with Y-27632 decreased the accumulation of neutrophils in the pleural cavity and was associated with an increase in apoptotic events and efferocytosis, as evaluated by an in vivo assay. In a model of gout, treatment with Y-27632 reduced neutrophil accumulation, IL-1β levels and hypernociception in the joint. These were associated with reduced MYPT and IκBα phosphorylation levels and increased apoptosis. Finally, inhibition of ROCK activity also induced apoptosis in human neutrophils and destabilized cytoskeleton, extending the observed effects to human cells. Taken together, these data show that inhibition of the ROCK pathway might represent a potential therapeutic target for neutrophilic inflammatory diseases.
Collapse
Affiliation(s)
- Izabela Galvão
- Departamento de Morfologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte 31270-901, Brazil
| | - Rayssa M Athayde
- Departamento de Morfologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte 31270-901, Brazil
| | - Denise A Perez
- Departamento de Morfologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte 31270-901, Brazil
| | - Alesandra C Reis
- Departamento de Morfologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte 31270-901, Brazil
| | - Luisa Rezende
- Departamento de Morfologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte 31270-901, Brazil
| | - Vivian Louise S de Oliveira
- Departamento de Bioquímica e Imunologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte 31270-901, Brazil
| | - Barbara M Rezende
- Departamento de Enfermagem Básica, Escola de Enfermagem, Universidade Federal de Minas Gerais, Belo Horizonte 30130-100, Brazil
| | - William A Gonçalves
- Departamento de Morfologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte 31270-901, Brazil
| | - Lirlândia P Sousa
- Departamento de Análises Clínicas e Toxicológicas, Faculdade de Farmácia; Universidade Federal de Minas Gerais, Belo Horizonte 312701-901, Brazil
| | - Mauro M Teixeira
- Departamento de Bioquímica e Imunologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte 31270-901, Brazil
| | - Vanessa Pinho
- Departamento de Morfologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte 31270-901, Brazil.
| |
Collapse
|
38
|
Han G, Lu K, Xu W, Zhang S, Huang J, Dai C, Sun G, Ye J. Annexin A1-mediated inhibition of inflammatory cytokines may facilitate the resolution of inflammation in acute radiation-induced lung injury. Oncol Lett 2019; 18:321-329. [PMID: 31289503 DOI: 10.3892/ol.2019.10317] [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: 04/05/2018] [Accepted: 02/14/2019] [Indexed: 12/17/2022] Open
Abstract
The present study evaluated the role of annexin A1 (ANXA1) in the treatment of acute radiation-induced lung injury (RILI) and investigated the mechanism of its action. The expression of ANXA1, interleukin-6 (IL-6) and myeloperoxidase (MPO) in the plasma of patients with RILI prior to and following hormonotherapy was assessed by enzyme-linked immunosorbent assay. The association of plasma ANXA1 concentration with clinical effect, and the correlation between the expression of ANXA1 and that of IL-6 and MPO were evaluated. ANXA1 was overexpressed or knocked down in a macrophage cell line, and its impact on IL-6 and MPO expression was measured. Following glucocorticoid hormonotherapy, patients with RILI exhibited a higher plasma concentration of ANXA1 compared with that prior to treatment, while IL-6 and MPO levels were lower. The concentration of ANXA1 in plasma was negatively correlated with IL-6 and MPO levels, with a correlation coefficient of -0.492 and -0.437, respectively (P<0.001). The increasing concentration of ANXA1 in plasma following treatment was associated with the clinical effect in patients with RILI (P=0.007). The expression levels of of IL-6 and MPO were inhibited both in the cytoplasm and in the culture solution, when ANXA1 expression was upregulated in a macrophage cell line. In conclusion, ANXA1 inhibited the synthesis and secretion of IL-6 and MPO inflammatory cytokines, indicating that ANXA1 may have therapeutic potential as a treatment target for RILI.
