1
|
Mao X, Shi M, Zhang B, Fu R, Cai M, Yu S, Lin K, Zhang C, Li D, Chen G, Luo W. Integration of single-cell and bulk RNA sequencing revealed immune heterogeneity and its association with disease activity in rheumatoid arthritis patients. Immunol Res 2024:10.1007/s12026-024-09513-5. [PMID: 39009881 DOI: 10.1007/s12026-024-09513-5] [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: 05/22/2024] [Accepted: 06/27/2024] [Indexed: 07/17/2024]
Abstract
Rheumatoid arthritis (RA) is a chronic, inflammatory, systemic autoimmune disease characterized by cartilage, bone damage, synovial inflammation, hyperplasia, autoantibody production, and systemic features. To obtain an overall profile of the immune environment in RA patients and its association with clinical features, we performed single-cell transcriptome and T-cell receptor sequencing of mononuclear cells from peripheral blood (PBMC) and synovial fluid (SF) from RA patients, integrated with two large cohorts with bulk RNA sequencing for further validation and investigation. Dendritic cells (DCs) exhibited relatively high functional heterogeneity and tissue specificity in relation to both antigen presentation and proinflammatory functions. Peripheral helper T cells (TPHs) are likely to originate from synovial tissue, undergo activation and exhaustion, and are subsequently released into the peripheral blood. Notably, among all immune cell types, TPHs were found to have the most intense associations with disease activity. In addition, CD8 effector T cells could be clustered into two groups with different cytokine expressions and play distinct roles in RA development. By integrating single-cell data with bulk sequencing from two large cohorts, we identified interactions among TPHs, CD8 cells, CD16 monocytes, and DCs that strongly contribute to the proinflammatory local environment in RA joints. Of note, the swollen 28-joint counts exhibited a more pronounced association with this immune environment compared to other disease activity indexes. The immune environment alternated significantly from PBMCs to SF, which indicated that a series of immune cells was involved in proinflammatory responses in the local joints of RA patients. By integrating single-cell data with two large cohorts, we have uncovered associations between specific immune cell populations and clinical features. This integration provides a rapid and precise methodology for assessing local immune activation, offering valuable insights into the pathophysiological mechanisms at play in RA.
Collapse
Affiliation(s)
- Xiaofan Mao
- Institute of Translational Medicine, The First People's Hospital of Foshan, Foshan, China
| | - Maohua Shi
- Department of Rheumatology, The First People's Hospital of Foshan, Foshan, China
| | - Beiying Zhang
- Institute of Translational Medicine, The First People's Hospital of Foshan, Foshan, China
| | - Rongdang Fu
- Department of Hepatology, The First People's Hospital of Foshan, Foshan, China
| | - Mengyun Cai
- Institute of Translational Medicine, The First People's Hospital of Foshan, Foshan, China
| | - Sifei Yu
- Institute of Translational Medicine, The First People's Hospital of Foshan, Foshan, China
| | - Kairong Lin
- Institute of Translational Medicine, The First People's Hospital of Foshan, Foshan, China
| | - Chuling Zhang
- Institute of Translational Medicine, The First People's Hospital of Foshan, Foshan, China
| | - Dingru Li
- School of Biology and Biological Engineering, South China University of Technology, Guangzhou, China
| | - Guoqiang Chen
- Department of Rheumatology, The First People's Hospital of Foshan, Foshan, China.
| | - Wei Luo
- Institute of Translational Medicine, The First People's Hospital of Foshan, Foshan, China.
| |
Collapse
|
2
|
Lucchi DBM, Sasso GRS, Sena LS, Franco PC, Lice I, Borges FT, Oliani SM, Gil CD. Protective effects of annexin A1-derived peptide Ac 2-26 on liver and kidney injuries induced by cisplatin in rats. Life Sci 2022; 304:120677. [PMID: 35654117 DOI: 10.1016/j.lfs.2022.120677] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2022] [Revised: 05/14/2022] [Accepted: 05/27/2022] [Indexed: 10/18/2022]
Abstract
AIMS In this study we evaluated the effect of pharmacological treatment with annexin A1-derived peptide Ac2-26 in an experimental model of toxicity induced by cisplatin. MAIN METHODS Male rats were divided into Sham (control), Cisplatin (received intraperitoneal injections of 10 mg/kg/day of cisplatin for 3 days) and Ac2-26 (received intraperitoneal injections of 1 mg/kg/day of peptide, 15 min before cisplatin) groups. KEY FINDINGS After 6 h of the last dose of cisplatin, an acute inflammatory response was observed characterized by a marked increase in the number of neutrophils and GM-CSF, IL-1β, IL-6, IL-10 and TNF-α plasma levels. Treatment with Ac2-26 produced higher levels of GM-CSF, corroborating the high numbers of neutrophils, and the anti-inflammatory cytokine IL-4. Ac2-26 preserved the morphology of liver structures, preventing the damage caused by cisplatin, but did not reduce plasma levels of the hepatotoxicity biomarkers ARG1, GSTα and SDH. In the kidneys, the peptide maintained the markers of kidney damage CLU and KIM-1 at similar levels to the Sham group but did not avoid morphological changes caused by cisplatin. These effects of Ac2-26 were associated with the reduction of Fpr1 and Fpr2 levels in the organs studied. SIGNIFICANCE Pharmacological treatment with peptide Ac2-26 partially protects the liver and kidneys against the deleterious effects caused by cisplatin in this experimental model.
Collapse
Affiliation(s)
- Danilo B M Lucchi
- Department of Morphology and Genetics, Universidade Federal de São Paulo (UNIFESP), Sao Paulo, SP 04023-900, Brazil
| | - Gisela R S Sasso
- Department of Morphology and Genetics, Universidade Federal de São Paulo (UNIFESP), Sao Paulo, SP 04023-900, Brazil
| | - Letícia S Sena
- Department of Morphology and Genetics, Universidade Federal de São Paulo (UNIFESP), Sao Paulo, SP 04023-900, Brazil
| | - Paulo C Franco
- Department of Morphology and Genetics, Universidade Federal de São Paulo (UNIFESP), Sao Paulo, SP 04023-900, Brazil
| | - Izabella Lice
- Department of Morphology and Genetics, Universidade Federal de São Paulo (UNIFESP), Sao Paulo, SP 04023-900, Brazil
| | - Fernanda T Borges
- Department of Medicine, Nephology Division, Universidade Federal de São Paulo (UNIFESP), Sao Paulo, SP 04038-901, Brazil
| | - Sonia M Oliani
- Biosciences Graduate Program, Institute of Biosciences, Letters and Exact Sciences, Universidade Estadual Paulista (UNESP), Sao Jose do Rio Preto, SP 15054-000, Brazil; Advanced Research Center in Medicine (CEPAM) Unilago, São José do Rio Preto, SP 15030-070, Brazil
| | - Cristiane D Gil
- Department of Morphology and Genetics, Universidade Federal de São Paulo (UNIFESP), Sao Paulo, SP 04023-900, Brazil; Biosciences Graduate Program, Institute of Biosciences, Letters and Exact Sciences, Universidade Estadual Paulista (UNESP), Sao Jose do Rio Preto, SP 15054-000, Brazil.
| |
Collapse
|
3
|
Pharmacological treatment with annexin A1-derived peptide protects against cisplatin-induced hearing loss. Toxicol Lett 2022; 363:27-35. [PMID: 35561849 DOI: 10.1016/j.toxlet.2022.05.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2021] [Revised: 04/11/2022] [Accepted: 05/02/2022] [Indexed: 11/20/2022]
Abstract
Cisplatin is an antineoplastic agent widely used, and no effective treatments capable of preventing cisplatin-induced ototoxicity and neurotoxicity in humans have yet been identified. This study evaluated the effect of the anti-inflammatory annexin A1 (AnxA1)-derived peptide Ac2-26 in a cisplatin-induced ototoxicity model. Wistar rats received intraperitoneal injections of cisplatin (10mg/kg/day) for 3 days to induce hearing loss, and Ac2-26 (1mg/kg) was administered 15minutes before cisplatin administration. Control animals received an equal volume of saline. Hearing thresholds were measured by distortion product otoacoustic emissions (DPOAE) before and after treatments. Pharmacological treatment with Ac2-26 protected against cisplatin-induced hearing loss, as evidenced by DPOAE results showing similar signal-noise ratios between the control and Ac2-26-treated groups. These otoprotective effects of Ac2-26 were associated with an increased number of ganglion neurons compared with the untreated cisplatin group. Additionally, Ac2-26 treatment produced reduced immunoreactivity on cleaved caspase 3 and phosphorylated ERK levels in the ganglion neurons, compared to the untreated group, supporting the neuroprotective effects of the Ac2-26. Our results suggest that Ac2-26 has a substantial otoprotective effect in this cisplatin-induced ototoxicity model mediated by neuroprotection and the regulation of the ERK pathway.
Collapse
|
4
|
Li YZ, Wang YY, Huang L, Zhao YY, Chen LH, Zhang C. Annexin A Protein Family in Atherosclerosis. Clin Chim Acta 2022; 531:406-417. [PMID: 35562096 DOI: 10.1016/j.cca.2022.05.009] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2022] [Revised: 05/06/2022] [Accepted: 05/06/2022] [Indexed: 12/25/2022]
Abstract
Atherosclerosis, a silent chronic vascular pathology, is the cause of the majority of cardiovascular ischaemic events. Atherosclerosis is characterized by a series of deleterious changes in cellularity, including endothelial dysfunction, transmigration of circulating inflammatory cells into the arterial wall, pro-inflammatory cytokines production, lipid accumulation in the intima, vascular local inflammatory response, atherosclerosis-related cells apoptosis and autophagy. Proteins of Annexin A (AnxA) family, the well-known Ca2+ phospholipid-binding protein, have many functions in regulating inflammation-related enzymes and cell signaling transduction, thus influencing cell adhesion, migration, differentiation, proliferation and apoptosis. There is now accumulating evidence that some members of the AnxA family, such as AnxA1, AnxA2, AnxA5 and AnxA7, play major roles in the development of atherosclerosis. This article discusses the major roles of AnxA1, AnxA2, AnxA5 and AnxA7, and the multifaceted mechanisms of the main biological process in which they are involved in atherosclerosis. Considering these evidences, it has been proposed that AnxA are drivers- and not merely participator- on the road to atherosclerosis, thus the progression of atherosclerosis may be prevented by targeting the expression or function of the AnxA family proteins.
Collapse
Affiliation(s)
- Yong-Zhen Li
- Institute of Cardiovascular Disease, Key Lab for Arteriosclerology of Hunan province, Hengyang Medical College, University of South China, Hengyang, Hunan 421001, People's Republic of China
| | - Yan-Yue Wang
- Institute of Cardiovascular Disease, Key Lab for Arteriosclerology of Hunan province, Hengyang Medical College, University of South China, Hengyang, Hunan 421001, People's Republic of China
| | - Liang Huang
- Research Laboratory of Translational Medicine, Hengyang Medical College, University of South China, Hengyang, Hunan 421001, People's Republic of China
| | - Yu-Yan Zhao
- Institute of Cardiovascular Disease, Key Lab for Arteriosclerology of Hunan province, Hengyang Medical College, University of South China, Hengyang, Hunan 421001, People's Republic of China
| | - Lin-Hui Chen
- Institute of Cardiovascular Disease, Key Lab for Arteriosclerology of Hunan province, Hengyang Medical College, University of South China, Hengyang, Hunan 421001, People's Republic of China
| | - Chi Zhang
- Institute of Cardiovascular Disease, Key Lab for Arteriosclerology of Hunan province, Hengyang Medical College, University of South China, Hengyang, Hunan 421001, People's Republic of China.
| |
Collapse
|
5
|
Liver ischaemia-reperfusion injury: a new understanding of the role of innate immunity. Nat Rev Gastroenterol Hepatol 2022; 19:239-256. [PMID: 34837066 DOI: 10.1038/s41575-021-00549-8] [Citation(s) in RCA: 143] [Impact Index Per Article: 71.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 10/29/2021] [Indexed: 02/08/2023]
Abstract
Liver ischaemia-reperfusion injury (LIRI), a local sterile inflammatory response driven by innate immunity, is one of the primary causes of early organ dysfunction and failure after liver transplantation. Cellular damage resulting from LIRI is an important risk factor not only for graft dysfunction but also for acute and even chronic rejection and exacerbates the shortage of donor organs for life-saving liver transplantation. Hepatocytes, liver sinusoidal endothelial cells and Kupffer cells, along with extrahepatic monocyte-derived macrophages, neutrophils and platelets, are all involved in LIRI. However, the mechanisms underlying the responses of these cells in the acute phase of LIRI and how these responses are orchestrated to control and resolve inflammation and achieve homeostatic tissue repair are not well understood. Technological advances allow the tracking of cells to better appreciate the role of hepatic macrophages and platelets (such as their origin and immunomodulatory and tissue-remodelling functions) and hepatic neutrophils (such as their selective recruitment, anti-inflammatory and tissue-repairing functions, and formation of extracellular traps and reverse migration) in LIRI. In this Review, we summarize the role of macrophages, platelets and neutrophils in LIRI, highlight unanswered questions, and discuss prospects for innovative therapeutic regimens against LIRI in transplant recipients.
Collapse
|
6
|
Oliveira MP, Prates J, Gimenes AD, Correa SG, Oliani SM. Annexin A1 Mimetic Peptide Ac 2-26 Modulates the Function of Murine Colonic and Human Mast Cells. Front Immunol 2021; 12:689484. [PMID: 34557187 PMCID: PMC8452975 DOI: 10.3389/fimmu.2021.689484] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2021] [Accepted: 08/19/2021] [Indexed: 12/17/2022] Open
Abstract
Mast cells (MCs) are main effector cells in allergic inflammation and after activation, they release stored (histamine, heparin, proteases) and newly synthesized (lipid mediators and cytokines) substances. In the gastrointestinal tract the largest MC population is located in the lamina propria and submucosa whereas several signals such as the cytokine IL-4, seem to increase the granule content and to stimulate a remarkable expansion of intestinal MCs. The broad range of MC-derived bioactive molecules may explain their involvement in many different allergic disorders of the gastrointestinal tract. Annexin A1 (AnxA1) is a 37 KDa glucocorticoid induced monomeric protein selectively distributed in certain tissues. Its activity can be reproduced by mimetic peptides of the N-terminal portion, such as Ac2-26, that share the same receptor FPR-L1. Although previous reports demonstrated that AnxA1 inhibits MC degranulation in murine models, the effects of exogenous peptide Ac2-26 on intestinal MCs or the biological functions of the Ac2-26/FPR2 system in human MCs have been poorly studied. To determine the effects of Ac2-26 on the function of MCs toward the possibility of AnxA1-based therapeutics, we treated WT and IL-4 knockout mice with peptide Ac2-26, and we examined the spontaneous and compound 48/80 stimulated colonic MC degranulation and cytokine production. Moreover, in vitro, using human mast cell line HMC-1 we demonstrated that exogenous AnxA1 peptide is capable of interfering with the HMC-1 degranulation in a direct pathway through formyl peptide receptors (FPRs). We envisage that our results can provide therapeutic strategies to reduce the release of MC mediators in inflammatory allergic processes.
