1
|
Tian X, Chen J, Hong Y, Cao Y, Xiao J, Zhu Y. Exploring the Role of Macrophages and Their Associated Structures in Rheumatoid Arthritis. J Innate Immun 2025; 17:95-111. [PMID: 39938504 PMCID: PMC11820663 DOI: 10.1159/000543444] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2024] [Accepted: 01/02/2025] [Indexed: 02/14/2025] Open
Abstract
BACKGROUND Rheumatoid arthritis (RA) is a chronic, invasive autoimmune disease characterized by symmetrical polyarthritis involving synovial inflammation. Epidemiological studies indicate that the incidence of RA continues to rise, yet the pathogenesis of this disease remains not fully understood. A significant infiltration of macrophages is observed in the synovium of RA patients. It can be inferred that macrophages likely play a crucial role in the onset and progression of RA. SUMMARY This review aims to summarize the research progress on the mechanisms by which macrophages and their associated structures contribute to RA, as well as potential therapeutic approaches, aiming to provide new insights into the study of RA pathogenesis and its clinical treatment. KEY MESSAGES During the course of RA, besides the inherent roles of macrophages, these cells respond to microenvironmental changes such as pathogen invasion or tissue damage by undergoing polarization, pyroptosis, or forming macrophage extracellular traps (METs), all of which influence inflammatory responses and immune homeostasis, thereby mediating the occurrence and development of RA. Additionally, macrophages secrete exosomes, which participate in intercellular communication and signal transduction processes, thus contributing to the progression of RA. Therefore, it is critical to elucidate how macrophages and their related structures function in RA. BACKGROUND Rheumatoid arthritis (RA) is a chronic, invasive autoimmune disease characterized by symmetrical polyarthritis involving synovial inflammation. Epidemiological studies indicate that the incidence of RA continues to rise, yet the pathogenesis of this disease remains not fully understood. A significant infiltration of macrophages is observed in the synovium of RA patients. It can be inferred that macrophages likely play a crucial role in the onset and progression of RA. SUMMARY This review aims to summarize the research progress on the mechanisms by which macrophages and their associated structures contribute to RA, as well as potential therapeutic approaches, aiming to provide new insights into the study of RA pathogenesis and its clinical treatment. KEY MESSAGES During the course of RA, besides the inherent roles of macrophages, these cells respond to microenvironmental changes such as pathogen invasion or tissue damage by undergoing polarization, pyroptosis, or forming macrophage extracellular traps (METs), all of which influence inflammatory responses and immune homeostasis, thereby mediating the occurrence and development of RA. Additionally, macrophages secrete exosomes, which participate in intercellular communication and signal transduction processes, thus contributing to the progression of RA. Therefore, it is critical to elucidate how macrophages and their related structures function in RA.
Collapse
Affiliation(s)
- Xin Tian
- The Geriatrics, Graduate School of Anhui University of Chinese Medicine, Hefei, China
| | - Jingjing Chen
- The Geriatrics, Graduate School of Anhui University of Chinese Medicine, Hefei, China
| | - Yujie Hong
- The Geriatrics, Graduate School of Anhui University of Chinese Medicine, Hefei, China
| | - Yang Cao
- The Geriatrics, Graduate School of Anhui University of Chinese Medicine, Hefei, China
| | - Jing Xiao
- The Geriatrics, Graduate School of Anhui University of Chinese Medicine, Hefei, China
| | - Yan Zhu
- The Geriatrics, Graduate School of Anhui University of Chinese Medicine, Hefei, China
- The Geriatrics, The Second Affiliated Hospital of Anhui University of Chinese Medicine, Hefei, China
| |
Collapse
|
2
|
Huang X, Yi N, Zhu P, Gao J, Lv J. Sorafenib-induced macrophage extracellular traps via ARHGDIG/IL4/PADI4 axis confer drug resistance through inhibiting ferroptosis in hepatocellular carcinoma. Biol Direct 2024; 19:110. [PMID: 39529192 PMCID: PMC11555812 DOI: 10.1186/s13062-024-00560-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2024] [Accepted: 11/04/2024] [Indexed: 11/16/2024] Open
Abstract
BACKGROUND Hepatocellular carcinoma (HCC) is one of the most common as well as leading causes of mortality worldwide, and sorafenib is the first-line treatment in HCC patients. Unfortunately, drug resistance to sorafenib often develops. However, the underlying mechanism remains unclear. Here, we reveal the important role of macrophage extracellular traps (METs)-mediated crosstalk between macrophages and tumor cells in sorafenib resistance. METHODS METs in HCC tumor tissues were detected using immunofluorescence. The concentrations of MPO-DNA, elastase and cytokines were measured using ELISA. The mRNA expression levels of genes were confirmed by qRT-PCR. The siRNAs were conducted to knock ARHGDIG in Hepa1-6 and Hep3B cells. Western Blot assay was performed to determine protein expression of Rho GDP dissociation inhibitor gamma (ARHGDIG, or RHOGDI-3), PADI2, and PADI4. Cell viability and migration were evaluated by CCK-8 assay and transwell assay, respectively. Cell ferroptosis was assessed by measurement of Fe2+ concentration, flow cytometry assay of lipid ROS, and western blot assay of GPX4. The functions of sorafenib, DNase I, IL4 neutralization antibody and GPX4 in tumor growth were explored through in vivo experiments. RESULTS Sorafenib induced MET formation in M2 macrophages rather than M1 macrophages derived from both human and mice. In Hepa1-6 HCC mice, METs clearance by DNase I improved response to sorafenib therapy, detected by tumor weight, tumor growth curve, tumor volume, and survival. By screening candidate cytokines that affect macrophage function, we found that sorafenib-promoting IL4 secretion by HCC cells plays a crucial role in sorafenib-induced MET formation. Understanding the critical role of IL4 in sorafenib-induced MET formation led us to find that IL4 neutralization significantly improved the efficiency of sorafenib in HCC models. Mechanistically, we discovered that sorafenib increased the expression of ARHGDIG in HCC cells, which led to the release of IL4. In M2 macrophages, IL4 triggered MET formation by elevating the mRNA and protein expression of peptidyl arginine deiminase 4 (PADI4) rather than PADI2. In HCC models, GSK484 inhibition of PADI4 could consistently weaken sorafenib resistance and improve sorafenib efficiency. Importantly, we discovered that METs contribute to sorafenib resistance by inhibiting the ferroptosis of HCC cells. Meanwhile, PADI4 inhibition or DNase I could reverse the sorafenib resistance caused by METs-inhibiting ferroptosis of HCC cells. CONCLUSION Our study concludes that sorafenib-induced METs inhibit the ferroptosis of tumor cells, suggesting that targeting the IL4/PADI4/METs axis in HCC could reduce or prevent sorafenib resistance.
Collapse
Affiliation(s)
- Xiangbo Huang
- Department of Clinical Laboratory, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, People's Republic of China
- Key Clinical Laboratory of Henan Province, Zhengzhou, 450052, People's Republic of China
| | - Nan Yi
- Department of Clinical Laboratory, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, People's Republic of China
- Key Clinical Laboratory of Henan Province, Zhengzhou, 450052, People's Republic of China
| | - Pengfei Zhu
- Department of Clinical Laboratory, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, People's Republic of China
- Key Clinical Laboratory of Henan Province, Zhengzhou, 450052, People's Republic of China
| | - Jian Gao
- State Key Laboratory of Systems Medicine for Cancer, Shanghai Cancer Institute, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200032, People's Republic of China.
| | - Jun Lv
- Department of Infectious Diseases, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, People's Republic of China.