Collapse
Affiliation(s)
- Gaohua Han
- Department of Oncology, The Fifth Affiliated Hospital of Nantong University, Taizhou People's Hospital, Taizhou, Jiangsu 225300, P.R. China
| | - Kaijin Lu
- Department of Thoracic Surgery, The Fifth Affiliated Hospital of Nantong University, Taizhou People's Hospital, Taizhou, Jiangsu 225300, P.R. China
| | - Wansong Xu
- Radiation Therapy Center, The Fifth Affiliated Hospital of Nantong University, Taizhou People's Hospital, Taizhou, Jiangsu 225300, P.R. China
| | - Sihui Zhang
- Department of Oncology, The Fifth Affiliated Hospital of Nantong University, Taizhou People's Hospital, Taizhou, Jiangsu 225300, P.R. China
| | - Junxing Huang
- Department of Oncology, The Fifth Affiliated Hospital of Nantong University, Taizhou People's Hospital, Taizhou, Jiangsu 225300, P.R. China
| | - Chunlei Dai
- Medical Imaging Center, The Fifth Affiliated Hospital of Nantong University, Taizhou People's Hospital, Taizhou, Jiangsu 225300, P.R. China
| | - Guangzhi Sun
- Radiation Therapy Center, The Fifth Affiliated Hospital of Nantong University, Taizhou People's Hospital, Taizhou, Jiangsu 225300, P.R. China
| | - Jun Ye
- Central Laboratory, The Fifth Affiliated Hospital of Nantong University, Taizhou People's Hospital, Taizhou, Jiangsu 225300, P.R. China
| |
Collapse
|
39
|
Sugimoto MA, Vago JP, Perretti M, Teixeira MM. Mediators of the Resolution of the Inflammatory Response. Trends Immunol 2019; 40:212-227. [DOI: 10.1016/j.it.2019.01.007] [Citation(s) in RCA: 100] [Impact Index Per Article: 20.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2018] [Revised: 01/13/2019] [Accepted: 01/14/2019] [Indexed: 02/06/2023]
|
40
|
Hilliard KA, Brown CR. Treatment of Borrelia burgdorferi-Infected Mice with Apoptotic Cells Attenuates Lyme Arthritis via PPAR-γ. THE JOURNAL OF IMMUNOLOGY 2019; 202:1798-1806. [PMID: 30700583 DOI: 10.4049/jimmunol.1801179] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/24/2018] [Accepted: 01/02/2019] [Indexed: 12/25/2022]
Abstract
Infection of mice with Borrelia burgdorferi causes an inflammatory arthritis that peaks 3-4 wk postinfection and then spontaneously resolves. Although the recruitment of neutrophils is known to drive the development of arthritis, mechanisms of disease resolution remain unclear. Efficient clearance of apoptotic cells (AC) is likely an important component of arthritis resolution. In this article, we show the number of AC increases in the joints of B. burgdorferi-infected mice around day 21 postinfection and peaks around day 28. Injection of AC directly into the ankles of B. burgdorferi-infected mice limited ankle swelling but had no effect on spirochete clearance or arthritis severity scores. In vitro, addition of AC to bone marrow macrophage cultures decreased B. burgdorferi-induced TNF-α and KC and increased IL-10. In addition, phagocytosis of B. burgdorferi and neutrophil migration to LTB4 were inhibited by AC. Exogenous AC caused an increase in peroxisome proliferator-activated receptor-γ (PPAR-γ) expression both in vitro and in vivo during B. burgdorferi infection. The PPAR-γ agonist rosiglitazone elicited similar changes in macrophage cytokine production and neutrophil migration as exogenous AC. Addition of the PPAR-γ antagonist GW 9662 abrogated the effects of AC in vitro. Injection of rosiglitazone directly into the tibiotarsal joints of B. burgdorferi-infected mice decreased ankle swelling and immune cell recruitment, similar to the injection of AC. These results suggest that clearance of AC plays a role in the resolution of inflammation during experimental Lyme arthritis through the activation of PPAR-γ. PPAR-γ agonists, such as rosiglitazone, may therefore be effective treatments for inducing arthritis resolution.
Collapse
Affiliation(s)
- Kinsey A Hilliard
- Department of Veterinary Pathobiology, University of Missouri, Columbia, MO 65211
| | - Charles R Brown
- Department of Veterinary Pathobiology, University of Missouri, Columbia, MO 65211
| |
Collapse
|
41
|
Galvão I, Queiroz-Junior CM, de Oliveira VLS, Pinho V, Hirsch E, Teixeira MM. The Inhibition of Phosphoinositide-3 Kinases Induce Resolution of Inflammation in a Gout Model. Front Pharmacol 2019; 9:1505. [PMID: 30666201 PMCID: PMC6330337 DOI: 10.3389/fphar.2018.01505] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2018] [Accepted: 12/10/2018] [Indexed: 01/11/2023] Open
Abstract
Phosphoinositide-3 kinases (PI3Ks) are central signaling enzymes that are involved in many aspects of immune cell function. PI3Kγ and PI3Kδ are the major isoforms expressed in leukocytes. The role of PI3K isoforms in the resolution of inflammation is still poorly understood. Here, we investigated the contribution of PI3Kγ and PI3Kδ to the resolution of inflammation in a model of gout in mice. Methods and Results: Experiments were performed in wild-type male C57/Bl6 mice. Selective inhibitors of PI3K-γ (AS605240) or PI3Kδ (GSK045) were injected in the joint 12 h after injection of MSU crystals, hence at the peak of inflammation. Inhibition of either PI3K isoform decreased number of neutrophils that migrated in response to the injection of MSU crystals. This was associated with reduction of myeloperoxidase activity and IL-1β levels in periarticular tissues and reduction of histological score. Joint dysfunction, as seen by reduced mechanical hypernociception, was improved by treatment with either inhibitor. The decrease in neutrophil numbers was associated with enhanced apoptosis and efferocytosis of these cells. There was shortening of resolution intervals, suggesting inhibition of either isoform induced the resolution of neutrophilic inflammation. Blockade of PI3Kγ or PI3Kδ reduced Nuclear Factor kappa B (NF-κB) activation. A pan-PI3K inhibitor (CL27c) reduced inflammation induced by MSU crystals by a magnitude that was similar to that attained by the PI3Kγ or PI3Kδ selective inhibitors alone. Conclusion: Taken together, these results suggest that neutrophils can use PI3Kγ or PI3Kδ to remain in the cavity and blockade of either isoenzyme is sufficient to induce their apoptosis and resolve inflammation in a murine model of gout.