Collapse
Affiliation(s)
- Marcia Pereira Oliveira
- Laboratory of Interdisciplinary Medical Research, Oswaldo Cruz Institute, Fiocruz, Rio de Janeiro, Brazil
| | - Janesly Prates
- Department of Biology, Institute of Bioscience, Humanities and Exact Science, São Paulo State University (Unesp), São José do Rio Preto, Brazil
| | | | - Silvia Graciela Correa
- Departamento de Bioquímica Clinica-Centro de Investigaciones en Bioquímica Clínica e Inmunología (CIBICI - CONICET) - Facultad de Ciencias Quimicas- Universidad Nacional de Córdoba (UNC), Córdoba, Argentina
| | - Sonia Maria Oliani
- Department of Biology, Institute of Bioscience, Humanities and Exact Science, São Paulo State University (Unesp), São José do Rio Preto, Brazil
- Advanced Research Center in Medicine, CEPAM –Unilago, São José do Rio Preto, Brazil
- Federal University of São Paulo, Post Graduate Program in Structural and Functional Biology, Escola Paulista de Medicina (Unifesp-EPM), São Paulo, Brazil
| |
Collapse
|
7
|
Zhang Y, Ma S, Ke X, Yi Y, Yu H, Yu D, Li Q, Shang Y, Lu Y, Pei L. The mechanism of Annexin A1 to modulate TRPV1 and nociception in dorsal root ganglion neurons. Cell Biosci 2021; 11:167. [PMID: 34446102 PMCID: PMC8393810 DOI: 10.1186/s13578-021-00679-1] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2021] [Accepted: 08/15/2021] [Indexed: 12/15/2022] Open
Abstract
BACKGROUND Annexin A1 (ANXA1) exerts anti-nociceptive effect through ANXA1 receptor formyl peptide receptor 2 (FPR2/ALX (receptor for lipoxin A4), FPR2) at the dorsal root ganglia (DRG) level. However, the mechanisms remain elucidated. By using radiant heat, hot/cold plate, tail flick, von Frey, and Randall-Selitto tests to detect nociception in intact and chemical (capsaicin, menthol, mustard oil, formalin or CFA) injected AnxA1 conditional knockout (AnxA1-/-) mice, applying calcium imaging and patch clamp recordings in cultured DRG neurons to measure neuronal excitability, conducting immunofluorescence and western blotting to detect the protein levels of TRPV1, FPR2 and its downstream molecules, and performing double immunofluorescence and co-immunoprecipitation to investigate the interaction between Calmodulin (CaM) and TRPV1; we aim to uncover the molecular and cellular mechanisms of ANXA1's role in antinociception. RESULTS AnxA1-/- mice exhibited significant sensitivity to noxious heat (mean ± SD, 6.2 ± 1.0 s vs. 9.9 ± 1.6 s in Hargreaves test; 13.6 ± 1.5 s vs. 19.0 ± 1.9 s in hot plate test; n = 8; P < 0.001), capsaicin (101.0 ± 15.3 vs. 76.2 ± 10.9; n = 8; P < 0.01), formalin (early phase: 169.5 ± 32.8 s vs. 76.0 ± 21.9 s; n = 8; P < 0.05; late phase: 444.6 ± 40.1 s vs. 320.4 ± 33.6 s; n = 8; P < 0.01) and CFA (3.5 ± 0.8 s vs. 5.9 ± 1.4 s; n = 8; P < 0.01). In addition, we found significantly increased capsaicin induced Ca2+ response, TRPV1 currents and neuronal firing in AnxA1 deficient DRG neurons. Furthermore, ANXA1 mimic peptide Ac2-26 robustly increased intracellular Ca2+, inhibited TRPV1 current, activated PLCβ and promoted CaM-TRPV1 interaction. And these effects of Ac2-26 could be attenuated by FPR2 antagonist Boc2. CONCLUSIONS Selective deletion of AnxA1 in DRG neurons enhances TRPV1 sensitivity and deteriorates noxious heat or capsaicin induced nociception, while ANXA1 mimic peptide Ac2-26 desensitizes TRPV1 via FPR2 and the downstream PLCβ-Ca2+-CaM signal. This study may provide possible target for developing new analgesic drugs in inflammatory pain.
Collapse
Affiliation(s)
- Yufen Zhang
- Department of Neurobiology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China.,Collaborative Innovation Center for Brain Science, The Institute for Brain Research (IBR), Huazhong University of Science and Technology, Wuhan, 430030, China.,Department of Anesthesiology, School of Medicine, Washington University in Saint Loius, St. Loius, MO, 63110, USA
| | - Sehui Ma
- Department of Neurobiology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China.,Collaborative Innovation Center for Brain Science, The Institute for Brain Research (IBR), Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Xiao Ke
- Department of Neurobiology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China.,Collaborative Innovation Center for Brain Science, The Institute for Brain Research (IBR), Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Yao Yi
- Department of Neurobiology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China.,Collaborative Innovation Center for Brain Science, The Institute for Brain Research (IBR), Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Hongyan Yu
- Department of Neurobiology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China.,Collaborative Innovation Center for Brain Science, The Institute for Brain Research (IBR), Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Dian Yu
- Department of Neurobiology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China.,Collaborative Innovation Center for Brain Science, The Institute for Brain Research (IBR), Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Qiang Li
- Exchange, Development & Service Center for Science & Technology Talents, The Ministry of Science and Technology (Most), Beijing, 100045, China
| | - You Shang
- Collaborative Innovation Center for Brain Science, The Institute for Brain Research (IBR), Huazhong University of Science and Technology, Wuhan, 430030, China.,Department of Critical Care Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Youming Lu
- Collaborative Innovation Center for Brain Science, The Institute for Brain Research (IBR), Huazhong University of Science and Technology, Wuhan, 430030, China.,Department of Physiology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Lei Pei
- Department of Neurobiology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China. .,Collaborative Innovation Center for Brain Science, The Institute for Brain Research (IBR), Huazhong University of Science and Technology, Wuhan, 430030, China. .,Department of Anesthesiology, School of Medicine, Washington University in Saint Loius, St. Loius, MO, 63110, USA.
| |
Collapse
|
8
|
Yang A, Wu Y, Yu G, Wang H. Role of specialized pro-resolving lipid mediators in pulmonary inflammation diseases: mechanisms and development. Respir Res 2021; 22:204. [PMID: 34261470 PMCID: PMC8279385 DOI: 10.1186/s12931-021-01792-y] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2020] [Accepted: 06/30/2021] [Indexed: 12/12/2022] Open
Abstract
Inflammation is an essential mechanism of various diseases. The development and resolution of inflammation are complex immune-modulation processes which induce the involvement of various types of immune cells. Specialized pro-resolving lipid mediators (SPMs) have been demonstrated to be signaling molecules in inflammation. SPMs are involved in the pathophysiology of different diseases, especially respiratory diseases, including asthma, pneumonia, and chronic obstructive pulmonary disease. All of these diseases are related to the inflammatory response and its persistence. Therefore, a deeper understanding of the mechanisms and development of inflammation in respiratory disease, and the roles of the SPM family in the resolution process, might be useful in the quest for novel therapies and preventive measures for pulmonary diseases.
Collapse
Affiliation(s)
- Ailin Yang
- Department of Respiratory Medicine, Beijing Friendship Hospital, Capital Medical University, No. 95 Yong An Road, Xicheng, , Beijing, 100050, China
| | - Yanjun Wu
- Department of Respiratory Medicine, Beijing Friendship Hospital, Capital Medical University, No. 95 Yong An Road, Xicheng, , Beijing, 100050, China
| | - Ganggang Yu
- Department of Respiratory Medicine, Beijing Friendship Hospital, Capital Medical University, No. 95 Yong An Road, Xicheng, , Beijing, 100050, China.
| | - Haoyan Wang
- Department of Respiratory Medicine, Beijing Friendship Hospital, Capital Medical University, No. 95 Yong An Road, Xicheng, , Beijing, 100050, China.
| |
Collapse
|
9
|
Yang M, Bair JA, Hodges RR, Serhan CN, Dartt DA. Resolvin E1 Reduces Leukotriene B4-Induced Intracellular Calcium Increase and Mucin Secretion in Rat Conjunctival Goblet Cells. THE AMERICAN JOURNAL OF PATHOLOGY 2020; 190:1823-1832. [PMID: 32561135 DOI: 10.1016/j.ajpath.2020.06.001] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/12/2020] [Revised: 06/01/2020] [Accepted: 06/02/2020] [Indexed: 01/25/2023]
Abstract
Leukotriene B4 (LTB4) is a major proinflammatory mediator important in host defense, whereas resolvins (Rvs) are produced during the resolution phase of inflammation. The authors determined the actions of both RvE1 and RvD1 on LTB4-induced responses of goblet cells cultured from rat conjunctiva. The responses measured were an increase in the intracellular [Ca2+] ([Ca2+]i) and high-molecular-weight glycoprotein secretion. Treatment with RvE1 or RvD1 for 30 minutes significantly blocked the LTB4-induced [Ca2+]i increase. The actions of RvE1 on LTB4-induced [Ca2+]i increase were reversed by siRNA for the RvE1 receptor, and the actions of RvD1 were reversed by an RvD1 receptor inhibitor. The RvE1 and RvD1 block of LTB4-stimulated increase in [Ca2+]i was also reversed by an inhibitory peptide to β-adrenergic receptor kinase. LTB4 and block of the LTB4-stimulated increase in [Ca2+]i by RvE1 and RvD1 were partially mediated by the depletion of intracellular Ca2+ stores. RvE1, but not RvD1, counterregulated the LTB4-induced high-molecular-weight glycoprotein secretion. Thus, both RvE1 and RvD1 receptors directly inhibit LTB4 by phosphorylating the LTB4 receptor using β adrenergic receptor kinase. RvE1 receptor counterregulates the LTB4-induced increase in [Ca2+]i and secretion, whereas RvD1 receptor only counterregulates LTB4-induced [Ca2+]i increase.
Collapse
Affiliation(s)
- Menglu Yang
- Schepens Eye Research Institute, Massachusetts Eye and Ear Infirmary, Department of Ophthalmology, Harvard Medical School, Boston, Massachusetts
| | - Jeffrey A Bair
- Schepens Eye Research Institute, Massachusetts Eye and Ear Infirmary, Department of Ophthalmology, Harvard Medical School, Boston, Massachusetts
| | - Robin R Hodges
- Schepens Eye Research Institute, Massachusetts Eye and Ear Infirmary, Department of Ophthalmology, Harvard Medical School, Boston, Massachusetts
| | - Charles N Serhan
- Center for Experimental Therapeutics and Reperfusion Injury, Brigham and Women's Hospital and Department of Anaesthesia, Perioperative and Pain Medicine, Harvard Medical School, Boston, Massachusetts
| | - Darlene A Dartt
- Schepens Eye Research Institute, Massachusetts Eye and Ear Infirmary, Department of Ophthalmology, Harvard Medical School, Boston, Massachusetts.
| |
Collapse
|
10
|
The involvement of annexin A1 in human placental response to maternal Zika virus infection. Antiviral Res 2020; 179:104809. [PMID: 32360947 DOI: 10.1016/j.antiviral.2020.104809] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2019] [Revised: 04/02/2020] [Accepted: 04/25/2020] [Indexed: 12/22/2022]
Abstract
The association of Zika virus infection (ZIKV) with congenital malformation and neurological sequelae brought a significant global concern. Recent studies have shown that maternal viral infection leads to inflammation in the placental tissue. In this context, the antiinflammatory protein annexin 1 (ANXA1) has a major determination of the resolution of inflammation and it has been positively associated with antiparasitic activity in infected placental explants. Although these effects have been explored to some degree, ANXA1 expression and potential properties have not yet been fully elucidated in placentas infected with ZIKV. This study was conducted to evaluate the histopathology, inflammatory process and elucidate if ANXA1 were differently expressed in placentas of ZIKV-infected mothers. Three classification groups were used in this study: Neg/Neg (mother and placenta negative for the virus), Pos/Neg (infected mother, but no virus detected in placenta) and Pos/Pos (mother and placenta infected with ZIKV). ANXA1 was expressed in syncytiotrophoblast cells of all studied groups, and its expression was decreased in Pos/Neg group, which displayed also an increase of the inflammatory response, as evinced from the recruitment of inflammatory cells, increased levels of placenta cytokines, and evidence of impaired tissue repair. The presence of ZIKV in placentas of Pos/Pos group shows structural alterations, including detachment and disorganization of the trophoblastic epithelium. In summary, our results suggest that maternal infection with ZIKV, even without direct tissue infection, leads to a placental inflammatory response probably related to the modulation of ANXA1. After placental infection, structural changes - including inflammatory cells influx - are observed leading to placental dysfunction and reduced fetal weight. Our study sheds additional light on the outcomes of ZIKV infection in trophoblast and reveals a potential involvement of ANXA1 in the placental biology.
Collapse
|
11
|
Annexin A1 peptide is able to induce an anti-parasitic effect in human placental explants infected by Toxoplasma gondii. Microb Pathog 2018; 123:153-161. [PMID: 30003946 DOI: 10.1016/j.micpath.2018.07.005] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2018] [Revised: 06/18/2018] [Accepted: 07/08/2018] [Indexed: 12/31/2022]
Abstract
This study was conducted to investigate annexin A1 (ANXA1) functions in human placental explants infected with Toxoplasma gondii (T. gondii). We examined the first and third trimester placental explants infected with T. gondii (n = 7 placentas/group) to identify the number and location of parasites, ANXA1 protein, potential involvement of formyl peptide receptors (FPR1 and FPR2), and COX-2 expressions by immunohistochemistry. Treatments with Ac2-26 mimetic peptide of ANXA1 were performed to verify the parasitism rate (β-galactosidase assay), prostaglandin E2 levels (ELISA assay), and ANXA1, FPR1 and COX-2 expression in third trimester placentas. Placental explants of third trimester expressed less ANXA1 and were more permissive to T. gondii infection than first trimester placentas that expressed more ANXA1. Ac2-26 treatment increases endogenous ANXA1 and decreases parasitism rate, COX-2, and prostaglandin E2 levels. Altogether, these data provide further insight into the anti-parasitic and anti-inflammatory effects of ANXA1 in placentas infected with T. gondii.