| |
Collapse
|
3
|
Shen Y, Shi R, Lu S, Wang Y, Zhou Z, Wu C, You Q, Fan H, Wu J. Role of Peptidyl Arginine Deiminase 4-Dependent Macrophage Extracellular Trap Formation in Type 1 Diabetes Pathogenesis. Diabetes 2024; 73:1862-1874. [PMID: 39137121 DOI: 10.2337/db23-1000] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/15/2023] [Accepted: 08/06/2024] [Indexed: 08/15/2024]
Abstract
Excessive formation of macrophage extracellular trap (MET) has been implicated in several autoimmune disease pathogeneses; however, its impact on type 1 diabetes (T1D) and related mechanisms remains enigmatic. We demonstrated the pivotal role of peptidyl arginine deiminase 4 (PAD4) in driving profuse MET formation and macrophage M1 polarization in intestinal inflammation in NOD mice. Genetic knockout of PAD4 or adoptive transfer of METs altered the proportion of proinflammatory T cells in the intestine, subsequently influencing their migration to the pancreas. Combining RNA sequencing and CUT&Tag analysis, we found activated PAD4 transcriptionally regulated CXCL10 expression. This study comprehensively investigated how excessive PAD4-mediated MET formation in the colon increases the aggravation of intestinal inflammation and proinflammatory T-cell migration and finally is involved in T1D progression, suggesting that inhibition of MET formation may be a potential therapeutic target in T1D. ARTICLE HIGHLIGHTS
Collapse
Affiliation(s)
- Yiming Shen
- School of Life Science and Technology, China Pharmaceutical University, Nanjing, China
| | - Ruiya Shi
- School of Life Science and Technology, China Pharmaceutical University, Nanjing, China
| | - ShiPing Lu
- Center for Translational Research in Infection and Inflammation, Tulane University, New Orleans, LA
| | - Yan Wang
- School of Life Science and Technology, China Pharmaceutical University, Nanjing, China
| | - Ziqi Zhou
- School of Life Science and Technology, China Pharmaceutical University, Nanjing, China
| | - Chenhua Wu
- School of Life Science and Technology, China Pharmaceutical University, Nanjing, China
| | - Qi You
- School of Life Science and Technology, China Pharmaceutical University, Nanjing, China
| | - Hongye Fan
- School of Life Science and Technology, China Pharmaceutical University, Nanjing, China
| | - Jie Wu
- School of Life Science and Technology, China Pharmaceutical University, Nanjing, China
| |
Collapse
|
4
|
Lv X, Han Y, Li Y, Wang X, Zhang T, Wang X, Zhang Q, Yang D, Zhao J. Nonylphenol displays immunotoxicity by triggering hemocyte extracellular traps in Manila clam via ROS burst, ERK pathway and glycolysis. ECOTOXICOLOGY AND ENVIRONMENTAL SAFETY 2024; 285:117145. [PMID: 39357378 DOI: 10.1016/j.ecoenv.2024.117145] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/21/2024] [Revised: 09/28/2024] [Accepted: 09/30/2024] [Indexed: 10/04/2024]
Abstract
Nonylphenol (NP), an endocrine disruptor, has been demonstrated to be a harmful environmental contaminant and toxic to organisms. In this study, to address concerns regarding the immunotoxicity of NP, we treated clam Ruditapes philippinarum hemocytes with NP in vitro and explored the underlying mechanisms of NP-induced extracellular traps (ETs). NP could induce the formation of hemocytes ETs in a dose-dependent manner. Transcriptomics analysis revealed changes of signaling pathway involved in immunity and energy metabolism in hemocytes after NP stimulation. In this process, both reactive oxygen species (ROS) and myeloperoxidase (MPO) were up-regulated. Moreover, mitogen-activated protein kinase (MAPK) signaling pathway was proved to be activated in the formation of NP-induced ETs, manifested as enhanced phosphorylation of extracellular signal-regulated kinase (ERK) but not p38 or c-Jun N-terminal kinase (JNK). In the presence of U0126, an ERK phosphorylation inhibitor, the NP-induced expression of NADPH oxidase enzyme (NOX) was significantly decreased, which further alleviated the ROS production and ultimately limited the release of ETs. NP exposure increased glucose uptake, along with enhanced activities of glycolysis-related enzymes such as hexokinase (HK) and pyruvate kinase (PK). After inhibiting glycolysis by the inhibitor 2-DG, the formation of NP-induced ETs was significantly suppressed. ERK could regulate mTOR signaling and the PI3K/AKT pathway, potentially directing ETs formation by orchestrating the glycolysis through the activation of key transcription factors c-Myc and HIF-1α. Collectively, the results preliminary confirm that the ERK-NOX-ROS axis and glycolysis are involved in NP-induced ETs formation, contributing to the cellular immunotoxicity in clam.
Collapse
Affiliation(s)
- Xiaojing Lv
- Research and Development Center for Efficient Utilization of Coastal Bioresources, Yantai Institute of Coastal Zone Research, Chinese Academy of Sciences, Yantai 264003, PR China; Muping Coastal Environment Research Station, Yantai Institute of Coastal Zone Research, Chinese Academy of Sciences, Yantai 264003, PR China; University of Chinese Academy of Sciences, Beijing 100049, PR China
| | - Yijing Han
- School of Fisheries, Ludong University, Yantai 264025, PR China
| | - Yongxue Li
- Research and Development Center for Efficient Utilization of Coastal Bioresources, Yantai Institute of Coastal Zone Research, Chinese Academy of Sciences, Yantai 264003, PR China; Muping Coastal Environment Research Station, Yantai Institute of Coastal Zone Research, Chinese Academy of Sciences, Yantai 264003, PR China
| | - Xin Wang
- Research and Development Center for Efficient Utilization of Coastal Bioresources, Yantai Institute of Coastal Zone Research, Chinese Academy of Sciences, Yantai 264003, PR China; Muping Coastal Environment Research Station, Yantai Institute of Coastal Zone Research, Chinese Academy of Sciences, Yantai 264003, PR China
| | - Tianyu Zhang
- Research and Development Center for Efficient Utilization of Coastal Bioresources, Yantai Institute of Coastal Zone Research, Chinese Academy of Sciences, Yantai 264003, PR China; Muping Coastal Environment Research Station, Yantai Institute of Coastal Zone Research, Chinese Academy of Sciences, Yantai 264003, PR China
| | - Xiaodan Wang
- Research and Development Center for Efficient Utilization of Coastal Bioresources, Yantai Institute of Coastal Zone Research, Chinese Academy of Sciences, Yantai 264003, PR China; Muping Coastal Environment Research Station, Yantai Institute of Coastal Zone Research, Chinese Academy of Sciences, Yantai 264003, PR China
| | - Qianqian Zhang
- Research and Development Center for Efficient Utilization of Coastal Bioresources, Yantai Institute of Coastal Zone Research, Chinese Academy of Sciences, Yantai 264003, PR China; Muping Coastal Environment Research Station, Yantai Institute of Coastal Zone Research, Chinese Academy of Sciences, Yantai 264003, PR China
| | - Dinglong Yang
- Research and Development Center for Efficient Utilization of Coastal Bioresources, Yantai Institute of Coastal Zone Research, Chinese Academy of Sciences, Yantai 264003, PR China; Laboratory for Marine Biology and Biotechnology, Qingdao Marine Science and Technology Center, Qingdao 266237, PR China; Muping Coastal Environment Research Station, Yantai Institute of Coastal Zone Research, Chinese Academy of Sciences, Yantai 264003, PR China.
| | - Jianmin Zhao
- Research and Development Center for Efficient Utilization of Coastal Bioresources, Yantai Institute of Coastal Zone Research, Chinese Academy of Sciences, Yantai 264003, PR China; Laboratory for Marine Biology and Biotechnology, Qingdao Marine Science and Technology Center, Qingdao 266237, PR China; Muping Coastal Environment Research Station, Yantai Institute of Coastal Zone Research, Chinese Academy of Sciences, Yantai 264003, PR China.
| |
Collapse
|
5
|
Abacar K, Macleod T, Direskeneli H, McGonagle D. How underappreciated autoinflammatory (innate immunity) mechanisms dominate disparate autoimmune disorders. Front Immunol 2024; 15:1439371. [PMID: 39372419 PMCID: PMC11449752 DOI: 10.3389/fimmu.2024.1439371] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2024] [Accepted: 09/04/2024] [Indexed: 10/08/2024] Open
Abstract
Historically inflammation against self was considered autoimmune which stems back to the seminal observations by Ehrlich who described serum factors, now known to be autoantibodies produced by B lineage cells that mediate "horror autotoxicus". The 20th century elucidation of B- and T-cell adaptive immune responses cemented the understanding of the key role of adaptive immune responses in mediating pathology against self. However, Mechnikov shared the Nobel Prize for the discovery of phagocytosis, the most rudimentary aspect of innate immunity. Fast forward some 100 years and an immunogenetic understanding of innate immunity led to the categorising of innate immunopathology under the umbrella term 'auto inflammation' and terminology such as "horror autoinflammaticus" to highlight the schism from the classical adaptive immune understanding of autoimmunity. These concepts lead to calls for a two-tiered classification of inflammation against self, but just as innate and adaptive immunity are functionally integrated, so is immunopathology in many settings and the concept of an autoimmune to autoinflammation continuum emerged with overlaps between both. Herein we describe several historically designated disorders of adaptive immunity where innate immunity is key, including rheumatoid arthritis (RA), systemic lupus erythematosus (SLE), systemic juvenile idiopathic arthritis (sJIA) and adult-onset Still's disease (AOSD) where the immunopathology phenotype is strongly linked to major histocompatibility complex (MHC) class II associations and responds to drugs that target T-cells. We also consider MHC-I-opathies including psoriasis and Behcet's disease(BD) that are increasingly viewed as archetype CD8 T-cell related disorders. We also briefly review the key role of barrier dysfunction in eczema and ulcerative colitis (UC) where innate tissue permeability barrier dysfunction and microbial dysbiosis contributes to prominent adaptive immune pathological mechanisms. We also highlight the emerging roles of intermediate populations of lymphocytes including gamma delta (γδ) and mucosal-associated invariant T (MAIT) cells that represent a blend of adaptive immune plasticity and innate immune rapid responders that may also determine site specific patterns of inflammation.