Collapse
Affiliation(s)
- Izabela Galvão
- Departamento de Bioquímica e Imunologia, 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
| | - Vivian Louise Soares de Oliveira
- 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
| | - Emilio Hirsch
- Department of Molecular Biotechnology and Health Sciences, University of Turin, Turin, Italy
| | - Mauro Martins Teixeira
- Departamento de Bioquímica e Imunologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| |
Collapse
|
42
|
Fattori V, Zarpelon AC, Staurengo-Ferrari L, Borghi SM, Zaninelli TH, Da Costa FB, Alves-Filho JC, Cunha TM, Cunha FQ, Casagrande R, Arakawa NS, Verri WA. Budlein A, a Sesquiterpene Lactone From Viguiera robusta, Alleviates Pain and Inflammation in a Model of Acute Gout Arthritis in Mice. Front Pharmacol 2018; 9:1076. [PMID: 30319413 PMCID: PMC6167909 DOI: 10.3389/fphar.2018.01076] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2018] [Accepted: 09/05/2018] [Indexed: 01/27/2023] Open
Abstract
Background: Gout is the most common inflammatory arthritis worldwide. It is a painful inflammatory disease induced by the deposition of monosodium urate (MSU) crystals in the joints and peri-articular tissues. Sesquiterpene lactones (SLs) are secondary metabolite biosynthesized mainly by species from the family Asteraceae. It has been demonstrated that SLs present anti-inflammatory, analgesic, antitumoral, antiparasitic, and antimicrobial activities. In this study, we aimed at evaluating the efficacy of the SL budlein A in a model of acute gout arthritis in mice. Methods: Experiments were conducted in male Swiss or male LysM-eGFP mice. Animals were treated with budlein A (1 or 10 mg/kg) or vehicle 30 min before stimulus with MSU (100 μg/10 μL, intra-articular). Knee joint withdrawal threshold and edema were evaluated using electronic von Frey and caliper, respectively, 1-15 h after MSU injection. Leukocyte recruitment was determined by counting cells (Neubauer chamber), H&E staining, and using LysM-eGFP mice by confocal microscopy. Inflammasome components, Il-1β, and Tnf-α mRNA expression were determined by RT-qPCR. IL-1β and TNF-α production (in vitro) and NF-κB activation (in vitro and in vivo) were evaluated by ELISA. In vitro analysis using LPS-primed bone marrow-derived macrophages (BMDMs) was performed 5 h after stimulation with MSU crystals. For these experiments, BMDMs were either treated or pre-treated with budlein A at concentrations of 1, 3, or 10 μg/mL. Results: We demonstrated that budlein A reduced mechanical hypersensitivity and knee joint edema. Moreover, it reduced neutrophil recruitment, phagocytosis of MSU crystals by neutrophils, and Il-1β and Tnf-α mRNA expression in the knee joint. In vitro, budlein A decreased TNF-α production, which might be related to the inhibition of NF-κB activation. Furthermore, budlein A also reduced the IL-1β maturation, possibly by targeting inflammasome assembly in macrophages. Conclusion: Budlein A reduced pain and inflammation in a model of acute gout arthritis in mice. Therefore, it is likely that molecules with the ability of targeting NF-κB activation and inflammasome assembly, such as budlein A, are interesting approaches to treat gout flares.
Collapse
Affiliation(s)
- Victor Fattori
- Laboratory of Pain, Inflammation, Neuropathy, and Cancer, Department of Pathology, Londrina State University, Londrina, Brazil
| | - Ana C Zarpelon
- Laboratory of Pain, Inflammation, Neuropathy, and Cancer, Department of Pathology, Londrina State University, Londrina, Brazil
| | - Larissa Staurengo-Ferrari
- Laboratory of Pain, Inflammation, Neuropathy, and Cancer, Department of Pathology, Londrina State University, Londrina, Brazil
| | - Sergio M Borghi
- Laboratory of Pain, Inflammation, Neuropathy, and Cancer, Department of Pathology, Londrina State University, Londrina, Brazil
| | - Tiago H Zaninelli
- Laboratory of Pain, Inflammation, Neuropathy, and Cancer, Department of Pathology, Londrina State University, Londrina, Brazil
| | - Fernando B Da Costa
- AsterBioChem Research Team, School of Pharmaceutical Sciences of Ribeirão Preto, University of São Paulo, Ribeirão Preto, Brazil
| | - Jose C Alves-Filho
- Laboratory of Inflammation and Pain, Department of Pharmacology, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, Brazil
| | - Thiago M Cunha
- Laboratory of Inflammation and Pain, Department of Pharmacology, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, Brazil
| | - Fernando Q Cunha
- Laboratory of Inflammation and Pain, Department of Pharmacology, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, Brazil
| | - Rubia Casagrande
- Department of Pharmaceutical Sciences, Center of Health Sciences, Londrina State University, Londrina, Brazil
| | - Nilton S Arakawa