Collapse
|
12
|
Inhibition of the AnxA1/FPR1 autocrine axis reduces MDA-MB-231 breast cancer cell growth and aggressiveness in vitro and in vivo. BIOCHIMICA ET BIOPHYSICA ACTA-MOLECULAR CELL RESEARCH 2018; 1865:1368-1382. [PMID: 29932988 DOI: 10.1016/j.bbamcr.2018.06.010] [Citation(s) in RCA: 32] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/30/2017] [Revised: 06/14/2018] [Accepted: 06/17/2018] [Indexed: 12/18/2022]
Abstract
Breast Cancer (BC) is a highly heterogeneous disease whose most aggressive behavior is displayed by triple-negative breast cancer (TNBC), which lacks an efficient targeted therapy. Despite its controversial role, one of the proteins that having been linked with BC is Annexin A1 (AnxA1), which is a Ca+2 binding protein that acts modulating the immune system, cell membrane organization and vesicular trafficking. In this work we analyzed tissue microarrays of BC samples and observed a higher expression of AnxA1 in TNBCs and in lymph node metastasis. We also observed a positive correlation in primary tumors between expression levels of AnxA1 and its receptor, FPR1. Despite displaying a lesser strength, this correlation also exists in BC lymph node metastasis. In agreement, we have found that AnxA1 was highly expressed and secreted in the TNBC cell line MDA-MB-231 that also expressed high levels of FPR1. Furthermore, we demonstrated, by using the specific FPR1 inhibitor Cyclosporin H (CsH) and the immunosuppressive drug Cyclosporin A (CsA), the existence of an autocrine signaling of AnxA1 through the FPR1. Such signaling, elicited by AnxA1 upon its secretion, increased the aggressiveness and survival of MDA-MB-231 cells. In this manner, we demonstrated that CsA works very efficiently as an FPR1 inhibitor. Finally, by using CsA, we demonstrated that FPR1 inhibition decreased MDA-MB-231 tumor growth and metastasis formation in nude mice. These results indicate that FPR1 inhibition could be a potential intervention strategy to manage TNBCs displaying the characteristics of MDA-MB-231 cells. FPR1 inhibition can be efficiently achieved by CsA.
Collapse
|
13
|
Pietrani NT, Ferreira CN, Rodrigues KF, Perucci LO, Carneiro FS, Bosco AA, Oliveira MC, Pereira SS, Teixeira AL, Alvarez-Leite JI, Ferreira AV, Sousa LP, Gomes KB. Proresolving protein Annexin A1: The role in type 2 diabetes mellitus and obesity. Biomed Pharmacother 2018; 103:482-489. [PMID: 29677533 DOI: 10.1016/j.biopha.2018.04.024] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2017] [Revised: 03/26/2018] [Accepted: 04/03/2018] [Indexed: 01/22/2023] Open
Abstract
BACKGROUND Annexin A1 (AnxA1) is a protein involved in inflammation resolution that might be altered in obesity-associated type 2 diabetes mellitus (DM), which is a chronic inflammatory disease. The aim of this study was to evaluate AnxA1 serum levels in individuals with and without DM stratified according to the body mass index (BMI), and the dynamic of AnxA1 expression in adipose tissue from humans with obesity and non-obesity. METHODS Serum samples were obtained from 41 patients with DM (lean, overweight and obese) and 40 controls, and adipose tissue samples were obtained from 16 individuals with obesity (with or without DM), and 15 controls. RESULTS DM patients showed similar AnxA1 serum levels when compared to controls. However, when the individuals were stratified according to BMI, AnxA1 levels were higher in individuals with obesity than lean or overweight, and in overweight compared to lean individuals. Moreover, AnxA1 was correlated positively with IL-6 levels. AnxA1 levels were also positively correlated with BMI, waist circumference and waist-to-hip ratio. Furthermore, higher levels of cleaved AnxA1 were observed in adipose tissue from individuals with obesity, independently of DM status. CONCLUSIONS Enhanced levels of AnxA1 in serum of individuals with obesity suggest an attempt to counter-regulate the systemic inflammation process in this disease. However, the higher levels of cleaved AnxA1 in the adipose tissue of individuals with obesity could compromise its anti-inflammatory and proresolving actions, locally. Considering our data, AnxA1 cleavage in the adipose tissue, despite increased serum levels of this protein, and consequently the failure in inflammation resolution, suggests an important pathophysiological mechanism involved in inflammatory status observed in obesity.
Collapse
Affiliation(s)
- Nathalia T Pietrani
- Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| | - Cláudia N Ferreira
- Colégio Técnico - COLTEC- Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| | - Kathryna F Rodrigues
- Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| | - Luiza O Perucci
- Faculdade de Farmácia, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| | - Fernanda S Carneiro
- Faculdade de Farmácia, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| | - Adriana A Bosco
- Santa Casa de Belo Horizonte, Belo Horizonte, Minas Gerais, Brazil
| | - Marina C Oliveira
- Escola de Enfermagem, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| | - Solange S Pereira
- Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| | - Antônio L Teixeira
- Faculdade de Medicina, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| | - Jacqueline I Alvarez-Leite
- Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| | - Adaliene V Ferreira
- Escola de Enfermagem, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| | - Lirlândia P Sousa
- Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brazil; Faculdade de Farmácia, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| | - Karina B Gomes
- Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brazil; Faculdade de Farmácia, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brazil.
| |
Collapse
|
14
|
Drago F, Lombardi M, Prada I, Gabrielli M, Joshi P, Cojoc D, Franck J, Fournier I, Vizioli J, Verderio C. ATP Modifies the Proteome of Extracellular Vesicles Released by Microglia and Influences Their Action on Astrocytes. Front Pharmacol 2017; 8:910. [PMID: 29321741 PMCID: PMC5733563 DOI: 10.3389/fphar.2017.00910] [Citation(s) in RCA: 109] [Impact Index Per Article: 15.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2017] [Accepted: 11/29/2017] [Indexed: 11/23/2022] Open
Abstract
Extracellular ATP is among molecules promoting microglia activation and inducing the release of extracellular vesicles (EVs), which are potent mediators of intercellular communication between microglia and the microenvironment. We previously showed that EVs produced under ATP stimulation (ATP-EVs) propagate a robust inflammatory reaction among astrocytes and microglia in vitro and in mice with subclinical neuroinflammation (Verderio et al., 2012). However, the proteome of EVs released upon ATP stimulation has not yet been elucidated. In this study we applied a label free proteomic approach to characterize the proteome of EVs released constitutively and during microglia activation with ATP. We show that ATP drives sorting in EVs of a set of proteins implicated in cell adhesion/extracellular matrix organization, autophagy-lysosomal pathway and cellular metabolism, that may influence the response of recipient astrocytes to EVs. These data provide new clues to molecular mechanisms involved in microglia response to ATP and in microglia signaling to the environment via EVs.
Collapse
Affiliation(s)
- Francesco Drago
- Univ. Lille, INSERM, U1192 - Protéomique Réponse Inflammatoire Spectrométrie de Masse - PRISM, Lille, France.,Fondazione Istituto Oncologico del Mediterraneo, Viagrande, Italy
| | | | | | | | - Pooja Joshi
- Institute of Neuroscience (CNR), Milan, Italy
| | - Dan Cojoc
- Institute of Materials (CNR), Trieste, Italy
| | - Julien Franck
- Univ. Lille, INSERM, U1192 - Protéomique Réponse Inflammatoire Spectrométrie de Masse - PRISM, Lille, France
| | - Isabelle Fournier
- Univ. Lille, INSERM, U1192 - Protéomique Réponse Inflammatoire Spectrométrie de Masse - PRISM, Lille, France
| | - Jacopo Vizioli
- Univ. Lille, INSERM, U1192 - Protéomique Réponse Inflammatoire Spectrométrie de Masse - PRISM, Lille, France
| | - Claudia Verderio
- IRCCS Humanitas, Rozzano, Italy.,Institute of Neuroscience (CNR), Milan, Italy
| |
Collapse
|
15
|
Molás RB, de Paula-Silva M, Masood R, Ullah A, Gimenes AD, Oliani SM. Ac2-26 peptide and serine protease of Bothrops atrox similarly induces angiogenesis without triggering local and systemic inflammation in a murine model of dorsal skinfold chamber. Toxicon 2017; 137:7-14. [DOI: 10.1016/j.toxicon.2017.06.009] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2017] [Revised: 06/12/2017] [Accepted: 06/13/2017] [Indexed: 11/26/2022]
|
16
|
ANXA1Ac2–26 peptide, a possible therapeutic approach in inflammatory ocular diseases. Gene 2017; 614:26-36. [DOI: 10.1016/j.gene.2017.02.032] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2016] [Revised: 02/14/2017] [Accepted: 02/27/2017] [Indexed: 02/08/2023]
|
17
|
Ricci E, Ronchetti S, Pericolini E, Gabrielli E, Cari L, Gentili M, Roselletti E, Migliorati G, Vecchiarelli A, Riccardi C. Role of the glucocorticoid-induced leucine zipper gene in dexamethasone-induced inhibition of mouse neutrophil migration via control of annexin A1 expression. FASEB J 2017; 31:3054-3065. [PMID: 28373208 DOI: 10.1096/fj.201601315r] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2016] [Accepted: 03/13/2017] [Indexed: 12/15/2022]
Abstract
The glucocorticoid-induced leucine zipper (GILZ) gene is a pivotal mediator of the anti-inflammatory effects of glucocorticoids (GCs) that are known to regulate the function of both adaptive and innate immunity cells. Our aim was to investigate the role of GILZ in GC-induced inhibition of neutrophil migration, as this role has not been investigated before. We found that GILZ expression was induced by dexamethasone (DEX), a synthetic GC, in neutrophils, and that it regulated migration of these cells into inflamed tissues under DEX treatment. Of note, inhibition of neutrophil migration was not observed in GILZ-knockout mice with peritonitis that were treated by DEX. This was because DEX was unable to up-regulate annexin A1 (Anxa1) expression in the absence of GILZ. Furthermore, we showed that GILZ mediates Anxa1 induction by GCs by transactivating Anxa1 expression at the promoter level via binding with the transcription factor, PU.1. The present findings shed light on the role of GILZ in the mechanism of induction of Anxa1 by GCs. As Anxa1 is an important protein for the resolution of inflammatory response, GILZ may represent a new pharmacologic target for treatment of inflammatory diseases.-Ricci, E., Ronchetti, S., Pericolini, E., Gabrielli, E., Cari, L., Gentili, M., Roselletti, E., Migliorati, G., Vecchiarelli, A., Riccardi, C. Role of the glucocorticoid-induced leucine zipper gene in dexamethasone-induced inhibition of mouse neutrophil migration via control of annexin A1 expression.
Collapse
Affiliation(s)
- Erika Ricci
- Pharmacology Section, Department of Medicine, University of Perugia, Perugia, Italy
| | - Simona Ronchetti
- Pharmacology Section, Department of Medicine, University of Perugia, Perugia, Italy
| | - Eva Pericolini
- Microbiology Section, Department of Experimental Medicine, University of Perugia, Perugia, Italy.,Department of Diagnostic, Clinic, and Public Health Medicine, University of Modena and Reggio Emilia, Modena, Italy
| | - Elena Gabrielli
- Microbiology Section, Department of Experimental Medicine, University of Perugia, Perugia, Italy
| | - Luigi Cari
- Pharmacology Section, Department of Medicine, University of Perugia, Perugia, Italy
| | - Marco Gentili
- Pharmacology Section, Department of Medicine, University of Perugia, Perugia, Italy
| | - Elena Roselletti
- Microbiology Section, Department of Experimental Medicine, University of Perugia, Perugia, Italy
| | - Graziella Migliorati
- Pharmacology Section, Department of Medicine, University of Perugia, Perugia, Italy
| | - Anna Vecchiarelli
- Microbiology Section, Department of Experimental Medicine, University of Perugia, Perugia, Italy
| | - Carlo Riccardi
- Pharmacology Section, Department of Medicine, University of Perugia, Perugia, Italy;
| |
Collapse
|
18
|
Tu Y, Johnstone CN, Stewart AG. Annexin A1 influences in breast cancer: Controversies on contributions to tumour, host and immunoediting processes. Pharmacol Res 2017; 119:278-288. [PMID: 28212890 DOI: 10.1016/j.phrs.2017.02.011] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/13/2016] [Revised: 02/08/2017] [Accepted: 02/08/2017] [Indexed: 12/20/2022]
Abstract
Annexin A1 is a multifunctional protein characterised by its actions in modulating the innate and adaptive immune response. Accumulating evidence of altered annexin A1 expression in many human tumours raises interest in its functional role in cancer biology. In breast cancer, altered annexin A1 expression levels suggest a potential influence on tumorigenic and metastatic processes. However, reports of conflicting results reveal a relationship that is much more complex than first conceptualised. In this review, we explore the diverse actions of annexin A1 on breast tumour cells and various host cell types, including stromal immune and structural cells, particularly in the context of cancer immunoediting.
Collapse
Affiliation(s)
- Yan Tu
- Department of Pharmacology and Therapeutics, The University of Melbourne, Parkville, Melbourne, Australia
| | - Cameron N Johnstone
- Cancer & Inflammation Laboratory, Olivia Newton-John Cancer Research Institute, Heidelberg, Australia
| | - Alastair G Stewart
- Department of Pharmacology and Therapeutics, The University of Melbourne, Parkville, Melbourne, Australia.
| |
Collapse
|
19
|
The Dual Role of Neutrophils in Inflammatory Bowel Diseases. J Clin Med 2016; 5:jcm5120118. [PMID: 27999328 PMCID: PMC5184791 DOI: 10.3390/jcm5120118] [Citation(s) in RCA: 192] [Impact Index Per Article: 24.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2016] [Revised: 12/06/2016] [Accepted: 12/13/2016] [Indexed: 12/21/2022] Open
Abstract
Inflammatory bowel diseases (IBD), including Crohn’s disease and ulcerative colitis, are characterised by aberrant immunological responses leading to chronic inflammation without tissue regeneration. These two diseases are considered distinct entities, and there is some evidence that neutrophil behaviour, above all other aspects of immunity, clearly separate them. Neutrophils are the first immune cells recruited to the site of inflammation, and their action is crucial to limit invasion by microorganisms. Furthermore, they play an essential role in proper resolution of inflammation. When these processes are not tightly regulated, they can trigger positive feedback amplification loops that promote neutrophil activation, leading to significant tissue damage and evolution toward chronic disease. Defective chemotaxis, as observed in Crohn’s disease, can also contribute to the disease through impaired microbe elimination. In addition, through NET production, neutrophils may be involved in thrombo-embolic events frequently observed in IBD patients. While the role of neutrophils has been studied in different animal models of IBD for many years, their contribution to the pathogenesis of IBD remains poorly understood, and no molecules targeting neutrophils are used and validated for the treatment of these pathologies. Therefore, it is crucial to improve our understanding of their mode of action in these particular conditions in order to provide new therapeutic avenues for IBD.
Collapse
|
20
|
de Jong R, Leoni G, Drechsler M, Soehnlein O. The advantageous role of annexin A1 in cardiovascular disease. Cell Adh Migr 2016; 11:261-274. [PMID: 27860536 DOI: 10.1080/19336918.2016.1259059] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023] Open
Abstract
The inflammatory response protects the human body against infection and injury. However, uncontrolled and unresolved inflammation can lead to tissue damage and chronic inflammatory diseases. Therefore, active resolution of inflammation is essential to restore tissue homeostasis. This review focuses on the pro-resolving molecule annexin A1 (ANXA1) and its derived peptides. Mechanisms instructed by ANXA1 are multidisciplinary and affect leukocytes as well as endothelial cells and tissue resident cells like macrophages and mast cells. ANXA1 has an outstanding role in limiting leukocyte recruitment and different aspects of ANXA1 as modulator of the leukocyte adhesion cascade are discussed here. Additionally, this review details the therapeutic relevance of ANXA1 and its derived peptides in cardiovascular diseases since atherosclerosis stands out as a chronic inflammatory disease with impaired resolution and continuous leukocyte recruitment.