Collapse
Affiliation(s)
- Kerem Abacar
- Department of Internal Medicine, Division of Rheumatology, Marmara University School of Medicine, Istanbul, Türkiye
- Leeds Institute of Rheumatic and Musculoskeletal Medicine, University of Leeds, Leeds, United Kingdom
| | - Tom Macleod
- Department of Internal Medicine, Division of Rheumatology, Marmara University School of Medicine, Istanbul, Türkiye
| | - Haner Direskeneli
- Leeds Institute of Rheumatic and Musculoskeletal Medicine, University of Leeds, Leeds, United Kingdom
| | - Dennis McGonagle
- Department of Internal Medicine, Division of Rheumatology, Marmara University School of Medicine, Istanbul, Türkiye
- National Institute for Health Research, Leeds Biomedical Research Centre, Leeds Teaching Hospitals, Leeds, United Kingdom
| |
Collapse
|
6
|
Laha A, Nasra S, Bhatia D, Kumar A. Advancements in rheumatoid arthritis therapy: a journey from conventional therapy to precision medicine via nanoparticles targeting immune cells. NANOSCALE 2024; 16:14975-14993. [PMID: 39056352 DOI: 10.1039/d4nr02182g] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/28/2024]
Abstract
Rheumatoid arthritis (RA) is a progressive autoimmune disease that mainly affects the inner lining of the synovial joints and leads to chronic inflammation. While RA is not known as lethal, recent research indicates that it may be a silent killer because of its strong association with an increased risk of chronic lung and heart diseases. Patients develop these systemic consequences due to the regular uptake of heavy drugs such as disease-modifying antirheumatic medications (DMARDs), glucocorticoids (GCs), nonsteroidal anti-inflammatory medicines (NSAIDs), etc. Nevertheless, a number of these medications have off-target effects, which might cause adverse toxicity, and have started to become resistant in patients as well. Therefore, alternative and promising therapeutic techniques must be explored and adopted, such as post-translational modification inhibitors (like protein arginine deiminase inhibitors), RNA interference by siRNA, epigenetic drugs, peptide therapy, etc., specifically in macrophages, neutrophils, Treg cells and dendritic cells (DCs). As the target cells are specific, ensuring targeted delivery is also equally important, which can be achieved with the advent of nanotechnology. Furthermore, these nanocarriers have fewer off-site side effects, enable drug combinations, and allow for lower drug dosages. Among the nanoparticles that can be used for targeting, there are both inorganic and organic nanomaterials such as solid-lipid nanoparticles, liposomes, hydrogels, dendrimers, and biomimetics that have been discussed. This review highlights contemporary therapy options targeting macrophages, neutrophils, Treg cells, and DCs and explores the application of diverse nanotechnological techniques to enhance precision RA therapies.
Collapse
Affiliation(s)
- Anwesha Laha
- Biological and Life Sciences, School of Arts & Sciences, Ahmedabad University, Central Campus, Navrangpura, Ahmedabad 380009, Gujarat, India.
| | - Simran Nasra
- Biological and Life Sciences, School of Arts & Sciences, Ahmedabad University, Central Campus, Navrangpura, Ahmedabad 380009, Gujarat, India.
| | - Dhiraj Bhatia
- Indian Institute of Technology Gandhinagar, Palaj, Gandhinagar - 382055, Gujarat, India
| | - Ashutosh Kumar
- Biological and Life Sciences, School of Arts & Sciences, Ahmedabad University, Central Campus, Navrangpura, Ahmedabad 380009, Gujarat, India.
| |
Collapse
|
7
|
Gu W, Huang C, Chen G, Kong W, Zhao L, Jie H, Zhen G. The role of extracellular traps released by neutrophils, eosinophils, and macrophages in asthma. Respir Res 2024; 25:290. [PMID: 39080638 PMCID: PMC11290210 DOI: 10.1186/s12931-024-02923-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2024] [Accepted: 07/22/2024] [Indexed: 08/02/2024] Open
Abstract
Extracellular traps (ETs) are a specialized form of innate immune defense in which leukocytes release ETs composed of chromatin and active proteins to eliminate pathogenic microorganisms. In addition to the anti-infection effect of ETs, researchers have also discovered their involvement in the pathogenesis of inflammatory disease, tumors, autoimmune disease, and allergic disease. Asthma is a chronic airway inflammatory disease involving multiple immune cells. The increased level of ETs in asthma patients suggests that ETs play an important role in the pathogenesis of asthma. Here we review the research work on the formation mechanism, roles, and therapeutic strategies of ETs released by neutrophils, eosinophils, and macrophages in asthma.
Collapse
Affiliation(s)
- Wei Gu
- Division of Respiratory and Critical Care Medicine, Department of Internal Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
- Key Laboratory of Respiratory Diseases, National Health Commission of People's Republic of China, Wuhan, China
| | - Chunli Huang
- Division of Respiratory and Critical Care Medicine, Department of Internal Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
- Key Laboratory of Respiratory Diseases, National Health Commission of People's Republic of China, Wuhan, China
| | - Gongqi Chen
- Division of Respiratory and Critical Care Medicine, Department of Internal Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
- Key Laboratory of Respiratory Diseases, National Health Commission of People's Republic of China, Wuhan, China
| | - Weiqiang Kong
- Division of Respiratory and Critical Care Medicine, Department of Internal Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
- Key Laboratory of Respiratory Diseases, National Health Commission of People's Republic of China, Wuhan, China
| | - Lu Zhao
- Division of Respiratory and Critical Care Medicine, Department of Internal Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
- Key Laboratory of Respiratory Diseases, National Health Commission of People's Republic of China, Wuhan, China
| | - Huiru Jie
- Division of Respiratory and Critical Care Medicine, Department of Internal Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
- Key Laboratory of Respiratory Diseases, National Health Commission of People's Republic of China, Wuhan, China
| | - Guohua Zhen
- Division of Respiratory and Critical Care Medicine, Department of Internal Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China.
- Key Laboratory of Respiratory Diseases, National Health Commission of People's Republic of China, Wuhan, China.
| |
Collapse
|
8
|
Bircher JS, Denorme F, Cody MJ, de Araujo CV, Petrey AC, Middleton EA, Campbell RA, Yost CC. Neonatal NET-inhibitory factor inhibits macrophage extracellular trap formation. Blood Adv 2024; 8:3686-3690. [PMID: 38810257 PMCID: PMC11284700 DOI: 10.1182/bloodadvances.2024013094] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2024] [Revised: 05/06/2024] [Accepted: 05/19/2024] [Indexed: 05/31/2024] Open
Affiliation(s)
- Joseph S. Bircher
- Department of Pediatrics, The University of Utah, Salt Lake City, UT
| | - Frederik Denorme
- Department of Emergency Medicine, Washington University School of Medicine, St. Louis, MO
| | - Mark J. Cody
- Department of Pediatrics, The University of Utah, Salt Lake City, UT
- Molecular Medicine Program, The University of Utah, Salt Lake City, UT
| | - Claudia V. de Araujo
- Department of Pediatrics, The University of Utah, Salt Lake City, UT
- Molecular Medicine Program, The University of Utah, Salt Lake City, UT
| | - Aaron C. Petrey
- Molecular Medicine Program, The University of Utah, Salt Lake City, UT
| | - Elizabeth A. Middleton
- Molecular Medicine Program, The University of Utah, Salt Lake City, UT
- Department of Internal Medicine, The University of Utah, Salt Lake City, UT
| | - Robert A. Campbell
- Department of Emergency Medicine, Washington University School of Medicine, St. Louis, MO
| | - Christian C. Yost
- Department of Pediatrics, The University of Utah, Salt Lake City, UT
- Molecular Medicine Program, The University of Utah, Salt Lake City, UT
| |
Collapse
|
9
|
Baz AA, Hao H, Lan S, Li Z, Liu S, Jin X, Chen S, Chu Y. Emerging insights into macrophage extracellular traps in bacterial infections. FASEB J 2024; 38:e23767. [PMID: 38924166 DOI: 10.1096/fj.202400739r] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2024] [Revised: 06/11/2024] [Accepted: 06/12/2024] [Indexed: 06/28/2024]
Abstract
Macrophages possess a diverse range of well-defined capabilities and roles as phagocytes, encompassing the regulation of inflammation, facilitation of wound healing, maintenance of tissue homeostasis, and serving as a crucial element in the innate immune response against microbial pathogens. The emergence of extracellular traps is a novel strategy of defense that has been observed in several types of innate immune cells. In response to infection, macrophages are stimulated and produce macrophage extracellular traps (METs), which take the form of net-like structures, filled with strands of DNA and adorned with histones and other cellular proteins. METs not only capture and eliminate microorganisms but also play a role in the development of certain diseases such as inflammation and autoimmune disorders. The primary objective of this study is to examine the latest advancements in METs for tackling bacterial infections. We also delve into the current knowledge and tactics utilized by bacteria to elude or endure the effects of METs. Through this investigation, we hope to shed light on the intricate interactions between bacteria and the host's immune system, particularly in the context of microbicidal effector mechanisms of METs. The continued exploration of METs and their impact on host defense against various pathogens opens up new avenues for understanding and potentially manipulating the immune system's response to infections.