- Department of Pharmaceutical Sciences, Center of Health Sciences, Londrina State University, Londrina, Brazil
| | - Waldiceu A Verri
- Laboratory of Pain, Inflammation, Neuropathy, and Cancer, Department of Pathology, Londrina State University, Londrina, Brazil
| |
Collapse
|
43
|
Vidotto A, Polachini GM, de Paula-Silva M, Oliani SM, Henrique T, López RVM, Cury PM, Nunes FD, Góis-Filho JF, de Carvalho MB, Leopoldino AM, Tajara EH. Differentially expressed proteins in positive versus negative HNSCC lymph nodes. BMC Med Genomics 2018; 11:73. [PMID: 30157864 PMCID: PMC6114741 DOI: 10.1186/s12920-018-0382-6] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2017] [Accepted: 08/02/2018] [Indexed: 12/21/2022] Open
Abstract
Background Lymph node metastasis is one of the most important prognostic factors in head and neck squamous cell carcinomas (HNSCCs) and critical for delineating their treatment. However, clinical and histological criteria for the diagnosis of nodal status remain limited. In the present study, we aimed to characterize the proteomic profile of lymph node metastasis from HNSCC patients. Methods In the present study, we used one- and two-dimensional electrophoresis and mass spectrometry analysis to characterize the proteomic profile of lymph node metastasis from HNSCC. Results Comparison of metastatic and non-metastatic lymph nodes showed 52 differentially expressed proteins associated with neoplastic development and progression. The results reinforced the idea that tumors from different anatomical subsites have dissimilar behaviors, which may be influenced by micro-environmental factor including the lymphatic network. The expression pattern of heat shock proteins and glycolytic enzymes also suggested an effect of the lymph node environment in controlling tumor growth or in metabolic reprogramming of the metastatic cell. Our study, for the first time, provided direct evidence of annexin A1 overexpression in lymph node metastasis of head and neck cancer, adding information that may be useful for diagnosing aggressive disease. Conclusions In brief, this study contributed to our understanding of the metastatic phenotype of HNSCC and provided potential targets for diagnostic in this group of carcinomas. Electronic supplementary material The online version of this article (10.1186/s12920-018-0382-6) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Alessandra Vidotto
- Departamento de Biologia Molecular, Faculdade de Medicina (FAMERP), Av. Brigadeiro Faria Lima, 5416, Vila São Pedro, São José do Rio Preto, SP, CEP 15090-000, Brazil
| | - Giovana M Polachini
- Departamento de Biologia Molecular, Faculdade de Medicina (FAMERP), Av. Brigadeiro Faria Lima, 5416, Vila São Pedro, São José do Rio Preto, SP, CEP 15090-000, Brazil
| | - Marina de Paula-Silva
- Departamento de Biologia, Instituto de Biociências, Letras e Ciências Exatas (IBILCE), Universidade Estadual Paulista (UNESP), R. Cristóvão Colombo, 2265, São José do Rio Preto, SP, CEP 15054-000, Brazil
| | - Sonia M Oliani
- Departamento de Biologia, Instituto de Biociências, Letras e Ciências Exatas (IBILCE), Universidade Estadual Paulista (UNESP), R. Cristóvão Colombo, 2265, São José do Rio Preto, SP, CEP 15054-000, Brazil
| | - Tiago Henrique
- Departamento de Biologia Molecular, Faculdade de Medicina (FAMERP), Av. Brigadeiro Faria Lima, 5416, Vila São Pedro, São José do Rio Preto, SP, CEP 15090-000, Brazil
| | - Rossana V M López
- Instituto do Câncer de São Paulo Octavio Frias de Oliveira - ICESP, Av. Dr. Arnaldo, 251 - Cerqueira César, São Paulo, SP, CEP 01246-000, Brazil
| | - Patrícia M Cury
- Faculdade Ceres (Faceres), Av. Anísio Haddad, 6751, São José do Rio Preto, SP, CEP 15090-305, Brazil
| | - Fabio D Nunes
- Departamento de Estomatologia, Faculdade de Odontologia, Universidade de São Paulo, Av. Prof. Lineu Prestes, 2227, São Paulo, SP, CEP 05508-000, Brazil
| | - José F Góis-Filho
- Instituto do Câncer Arnaldo Vieira de Carvalho, R. Dr Cesário Mota Jr, 112, São Paulo, SP, CEP 01221-020, Brazil
| | - Marcos B de Carvalho
- Departamento de Cirurgia de Cabeça e Pescoço, Hospital Heliópolis, R. Cônego Xavier, 276, São Paulo, SP, CEP 04231-030, Brazil
| | - Andréia M Leopoldino
- Departamento de Análises Clínicas, Toxicológicas e Bromatológicas, Faculdade de Ciências Farmacêuticas, Universidade de São Paulo, Avenida do Café, s/n, Ribeirão Preto, SP, CEP 14040-903, Brazil
| | - Eloiza H Tajara
- Departamento de Biologia Molecular, Faculdade de Medicina (FAMERP), Av. Brigadeiro Faria Lima, 5416, Vila São Pedro, São José do Rio Preto, SP, CEP 15090-000, Brazil. .,Departamento de Genética e Biologia Evolutiva, Instituto de Biociências, Universidade de São Paulo, R. do Matão, 321, São Paulo, SP, CEP 05508-090, Brazil.