Collapse
Affiliation(s)
- Renske de Jong
- a Institute for Cardiovascular Prevention , Ludwig-Maximilians University , Munich , Germany.,b Department of Pathology , Academic Medical Center, Amsterdam University , Amsterdam , the Netherlands
| | - Giovanna Leoni
- a Institute for Cardiovascular Prevention , Ludwig-Maximilians University , Munich , Germany.,b Department of Pathology , Academic Medical Center, Amsterdam University , Amsterdam , the Netherlands
| | - Maik Drechsler
- a Institute for Cardiovascular Prevention , Ludwig-Maximilians University , Munich , Germany.,b Department of Pathology , Academic Medical Center, Amsterdam University , Amsterdam , the Netherlands.,c DZHK (German Centre for Cardiovascular Research), partner site Munich Heart Alliance , Munich , Germany
| | - Oliver Soehnlein
- a Institute for Cardiovascular Prevention , Ludwig-Maximilians University , Munich , Germany.,b Department of Pathology , Academic Medical Center, Amsterdam University , Amsterdam , the Netherlands.,c DZHK (German Centre for Cardiovascular Research), partner site Munich Heart Alliance , Munich , Germany
| |
Collapse
|
21
|
Torres LS, Okumura JV, Silva DGH, Mimura KKO, Belini-Júnior É, Oliveira RG, Lobo CLC, Oliani SM, Bonini-Domingos CR. Inflammation in Sickle Cell Disease: Differential and Down-Expressed Plasma Levels of Annexin A1 Protein. PLoS One 2016; 11:e0165833. [PMID: 27802331 PMCID: PMC5089686 DOI: 10.1371/journal.pone.0165833] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2016] [Accepted: 10/18/2016] [Indexed: 01/09/2023] Open
Abstract
Sickle cell disease (SCD) is an inherited hemolytic anemia whose pathophysiology is driven by polymerization of the hemoglobin S (Hb S), leading to hemolysis and vaso-occlusive events. Inflammation is a fundamental component in these processes and a continuous inflammatory stimulus can lead to tissue damages. Thus, pro-resolving pathways emerge in order to restore the homeostasis. For example there is the annexin A1 (ANXA1), an endogenous anti-inflammatory protein involved in reducing neutrophil-endothelial interactions, accelerating neutrophil apoptosis and stimulating macrophage efferocytosis. We investigated the expression of ANXA1 in plasma of SCD patients and its relation with anemic, hemolytic and inflammatory parameters of the disease. Three SCD genotypes were considered: the homozygous inheritance for Hb S (Hb SS) and the association between Hb S and the hemoglobin variants D-Punjab (Hb SD) and C (Hb SC). ANXA1 and proinflammatory cytokines were quantified by ELISA in plasma of SCD patients and control individuals without hemoglobinopathies. Hematological and biochemical parameters were analyzed by flow cytometry and spectrophotometer. The plasma levels of ANXA1 were about three-fold lesser in SCD patients compared to the control group, and within the SCD genotypes the most elevated levels were found in Hb SS individuals (approximately three-fold higher). Proinflammatory cytokines were higher in SCD groups than in the control individuals. Anemic and hemolytic markers were higher in Hb SS and Hb SD genotypes compared to Hb SC patients. White blood cells and platelets count were higher in Hb SS genotype and were positively correlated to ANXA1 levels. We found that ANXA1 is down-regulated and differentially expressed within the SCD genotypes. Its expression seems to depend on the inflammatory, hemolytic and vaso-occlusive characteristics of the diseased. These data may lead to new biological targets for therapeutic intervention in SCD.
Collapse
Affiliation(s)
- Lidiane S. Torres
- Laboratory of Hemoglobin and Hematologic Genetic Diseases, Department of Biology, São Paulo State University (UNESP), São José do Rio Preto, São Paulo, Brazil
- * E-mail: (LST); (SMO)
| | - Jéssika V. Okumura
- Laboratory of Hemoglobin and Hematologic Genetic Diseases, Department of Biology, São Paulo State University (UNESP), São José do Rio Preto, São Paulo, Brazil
| | - Danilo G. H. Silva
- Laboratory of Hemoglobin and Hematologic Genetic Diseases, Department of Biology, São Paulo State University (UNESP), São José do Rio Preto, São Paulo, Brazil
| | - Kallyne K. O. Mimura
- Laboratory of Immunomorphology, Department of Biology, São Paulo State University (UNESP), São José do Rio Preto, São Paulo, Brazil
| | - Édis Belini-Júnior
- Laboratory of Hemoglobin and Hematologic Genetic Diseases, Department of Biology, São Paulo State University (UNESP), São José do Rio Preto, São Paulo, Brazil
| | - Renan G. Oliveira
- Laboratory of Hemoglobin and Hematologic Genetic Diseases, Department of Biology, São Paulo State University (UNESP), São José do Rio Preto, São Paulo, Brazil
| | - Clarisse L. C. Lobo
- Institute of Hematology Arthur de Siqueira Cavalcanti (HEMORIO), Rio de Janeiro, Rio de Janeiro, Brazil
| | - Sonia M. Oliani
- Laboratory of Immunomorphology, Department of Biology, São Paulo State University (UNESP), São José do Rio Preto, São Paulo, Brazil
- * E-mail: (LST); (SMO)
| | - Claudia R. Bonini-Domingos
- Laboratory of Hemoglobin and Hematologic Genetic Diseases, Department of Biology, São Paulo State University (UNESP), São José do Rio Preto, São Paulo, Brazil
| |
Collapse
|
22
|
Fernández C, Jaimes J, Ortiz MC, Ramírez JD. Host and Toxoplasma gondii genetic and non-genetic factors influencing the development of ocular toxoplasmosis: A systematic review. INFECTION GENETICS AND EVOLUTION 2016; 44:199-209. [PMID: 27389360 DOI: 10.1016/j.meegid.2016.06.053] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/12/2016] [Revised: 06/29/2016] [Accepted: 06/30/2016] [Indexed: 12/13/2022]
Abstract
Toxoplasmosis is a cosmopolitan infection caused by the apicomplexan parasite Toxoplasma gondii. This infectious disease is widely distributed across the world where cats play an important role in its spread. The symptomatology caused by this parasite is diverse but the ocular affectation emerges as the most important clinical phenotype. Therefore, we conducted a systematic review of the current knowledge of ocular toxoplasmosis from the genetic diversity of the pathogen towards the treatment available for this infection. This review represents an update to the scientific community regarding the genetic diversity of the parasite, the genetic factors of the host, the molecular pathogenesis and its association with disease, the available diagnostic tools and the available treatment of patients undergoing ocular toxoplamosis. This review will be an update for the scientific community in order to encourage researchers to deploy cutting-edge investigation across this field.
Collapse
Affiliation(s)
- Carolina Fernández
- Escuela de Medicina y Ciencias de la Salud, Universidad del Rosario, Bogotá, Colombia; Grupo de Investigaciones Microbiológicas - UR (GIMUR), Programa de Biología, Facultad de Ciencias Naturales y Matemáticas, Universidad del Rosario, Bogotá, Colombia
| | - Jesús Jaimes
- Escuela de Medicina y Ciencias de la Salud, Universidad del Rosario, Bogotá, Colombia; Grupo de Investigaciones Microbiológicas - UR (GIMUR), Programa de Biología, Facultad de Ciencias Naturales y Matemáticas, Universidad del Rosario, Bogotá, Colombia
| | - María Camila Ortiz
- Escuela de Medicina y Ciencias de la Salud, Universidad del Rosario, Bogotá, Colombia; Grupo de Investigaciones Microbiológicas - UR (GIMUR), Programa de Biología, Facultad de Ciencias Naturales y Matemáticas, Universidad del Rosario, Bogotá, Colombia
| | - Juan David Ramírez
- Grupo de Investigaciones Microbiológicas - UR (GIMUR), Programa de Biología, Facultad de Ciencias Naturales y Matemáticas, Universidad del Rosario, Bogotá, Colombia.
| |
Collapse
|
23
|
Sugimoto MA, Sousa LP, Pinho V, Perretti M, Teixeira MM. Resolution of Inflammation: What Controls Its Onset? Front Immunol 2016. [PMID: 27199985 DOI: 10.3389/fimmu.2016.00.00160] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/04/2023] Open
Abstract
An effective resolution program may be able to prevent the progression from non-resolving acute inflammation to persistent chronic inflammation. It has now become evident that coordinated resolution programs initiate shortly after inflammatory responses begin. In this context, several mechanisms provide the fine-tuning of inflammation and create a favorable environment for the resolution phase to take place and for homeostasis to return. In this review, we focus on the events required for an effective transition from the proinflammatory phase to the onset and establishment of resolution. We suggest that several mediators that promote the inflammatory phase of inflammation can simultaneously initiate a program for active resolution. Indeed, several events enact a decrease in the local chemokine concentration, a reduction which is essential to inhibit further infiltration of neutrophils into the tissue. Interestingly, although neutrophils are cells that characteristically participate in the active phase of inflammation, they also contribute to the onset of resolution. Further understanding of the molecular mechanisms that initiate resolution may be instrumental to develop pro-resolution strategies to treat complex chronic inflammatory diseases, in humans. The efforts to develop strategies based on resolution of inflammation have shaped a new area of pharmacology referred to as "resolution pharmacology."
Collapse
Affiliation(s)
- Michelle A Sugimoto
- Laboratório de Sinalização 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, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Lirlândia P Sousa
- Laboratório de Sinalização 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, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Vanessa Pinho
- Laboratório de Imunofarmacologia, Departamento de Bioquímica e Imunologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil; Laboratório de Resolução da Resposta Inflamatória, Departamento de Morfologia, Instituto de Ciências Biológicas, 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 , UK
| | - Mauro M Teixeira
- Laboratório de Imunofarmacologia, Departamento de Bioquímica e Imunologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais , Belo Horizonte , Brazil
| |
Collapse
|
24
|
Sugimoto MA, Sousa LP, Pinho V, Perretti M, Teixeira MM. Resolution of Inflammation: What Controls Its Onset? Front Immunol 2016; 7:160. [PMID: 27199985 PMCID: PMC4845539 DOI: 10.3389/fimmu.2016.00160] [Citation(s) in RCA: 409] [Impact Index Per Article: 51.1] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2016] [Accepted: 04/12/2016] [Indexed: 12/12/2022] Open
Abstract
An effective resolution program may be able to prevent the progression from non-resolving acute inflammation to persistent chronic inflammation. It has now become evident that coordinated resolution programs initiate shortly after inflammatory responses begin. In this context, several mechanisms provide the fine-tuning of inflammation and create a favorable environment for the resolution phase to take place and for homeostasis to return. In this review, we focus on the events required for an effective transition from the proinflammatory phase to the onset and establishment of resolution. We suggest that several mediators that promote the inflammatory phase of inflammation can simultaneously initiate a program for active resolution. Indeed, several events enact a decrease in the local chemokine concentration, a reduction which is essential to inhibit further infiltration of neutrophils into the tissue. Interestingly, although neutrophils are cells that characteristically participate in the active phase of inflammation, they also contribute to the onset of resolution. Further understanding of the molecular mechanisms that initiate resolution may be instrumental to develop pro-resolution strategies to treat complex chronic inflammatory diseases, in humans. The efforts to develop strategies based on resolution of inflammation have shaped a new area of pharmacology referred to as “resolution pharmacology.”
Collapse
Affiliation(s)
- Michelle A Sugimoto
- Laboratório de Sinalização 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, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Lirlândia P Sousa
- Laboratório de Sinalização 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, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Vanessa Pinho
- Laboratório de Imunofarmacologia, Departamento de Bioquímica e Imunologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil; Laboratório de Resolução da Resposta Inflamatória, Departamento de Morfologia, Instituto de Ciências Biológicas, 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 , UK
| | - Mauro M Teixeira
- Laboratório de Imunofarmacologia, Departamento de Bioquímica e Imunologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais , Belo Horizonte , Brazil
| |
Collapse
|
25
|
Annexin A1 and the Resolution of Inflammation: Modulation of Neutrophil Recruitment, Apoptosis, and Clearance. J Immunol Res 2016; 2016:8239258. [PMID: 26885535 PMCID: PMC4738713 DOI: 10.1155/2016/8239258] [Citation(s) in RCA: 222] [Impact Index Per Article: 27.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2015] [Accepted: 12/01/2015] [Indexed: 12/13/2022] Open
Abstract
Neutrophils (also named polymorphonuclear leukocytes or PMN) are essential components of the immune system, rapidly recruited to sites of inflammation, providing the first line of defense against invading pathogens. Since neutrophils can also cause tissue damage, their fine-tuned regulation at the inflammatory site is required for proper resolution of inflammation. Annexin A1 (AnxA1), also known as lipocortin-1, is an endogenous glucocorticoid-regulated protein, which is able to counterregulate the inflammatory events restoring homeostasis. AnxA1 and its mimetic peptides inhibit neutrophil tissue accumulation by reducing leukocyte infiltration and activating neutrophil apoptosis. AnxA1 also promotes monocyte recruitment and clearance of apoptotic leukocytes by macrophages. More recently, some evidence has suggested the ability of AnxA1 to induce macrophage reprogramming toward a resolving phenotype, resulting in reduced production of proinflammatory cytokines and increased release of immunosuppressive and proresolving molecules. The combination of these mechanisms results in an effective resolution of inflammation, pointing to AnxA1 as a promising tool for the development of new therapeutic strategies to treat inflammatory diseases.
Collapse
|
26
|
Prates J, Franco-Salla GB, Dinarte Dos Santos AR, da Silva WA, da Cunha BR, Tajara EH, Oliani SM, Rodrigues-Lisoni FC. ANXA1Ac₂₋₂₆ peptide reduces ID1 expression in cervical carcinoma cultures. Gene 2015; 570:248-54. [PMID: 26072160 DOI: 10.1016/j.gene.2015.06.021] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2015] [Revised: 05/04/2015] [Accepted: 06/08/2015] [Indexed: 11/30/2022]
Abstract
Cervical cancer is the second most frequent cancer in women worldwide and is associated with genetic alterations, infection with human papilloma virus (HPV), angiogenesis and inflammatory processes. The idea that inflammation is involved in tumorigenesis is supported by the frequent appearance of cancer in areas of chronic inflammation. On the other hand, the inflammatory response is controlled by the action of anti-inflammatory mediators, among these mediators, annexin A1 (ANXA1), a 37 kDa protein was detected as a modulator of inflammatory processes and is expressed by tumor cells. The study was carried out on the epithelial cancer cell line (SiHa) treated with the peptide of annexin A1 (ANXA1Ac2-26). We combined subtraction hybridization approach, Ingenuity Systems software and quantitative PCR, in order to evaluate gene expression influenced by ANXA1. We observed that ANXA1Ac2-26 inhibited proliferation in SiHa cells after 72h. In these cells, 55 genes exhibited changes in expression levels in response to peptide treatment. Six genes were selected and the expression results of 5 up-regulated genes (TPT1, LDHA, NCOA3, HIF1A, RAB13) and one down-regulated gene (ID1) were research by real time quantitative PCR. Four more genes (BMP4, BMPR1B, SMAD1 and SMAD4) of the ID1 pathway were investigated and only one (BMPR1B) shows the same down regulation. The data indicate the involvement of ANXA1Ac2-26 in the altered expression of genes involved in tumorigenic processes, which could potentially be applied as a therapeutic indicator of cervical cancer.