Collapse
Affiliation(s)
- Ahmed Adel Baz
- State Key Laboratory for Animal Disease Control and Prevention, College of Veterinary Medicine, Lanzhou University, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou, China
- Gansu Province Research Center for Basic Disciplines of Pathogen Biology, Lanzhou, China
- Key Laboratory of Veterinary Etiological Biology, Key Laboratory of Ruminant Disease Prevention and Control (West), Ministry of Agricultural and Rural Affairs, Lanzhou, China
- Botany and Microbiology Department, Faculty of Science, Al-Azhar University, Assiut, Egypt
| | - Huafang Hao
- State Key Laboratory for Animal Disease Control and Prevention, College of Veterinary Medicine, Lanzhou University, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou, China
- Gansu Province Research Center for Basic Disciplines of Pathogen Biology, Lanzhou, China
- Key Laboratory of Veterinary Etiological Biology, Key Laboratory of Ruminant Disease Prevention and Control (West), Ministry of Agricultural and Rural Affairs, Lanzhou, China
| | - Shimei Lan
- State Key Laboratory for Animal Disease Control and Prevention, College of Veterinary Medicine, Lanzhou University, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou, China
- Gansu Province Research Center for Basic Disciplines of Pathogen Biology, Lanzhou, China
- Key Laboratory of Veterinary Etiological Biology, Key Laboratory of Ruminant Disease Prevention and Control (West), Ministry of Agricultural and Rural Affairs, Lanzhou, China
| | - Zhangcheng Li
- State Key Laboratory for Animal Disease Control and Prevention, College of Veterinary Medicine, Lanzhou University, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou, China
- Gansu Province Research Center for Basic Disciplines of Pathogen Biology, Lanzhou, China
- Key Laboratory of Veterinary Etiological Biology, Key Laboratory of Ruminant Disease Prevention and Control (West), Ministry of Agricultural and Rural Affairs, Lanzhou, China
| | - Shuang Liu
- State Key Laboratory for Animal Disease Control and Prevention, College of Veterinary Medicine, Lanzhou University, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou, China
- Gansu Province Research Center for Basic Disciplines of Pathogen Biology, Lanzhou, China
- Key Laboratory of Veterinary Etiological Biology, Key Laboratory of Ruminant Disease Prevention and Control (West), Ministry of Agricultural and Rural Affairs, Lanzhou, China
| | - Xiangrui Jin
- State Key Laboratory for Animal Disease Control and Prevention, College of Veterinary Medicine, Lanzhou University, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou, China
- Gansu Province Research Center for Basic Disciplines of Pathogen Biology, Lanzhou, China
- Key Laboratory of Veterinary Etiological Biology, Key Laboratory of Ruminant Disease Prevention and Control (West), Ministry of Agricultural and Rural Affairs, Lanzhou, China
| | - Shengli Chen
- State Key Laboratory for Animal Disease Control and Prevention, College of Veterinary Medicine, Lanzhou University, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou, China
- Gansu Province Research Center for Basic Disciplines of Pathogen Biology, Lanzhou, China
- Key Laboratory of Veterinary Etiological Biology, Key Laboratory of Ruminant Disease Prevention and Control (West), Ministry of Agricultural and Rural Affairs, Lanzhou, China
| | - Yuefeng Chu
- State Key Laboratory for Animal Disease Control and Prevention, College of Veterinary Medicine, Lanzhou University, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou, China
- Gansu Province Research Center for Basic Disciplines of Pathogen Biology, Lanzhou, China
- Key Laboratory of Veterinary Etiological Biology, Key Laboratory of Ruminant Disease Prevention and Control (West), Ministry of Agricultural and Rural Affairs, Lanzhou, China
| |
Collapse
|
10
|
Carvalho WA, Gaspar EB, Domingues R, Regitano LCA, Cardoso FF. Genetic factors underlying host resistance to Rhipicephalus microplus tick infestation in Braford cattle: a systems biology perspective. Mamm Genome 2024; 35:186-200. [PMID: 38480585 DOI: 10.1007/s00335-024-10030-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2023] [Accepted: 01/29/2024] [Indexed: 05/29/2024]
Abstract
Approximately 80% of the world's cattle are raised in regions with a high risk of tick-borne diseases, resulting in significant economic losses due to parasitism by Rhipicephalus (Boophilus) microplus. However, the lack of a systemic biology approach hampers a comprehensive understanding of tick-host interactions that mediate tick resistance phenotypes. Here, we conducted a genome-wide association study (GWAS) of 2933 Braford cattle and found 340 single-nucleotide polymorphisms (SNPs) associated with tick counts. Gene expression analyses were performed on skin samples obtained from previously tick-exposed heifers with extremely high or low estimated breeding values for R. microplus counts. Evaluations were performed both before and after artificial infestation with ticks. Differentially expressed genes were found within 1-Mb windows centered at significant SNPs from GWAS. A total of 330 genes were related to the breakdown of homeostasis that was induced by larval attachment to bovine skin. Enrichment analysis pointed to a key role of proteolysis and signal transduction via JAK/STAT, NFKB and WNT/beta catenin signaling pathways. Integrative analysis on matrixEQTL revealed two cis-eQTLs and four significant SNPs in the genes peptidyl arginine deiminase type IV (PADI4) and LOC11449251. The integration of genomic data from QTL maps and transcriptome analyses has identified a set of twelve key genes that show significant associations with tick loads. These genes could be key candidates to improve the accuracy of genomic predictions for tick resistance in Braford cattle.
Collapse
|
11
|
Krishnan J, Hennen EM, Ao M, Kirabo A, Ahmad T, de la Visitación N, Patrick DM. NETosis Drives Blood Pressure Elevation and Vascular Dysfunction in Hypertension. Circ Res 2024; 134:1483-1494. [PMID: 38666386 PMCID: PMC11116040 DOI: 10.1161/circresaha.123.323897] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/25/2023] [Revised: 03/18/2024] [Accepted: 04/08/2024] [Indexed: 04/28/2024]
Abstract
BACKGROUND Neutrophil extracellular traps (NETs) are composed of DNA, enzymes, and citrullinated histones that are expelled by neutrophils in the process of NETosis. NETs accumulate in the aorta and kidneys in hypertension. PAD4 (protein-arginine deiminase-4) is a calcium-dependent enzyme that is essential for NETosis. TRPV4 (transient receptor potential cation channel subfamily V member 4) is a mechanosensitive calcium channel expressed in neutrophils. Thus, we hypothesize that NETosis contributes to hypertension via NET-mediated endothelial cell (EC) dysfunction. METHODS NETosis-deficient Padi4-/- mice were treated with Ang II (angiotensin II). Blood pressure was measured by radiotelemetry, and vascular reactivity was measured with wire myography. Neutrophils were cultured with or without ECs and exposed to normotensive or hypertensive uniaxial stretch. NETosis was measured by flow cytometry. ECs were treated with citrullinated histone H3, and gene expression was measured by quantitative reverse transcription PCR. Aortic rings were incubated with citrullinated histone H3, and wire myography was performed to evaluate EC function. Neutrophils were treated with the TRPV4 agonist GSK1016790A. Calcium influx was measured using Fluo-4 dye, and NETosis was measured by immunofluorescence. RESULTS Padi4-/- mice exhibited attenuated hypertension, reduced aortic inflammation, and improved EC-dependent vascular relaxation in response to Ang II. Coculture of neutrophils with ECs and exposure to hypertensive uniaxial stretch increased NETosis and accumulation of neutrophil citrullinated histone H3. Histone H3 and citrullinated histone H3 exposure attenuates EC-dependent vascular relaxation. Treatment of neutrophils with the TRPV4 agonist GSK1016790A increases intracellular calcium and NETosis. CONCLUSIONS These observations identify a role of NETosis in the pathogenesis of hypertension. Moreover, they define an important role of EC stretch and TRPV4 as initiators of NETosis. Finally, they define a role of citrullinated histones as drivers of EC dysfunction in hypertension.