| |
Collapse
|
44
|
Formyl peptide receptor activation inhibits the expansion of effector T cells and synovial fibroblasts and attenuates joint injury in models of rheumatoid arthritis. Int Immunopharmacol 2018; 61:140-149. [PMID: 29879657 DOI: 10.1016/j.intimp.2018.05.028] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2017] [Revised: 05/25/2018] [Accepted: 05/25/2018] [Indexed: 12/12/2022]
Abstract
The effects of formyl peptide receptors (FPRs) on effector T cells and inflammation-causing tissue-resident cells are not well known. Here, we explored the effect of FPR activation on efferent T cell responses in models of rheumatoid arthritis (RA) and on the expansion of fibroblast-like synoviocytes (FLS). Compound 43 (Cpd43; FPR1/2 agonist) was administered to mice with collagen-induced arthritis (CIA) or antigen-induced arthritis (AIA) after disease onset. Joint inflammation/damage and immunity were assessed. FLS were cultured with Cpd43 to test its effects on cell apoptosis and proliferation. To explore the effects of endogenous FPR2 ligands on FLS proliferation, FLS FPR2 was blocked or Annexin A1 (AnxA1) expression silenced. Cpd43 reduced arthritis severity in both models. In CIA, Cpd43 decreased CD4 T cell proliferation and survival and increased the production of the protective cytokine, IFNγ, in lymph nodes. In AIA, Cpd43 increased CD4 apoptosis and production of the anti-inflammatory IL-4, while augmenting the proportion of splenic regulatory T cells and their expression of IL-2Rα. In both models, Cpd43 increased CD4 IL-17A production, without affecting humoral immunity. FPR2 inhibitors reversed Cpd43-mediated effects on AIA and T cell immunity. Cpd43 decreased TNF-induced FLS proliferation and augmented FLS apoptosis in association with intracellular FPR2 accumulation, while endogenous AnxA1 and FPR2 reduced FLS proliferation via the ERK and NFκB pathways. Overall, FPR activation inhibits the expansion of arthritogenic effector CD4 T cells and FLS, and reduces joint injury in experimental arthritis. This suggests the therapeutic potential of FPR ligation for the treatment of RA.
Collapse
|
45
|
Chen YC, Lin MC, Lee CH, Liu SF, Wang CC, Fang WF, Chao TY, Wu CC, Wei YF, Chang HC, Tsen CC, Chen HC. Defective formyl peptide receptor 2/3 and annexin A1 expressions associated with M2a polarization of blood immune cells in patients with chronic obstructive pulmonary disease. J Transl Med 2018; 16:69. [PMID: 29544524 PMCID: PMC5856198 DOI: 10.1186/s12967-018-1435-5] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2017] [Accepted: 03/05/2018] [Indexed: 12/14/2022] Open
Abstract
Background Controversy exists in previous studies on macrophage M1/M2 polarization in chronic obstructive pulmonary disease (COPD). We hypothesized that formyl peptide receptor (FPR), a marker of efferocytosis and mediator of M1/M2 polarization, may be involved in the development of COPD. Methods We examined FPR 1/2/3 expressions of blood M1/M2a monocyte, neutrophil, natural killer (NK) cell, NK T cell, T helper (Th) cell, and T cytotoxic (Tc) cell by flowcytometry method in 40 patients with cigarette smoking-related COPD and 16 healthy non-smokers. Serum levels of five FPR ligands were measured by ELISA method. Results The COPD patients had lower M2a percentage and higher percentages of NK, NK T, Th, and Tc cells than the healthy non-smokers. FPR2 expressions on Th/Tc cells, FPR3 expressions of M1, M2a, NK, NK T, Th, and Tc cells, and serum annexin A1 (an endogenous FPR2 ligand) levels were all decreased in the COPD patients as compared with that in the healthy non-smokers. FPR1 expression on neutrophil was increased in the COPD patient with a high MMRC dyspnea scale, while FPR2 expression on neutrophil and annexin A1 were both decreased in the COPD patients with a history of frequent moderate exacerbation (≥ 2 events in the past 1 year). In 10 COPD patients whose blood samples were collected again after 1-year treatment, M2a percentage, FPR3 expressions of M1/NK/Th cells, FPR2 expression on Th cell, and FPR1 expression on neutrophil were all reversed to normal, in parallel with partial improvement in small airway dysfunction. Conclusions Our findings provide evidence for defective FPR2/3 and annexin A1 expressions that, associated with decreased M2a polarization, might be involved in the development of cigarette smoking induced persistent airflow limitation in COPD.