Collapse
Affiliation(s)
- Janesly Prates
- Department of Biology, Institute of Biosciences, Letters and Science - IBILCE/UNESP, São José do Rio Preto, SP, Brazil
| | - Gabriela Bueno Franco-Salla
- Department of Biology, Institute of Biosciences, Letters and Science - IBILCE/UNESP, São José do Rio Preto, SP, Brazil
| | - Anemari Ramos Dinarte Dos Santos
- Department of Clinical Medical, Foundation Blood Center of Ribeirão Preto, Faculty of Medicine of Ribeirão Preto, University of São Paulo - FCFRP/USP, Ribeirão Preto, SP, Brazil
| | - Wilson Araújo da Silva
- Department of Clinical Medical, Foundation Blood Center of Ribeirão Preto, Faculty of Medicine of Ribeirão Preto, University of São Paulo - FCFRP/USP, Ribeirão Preto, SP, Brazil
| | - Bianca Rodrigues da Cunha
- Department of Molecular, Biology Faculty of Medicine of São José do Rio Preto - FAMERP, São José do Rio Preto, SP, Brazil
| | - Eloiza Helena Tajara
- Department of Molecular, Biology Faculty of Medicine of São José do Rio Preto - FAMERP, São José do Rio Preto, SP, Brazil
| | - Sonia Maria Oliani
- Department of Biology, Institute of Biosciences, Letters and Science - IBILCE/UNESP, São José do Rio Preto, SP, Brazil
| | | |
Collapse
|
27
|
Sena AA, Pedrotti LP, Barrios BE, Cejas H, Balderramo D, Diller A, Correa SG. Lack of TNFRI signaling enhances annexin A1 biological activity in intestinal inflammation. Biochem Pharmacol 2015; 98:422-31. [PMID: 26386311 DOI: 10.1016/j.bcp.2015.09.009] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2015] [Accepted: 09/11/2015] [Indexed: 12/19/2022]
Abstract
We evaluated whether the lack of TNF-α signaling increases mucosal levels of annexin A1 (AnxA1); the hypothesis stems from previous findings showing that TNF-α neutralization in Crohn's disease patients up-regulates systemic AnxA1 expression. Biopsies from healthy volunteers and patients under anti-TNF-α therapy with remittent ulcerative colitis (UC) showed higher AnxA1 expression than those with active disease. We also evaluated dextran sulfate sodium (DSS)-acute colitis in TNF-α receptor 1 KO (TNFR1-/-) strain with impaired TNF-α signaling and C57BL/6 (WT) mice. Although both strains developed colitis, TNFR1-/- mice showed early clinical recovery, lower myeloperoxidase (MPO) activity and milder histopathological alterations. Colonic epithelium from control and DSS-treated TNFR1-/- mice showed intense AnxA1 expression and AnxA1+ CD4+ and CD8+ T cells were more frequent in TNFR1-/- animals, suggesting an extra supply of AnxA1. The pan antagonist of AnxA1 receptors exacerbated the colitis outcome in TNFR1-/- mice, supporting the pivotal role of AnxA1 in the early recovery. Our findings demonstrate that the TNF-α signaling reduction favors the expression and biological activity of AnxA1 in inflamed intestinal mucosa.
Collapse
Affiliation(s)
- Angela A Sena
- Immunology, Department of Clinical Biochemistry, CIBICI (CONICET), Faculty of Chemical Sciences, National University of Cordoba, Córdoba, Argentina
| | - Luciano P Pedrotti
- Immunology, Department of Clinical Biochemistry, CIBICI (CONICET), Faculty of Chemical Sciences, National University of Cordoba, Córdoba, Argentina
| | - Bibiana E Barrios
- Immunology, Department of Clinical Biochemistry, CIBICI (CONICET), Faculty of Chemical Sciences, National University of Cordoba, Córdoba, Argentina
| | - Hugo Cejas
- Immunology, Department of Clinical Biochemistry, CIBICI (CONICET), Faculty of Chemical Sciences, National University of Cordoba, Córdoba, Argentina
| | - Domingo Balderramo
- Gastroenterology Department, Hospital Privado, Centro Médico, Córdoba, Argentina
| | - Ana Diller
- Pathology Department, Hospital Privado, Centro Médico, Córdoba, Argentina
| | - Silvia G Correa
- Immunology, Department of Clinical Biochemistry, CIBICI (CONICET), Faculty of Chemical Sciences, National University of Cordoba, Córdoba, Argentina.
| |
Collapse
|
28
|
Stuqui B, de Paula-Silva M, Carlos CP, Ullah A, Arni RK, Gil CD, Oliani SM. Ac2-26 Mimetic Peptide of Annexin A1 Inhibits Local and Systemic Inflammatory Processes Induced by Bothrops moojeni Venom and the Lys-49 Phospholipase A2 in a Rat Model. PLoS One 2015; 10:e0130803. [PMID: 26147724 PMCID: PMC4492549 DOI: 10.1371/journal.pone.0130803] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2014] [Accepted: 05/25/2015] [Indexed: 11/19/2022] Open
Abstract
Annexin A1 (AnxA1) is an endogenous glucocorticoid regulated protein that modulates anti-inflammatory process and its therapeutic potential has recently been recognized in a range of systemic inflammatory disorders. The effect of the N-terminal peptide Ac2-26 of AnxA1 on the toxic activities of Bothrops moojeni crude venom (CV) and its myotoxin II (MjTX-II) were evaluated using a peritonitis rat model. Peritonitis was induced by the intraperitoneal injection of either CV or MjTX-II, a Lys-49 phospholipase A2. Fifteen minutes after the injection, the rats were treated with either Ac2-26 or PBS. Four hours later, the CV and MjTX-II-induced peritonitis were characterized by neutrophilia (in the peritoneal exudate, blood and mesentery) and increased number of mesenteric degranulated mast cells and macrophages. At 24 hours post-injection, the local inflammatory response was attenuated in the CV-induced peritonitis while the MjTX-II group exhibited neutrophilia (peritoneal exudates and blood). Ac2-26 treatment prevented the influx of neutrophils in MjTX-II-induced peritonitis and diminished the proportion of mesenteric degranulated mast cells and macrophages in CV-induced peritonitis. Additionally, CV and MjTX-II promoted increased levels of IL-1β and IL-6 in the peritoneal exudates which were significantly reduced after Ac2-26 treatment. At 4 and 24 hours, the endogenous expression of AnxA1 was upregulated in the mesenteric neutrophils (CV and MjTX-II groups) and mast cells (CV group). In the kidneys, CV and MjTX-II administrations were associated with an increased number of macrophages and morphological alterations in the juxtamedullary nephrons in proximal and distal tubules. Ac2-26 promoted significant recovery of the juxtamedullary structures, decreased the number of macrophages and diminished the AnxA1 in epithelial cells from distal tubules and renal capsules. Our results show that Ac2-26 treatment significantly attenuates local and systemic inflammatory processes and indicate this peptide as a potential target for the development of new therapeutic strategies for the snakebite envenomation treatment.
Collapse
Affiliation(s)
- Bruna Stuqui
- Laboratory of Immunomorphology, Department of Biology, São Paulo State University (UNESP), São José do Rio Preto, São Paulo, Brazil
| | - Marina de Paula-Silva
- Laboratory of Immunomorphology, Department of Biology, São Paulo State University (UNESP), São José do Rio Preto, São Paulo, Brazil
| | - Carla Patrícia Carlos
- Laboratory of Immunomorphology, Department of Biology, São Paulo State University (UNESP), São José do Rio Preto, São Paulo, Brazil
| | - Anwar Ullah
- Multiuser Center for Biomolecular Innovation, Department of Physics, São Paulo State University (UNESP), São José do Rio Preto, São Paulo, Brazil
| | - Raghuvir Krishnaswamy Arni
- Multiuser Center for Biomolecular Innovation, Department of Physics, São Paulo State University (UNESP), São José do Rio Preto, São Paulo, Brazil
| | - Cristiane Damas Gil
- Department of Morphology and Genetics, São Paulo Federal University (UNIFESP), São Paulo, Brazil
| | - Sonia Maria Oliani
- Laboratory of Immunomorphology, Department of Biology, São Paulo State University (UNESP), São José do Rio Preto, São Paulo, Brazil
- * E-mail:
| |
Collapse
|
29
|
The resolution of inflammation: Principles and challenges. Semin Immunol 2015; 27:149-60. [PMID: 25911383 DOI: 10.1016/j.smim.2015.03.014] [Citation(s) in RCA: 263] [Impact Index Per Article: 29.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/11/2015] [Revised: 03/27/2015] [Accepted: 03/30/2015] [Indexed: 12/11/2022]
Abstract
The concept that chemokines, cytokines and pro-inflammatory mediators act in a co-ordinated fashion to drive the initiation of the inflammatory reaction is well understood. The significance of such networks acting during the resolution of inflammation however is poorly appreciated. In recent years, specific pro-resolving mediators were discovered which activate resolution pathways to return tissues to homeostasis. These mediators are diverse in nature, and include specialized lipid mediators (lipoxins, resolvins, protectins and maresins) proteins (annexin A1, galectins) and peptides, gaseous mediators including hydrogen sulphide, a purine (adenosine), as well as neuromodulator release under the control of the vagus nerve. Functionally, they can act to limit further leukocyte recruitment, induce neutrophil apoptosis and enhance efferocytosis by macrophages. They can also switch macrophages from classical to alternatively activated cells, promote the return of non-apoptotic cells to the lymphatics and help initiate tissue repair mechanisms and healing. Within this review we highlight the essential cellular aspects required for successful tissue resolution, briefly discuss the pro-resolution mediators that drive these processes and consider potential challenges faced by researchers in the quest to discover how inflammation resolves and why chronic inflammation persists.
Collapse
|
30
|
Gimenes AD, Andrade TRM, Mello CB, Ramos L, Gil CD, Oliani SM. Beneficial effect of annexin A1 in a model of experimental allergic conjunctivitis. Exp Eye Res 2015; 134:24-32. [PMID: 25795053 DOI: 10.1016/j.exer.2015.03.013] [Citation(s) in RCA: 40] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2014] [Revised: 03/12/2015] [Accepted: 03/16/2015] [Indexed: 01/01/2023]
Abstract
Annexin A1 (ANXA1), a 37 kDa glucocorticoid-regulated protein, is a potent anti-inflammatory mediator effective in terminating acute inflammatory response, and its role in allergic settings has been poorly studied. The aim of this investigation was to evaluate the mechanism of action of ANXA1 in intraocular inflammation using a classical model of ovalbumin (OVA)-induced allergic conjunctivitis (AC). OVA-immunised Balb/c mice, wild-type (WT) and ANXA1-deficient (AnxA1(-/-)), were challenged with eye drops containing OVA on days 14-16 with a subset of WT animals pretreated intraperitoneally with the peptide Ac2-26 (N-terminal region of ANXA1) or dexamethasone (DEX). After 24 h of the last ocular challenge, WT mice treated with Ac2-26 and DEX had significantly reduced clinical signs of conjunctivitis (chemosis, conjunctival hyperaemia, lid oedema and tearing), plasma IgE levels, leukocyte (eosinophil and neutrophil) influx and mast cell degranulation in the conjunctiva compared to WT controls. These anti-inflammatory effects of DEX were associated with high endogenous levels of ANXA1 in the ocular tissues as detected by immunohistochemistry. Additionally, Ac2-26 administration was effective to reduce IL-2, IL-4, IL-10, IL-13, eotaxin and RANTES in the eye and lymph nodes compared to untreated WT animals. The lack of ANXA1 produced an exacerbated allergic response as detected by the density of the inflammatory cell influx to the conjunctiva and the cytokine/chemokine release. These different effects observed for Ac2-26 were correlated with diminished level of activated ERK at 24 h in the ocular tissues compared to untreated OVA group. Our findings demonstrate the protective effect of ANXA1 during the inflammatory allergic response suggesting this protein as a potential target for new ocular inflammation therapies.
Collapse
Affiliation(s)
- Alexandre D Gimenes
- UNIFESP - Universidade Federal de São Paulo, Laboratório de Histologia, Departamento de Morfologia e Genética, 04023-900 São Paulo, São Paulo, Brazil
| | - Teresa Raquel M Andrade
- UNIFESP - Universidade Federal de São Paulo, Laboratório de Histologia, Departamento de Morfologia e Genética, 04023-900 São Paulo, São Paulo, Brazil
| | - Cláudia B Mello
- UNESP - Universidade Estadual Paulista, Laboratório de Imunomorfologia, Departamento de Biologia, 15054-000 São José do Rio Preto, São Paulo, Brazil
| | - Lisandra Ramos
- UNIFESP - Universidade Federal de São Paulo, Laboratório de Histologia, Departamento de Morfologia e Genética, 04023-900 São Paulo, São Paulo, Brazil
| | - Cristiane D Gil
- UNIFESP - Universidade Federal de São Paulo, Laboratório de Histologia, Departamento de Morfologia e Genética, 04023-900 São Paulo, São Paulo, Brazil
| | - Sonia M Oliani
- UNIFESP - Universidade Federal de São Paulo, Laboratório de Histologia, Departamento de Morfologia e Genética, 04023-900 São Paulo, São Paulo, Brazil; UNESP - Universidade Estadual Paulista, Laboratório de Imunomorfologia, Departamento de Biologia, 15054-000 São José do Rio Preto, São Paulo, Brazil.
| |
Collapse
|
31
|
Schepetkin IA, Khlebnikov AI, Giovannoni MP, Kirpotina LN, Cilibrizzi A, Quinn MT. Development of small molecule non-peptide formyl peptide receptor (FPR) ligands and molecular modeling of their recognition. Curr Med Chem 2015; 21:1478-504. [PMID: 24350845 DOI: 10.2174/0929867321666131218095521] [Citation(s) in RCA: 40] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2013] [Revised: 10/14/2013] [Accepted: 12/10/2013] [Indexed: 02/07/2023]
Abstract
Formyl peptide receptors (FPRs) are G protein-coupled receptors (GPCRs) expressed on a variety of cell types. These receptors play an important role in the regulation of inflammatory reactions and sensing cellular damage. They have also been implicated in the pathogenesis of various diseases, including neurodegenerative diseases, cataract formation, and atherogenesis. Thus, FPR ligands, both agonists and antagonists, may represent novel therapeutics for modulating host defense and innate immunity. A variety of molecules have been identified as receptor subtype-selective and mixed FPR agonists with potential therapeutic value during last decade. This review describes our efforts along with recent advances in the identification, optimization, biological evaluation, and structure-activity relationship (SAR) analysis of small molecule non-peptide FPR agonists and antagonists, including chiral molecules. Questions regarding the interaction at the molecular level of benzimidazoles, pyrazolones, pyridazin-3(2H)-ones, N-phenylureas and other derivatives with FPR1 and FPR2 are discussed. Application of computational models for virtual screening and design of FPR ligands is also considered.