Collapse
Affiliation(s)
- Jaya Krishnan
- Division of Clinical Pharmacology, Department of Medicine (J.K., A.K., T.A., N.d.l.V., D.M.P.), Vanderbilt University Medical Center, Nashville, TN
| | - Elizabeth M. Hennen
- Department of Biomedical Engineering, Vanderbilt University, Nashville, TN (E.M.H.)
| | - Mingfang Ao
- Department of Anesthesiology (M.A.), Vanderbilt University Medical Center, Nashville, TN
| | - Annet Kirabo
- Division of Clinical Pharmacology, Department of Medicine (J.K., A.K., T.A., N.d.l.V., D.M.P.), Vanderbilt University Medical Center, Nashville, TN
- Vanderbilt Center for Immunobiology (A.K.)
- Vanderbilt Institute for Infection, Immunology and Inflammation, Nashville, TN (A.K.)
- Vanderbilt Institute for Global Health, Nashville, TN (A.K.)
| | - Taseer Ahmad
- Division of Clinical Pharmacology, Department of Medicine (J.K., A.K., T.A., N.d.l.V., D.M.P.), Vanderbilt University Medical Center, Nashville, TN
- Department of Pharmacology, College of Pharmacy, University of Sargodha, Sargodha, Pakistan (T.A.)
| | - Néstor de la Visitación
- Division of Clinical Pharmacology, Department of Medicine (J.K., A.K., T.A., N.d.l.V., D.M.P.), Vanderbilt University Medical Center, Nashville, TN
- Division of Cardiovascular Medicine, Department of Medicine (D.M.P.), Vanderbilt University Medical Center, Nashville, TN
| | - David M. Patrick
- Division of Clinical Pharmacology, Department of Medicine (J.K., A.K., T.A., N.d.l.V., D.M.P.), Vanderbilt University Medical Center, Nashville, TN
- Department of Veterans Affairs, Nashville, TN (D.M.P.)
| |
Collapse
|
12
|
Wang X, Yuan W, Yang C, Wang Z, Zhang J, Xu D, Sun X, Sun W. Emerging role of gut microbiota in autoimmune diseases. Front Immunol 2024; 15:1365554. [PMID: 38765017 PMCID: PMC11099291 DOI: 10.3389/fimmu.2024.1365554] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2024] [Accepted: 04/22/2024] [Indexed: 05/21/2024] Open
Abstract
Accumulating studies have indicated that the gut microbiota plays a pivotal role in the onset of autoimmune diseases by engaging in complex interactions with the host. This review aims to provide a comprehensive overview of the existing literatures concerning the relationship between the gut microbiota and autoimmune diseases, shedding light on the complex interplay between the gut microbiota, the host and the immune system. Furthermore, we aim to summarize the impacts and potential mechanisms that underlie the interactions between the gut microbiota and the host in autoimmune diseases, primarily focusing on systemic lupus erythematosus, rheumatoid arthritis, Sjögren's syndrome, type 1 diabetes mellitus, ulcerative colitis and psoriasis. The present review will emphasize the clinical significance and potential applications of interventions based on the gut microbiota as innovative adjunctive therapies for autoimmune diseases.
Collapse
Affiliation(s)
- Xinyi Wang
- School of Clinical and Basic Medical Sciences, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, China
| | - Wei Yuan
- Department of Radiation Oncology, The First Affiliated Hospital of Shandong Second Medical University, Weifang, China
| | - Chunjuan Yang
- Department of Central Laboratory, The First Affiliated Hospital of Shandong Second Medical University, Weifang, China
- Department of Rheumatology, The First Affiliated Hospital of Shandong Second Medical University, Weifang, China
| | - Zhangxue Wang
- Department of Rheumatology, The First Affiliated Hospital of Shandong Second Medical University, Weifang, China
| | - Jin Zhang
- Department of Rheumatology, The First Affiliated Hospital of Shandong Second Medical University, Weifang, China
| | - Donghua Xu
- Department of Central Laboratory, The First Affiliated Hospital of Shandong Second Medical University, Weifang, China
- Department of Rheumatology, The First Affiliated Hospital of Shandong Second Medical University, Weifang, China
| | - Xicai Sun
- Department of Hospital Office, The First Affiliated Hospital of Shandong Second Medical University, Weifang, China
| | - Wenchang Sun
- Department of Central Laboratory, The First Affiliated Hospital of Shandong Second Medical University, Weifang, China
| |
Collapse
|
13
|
Ahluwalia K, Du Z, Martinez-Camarillo JC, Naik A, Thomas BB, Pollalis D, Lee SY, Dave P, Zhou E, Li Z, Chester C, Humayun MS, Louie SG. Unveiling Drivers of Retinal Degeneration in RCS Rats: Functional, Morphological, and Molecular Insights. Int J Mol Sci 2024; 25:3749. [PMID: 38612560 PMCID: PMC11011632 DOI: 10.3390/ijms25073749] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2024] [Revised: 03/12/2024] [Accepted: 03/22/2024] [Indexed: 04/14/2024] Open
Abstract
Retinal degenerative diseases, including age-related macular degeneration and retinitis pigmentosa, significantly contribute to adult blindness. The Royal College of Surgeons (RCS) rat is a well-established disease model for studying these dystrophies; however, molecular investigations remain limited. We conducted a comprehensive analysis of retinal degeneration in RCS rats, including an immunodeficient RCS (iRCS) sub-strain, using ocular coherence tomography, electroretinography, histology, and molecular dissection using transcriptomics and immunofluorescence. No significant differences in retinal degeneration progression were observed between the iRCS and immunocompetent RCS rats, suggesting a minimal role of adaptive immune responses in disease. Transcriptomic alterations were primarily in inflammatory signaling pathways, characterized by the strong upregulation of Tnfa, an inflammatory signaling molecule, and Nox1, a contributor to reactive oxygen species (ROS) generation. Additionally, a notable decrease in Alox15 expression was observed, pointing to a possible reduction in anti-inflammatory and pro-resolving lipid mediators. These findings were corroborated by immunostaining, which demonstrated increased photoreceptor lipid peroxidation (4HNE) and photoreceptor citrullination (CitH3) during retinal degeneration. Our work enhances the understanding of molecular changes associated with retinal degeneration in RCS rats and offers potential therapeutic targets within inflammatory and oxidative stress pathways for confirmatory research and development.
Collapse
Affiliation(s)
- Kabir Ahluwalia
- Mann School of Pharmacy and Pharmaceutical Sciences, University of Southern California, Los Angeles, CA 90089, USA; (K.A.); (A.N.); (P.D.); (E.Z.); (Z.L.); (C.C.)
| | - Zhaodong Du
- USC Ginsburg Institute of for Biomedical Therapeutics, University of Southern California, Los Angeles, CA 90033, USA; (Z.D.); (J.C.M.-C.); (B.B.T.); (D.P.); (S.Y.L.); (M.S.H.)
| | - Juan Carlos Martinez-Camarillo
- USC Ginsburg Institute of for Biomedical Therapeutics, University of Southern California, Los Angeles, CA 90033, USA; (Z.D.); (J.C.M.-C.); (B.B.T.); (D.P.); (S.Y.L.); (M.S.H.)
- Roski Eye Institute, Keck School of Medicine, University of Southern California, Los Angeles, CA 90033, USA
| | - Aditya Naik
- Mann School of Pharmacy and Pharmaceutical Sciences, University of Southern California, Los Angeles, CA 90089, USA; (K.A.); (A.N.); (P.D.); (E.Z.); (Z.L.); (C.C.)
| | - Biju B. Thomas
- USC Ginsburg Institute of for Biomedical Therapeutics, University of Southern California, Los Angeles, CA 90033, USA; (Z.D.); (J.C.M.-C.); (B.B.T.); (D.P.); (S.Y.L.); (M.S.H.)
- Roski Eye Institute, Keck School of Medicine, University of Southern California, Los Angeles, CA 90033, USA
| | - Dimitrios Pollalis
- USC Ginsburg Institute of for Biomedical Therapeutics, University of Southern California, Los Angeles, CA 90033, USA; (Z.D.); (J.C.M.-C.); (B.B.T.); (D.P.); (S.Y.L.); (M.S.H.)
- Roski Eye Institute, Keck School of Medicine, University of Southern California, Los Angeles, CA 90033, USA
| | - Sun Young Lee
- USC Ginsburg Institute of for Biomedical Therapeutics, University of Southern California, Los Angeles, CA 90033, USA; (Z.D.); (J.C.M.-C.); (B.B.T.); (D.P.); (S.Y.L.); (M.S.H.)