Collapse
Affiliation(s)
- Yung-Che Chen
- Division of Pulmonary and Critical Care Medicine, Kaohsiung Chang Gung Memorial Hospital and Chang Gung University College of Medicine, Kaohsiung, Taiwan.,Medical Department, College of Medicine, Chang Gung University, Taoyuan, Taiwan
| | - Meng-Chih Lin
- Division of Pulmonary and Critical Care Medicine, Kaohsiung Chang Gung Memorial Hospital and Chang Gung University College of Medicine, Kaohsiung, Taiwan. .,Medical Department, College of Medicine, Chang Gung University, Taoyuan, Taiwan. .,Division of Pulmonary and Critical Care Medicine, Department of Medicine, Kaohsiung Chang Gung Memorial Hospital and Chang Gung University College of Medicine, 123, Ta-Pei Rd, Niao-Sung District, Kaohsiung, Taiwan.
| | - Chih-Hung Lee
- Medical Department, College of Medicine, Chang Gung University, Taoyuan, Taiwan.,Department of Dermatology, Kaohsiung Chang Gung Memorial Hospital and Chang Gung University College of Medicine, Kaohsiung, Taiwan
| | - Shih-Feng Liu
- Division of Pulmonary and Critical Care Medicine, Kaohsiung Chang Gung Memorial Hospital and Chang Gung University College of Medicine, Kaohsiung, Taiwan.,Department of Respiratory Therapy, Kaohsiung Chang Gung Memorial Hospital and Chang Gung University College of Medicine, Kaohsiung, Taiwan
| | - Chin-Chou Wang
- Division of Pulmonary and Critical Care Medicine, Kaohsiung Chang Gung Memorial Hospital and Chang Gung University College of Medicine, Kaohsiung, Taiwan.,Chang Gung University of Science and Technology, Chia-yi, Taiwan
| | - Wen-Feng Fang
- Division of Pulmonary and Critical Care Medicine, Kaohsiung Chang Gung Memorial Hospital and Chang Gung University College of Medicine, Kaohsiung, Taiwan.,Chang Gung University of Science and Technology, Chia-yi, Taiwan
| | - Tung-Ying Chao
- Division of Pulmonary and Critical Care Medicine, Kaohsiung Chang Gung Memorial Hospital and Chang Gung University College of Medicine, Kaohsiung, Taiwan
| | - Chao-Chien Wu
- Division of Pulmonary and Critical Care Medicine, Kaohsiung Chang Gung Memorial Hospital and Chang Gung University College of Medicine, Kaohsiung, Taiwan
| | - Yu-Feng Wei
- Department of Internal Medicine, E-Da Hospital, I-Shou University, Kaohsiung, Taiwan
| | - Huang-Chih Chang
- Division of Pulmonary and Critical Care Medicine, Kaohsiung Chang Gung Memorial Hospital and Chang Gung University College of Medicine, Kaohsiung, Taiwan
| | - Chia-Cheng Tsen
- Division of Pulmonary and Critical Care Medicine, Kaohsiung Chang Gung Memorial Hospital and Chang Gung University College of Medicine, Kaohsiung, Taiwan
| | - Hung-Chen Chen
- Division of Pulmonary and Critical Care Medicine, Kaohsiung Chang Gung Memorial Hospital and Chang Gung University College of Medicine, Kaohsiung, Taiwan
| | | |
Collapse
|
46
|
Abstract
The acute symptoms of gout are triggered by the inflammatory response to monosodium urate crystals, mediated principally by macrophages and neutrophils. Innate immune pathways are of key importance in the pathogenesis of gout, in particular the activation of the NLRP3 inflammasome, which leads to the release of IL-1β and other pro-inflammatory cytokines. The orchestration of this pro-inflammatory cascade involves multiple intracellular and extracellular receptors and enzymes interacting with environmental influences that modulate the inflammatory state. Furthermore, the resolution of inflammation in gout is becoming better understood. This Review highlights recent advances in our understanding of both positive and negative regulatory pathways, as well as the genetic and environmental factors that modulate the inflammatory response. Some of these pathways can be manipulated and present novel therapeutic opportunities for the treatment of acute gout attacks.
Collapse
Affiliation(s)
- Alexander K So
- Service of Rheumatology, Centre Hospitalier Universitaire Vaudois and University of Lausanne, Avenue Pierre Decker 4, 1011 Lausanne, Switzerland
| | - Fabio Martinon
- Department of Biochemistry, University of Lausanne, 155 Chemin des Boveresses, 1066 Epalinges, Switzerland
| |
Collapse
|
47
|
Abstract
Acute gout arthritis flares contribute dominantly to gout-specific impaired health-related quality of life, representing a progressively increasing public health problem. Flares can be complex and expensive to treat, partly due to the frequent comorbidities. Unmet needs in gout management are more pressing given the markedly increasing gout flare hospital admission rates. In addition, chronic gouty arthritis can cause joint damage and functional impairment. This review addresses new knowledge on the basis for the marked, inherent variability of responses to deposited urate crystals, including the unpredictable and self-limited aspects of many gout flares. Specific topics reviewed include how innate immunity and two-signal inflammasome activation intersect with diet, metabolism, nutritional biosensing, the microbiome, and the phagocyte cytoskeleton and cell fate. The paper discusses the roles of endogenous constitutive regulators of inflammation, including certain nutritional biosensors, and emerging genetic and epigenetic factors. Recent advances in the basis of variability in responses to urate crystals in gout provide information about inflammatory arthritis, and have identified potential new targets and strategies for anti-inflammatory prevention and treatment of gouty arthritis.