Collapse
Affiliation(s)
| | | | | | | | | | - M T Quinn
- Department of Immunology and Infectious Diseases, Montana State University, Bozeman, MT 59717, USA.
| |
Collapse
|
32
|
Gastardelo TS, Cunha BR, Raposo LS, Maniglia JV, Cury PM, Lisoni FCR, Tajara EH, Oliani SM. Inflammation and cancer: role of annexin A1 and FPR2/ALX in proliferation and metastasis in human laryngeal squamous cell carcinoma. PLoS One 2014; 9:e111317. [PMID: 25490767 PMCID: PMC4260827 DOI: 10.1371/journal.pone.0111317] [Citation(s) in RCA: 56] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2013] [Accepted: 09/30/2014] [Indexed: 11/19/2022] Open
Abstract
The anti-inflammatory protein annexin A1 (ANXA1) has been associated with cancer progression and metastasis, suggesting its role in regulating tumor cell proliferation. We investigated the mechanism of ANXA1 interaction with formylated peptide receptor 2 (FPR2/ALX) in control, peritumoral and tumor larynx tissue samples from 20 patients, to quantitate the neutrophils and mast cells, and to evaluate the protein expression and co-localization of ANXA1/FPR2 in these inflammatory cells and laryngeal squamous cells by immunocytochemistry. In addition, we performed in vitro experiments to further investigate the functional role of ANXA1/FPR2 in the proliferation and metastasis of Hep-2 cells, a cell line from larynx epidermoid carcinoma, after treatment with ANXA12–26 (annexin A1 N-terminal-derived peptide), Boc2 (antagonist of FPR) and/or dexamethasone. Under these treatments, the level of Hep-2 cell proliferation, pro-inflammatory cytokines, ANXA1/FPR2 co-localization, and the prostaglandin signalling were analyzed using ELISA, immunocytochemistry and real-time PCR. An influx of neutrophils and degranulated mast cells was detected in tumor samples. In these inflammatory cells of peritumoral and tumor samples, ANXA1/FPR2 expression was markedly exacerbated, however, in laryngeal carcinoma cells, this expression was down-regulated. ANXA12–26 treatment reduced the proliferation of the Hep-2 cells, an effect that was blocked by Boc2, and up-regulated ANXA1/FPR2 expression. ANXA12–26 treatment also reduced the levels of pro-inflammatory cytokines and affected the expression of metalloproteinases and EP receptors, which are involved in the prostaglandin signalling. Overall, this study identified potential roles for the molecular mechanism of the ANXA1/FPR2 interaction in laryngeal cancer, including its relationship with the prostaglandin pathway, providing promising starting points for future research. ANXA1 may contribute to the regulation of tumor growth and metastasis through paracrine mechanisms that are mediated by FPR2/ALX. These data may lead to new biological targets for therapeutic intervention in human laryngeal cancer.
Collapse
MESH Headings
- Aged
- Aged, 80 and over
- Amino Acid Sequence
- Annexin A1/chemistry
- Annexin A1/metabolism
- Carcinoma, Squamous Cell/immunology
- Carcinoma, Squamous Cell/metabolism
- Carcinoma, Squamous Cell/pathology
- Cell Degranulation/drug effects
- Cell Line, Tumor
- Cell Proliferation/drug effects
- Humans
- Inflammation/immunology
- Inflammation/metabolism
- Inflammation/pathology
- Laryngeal Neoplasms/immunology
- Laryngeal Neoplasms/metabolism
- Laryngeal Neoplasms/pathology
- Male
- Mast Cells/cytology
- Mast Cells/drug effects
- Metalloproteases/metabolism
- Middle Aged
- Molecular Sequence Data
- Neoplasm Metastasis
- Neutrophils/drug effects
- Neutrophils/immunology
- Peptide Fragments/chemistry
- Peptide Fragments/pharmacology
- Prostaglandins/metabolism
- Receptors, Formyl Peptide/metabolism
- Receptors, Lipoxin/metabolism
- Receptors, Prostaglandin E, EP3 Subtype/metabolism
- Receptors, Prostaglandin E, EP4 Subtype/metabolism
- Signal Transduction/drug effects
- Tumor Microenvironment/drug effects
- Up-Regulation/drug effects
Collapse
Affiliation(s)
- Thaís Santana Gastardelo
- From the Post-graduation in Structural and Functional Biology, Federal University of São Paulo (UNIFESP), Paulista School of Medicine (EPM), São Paulo, SP, Brazil
| | - Bianca Rodrigues Cunha
- Department of Molecular Biology, Faculty of Medicine (FAMERP), São José do Rio Preto, SP, Brazil
| | - Luís Sérgio Raposo
- Department of Otorhinolaringology, Faculty of Medicine (FAMERP), São José do Rio Preto, SP, Brazil
| | - José Victor Maniglia
- Department of Otorhinolaringology, Faculty of Medicine (FAMERP), São José do Rio Preto, SP, Brazil
| | - Patrícia Maluf Cury
- Department of Pathology, Faculty of Medicine (FAMERP), São José do Rio Preto, SP, Brazil
| | | | - Eloiza Helena Tajara
- Department of Molecular Biology, Faculty of Medicine (FAMERP), São José do Rio Preto, SP, Brazil
- Department of Genetics and Evolutionary Biology, Institute of Biosciences, University of São Paulo (USP), São Paulo, SP, Brazil
| | - Sonia Maria Oliani
- From the Post-graduation in Structural and Functional Biology, Federal University of São Paulo (UNIFESP), Paulista School of Medicine (EPM), São Paulo, SP, Brazil
- Department of Biology, Instituto de Biociências, Letras e Ciências Exatas (IBILCE), São Paulo State University (UNESP), São José do Rio Preto, SP, Brazil
- * E-mail:
| |
Collapse
|
33
|
Qin C, Yang YH, May L, Gao X, Stewart AG, Tu Y, Woodman OL, Ritchie RH. Cardioprotective potential of annexin-A1 mimetics in myocardial infarction. Pharmacol Ther 2014; 148:47-65. [PMID: 25460034 DOI: 10.1016/j.pharmthera.2014.11.012] [Citation(s) in RCA: 58] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2014] [Accepted: 11/14/2014] [Indexed: 12/15/2022]
Abstract
Myocardial infarction (MI) and its resultant heart failure remains a major cause of death in the world. The current treatments for patients with MI are revascularization with thrombolytic agents or interventional procedures. These treatments have focused on restoring blood flow to the ischemic tissue to prevent tissue necrosis and preserve organ function. The restoration of blood flow after a period of ischemia, however, may elicit further myocardial damage, called reperfusion injury. Pharmacological interventions, such as antioxidant and Ca(2+) channel blockers, have shown premises in experimental settings; however, clinical studies have shown limited success. Thus, there is a need for the development of novel therapies to treat reperfusion injury. The therapeutic potential of glucocorticoid-regulated anti-inflammatory mediator annexin-A1 (ANX-A1) has recently been recognized in a range of systemic inflammatory disorders. ANX-A1 binds to and activates the family of formyl peptide receptors (G protein-coupled receptor family) to inhibit neutrophil activation, migration and infiltration. Until recently, studies on the cardioprotective actions of ANX-A1 and its peptide mimetics (Ac2-26, CGEN-855A) have largely focused on its anti-inflammatory effects as a mechanism of preserving myocardial viability following I-R injury. Our laboratory provided the first evidence of the direct protective action of ANX-A1 on myocardium, independent of inflammatory cells in vitro. We now review the potential for ANX-A1 based therapeutics to be seen as a "triple shield" therapy against myocardial I-R injury, limiting neutrophil infiltration and preserving both cardiomyocyte viability and contractile function. This novel therapy may thus represent a valuable clinical approach to improve outcome after MI.
Collapse
Affiliation(s)
- Chengxue Qin
- Baker IDI Heart & Diabetes Institute, Melbourne, Victoria, Australia; Department of Pharmacology and Therapeutics, The University of Melbourne, Parkville, Victoria, Australia
| | - Yuan H Yang
- Centre for Inflammatory Diseases Monash University and Monash Medical Centre, Clayton, Victoria, Australia
| | - Lauren May
- Department of Drug Discovery Biology, Monash Institute of Pharmaceutical Sciences, and Department of Pharmacology, Monash University, Parkville, Victoria, Australia
| | - Xiaoming Gao
- Baker IDI Heart & Diabetes Institute, Melbourne, Victoria, Australia
| | - Alastair G Stewart
- Department of Pharmacology and Therapeutics, The University of Melbourne, Parkville, Victoria, Australia
| | - Yan Tu
- Department of Pharmacology and Therapeutics, The University of Melbourne, Parkville, Victoria, Australia
| | - Owen L Woodman
- School of Medical Sciences, RMIT University, Bundoora 3083, Victoria, Australia
| | - Rebecca H Ritchie
- Baker IDI Heart & Diabetes Institute, Melbourne, Victoria, Australia; Department of Pharmacology and Therapeutics, The University of Melbourne, Parkville, Victoria, Australia; Department of Medicine, Monash University, Clayton, Victoria, Australia.
| |
Collapse
|
34
|
Schepetkin IA, Kirpotina LN, Khlebnikov AI, Cheng N, Ye RD, Quinn MT. Antagonism of human formyl peptide receptor 1 (FPR1) by chromones and related isoflavones. Biochem Pharmacol 2014; 92:627-41. [PMID: 25450672 DOI: 10.1016/j.bcp.2014.09.027] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2014] [Revised: 09/25/2014] [Accepted: 09/26/2014] [Indexed: 12/24/2022]
Abstract
Formyl peptide receptors (FPRs) are G protein-coupled receptors (GPCRs) expressed on a variety of cell types. Because FPRs play an important role in the regulation of inflammatory reactions implicated in disease pathogenesis, FPR antagonists may represent novel therapeutics for modulating innate immunity. Previously, 4H-chromones were reported to be potent and competitive FPR1 antagonists. In the present studies, 96 additional chromone analogs, including related synthetic and natural isoflavones were evaluated for FPR1 antagonist activity. We identified a number of novel competitive FPR1 antagonists that inhibited fMLF-induced intracellular Ca2+ mobilization in FPR1-HL60 cells and effectively competed with WKYMVm-FITC for binding to FPR1 in FPR1-HL60 and FPR1-RBL cells. Compound 10 (6-hexyl-2-methyl-3-(1-methyl-1H-benzimidazol-2-yl)-4-oxo-4H-chromen-7-yl acetate) was found to be the most potent FPR1-specific antagonist, with binding affinity Ki∼100 nM. These chromones inhibited Ca2+ flux and chemotaxis in human neutrophils with nanomolar-micromolar IC50 values. In addition, the most potent novel FPR1 antagonists inhibited fMLF-induced phosphorylation of extracellular signal-regulated kinases (ERK1/2) in FPR1-RBL cells. These antagonists were specific for FPR1 and did not inhibit WKYMVM/WKYMVm-induced intracellular Ca2+ mobilization in FPR2-HL60 cells, FPR3-HL60 cells, RBL cells transfected with murine Fpr1, or interleukin 8-induced Ca2+ flux in human neutrophils and RBL cells transfected with CXC chemokine receptor 1 (CXCR1). Moreover, pharmacophore modeling showed that the active chromones had a significantly higher degree of similarity with the pharmacophore template as compared to inactive analogs. Thus, the chromone/isoflavone scaffold represents a relevant backbone for development of novel FPR1 antagonists.
Collapse
Affiliation(s)
- Igor A Schepetkin
- Department of Microbiology and Immunology, Montana State University, Bozeman, MT 59717, USA
| | - Liliya N Kirpotina
- Department of Microbiology and Immunology, Montana State University, Bozeman, MT 59717, USA
| | - Andrei I Khlebnikov
- Department of Chemistry, Altai State Technical University, Barnaul, Russia; Department of Biotechnology and Organic Chemistry, Tomsk Polytechnic University, Tomsk, Russia
| | - Ni Cheng
- Department of Pharmacology, University of Illinois, Chicago, IL, USA
| | - Richard D Ye
- Department of Pharmacology, University of Illinois, Chicago, IL, USA
| | - Mark T Quinn
- Department of Microbiology and Immunology, Montana State University, Bozeman, MT 59717, USA.
| |
Collapse
|
35
|
Brancaleone V, Mitidieri E, Flower RJ, Cirino G, Perretti M. Annexin A1 mediates hydrogen sulfide properties in the control of inflammation. J Pharmacol Exp Ther 2014; 351:96-104. [PMID: 25077524 DOI: 10.1124/jpet.114.217034] [Citation(s) in RCA: 46] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Hydrogen sulfide (H2S) is a gaseous mediator synthesized in mammalian tissues by three main enzymes-cystathionine-β-synthase (CBS), cystathionine-γ-lyase (CSE), and 3-mercaptopyruvate-sulfurtransferase-and its levels increase under inflammatory conditions or sepsis. Since H2S and H2S-releasing molecules afford inhibitory properties in leukocyte trafficking, we tested whether endogenous annexin A1 (AnxA1), a glucocorticoid-regulated inhibitor of inflammation acting through formylated-peptide receptor 2 (ALX), could display intermediary functions in the anti-inflammatory profile of H2S. We first investigated whether endogenous AnxA1 could modulate H2S biosynthesis. To this end, a marked increase in CBS and/or CSE gene products was quantified by quantitative real-time polymerase chain reaction in aortas, kidneys, and spleens collected from AnxA1(-/-) mice, as compared with wild-type animals. When lipopolysaccharide-stimulated bone marrow-derived macrophages were studied, H2S-donor sodium hydrosulfide (NaHS) counteracted the increased expression of inducible nitric oxide synthase and cyclooxygenase 2 mRNA evoked by the endotoxin, yet it was inactive in macrophages harvested from AnxA1(-/-) mice. Next we studied the effect of in vivo administration of NaHS in a model of interleukin-1β (IL-1β)-induced mesenteric inflammation. AnxA1(+/+) mice treated with NaHS (100 μmol/kg) displayed inhibition of IL-1β-induced leukocyte adhesion/emigration in the inflamed microcirculation, not observed in AnxA1(-/-) animals. These results were translated by testing human neutrophils, where NaHS (10-100 μM) prompted an intense mobilization (>50%) of AnxA1 from cytosol to cell surface, an event associated with inhibition of cell/endothelium interaction under flow. Taken together, these data strongly indicate the existence of a positive interlink between AnxA1 and H2S pathway, with nonredundant functions in the control of experimental inflammation.