- Roski Eye Institute, Keck School of Medicine, University of Southern California, Los Angeles, CA 90033, USA
- Department of Physiology & Neuroscience, Keck School of Medicine, University of Southern California, Los Angeles, CA 90089, USA
| | - Priyal Dave
- Mann School of Pharmacy and Pharmaceutical Sciences, University of Southern California, Los Angeles, CA 90089, USA; (K.A.); (A.N.); (P.D.); (E.Z.); (Z.L.); (C.C.)
| | - Eugene Zhou
- Mann School of Pharmacy and Pharmaceutical Sciences, University of Southern California, Los Angeles, CA 90089, USA; (K.A.); (A.N.); (P.D.); (E.Z.); (Z.L.); (C.C.)
| | - Zeyang Li
- Mann School of Pharmacy and Pharmaceutical Sciences, University of Southern California, Los Angeles, CA 90089, USA; (K.A.); (A.N.); (P.D.); (E.Z.); (Z.L.); (C.C.)
| | - Catherine Chester
- Mann School of Pharmacy and Pharmaceutical Sciences, University of Southern California, Los Angeles, CA 90089, USA; (K.A.); (A.N.); (P.D.); (E.Z.); (Z.L.); (C.C.)
| | - Mark S. Humayun
- USC Ginsburg Institute of for Biomedical Therapeutics, University of Southern California, Los Angeles, CA 90033, USA; (Z.D.); (J.C.M.-C.); (B.B.T.); (D.P.); (S.Y.L.); (M.S.H.)
- Roski Eye Institute, Keck School of Medicine, University of Southern California, Los Angeles, CA 90033, USA
| | - Stan G. Louie
- Mann School of Pharmacy and Pharmaceutical Sciences, University of Southern California, Los Angeles, CA 90089, USA; (K.A.); (A.N.); (P.D.); (E.Z.); (Z.L.); (C.C.)
- USC Ginsburg Institute of for Biomedical Therapeutics, University of Southern California, Los Angeles, CA 90033, USA; (Z.D.); (J.C.M.-C.); (B.B.T.); (D.P.); (S.Y.L.); (M.S.H.)
| |
Collapse
|
14
|
Conedera FM, Kokona D, Zinkernagel MS, Stein JV, Lin CP, Alt C, Enzmann V. Macrophages coordinate immune response to laser-induced injury via extracellular traps. J Neuroinflammation 2024; 21:68. [PMID: 38500151 PMCID: PMC10949579 DOI: 10.1186/s12974-024-03064-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2024] [Accepted: 03/13/2024] [Indexed: 03/20/2024] Open
Abstract
BACKGROUND Retinal degeneration results from disruptions in retinal homeostasis due to injury, disease, or aging and triggers peripheral leukocyte infiltration. Effective immune responses rely on coordinated actions of resident microglia and recruited macrophages, critical for tissue remodeling and repair. However, these phagocytes also contribute to chronic inflammation in degenerated retinas, yet the precise coordination of immune response to retinal damage remains elusive. Recent investigations have demonstrated that phagocytic cells can produce extracellular traps (ETs), which are a source of self-antigens that alter the immune response, which can potentially lead to tissue injury. METHODS Innovations in experimental systems facilitate real-time exploration of immune cell interactions and dynamic responses. We integrated in vivo imaging with ultrastructural analysis, transcriptomics, pharmacological treatments, and knockout mice to elucidate the role of phagocytes and their modulation of the local inflammatory response through extracellular traps (ETs). Deciphering these mechanisms is essential for developing novel and enhanced immunotherapeutic approaches that can redirect a specific maladaptive immune response towards favorable wound healing in the retina. RESULTS Our findings underscore the pivotal role of innate immune cells, especially macrophages/monocytes, in regulating retinal repair and inflammation. The absence of neutrophil and macrophage infiltration aids parenchymal integrity restoration, while their depletion, particularly macrophages/monocytes, impedes vascular recovery. We demonstrate that macrophages/monocytes, when recruited in the retina, release chromatin and granular proteins, forming ETs. Furthermore, the pharmacological inhibition of ETosis support retinal and vascular repair, surpassing the effects of blocking innate immune cell recruitment. Simultaneously, the absence of ETosis reshapes the inflammatory response, causing neutrophils, helper, and cytotoxic T-cells to be restricted primarily in the superficial capillary plexus instead of reaching the damaged photoreceptor layer. CONCLUSIONS Our data offer novel insights into innate immunity's role in responding to retinal damage and potentially help developing innovative immunotherapeutic approaches that can shift the immune response from maladaptive to beneficial for retinal regeneration.
Collapse
Affiliation(s)
- Federica M Conedera
- Department of Oncology, Microbiology and Immunology, University of Fribourg, Fribourg, Switzerland.
- Department of Ophthalmology, Bern University Hospital and Department of BioMedical Research, University of Bern, Bern, Switzerland.
| | - Despina Kokona
- Department of Ophthalmology, Bern University Hospital and Department of BioMedical Research, University of Bern, Bern, Switzerland
| | - Martin S Zinkernagel
- Department of Ophthalmology, Bern University Hospital and Department of BioMedical Research, University of Bern, Bern, Switzerland
| | - Jens V Stein
- Department of Oncology, Microbiology and Immunology, University of Fribourg, Fribourg, Switzerland
| | - Charles P Lin
- Center for Systems Biology and Wellman Center for Photomedicine, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| | - Clemens Alt
- Center for Systems Biology and Wellman Center for Photomedicine, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| | - Volker Enzmann
- Department of Ophthalmology, Bern University Hospital and Department of BioMedical Research, University of Bern, Bern, Switzerland
| |
Collapse
|
15
|
Bashar SJ, Holmes CL, Shelef MA. Macrophage extracellular traps require peptidylarginine deiminase 2 and 4 and are a source of citrullinated antigens bound by rheumatoid arthritis autoantibodies. Front Immunol 2024; 15:1167362. [PMID: 38476240 PMCID: PMC10927735 DOI: 10.3389/fimmu.2024.1167362] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2023] [Accepted: 01/29/2024] [Indexed: 03/14/2024] Open
Abstract
Introduction Anti-citrullinated protein antibodies (ACPAs) are a hallmark of rheumatoid arthritis, but the sources of citrullinated antigens as well as which peptidylarginine deiminases (PADs) are required for their production remain incompletely defined. Here, we investigated if macrophage extracellular traps (METs) could be a source of citrullinated proteins bound by APCAs, and if their formation requires PAD2 or PAD4. Methods Thioglycolate-induced peritoneal macrophages from wild-type, PAD2-/-, and PAD4-/- mice or human peripheral blood-derived M1 macrophages were activated with a variety of stimulants, then fixed and stained with DAPI and either anti-citrullinated histone H4 (citH4) antibody or sera from ACPA+ or ACPA- rheumatoid arthritis subjects. METs were visualized by immunofluorescence, confirmed to be extracellular using DNase, and quantified. Results We found that ionomycin and monosodium urate crystals reliably induced murine citH4+ METs, which were reduced in the absence of PAD2 and lost in the absence of PAD4. Also, IgG from ACPA+, but not ACPA-, rheumatoid arthritis sera bound to murine METs, and in the absence of PAD2 or PAD4, ACPA-bound METs were lost. Finally, ionomycin induced human METs that are citH4+ and ACPA-bound. Discussion Thus, METs may contribute to the pool of citrullinated antigens bound by ACPAs in a PAD2- and PAD4-dependent manner, providing new insights into the targets of immune tolerance loss in rheumatoid arthritis.
Collapse
Affiliation(s)
- S. Janna Bashar
- Department of Medicine, University of Wisconsin-Madison, Madison, WI, United States
| | - Caitlyn L. Holmes
- Department of Medicine, University of Wisconsin-Madison, Madison, WI, United States
| | - Miriam A. Shelef
- Department of Medicine, University of Wisconsin-Madison, Madison, WI, United States
- William S. Middleton Memorial Veteran’s Hospital, Madison, WI, United States
| |
Collapse
|
16
|
Chen P, Luo Z, Lu C, Jian G, Qi X, Xiong H. Gut-immunity-joint axis: a new therapeutic target for gouty arthritis. Front Pharmacol 2024; 15:1353615. [PMID: 38464719 PMCID: PMC10920255 DOI: 10.3389/fphar.2024.1353615] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2023] [Accepted: 02/05/2024] [Indexed: 03/12/2024] Open
Abstract
Gouty arthritis (GA) is an inflammatory disease characterized by pain. The primary goal of current treatment strategies during GA flares remains the reduction of inflammation and pain. Research suggests that the gut microbiota and microbial metabolites contribute to the modulation of the inflammatory mechanism associated with GA, particularly through their effect on macrophage polarization. The increasing understanding of the gut-joint axis emphasizes the importance of this interaction. The primary objective of this review is to summarize existing research on the gut-immune-joint axis in GA, aiming to enhance understanding of the intricate processes and pathogenic pathways associated with pain and inflammation in GA, as documented in the published literature. The refined comprehension of the gut-joint axis may potentially contribute to the future development of analgesic drugs targeting gut microbes for GA.