Collapse
Affiliation(s)
- Robert Terkeltaub
- VA San Diego Healthcare System, 111K, 3350 La Jolla Village Drive, San Diego, CA, 92161, USA. .,Department of Medicine, University of California San Diego, San Diego, CA, USA.
| |
Collapse
|
48
|
Perretti M, Di Filippo C, D’Amico M, Dalli J. Characterizing the anti-inflammatory and tissue protective actions of a novel Annexin A1 peptide. PLoS One 2017; 12:e0175786. [PMID: 28407017 PMCID: PMC5391094 DOI: 10.1371/journal.pone.0175786] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2017] [Accepted: 03/31/2017] [Indexed: 12/13/2022] Open
Abstract
Inflammation in now appreciated to be at the centre of may diseases that affect Western civilization. Current therapeutics for managing these conditions may interfere with the host response leading to immune suppression. We recently developed an annexin (Anx) A1-derived peptide, coined CR-AnxA12-50, which displays potent pro-resolving and tissue protective actions. Herein, we designed a novel peptide using CR-AnxA12-50 as a template that was significantly more resistant to neutrophil-mediated degradation. This peptide, termed CR-AnxA12-48, retained high affinity and specificity to the pro-resolving Lipoxin A4 receptor (ALX) with an IC50 of ~20nM. CR-AnxA12-48 dose dependently (100fM-10nM) promoted the efferocytosis of apoptotic neutrophils, an action that was mediated by the murine orthologue of human ALX. The neutrophil-directed actions were also retained with human primary cells were CR-AnxA12-48 reduced human neutrophil recruitment to activated endothelial cells at concentrations as low as 100 pM. This protective action was mediated by human ALX, since incubation of neutrophils with an anti-ALX antibody reversed this anti-inflammatory actions of CR-AnxA12-48. Administration of this peptide to mice during dermal inflammation led to a significant and dose dependent decrease in neutrophil recruitment. This reduction in neutrophil numbers was more pronounced than that displayed by the parent peptide CR-AnxA12-50. CR-AnxA12-48 was also cardioprotecitve reducing infarct size and systemic chemokine (C-C motif) ligand 5 concentration following ischemia reperfusion injury. These findings identify CR-AnxA12-48 as a new ALX agonist that regulates phagocyte responses and displays tissue-protective actions.
Collapse
Affiliation(s)
- Mauro Perretti
- The William Harvey Research Institute, Barts and The London School of Medicine, Queen Mary University of London, Charterhouse Square, London, United Kingdom
| | - Clara Di Filippo
- Department of Experimental Medicine, Second University of Naples, Naples, Italy
| | - Michele D’Amico
- Department of Experimental Medicine, Second University of Naples, Naples, Italy
| | - Jesmond Dalli
- The William Harvey Research Institute, Barts and The London School of Medicine, Queen Mary University of London, Charterhouse Square, London, United Kingdom
| |
Collapse
|
49
|
Kourtzelis I, Mitroulis I, von Renesse J, Hajishengallis G, Chavakis T. From leukocyte recruitment to resolution of inflammation: the cardinal role of integrins. J Leukoc Biol 2017; 102:677-683. [PMID: 28292945 DOI: 10.1189/jlb.3mr0117-024r] [Citation(s) in RCA: 80] [Impact Index Per Article: 11.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2017] [Revised: 02/21/2017] [Accepted: 02/23/2017] [Indexed: 12/23/2022] Open
Abstract
Integrins constitute a large group of adhesion receptors that are formed as heterodimers of α and β subunits. Their presence and activation status on the surface of leukocytes modulate a broad spectrum of processes in inflammation and immunity. This mini review critically outlines research advances with regard to the function of leukocyte integrins in regulating and integrating the onset and resolution of acute inflammation. Specifically, we summarize and discuss relevant, current literature that supports the multifunctional role of integrins and their partners. The latter include molecules that physically associate with integrins or regulate their activity in the context of the following: 1) leukocyte recruitment to an inflamed tissue, 2) recognition and phagocytosis of apoptotic neutrophils (efferocytosis), and 3) egress of efferocytic macrophages from the inflamed site to lymphoid tissues. The understanding of the fine-tuning mechanisms of the aforementioned processes by integrins and their functional partners may enable the design of therapeutic tools to counteract destructive inflammation and promote more efficient resolution of inflammation.