Collapse
Affiliation(s)
- Vincenzo Brancaleone
- Department of Science, University of Basilicata, Potenza, Italy (V.B.); Department of Pharmacy, University of Naples, Naples, Italy (E.M., G.C.); and William Harvey Research Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London, UK (V.B., R.J.F., M.P.)
| | - Emma Mitidieri
- Department of Science, University of Basilicata, Potenza, Italy (V.B.); Department of Pharmacy, University of Naples, Naples, Italy (E.M., G.C.); and William Harvey Research Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London, UK (V.B., R.J.F., M.P.)
| | - Roderick J Flower
- Department of Science, University of Basilicata, Potenza, Italy (V.B.); Department of Pharmacy, University of Naples, Naples, Italy (E.M., G.C.); and William Harvey Research Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London, UK (V.B., R.J.F., M.P.)
| | - Giuseppe Cirino
- Department of Science, University of Basilicata, Potenza, Italy (V.B.); Department of Pharmacy, University of Naples, Naples, Italy (E.M., G.C.); and William Harvey Research Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London, UK (V.B., R.J.F., M.P.)
| | - Mauro Perretti
- Department of Science, University of Basilicata, Potenza, Italy (V.B.); Department of Pharmacy, University of Naples, Naples, Italy (E.M., G.C.); and William Harvey Research Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London, UK (V.B., R.J.F., M.P.)
| |
Collapse
|
36
|
Chen L, Lv F, Pei L. Annexin 1: a glucocorticoid-inducible protein that modulates inflammatory pain. Eur J Pain 2013; 18:338-47. [PMID: 23904250 DOI: 10.1002/j.1532-2149.2013.00373.x] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/26/2013] [Indexed: 12/16/2022]
Abstract
Annexin 1, a glucocorticoid (GC)-inducible protein, can play an important role via formyl peptide receptor like 1 (FPR2/ALX, also known as FPRL1) in inflammatory pain modulation. The aim of this review is to analyze different lines of evidence for the role of ANXA1 with different mechanisms on inflammatory pain and describe the profile of ANXA1 as a potential analgesic. A Medline (PUBMED) search using the terms 'Annexin 1 distribution OR expression, FPR2/ALX distribution OR expression, Annexin 1 AND pain, Annexin 1 AND FPR2/ALX AND pain' was performed. Articles with a publication date up to Nov. 1st, 2012 were included. The antinociception of ANXA1 has been evaluated in diverse pain models. It has been suggested that ANXA1 may exerts its action via: (1) inhibiting vital cytokines involved in pain transmission, (2) inhibiting neutrophil accumulation through preventing transendothelial migration via an interaction with formyl peptide receptors, (3) facilitating tonic opioid release from neutrophil in inflammatory site, (4) interrupting the peripheral nociceptive transmission by suppressing neuronal excitability. In general, ANXA1 is a potential mediator for anti-nociception and the role with its receptor constitute attractive targets for developing anesthesia and analgesic drugs, and their interaction may prove to be a useful strategy to treat inflammatory pain.
Collapse
Affiliation(s)
- L Chen
- Department of Neurology of the First People's Hospital of Jingzhou, The first affiliated hospital of Yangtze University, Jingzhou, China
| | | | | |
Collapse
|
37
|
Teixeira RAP, Mimura KKO, Araujo LP, Greco KV, Oliani SM. The essential role of annexin A1 mimetic peptide in the skin allograft survival. J Tissue Eng Regen Med 2013; 10:E44-53. [PMID: 23897745 DOI: 10.1002/term.1773] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2012] [Revised: 04/15/2013] [Accepted: 04/16/2012] [Indexed: 12/11/2022]
Abstract
Immunosuppressive drugs have a critical role in inhibiting tissue damage and allograft rejection. Studies have demonstrated the anti-inflammatory effects of the annexin A1 (AnxA1) in the regulation of transmigration and apoptosis of leucocytes. In the present study, an experimental skin allograft model was used to evaluate a potential protective effect of AnxA1 in transplantation survival. Mice were used for the skin allograft model and pharmacological treatments were carried out using either the AnxA1 mimetic peptide Ac2-26, with or without cyclosporine A (CsA), starting 3 days before surgery until rejection. Graft survival, skin histopathology, leucocyte transmigration and expression of AnxA1 and AnxA5 post-transplantation were analysed. Pharmacological treatment with Ac2-26 increased skin allograft survival related with inhibition of neutrophil transmigration and induction of apoptosis, thereby reducing the tissue damage compared with control animals. Moreover, AnxA1 and AnxA5 expression increased after Ac2-26 treatment in neutrophils. Interestingly, the combination of Ac2-26 and cyclosporine A showed similar survival of transplants when compared with the cyclosporine A group, which could be attributed to a synergistic effect of both drugs. Investigations in vitro revealed that cyclosporine A inhibited extracellular-signal-regulated kinase (ERK) phosphorylation induced by Ac2-26 in neutrophils. Overall, the results suggest that AnxA1 has an essential role in augmenting the survival of skin allograft, mainly owing to inhibition of neutrophil transmigration and enhancement of apoptosis. This effect may lead to the development of new therapeutic approaches relevant to transplant rejection.
Collapse
Affiliation(s)
| | | | - Leandro Pires Araujo
- Post-Graduation in Structural and Functional Biology, Federal University of São Paulo (UNIFESP), São Paulo, Brazil
| | - Karin Vicente Greco
- Department of Surgical Research, Northwick Park Institute for Medical Research - University College London, London, UK
| | - Sonia Maria Oliani
- Post-Graduation in Structural and Functional Biology, Federal University of São Paulo (UNIFESP), São Paulo, Brazil.,Department of Biology, Instituto de Biociências, Letras e Ciências Exatas (IBILCE), São Paulo State University (UNESP), São José do Rio Preto, Brazil
| |
Collapse
|
38
|
Girol AP, Mimura KKO, Drewes CC, Bolonheis SM, Solito E, Farsky SHP, Gil CD, Oliani SM. Anti-inflammatory mechanisms of the annexin A1 protein and its mimetic peptide Ac2-26 in models of ocular inflammation in vivo and in vitro. THE JOURNAL OF IMMUNOLOGY 2013; 190:5689-701. [PMID: 23645879 DOI: 10.4049/jimmunol.1202030] [Citation(s) in RCA: 86] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Annexin A1 (AnxA1) is a protein that displays potent anti-inflammatory properties, but its expression in eye tissue and its role in ocular inflammatory diseases have not been well studied. We investigated the mechanism of action and potential uses of AnxA1 and its mimetic peptide (Ac2-26) in the endotoxin-induced uveitis (EIU) rodent model and in human ARPE-19 cells activated by LPS. In rats, analysis of untreated EIU after 24 and 48 h or EIU treated with topical applications or with a single s.c. injection of Ac2-26 revealed the anti-inflammatory actions of Ac2-26 on leukocyte infiltration and on the release of inflammatory mediators; the systemic administration of Boc2, a formylated peptide receptor (fpr) antagonist, abrogated the peptide's protective effects. Moreover, AnxA1(-/-) mice exhibited exacerbated EIU compared with wild-type animals. Immunohistochemical studies of ocular tissue showed a specific AnxA1 posttranslational modification in EIU and indicated that the fpr2 receptor mediated the anti-inflammatory actions of AnxA1. In vitro studies confirmed the roles of AnxA1 and fpr2 and the protective effects of Ac2-26 on the release of chemical mediators in ARPE-19 cells. Molecular analysis of NF-κB translocation and IL-6, IL-8, and cyclooxygenase-2 gene expression indicated that the protective effects of AnxA1 occur independently of the NF-κB signaling pathway and possibly in a posttranscriptional manner. Together, our data highlight the role of AnxA1 in ocular inflammation, especially uveitis, and suggest the use of AnxA1 or its mimetic peptide Ac2-26 as a therapeutic approach.
Collapse
Affiliation(s)
- Ana P Girol
- Department of Biology, Instituto de Biociências, Letras e Ciências Exatas, São Paulo State University, São José do Rio Preto 15054-000, Brazil
| | | | | | | | | | | | | | | |
Collapse
|
39
|
Bowyer JF, Patterson TA, Saini UT, Hanig JP, Thomas M, Camacho L, George NI, Chen JJ. Comparison of the global gene expression of choroid plexus and meninges and associated vasculature under control conditions and after pronounced hyperthermia or amphetamine toxicity. BMC Genomics 2013; 14:147. [PMID: 23497014 PMCID: PMC3602116 DOI: 10.1186/1471-2164-14-147] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2012] [Accepted: 02/21/2013] [Indexed: 01/01/2023] Open
Abstract
BACKGROUND The meninges (arachnoid and pial membranes) and associated vasculature (MAV) and choroid plexus are important in maintaining cerebrospinal fluid (CSF) generation and flow. MAV vasculature was previously observed to be adversely affected by environmentally-induced hyperthermia (EIH) and more so by a neurotoxic amphetamine (AMPH) exposure. Herein, microarray and RT-PCR analysis was used to compare the gene expression profiles between choroid plexus and MAV under control conditions and at 3 hours and 1 day after EIH or AMPH exposure. Since AMPH and EIH are so disruptive to vasculature, genes related to vasculature integrity and function were of interest. RESULTS Our data shows that, under control conditions, many of the genes with relatively high expression in both the MAV and choroid plexus are also abundant in many epithelial tissues. These genes function in transport of water, ions, and solutes, and likely play a role in CSF regulation. Most genes that help form the blood-brain barrier (BBB) and tight junctions were also highly expressed in MAV but not in choroid plexus. In MAV, exposure to EIH and more so to AMPH decreased the expression of BBB-related genes such as Sox18, Ocln, and Cldn5, but they were much less affected in the choroid plexus. There was a correlation between the genes related to reactive oxidative stress and damage that were significantly altered in the MAV and choroid plexus after either EIH or AMPH. However, AMPH (at 3 hr) significantly affected about 5 times as many genes as EIH in the MAV, while in the choroid plexus EIH affected more genes than AMPH. Several unique genes that are not specifically related to vascular damage increased to a much greater extent after AMPH compared to EIH in the MAV (Lbp, Reg3a, Reg3b, Slc15a1, Sct and Fst) and choroid plexus (Bmp4, Dio2 and Lbp). CONCLUSIONS Our study indicates that the disruption of choroid plexus function and damage produced by AMPH and EIH is significant, but the changes may not be as pronounced as they are in the MAV, particularly for AMPH. Expression profiles in the MAV and choroid plexus differed to some extent and differences were not restricted to vascular related genes.
Collapse
Affiliation(s)
- John F Bowyer
- Division of Neurotoxicology, National Center for Toxicological Research, U,S, Food and Drug Administration, Jefferson, AR 72079-9502, USA.
| | | | | | | | | | | | | | | |
Collapse
|
40
|
D'Acquisto F, Piras G, Rattazzi L. Pro-inflammatory and pathogenic properties of Annexin-A1: the whole is greater than the sum of its parts. Biochem Pharmacol 2013; 85:1213-8. [PMID: 23435354 DOI: 10.1016/j.bcp.2013.02.011] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2012] [Revised: 02/05/2013] [Accepted: 02/05/2013] [Indexed: 01/01/2023]
Abstract
According to Aristotle, "The whole is greater than the sum of its parts" and yet, although a long time has passed still, we seem to struggle to accept this universal concept. Searching in the literature for the biological function of Annexin-A1, one would find a wealth of information on its homeostatic and protective anti-inflammatory effects. However, very little has been said on its emerging role in a wide variety of pathological conditions ranging from cancer to autoimmunity. In this commentary, we will focus our attention on this novel pro-inflammatory and pathogenic "dark side" of Annexin-A1. We will summarize our current understanding of the signaling pathways regulated by this protein and link it to clinical and experimental evidences. Finally we will discuss assets and limitations of Annexin-A1 therapeutic strategies. Most importantly, we hope that this commentary will provide scientific support to "controversial" findings one might encounter while studying this fascinating protein.
Collapse
Affiliation(s)
- Fulvio D'Acquisto
- William Harvey Research Institute, Barts and the London School of Medicine and Dentistry, Queen Mary University of London, London, UK.
| | | | | |
Collapse
|
41
|
Expression of annexin-A1 and galectin-1 anti-inflammatory proteins and mRNA in chronic gastritis and gastric cancer. Mediators Inflamm 2013; 2013:152860. [PMID: 23431236 PMCID: PMC3574744 DOI: 10.1155/2013/152860] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2012] [Accepted: 12/28/2012] [Indexed: 12/24/2022] Open
Abstract
Objective. The anti-inflammatory proteins annexin-A1 and galectin-1 have been associated with tumor progression. This scenario prompted us to investigate the relationship between the gene and protein expression of annexin-A1 (ANXA1/AnxA1) and galectin-1 (LGALS1/Gal-1) in an inflammatory gastric lesion as chronic gastritis (CG) and gastric adenocarcinoma (GA) and its association with H. pylori infection. Methods. We analyzed 40 samples of CG, 20 of GA, and 10 of normal mucosa (C) by the quantitative real-time PCR (qPCR) technique and the immunohistochemistry assay. Results. High ANXA1 mRNA expression levels were observed in 90% (36/40) of CG cases (mean relative quantification RQ = 4.26 ± 2.03) and in 80% (16/20) of GA cases (mean RQ = 4.38 ± 4.77). However, LGALS1 mRNA levels were high (mean RQ = 2.44 ± 3.26) in 60% (12/20) of the GA cases, while low expression was found in CG (mean RQ = 0.43 ± 3.13; P < 0.01). Normal mucosa showed modest immunoreactivity in stroma but not in epithelium, while stroma and epithelium displayed an intense immunostaining in CG and GA for both proteins. Conclusion. These results have provided evidence that galectin-1 and mainly annexin-A1 are overexpressed in both gastritis and gastric cancer, suggesting a strong association of these proteins with chronic gastric inflammation and carcinogenesis.
Collapse
|
42
|
Jia Y, Morand EF, Song W, Cheng Q, Stewart A, Yang YH. Regulation of lung fibroblast activation by annexin A1. J Cell Physiol 2013; 228:476-84. [PMID: 22777765 DOI: 10.1002/jcp.24156] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
Annexin-A1 (AnxA1) is a glucocorticoid-induced protein with multiple actions in the regulation of inflammatory cell activation. The contribution of AnxA1 to human cell biology is not well understood. We investigated the contribution of AnxA1 and its receptor, formyl-peptide receptor 2 (FPR2), to the regulation of inflammatory responses in human normal lung fibroblasts (NLF). Silencing constitutive AnxA1 expression in NLF using small interfering RNA (siRNA) was associated with moderate but significant increases in tumor necrosis factor (TNF)-induced proliferation and interleukin (IL)-6 production, accompanied by reduction of ERK and NF-κB activity. AnxA1 regulation of ERK and NF-κB activation was associated with effects on proliferation. Blocking FPR2 using the specific antagonist WRW4 mimicked the effects of AnxA1 silencing on TNF-induced proliferation, IL-6, ERK, and NF-κB activation. AnxA1 silencing also impaired inhibitory effects of glucocorticoid on IL-6 production and on the expression of glucocorticoid-induced leucine zipper (GILZ), but blocking FPR2 failed to mimic these effects of AnxA1 silencing. These data suggest that AnxA1 regulates TNF-induced proliferation and inflammatory responses in lung fibroblasts, via effects on the ERK and NF-κB pathways, which depend on FPR2. AnxA1 also mediates effects of glucocorticoids and GILZ expression, but these effects appear independent of FPR2. These findings suggest that mimicking AnxA1 actions might have therapeutic potential in chronic inflammatory lung diseases.