Collapse
Affiliation(s)
- Pei Chen
- Hunan University of Chinese Medicine, Changsha, Hunan, China
- The Second Hospital of Hunan University of Chinese Medicine, Changsha, Hunan, China
- The First Hospital of Hunan University Chinese Medicine, Changsha, Hunan, China
| | - Zhiqiang Luo
- Hunan University of Chinese Medicine, Changsha, Hunan, China
- The Second Hospital of Hunan University of Chinese Medicine, Changsha, Hunan, China
| | - Chengyin Lu
- Hunan University of Chinese Medicine, Changsha, Hunan, China
- The Second Hospital of Hunan University of Chinese Medicine, Changsha, Hunan, China
| | - Gonghui Jian
- Hunan University of Chinese Medicine, Changsha, Hunan, China
- College of Integrative Chinese and Western Medicine, Hunan University of Chinese Medicine, Changsha, Hunan, China
| | - Xinyu Qi
- Hunan University of Chinese Medicine, Changsha, Hunan, China
| | - Hui Xiong
- Hunan University of Chinese Medicine, Changsha, Hunan, China
- The First Hospital of Hunan University Chinese Medicine, Changsha, Hunan, China
| |
Collapse
|
17
|
Weng W, Liu Y, Hu Z, Li Z, Peng X, Wang M, Dong B, Zhong S, Jiang Y, Pan Y. Macrophage extracellular traps promote tumor-like biologic behaviors of fibroblast-like synoviocytes through cGAS-mediated PI3K/Akt signaling pathway in patients with rheumatoid arthritis. J Leukoc Biol 2024; 115:116-129. [PMID: 37648663 DOI: 10.1093/jleuko/qiad102] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2023] [Revised: 08/03/2023] [Accepted: 08/08/2023] [Indexed: 09/01/2023] Open
Abstract
Rheumatoid arthritis is an autoimmune disease characterized by synovium hyperplasia and bone destruction. Macrophage extracellular traps are released from macrophages under various stimuli and may generate stable autoantigen-DNA complexes, as well as aggravate autoantibody generation and autoimmune responses. We aimed to investigate the role of macrophage extracellular traps on the biologic behaviors of rheumatoid arthritis fibroblast-like synoviocytes. Synovial tissues and fibroblast-like synoviocytes were obtained from patients with rheumatoid arthritis. Extracellular traps in synovium and synovial fluids were detected by immunofluorescence, immunohistochemistry, and SYTOX Green staining. Cell viability, migration, invasion, and cytokine expression of rheumatoid arthritis fibroblast-like synoviocytes were assessed by CCK-8, wound-healing assay, Transwell assays, and quantitative real-time polymerase chain reaction, respectively. RNA sequencing analysis was performed to explore the underlying mechanism, and Western blot was used to validate the active signaling pathways. We found that extracellular trap formation was abundant in rheumatoid arthritis and positively correlated to anti-CCP. Rheumatoid arthritis fibroblast-like synoviocytes stimulated with purified macrophage extracellular traps demonstrated the obvious promotion in tumor-like biologic behaviors. The DNA sensor cGAS in rheumatoid arthritis fibroblast-like synoviocytes was activated after macrophage extracellular trap stimuli. RNA sequencing revealed that differential genes were significantly enriched in the PI3K/Akt signaling pathway, and cGAS inhibitor RU.521 effectively reversed the promotion of tumor-like biologic behaviors in macrophage extracellular trap-treated rheumatoid arthritis fibroblast-like synoviocytes and downregulated the PI3K/Akt activation. In summary, our study demonstrates that macrophage extracellular traps promote the pathogenically biological behaviors of rheumatoid arthritis fibroblast-like synoviocytes through cGAS-mediated activation of the PI3K/Akt signaling pathway. These findings provide a novel insight into the pathogenesis of rheumatoid arthritis and the mechanisms of macrophages in modulating rheumatoid arthritis fibroblast-like synoviocyte tumor-like behaviors.
Collapse
Affiliation(s)
- Weizhen Weng
- Department of Rheumatology, The Third Affiliated Hospital of Sun Yat-sen University, 600 Tianhe Road, Tianhe District, Guangzhou, China
- Department of Infectious Disease, The Third People's Hospital of Shenzhen, 29 Bulang Road, Longgang district, Shenzhen, China
| | - Yan Liu
- Department of Rheumatology, The Third Affiliated Hospital of Sun Yat-sen University, 600 Tianhe Road, Tianhe District, Guangzhou, China
| | - Zuoyu Hu
- Nanfang Hospital, Southern Medical University, 1838 North Guangzhou Avenue, Baiyun District, Guangzhou, China
| | - Zhihui Li
- Department of Infectious Diseases, The Third Affiliated Hospital of Sun Yat-sen University, 600 Tianhe Road, Tianhe District, Guangzhou, China
| | - Xiaohua Peng
- Department of Gastroenterology, The Seventh Affiliated Hospital of Sun Yat-Sen University, 628 Zhenyuan Road, Guangming District, Shenzhen, China
| | - Manli Wang
- Medical Research Center, The Eighth Affiliated Hospital of Sun Yat-sen University, 3025 Shennan Road, Futian District, Shenzhen, China
| | - Bo Dong
- Department of Rheumatology, The Third Affiliated Hospital of Sun Yat-sen University, 600 Tianhe Road, Tianhe District, Guangzhou, China
| | - Shuyuan Zhong
- Department of Rheumatology, The Third Affiliated Hospital of Sun Yat-sen University, 600 Tianhe Road, Tianhe District, Guangzhou, China
| | - Yutong Jiang
- Department of Rheumatology, The Third Affiliated Hospital of Sun Yat-sen University, 600 Tianhe Road, Tianhe District, Guangzhou, China
| | - Yunfeng Pan
- Department of Rheumatology, The Third Affiliated Hospital of Sun Yat-sen University, 600 Tianhe Road, Tianhe District, Guangzhou, China
| |
Collapse
|
18
|
Cheng JW, Yu Y, Zong SY, Cai WW, Wang Y, Song YN, Xian H, Wei F. Berberine ameliorates collagen-induced arthritis in mice by restoring macrophage polarization via AMPK/mTORC1 pathway switching glycolytic reprogramming. Int Immunopharmacol 2023; 124:111024. [PMID: 37827054 DOI: 10.1016/j.intimp.2023.111024] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2023] [Revised: 09/29/2023] [Accepted: 10/03/2023] [Indexed: 10/14/2023]
Abstract
Dysfunction of macrophage polarization majorly contributes to the progression of rheumatoid arthritis (RA). Polarization and functions of activated macrophages are closely associated with the reprogramming of intracellular metabolisms. Previously, we demonstrated that the anti-arthritis effect of berberine (BBR) in rats with adjuvant-induced arthritis (AA) may be related to AMP-activated protein kinase (AMPK) activation (a key regulator in the biological energy metabolism), and balanced macrophage polarization. However, the specific molecular mechanism of BBR in macrophage metabolism is yet to be elucidated. In this study, we clarified that BBR ameliorated articular inflammation and restored M1/M2 ratio in collagen-induced arthritis (CIA) mice in an AMPK-dependent manner. Mechanistically, BBR reversed the effects of mTORC1 agonist leucine (Leu) on regulating macrophage polarization through activation of AMPK to switch glycolytic reprogramming. Furthermore, BBR inhibition of mTORC1 rely on activation of AMPK to phosphorylate raptor and TSC2 instead of destroying its structure. Our study revealed that the activation of AMPK is required for the BBR-mediated anti-arthritis effect by downregulating mTORC1/HIF-1α and inhibiting the glycolysis in M1 macrophages.