Collapse
Affiliation(s)
- Ioannis Kourtzelis
- Institute for Clinical Chemistry and Laboratory Medicine, Faculty of Medicine, Technische Universität Dresden, Dresden, Germany; and
| | - Ioannis Mitroulis
- Institute for Clinical Chemistry and Laboratory Medicine, Faculty of Medicine, Technische Universität Dresden, Dresden, Germany; and
| | - Janusz von Renesse
- Institute for Clinical Chemistry and Laboratory Medicine, Faculty of Medicine, Technische Universität Dresden, Dresden, Germany; and
| | - George Hajishengallis
- Department of Microbiology, University of Pennsylvania, School of Dental Medicine, Philadelphia, Pennsylvania, USA
| | - Triantafyllos Chavakis
- Institute for Clinical Chemistry and Laboratory Medicine, Faculty of Medicine, Technische Universität Dresden, Dresden, Germany; and
| |
Collapse
|
50
|
Oliveira LG, Souza-Testasicca MC, Vago JP, Figueiredo AB, Canavaci AMC, Perucci LO, Ferreira TPT, Coelho EAF, Gonçalves DU, Rocha MOC, E Silva PMR, Ferreira CN, Queiroz-Junior C, Sousa LP, Fernandes AP. Annexin A1 Is Involved in the Resolution of Inflammatory Responses during Leishmania braziliensis Infection. THE JOURNAL OF IMMUNOLOGY 2017; 198:3227-3236. [PMID: 28289158 DOI: 10.4049/jimmunol.1602028] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/30/2016] [Accepted: 02/13/2017] [Indexed: 12/30/2022]
Abstract
Leishmaniases are diseases caused by several Leishmania species. Leishmania (Viannia) braziliensis can cause localized cutaneous leishmaniasis (LCL), which heals spontaneously, or mucosal leishmaniasis (ML), characterized by chronic and intense inflammation and scanty parasitism. Annexin A1 (AnxA1) is a protein involved in modulation and resolution of inflammation through multiple mechanisms. In the present study, the role of AnxA1 was investigated in L. braziliensis-infected BALB/c mice. AnxA1 levels increased at the peak of tissue lesion and parasitism in infected mice. AnxA1 increased also after L. braziliensis infection of BALB/c (wild-type [WT]) bone marrow derived macrophages. Despite a lower parasite intake, parasite burden in bone marrow-derived macrophages from AnxA1-/- mice was similar to WT and associated with an early increase of TNF-α and, later, of IL-10. AnxA1-/- mice controlled tissue parasitism similarly to WT animals, but they developed significantly larger lesions at later stages of infection, with a more pronounced inflammatory infiltrate and increased specific production of IFN-γ, IL-4, and IL-10. AnxA1-/- mice also presented higher phosphorylation levels of ERK-1/2 and p65/RelA (NF-κB) and inducible NO synthase expression, suggesting that AnxA1 may be involved in modulation of inflammation in this model of experimental leishmaniasis. Finally, assessment of AnxA1 levels in sera from patients with LCL or ML revealed that ML patients had higher levels of serum AnxA1 than did LCL patients or control subjects. Collectively, these data indicate that AnxA1 is actively expressed during L. braziliensis infection. In the absence of AnxA1, mice are fully able to control parasite replication, but they present more intense inflammatory responses and delayed ability to resolve their lesion size.
Collapse
Affiliation(s)
- Leandro G Oliveira
- Departamento de Análises Clínicas e Toxicológicas, Faculdade de Farmácia, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais 31270-901, Brazil
| | - Míriam C Souza-Testasicca
- Departamento de Análises Clínicas e Toxicológicas, Faculdade de Farmácia, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais 31270-901, Brazil
| | - Juliana P Vago
- Departamento de Análises Clínicas e Toxicológicas, Faculdade de Farmácia, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais 31270-901, Brazil.,Departamento de Morfologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais 31270-901 Brazil
| | - Amanda Braga Figueiredo
- Laboratório de Imunoparasitologia, Núcleo de Pesquisas em Ciências Biológicas, Universidade Federal de Ouro Preto, Ouro Preto, Minas Gerais 35400-000, Brazil
| | - Adriana M C Canavaci
- Departamento de Análises Clínicas e Toxicológicas, Faculdade de Farmácia, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais 31270-901, Brazil
| | - Luiza Oliveira Perucci
- Departamento de Análises Clínicas e Toxicológicas, Faculdade de Farmácia, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais 31270-901, Brazil
| | | | - Eduardo A F Coelho
- Programa de Pós-Graduação em Ciências Saúde: Infectologia e Medicina Tropical, Faculdade de Medicina, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais 31270-901, Brazil; and
| | - Denise Utsch Gonçalves
- Programa de Pós-Graduação em Ciências Saúde: Infectologia e Medicina Tropical, Faculdade de Medicina, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais 31270-901, Brazil; and
| | - Manoel Otávio C Rocha
- Programa de Pós-Graduação em Ciências Saúde: Infectologia e Medicina Tropical, Faculdade de Medicina, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais 31270-901, Brazil; and
| | - Patrícia M R E Silva
- Laboratório de Inflamação, Instituto Oswaldo Cruz/FIOCRUZ, Rio de Janeiro 21040-360, Brazil
| | - Cláudia N Ferreira
- Setor de Patologia Clínica, Colégio Técnico, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais 31270-901 Brazil
| | - Celso Queiroz-Junior
- Departamento de Morfologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais 31270-901 Brazil
| | - Lirlândia P Sousa
- Departamento de Análises Clínicas e Toxicológicas, Faculdade de Farmácia, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais 31270-901, Brazil.,Departamento de Morfologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais 31270-901 Brazil
| | - Ana Paula Fernandes
- Departamento de Análises Clínicas e Toxicológicas, Faculdade de Farmácia, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais 31270-901, Brazil;
| |
Collapse
|