Collapse
Affiliation(s)
- Yuan Jia
- Centre for Inflammatory Diseases, Southern Clinical School, Monash University Faculty of Medicine Nursing and Health Sciences, Monash Medical Centre, Clayton, Victoria, Australia
| | | | | | | | | | | |
Collapse
|
43
|
Vong L, Ferraz JGP, Dufton N, Panaccione R, Beck PL, Sherman PM, Perretti M, Wallace JL. Up-regulation of Annexin-A1 and lipoxin A(4) in individuals with ulcerative colitis may promote mucosal homeostasis. PLoS One 2012; 7:e39244. [PMID: 22723974 PMCID: PMC3377644 DOI: 10.1371/journal.pone.0039244] [Citation(s) in RCA: 76] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2012] [Accepted: 05/17/2012] [Indexed: 12/16/2022] Open
Abstract
Background One of the characteristics of an active episode of ulcerative colitis (UC) is the intense mucosal infiltration of leukocytes. The pro-resolution mediators Annexin-A1 (AnxA1) and lipoxin A4 (LXA4) exert counter-regulatory effects on leukocyte recruitment, however to date, the dual/cumulative effects of these formyl peptide receptor-2 (FPR2/ALX) agonists in the context of human intestinal diseases are unclear. To define the contribution of these mediators, we measured their expression in biopsies from individuals with UC. Methods Colonic mucosal biopsies were collected from two broad patient groups: healthy volunteers without (‘Ctrl’ n = 20) or with a prior history of UC (‘hx of UC’ n = 5); individuals with UC experiencing active disease (‘active’ n = 8), or in medically-induced remission (‘remission’ n = 16). We assessed the mucosal expression of LXA4, AnxA1, and the FPR2/ALX receptor in each patient group using a combination of fluorescence microscopy, biochemical and molecular analyses. Results Mucosal expression of LXA4 was elevated exclusively in biopsies from individuals in remission (3-fold, P<0.05 vs. Ctrl). Moreover, in this same group we observed an upregulation of AnxA1 protein expression (2.5-fold increase vs. Ctrl, P<.01), concurrent with an increased level of macrophage infiltration, and an elevation in FPR2/ALX mRNA (7-fold increase vs. Ctrl, P<.05). Importantly, AnxA1 expression was not limited to cells infiltrating the lamina propria but was also detected in epithelial cells lining the intestinal crypts. Conclusions Our results demonstrate a specific up-regulation of this pro-resolution circuit in individuals in remission from UC, and suggest a significant role for LXA4 and AnxA1 in promoting mucosal homeostasis.
Collapse
Affiliation(s)
- Linda Vong
- Hospital for Sick Children, Research Institute, Toronto, Ontario, Canada.
| | | | | | | | | | | | | | | |
Collapse
|
44
|
Mimura KK, Tedesco RC, Calabrese KS, Gil CD, Oliani SM. The involvement of anti-inflammatory protein, annexin A1, in ocular toxoplasmosis. Mol Vis 2012; 18:1583-93. [PMID: 22740770 PMCID: PMC3382303] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2012] [Accepted: 06/12/2012] [Indexed: 11/13/2022] Open
Abstract
PURPOSE The aim of this study was to evaluate the expression of the protein annexin A1 (ANXA1), a potent endogenous regulator of the inflammatory process, in ocular toxoplasmosis. METHODS C57BL/6 female mice were infected using intravitreal injections of either 10(6) tachyzoites of Toxoplasma gondii (RH strain; T. gondii) or PBS only (control groups). After 24, 48, and 72 h, animals were sacrificed and their eyes were harvested for histopathological, immunohistochemical, and ultrastructural immunocytochemical analysis of ANXA1. Human retinal pigment epithelial (RPE) cells (ARPE-19) were infected in vitro with T. gondii and collected after 60, 120, 240 min, and 24 h. RESULTS Compared with non-infected eyes, an intense inflammatory response was observed in the anterior (24 h after infection) and posterior segments (72 h after infection) of the infected eye, characterized by neutrophil infiltration and by the presence of tachyzoites and their consequent destruction along with disorganization of normal retina architecture and RPE vacuolization. T. gondii infection was associated with a significant increase of ANXA1 expression in the neutrophils at 24, 48, and 72 h, and in the RPE at 48 and 72 h. In vitro studies confirmed an upregulation of ANXA1 levels in RPE cells, after 60 and 120 min of infection with T. gondii. CONCLUSIONS The positive modulation of endogenous ANXA1 in the inflammatory and RPE cells during T. gondii infection suggests that this protein may serve as a therapeutic target in ocular toxoplasmosis.
Collapse
Affiliation(s)
- Kallyne K. Mimura
- From the Post-Graduation Structural and Functional Biology, Federal University of São Paulo (UNIFESP), São Paulo, SP, Brazil
| | - Roberto C. Tedesco
- Department of Morphology and Genetics, Federal University of São Paulo (UNIFESP), São Paulo, SP, Brazil
| | - Katia S. Calabrese
- Laboratório de Imunomodulação e Protozoologia, Instituto Oswaldo Cruz/FIOCRUZ, Rio de Janeiro, RJ, Brazil
| | - Cristiane D. Gil
- Department of Morphology and Genetics, Federal University of São Paulo (UNIFESP), São Paulo, SP, Brazil
| | - Sonia M. Oliani
- From the Post-Graduation Structural and Functional Biology, Federal University of São Paulo (UNIFESP), São Paulo, SP, Brazil,Department of Biology, Instituto de Biociências, Letras e Ciências Exatas (IBILCE), São Paulo State University (UNESP), São José do Rio Preto, SP, Brazil
| |
Collapse
|
45
|
Patel HB, Kornerup KN, Sampaio ALF, D'Acquisto F, Seed MP, Girol AP, Gray M, Pitzalis C, Oliani SM, Perretti M. The impact of endogenous annexin A1 on glucocorticoid control of inflammatory arthritis. Ann Rheum Dis 2012; 71:1872-80. [PMID: 22562975 PMCID: PMC3440300 DOI: 10.1136/annrheumdis-2011-201180] [Citation(s) in RCA: 66] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
Abstract
Objectives To establish the role and effect of glucocorticoids and the endogenous annexin A1 (AnxA1) pathway in inflammatory arthritis. Methods Ankle joint mRNA and protein expression of AnxA1 and its receptors were analysed in naive and arthritic mice by real-time PCR and immunohistochemistry. Inflammatory arthritis was induced with the K/BxN arthritogenic serum in AnxA1+/+ and AnxA1−/− mice; in some experiments, animals were treated with dexamethasone (Dex) or with human recombinant AnxA1 or a protease-resistant mutant (termed SuperAnxA1). Readouts were arthritic score, disease incidence, paw oedema and histopathology, together with pro-inflammatory gene expression. Results All elements of the AnxA1 pathway could be detected in naive joints, with augmentation during ongoing disease, due to the infiltration of immune cells. No difference in arthritis intensity of profile could be observed between AnxA1+/+ and AnxA1−/− mice. Treatment of mice with Dex (10 µg intraperitoneally daily from day 2) afforded potent antiarthritic effects highly attenuated in the knockouts: macroscopic changes were mirrored by histopathological findings and pro-inflammatory gene (eg, Nos2) expression. Presence of proteinase 3 mRNA in the arthritic joints led the authors to test AnxA1 and the mutant SuperAnxA1 (1 µg intraperitoneally daily in both cases from day 2), with the latter one being able to accelerate the resolving phase of the disease. Conclusion AnxA1 is an endogenous determinant for the therapeutic efficacy of Dex in inflammatory arthritis. Such an effect can be partially mimicked by application of SuperAnxA1 which may represent the starting point for novel antiarthritic therapeutic strategies.
Collapse
Affiliation(s)
- Hetal B Patel
- William Harvey Research Institute, Barts and The London School of Medicine, London UK
| | | | | | | | | | | | | | | | | | | |
Collapse
|
46
|
Waechter V, Schmid M, Herova M, Weber A, Günther V, Marti-Jaun J, Wüst S, Rösinger M, Gemperle C, Hersberger M. Characterization of the Promoter and the Transcriptional Regulation of the Lipoxin A4 Receptor (FPR2/ALX) Gene in Human Monocytes and Macrophages. THE JOURNAL OF IMMUNOLOGY 2012; 188:1856-67. [DOI: 10.4049/jimmunol.1101788] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/17/2023]
|
47
|
Blume KE, Soeroes S, Keppeler H, Stevanovic S, Kretschmer D, Rautenberg M, Wesselborg S, Lauber K. Cleavage of annexin A1 by ADAM10 during secondary necrosis generates a monocytic "find-me" signal. THE JOURNAL OF IMMUNOLOGY 2011; 188:135-45. [PMID: 22116825 DOI: 10.4049/jimmunol.1004073] [Citation(s) in RCA: 67] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Annexin A1 is an intracellular calcium/phospholipid-binding protein that is involved in membrane organization and the regulation of the immune system. It has been attributed an anti-inflammatory role at various control levels, and recently we could show that annexin A1 externalization during secondary necrosis provides an important fail-safe mechanism counteracting inflammatory responses when the timely clearance of apoptotic cells has failed. As such, annexin A1 promotes the engulfment of dying cells and dampens the postphagocytic production of proinflammatory cytokines. In our current follow-up study, we report that exposure of annexin A1 during secondary necrosis coincided with proteolytic processing within its unique N-terminal domain by ADAM10. Most importantly, we demonstrate that the released peptide and culture supernatants of secondary necrotic, annexin A1-externalizing cells induced chemoattraction of monocytes, which was clearly reduced in annexin A1- or ADAM10-knockdown cells. Thus, altogether our findings indicate that annexin A1 externalization and its proteolytic processing into a chemotactic peptide represent final events during apoptosis, which after the transition to secondary necrosis contribute to the recruitment of monocytes and the prevention of inflammation.
Collapse
Affiliation(s)
- Karin E Blume
- Department of Internal Medicine I, Eberhard Karls University, Tuebingen, Germany
| | | | | | | | | | | | | | | |
Collapse
|
48
|
Araujo LP, Truzzi RR, Mendes GEF, Luz MAM, Burdmann EA, Oliani SM. Annexin A1 protein attenuates cyclosporine-induced renal hemodynamics changes and macrophage infiltration in rats. Inflamm Res 2011; 61:189-96. [PMID: 22101490 DOI: 10.1007/s00011-011-0400-z] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2011] [Revised: 10/30/2011] [Accepted: 11/02/2011] [Indexed: 01/27/2023] Open
Abstract
BACKGROUND Cyclosporine (CsA) remains an important immunosuppressant for transplantation and for treatment of autoimmune diseases. The most troublesome side effect of CsA is renal injury. Acute CsA-induced nephrotoxicity is characterized by reduced renal blood flow (RBF) and glomerular filtration rate (GFR) due to afferent arteriole vasoconstriction. Annexin A1 (ANXA1) is a potent anti-inflammatory protein with protective effect in renal ischemia/reperfusion injury. Here we study the effects of ANXA1 treatment in an experimental model of acute CsA nephrotoxicity. METHODS Salt-depleted rats were randomized to treatment with VH (vehicles 1 mL/kg body weight/day), ANXA1 (Ac2-26 peptide 1 mg/kg body weight/day intraperitoneally), CsA (20 mg/kg body weight/day subcutaneously) and CsA + ANXA1 (combination) for seven days. We compared renal function and hemodynamics, renal histopathology, renal tissue macrophage infiltration and renal ANXA1 expression between the four groups. RESULTS CsA significantly impaired GFR and RBF, caused tubular dilation and macrophage infiltration and increased ANXA1 renal tissue expression. Treatment with ANXA1 attenuated CSA-induced hemodynamic changes, tubular injury and macrophage infiltration. CONCLUSION ANXA1 treatment attenuated renal hemodynamic injury and inflammation in an acute CsA nephrotoxicity model.
Collapse
Affiliation(s)
- Leandro Pires Araujo
- Post-Graduation Program in Morphology, Federal University of São Paulo, São Paulo, SP, Brazil
| | | | | | | | | | | |
Collapse
|
49
|
Dalli J, Jones CP, Cavalcanti DM, Farsky SH, Perretti M, Rankin SM. Annexin A1 regulates neutrophil clearance by macrophages in the mouse bone marrow. FASEB J 2011; 26:387-96. [PMID: 21957127 PMCID: PMC3250241 DOI: 10.1096/fj.11-182089] [Citation(s) in RCA: 65] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Under homeostatic conditions, a proportion of senescent CXCR4(hi) neutrophils home from the circulation back to the bone marrow, where they are phagocytosed by bone marrow macrophages. In this study, we have identified an unexpected role for the anti-inflammatory molecule annexin A1 (AnxA1) as a critical regulator of this process. We first observed that AnxA1(-/-) mice have significantly increased neutrophil numbers in their bone marrow while having normal levels of GM and G colony-forming units, monocytes, and macrophages. Although AnxA1(-/-) mice have more neutrophils in the bone marrow, a greater proportion of these cells are senescent, as determined by their higher levels of CXCR4 expression and annexin V binding. Consequently, bone marrow neutrophils from AnxA1(-/-) mice exhibit a reduced migratory capacity in vitro. Studies conducted in vitro also show that expression of AnxA1 is required for bone marrow macrophages, but not peritoneal macrophages, to phagocytose apoptotic neutrophils. Moreover, in vivo experiments indicate a defect in clearance of wild-type neutrophils in the bone marrow of AnxA1(-/-) mice. Thus, we conclude that expression of AnxA1 by resident macrophages is a critical determinant for neutrophil clearance in the bone marrow.
Collapse
Affiliation(s)
- Jesmond Dalli
- The William Harvey Research Institute, Barts and The London School of Medicine, London, UK
| | | | | | | | | | | |
Collapse
|
50
|
Zhang XN, Zhang XY, Cao XH. Advances in understanding the relationship between annexin A1 and gastrointestinal cancer. Shijie Huaren Xiaohua Zazhi 2011; 19:2160-2165. [DOI: 10.11569/wcjd.v19.i20.2160] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Annexin A1 is a member of the annexin family of calcium-dependent phospholipid-binding proteins and participates in many important life processes, such as cellular signal transduction, proliferation, differentiation and apoptosis. Recent studies have shown that the expression levels of annexin A1 vary among different tumor tissues and different tumor subtypes and may be associated with the development, invasion and metastasis of malignant tumors. Understanding the relationship between annexin A1 and tumors has important implications for the early diagnosis and treatment of tumors.
Collapse
|