Collapse
Affiliation(s)
- Jing-Wen Cheng
- School of Pharmacy, Bengbu Medical College, No. 2600, Donghai Avenue, Bengbu, Anhui, China
| | - Yun Yu
- School of Pharmacy, Bengbu Medical College, No. 2600, Donghai Avenue, Bengbu, Anhui, China
| | - Shi-Ye Zong
- School of Pharmacy, Bengbu Medical College, No. 2600, Donghai Avenue, Bengbu, Anhui, China
| | - Wei-Wei Cai
- School of Pharmacy, Bengbu Medical College, No. 2600, Donghai Avenue, Bengbu, Anhui, China
| | - Ying Wang
- School of Pharmacy, Bengbu Medical College, No. 2600, Donghai Avenue, Bengbu, Anhui, China; Biochemical Engineering Center of Anhui, Bengbu, Anhui, China
| | - Yi-Ning Song
- School of Pharmacy, Bengbu Medical College, No. 2600, Donghai Avenue, Bengbu, Anhui, China; Biochemical Engineering Center of Anhui, Bengbu, Anhui, China
| | - Hao Xian
- School of Pharmacy, Bengbu Medical College, No. 2600, Donghai Avenue, Bengbu, Anhui, China
| | - Fang Wei
- School of Pharmacy, Bengbu Medical College, No. 2600, Donghai Avenue, Bengbu, Anhui, China; Biochemical Engineering Center of Anhui, Bengbu, Anhui, China.
| |
Collapse
|
19
|
Kimura A, Takagi T, Thamamongood T, Sakamoto S, Ito T, Seki I, Okamoto M, Aono H, Serada S, Naka T, Imataka H, Miyake K, Ueda T, Miyanokoshi M, Wakasugi K, Iwamoto N, Ohmagari N, Iguchi T, Nitta T, Takayanagi H, Yamashita H, Kaneko H, Tsuchiya H, Fujio K, Handa H, Suzuki H. Extracellular aaRSs drive autoimmune and inflammatory responses in rheumatoid arthritis via the release of cytokines and PAD4. Ann Rheum Dis 2023; 82:1153-1161. [PMID: 37400117 DOI: 10.1136/ard-2023-224055] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2023] [Accepted: 05/23/2023] [Indexed: 07/05/2023]
Abstract
OBJECTIVES Recent studies demonstrate that extracellular-released aminoacyl-tRNA synthetases (aaRSs) play unique roles in immune responses and diseases. This study aimed to understand the role of extracellular aaRSs in the pathogenesis of rheumatoid arthritis (RA). METHODS Primary macrophages and fibroblast-like synoviocytes were cultured with aaRSs. aaRS-induced cytokine production including IL-6 and TNF-α was detected by ELISA. Transcriptomic features of aaRS-stimulated macrophages were examined using RNA-sequencing. Serum and synovial fluid (SF) aaRS levels in patients with RA were assessed using ELISA. Peptidyl arginine deiminase (PAD) 4 release from macrophages stimulated with aaRSs was detected by ELISA. Citrullination of aaRSs by themselves was examined by immunoprecipitation and western blotting. Furthermore, aaRS inhibitory peptides were used for inhibition of arthritis in two mouse RA models, collagen-induced arthritis and collagen antibody-induced arthritis. RESULTS All 20 aaRSs functioned as alarmin; they induced pro-inflammatory cytokines through the CD14-MD2-TLR4 axis. Stimulation of macrophages with aaRSs displayed persistent innate inflammatory responses. Serum and SF levels of many aaRSs increased in patients with RA compared with control subjects. Furthermore, aaRSs released PAD4 from living macrophages, leading to their citrullination. We demonstrate that aaRS inhibitory peptides suppress cytokine production and PAD4 release by aaRSs and alleviate arthritic symptoms in a mouse RA model. CONCLUSIONS Our findings uncovered the significant role of aaRSs as a novel alarmin in RA pathogenesis, indicating that their blocking agents are potent antirheumatic drugs.
Collapse
Affiliation(s)
- Akihiro Kimura
- Dep of Immunology and Pathology, Research Center for Hepatitis and Immunology, Research Institute, National Center for Global Health and Medicine, Ichikawa-shi, Chiba, Japan
| | - Takeshi Takagi
- School of Life Science and Technology, Tokyo Institute of Technology, Yokohama, Kanagawa, Japan
| | - Thiprampai Thamamongood
- National Center for Genetic Engineering and Biotechnology, National Science and Technology Development Agency, Pathumthani, Thailand
| | - Satoshi Sakamoto
- School of Life Science and Technology, Tokyo Institute of Technology, Yokohama, Kanagawa, Japan
| | - Takumi Ito
- Center for Future Medical Research, Tokyo Medical University, Shinjuku-ku, Tokyo, Japan
| | - Iwao Seki
- Research and Development Department, AYUMI Pharmaceutical Corporation, Chuo-ku, Tokyo, Japan
| | - Masahiro Okamoto
- Research and Development Department, AYUMI Pharmaceutical Corporation, Chuo-ku, Tokyo, Japan
| | - Hiroyuki Aono
- Research and Development Department, AYUMI Pharmaceutical Corporation, Chuo-ku, Tokyo, Japan
| | - Satoshi Serada
- Institute for Biomedical Sciences Molecular Pathophysiology, Iwate Medical University, Morioka, Iwate, Japan
| | - Tetsuji Naka
- Institute for Biomedical Sciences Molecular Pathophysiology, Iwate Medical University, Morioka, Iwate, Japan
| | - Hiroaki Imataka
- Department of Applied Chemistry, Graduate School of Engineering, University of Hyogo, Himeji, Hyogo, Japan
| | - Kensuke Miyake
- Division of Innate Immunity, The Institute of Medical Science, The University of Tokyo, Minato-ku, Tokyo, Japan
| | - Takuya Ueda
- Department of Integrative Bioscience and Biomedical Engineering, Graduate School of Science and Engineering, Waseda University, Shinjuku-ku, Tokyo, Japan
| | - Miki Miyanokoshi
- Department of Life Sciences, Graduate School of Arts and Sciences, The University of Tokyo, Meguro-ku, Tokyo, Japan
| | - Keisuke Wakasugi
- Department of Life Sciences, Graduate School of Arts and Sciences, The University of Tokyo, Meguro-ku, Tokyo, Japan
| | - Noriko Iwamoto
- Disease Control and Prevention Center, National Center for Global Health and Medicine, Shinjuku-ku, Tokyo, Japan
| | - Norio Ohmagari
- Disease Control and Prevention Center, National Center for Global Health and Medicine, Shinjuku-ku, Tokyo, Japan
| | - Takahiro Iguchi
- Department of Immunology, Graduate School of Medicine and Faculty of Medicine, The University of Tokyo, Bunkyo-ku, Tokyo, Japan
| | - Takeshi Nitta
- Department of Immunology, Graduate School of Medicine and Faculty of Medicine, The University of Tokyo, Bunkyo-ku, Tokyo, Japan
| | - Hiroshi Takayanagi
- Department of Immunology, Graduate School of Medicine and Faculty of Medicine, The University of Tokyo, Bunkyo-ku, Tokyo, Japan
| | - Hiroyuki Yamashita
- Division of Rheumatic Diseases, National Center for Global Health and Medicine, Shinjuku-ku, Tokyo, Japan
| | - Hiroshi Kaneko
- Division of Rheumatic Diseases, National Center for Global Health and Medicine, Shinjuku-ku, Tokyo, Japan
| | - Haruka Tsuchiya
- Department of Allergy and Rheumatology, Graduate School of Medicine, The University of Tokyo, Bunkyo-ku, Tokyo, Japan
| | - Keishi Fujio
- Department of Allergy and Rheumatology, Graduate School of Medicine, The University of Tokyo, Bunkyo-ku, Tokyo, Japan
| | - Hiroshi Handa
- Center for Future Medical Research, Tokyo Medical University, Shinjuku-ku, Tokyo, Japan
| | - Harumi Suzuki
- Dep of Immunology and Pathology, Research Center for Hepatitis and Immunology, Research Institute, National Center for Global Health and Medicine, Ichikawa-shi, Chiba, Japan
| |
Collapse
|
20
|
Chang Y, Ou Q, Zhou X, Nie K, Liu J, Zhang S. Global research trends and focus on the link between rheumatoid arthritis and neutrophil extracellular traps: a bibliometric analysis from 1985 to 2023. Front Immunol 2023; 14:1205445. [PMID: 37680637 PMCID: PMC10481536 DOI: 10.3389/fimmu.2023.1205445] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2023] [Accepted: 08/11/2023] [Indexed: 09/09/2023] Open
Abstract
Rheumatoid arthritis (RA) is an autoimmune disease that currently has an unknown cause and pathogenesis, and is associated with many complications and a high disability rate. The neutrophil extracellular trap network (NETs) is a newly discovered mechanism that allows neutrophils to capture and kill pathogens. Multiple studies in recent years have highlighted its relevance to the progression of rheumatoid arthritis. Despite the growing number of studies indicating the crucial role of NETs in RA, there has been no bibliometric review of research hotspots and trends in this area. In this study, we retrieved articles related to NETs in RA from the Web of Science Core Collection (WoSCC) database from 1985 to 2023 and used visualization tools such as Citespace, VOSviewer, Tableau Public, and Microsoft Office Excel 2021 to analyze the data. After screening, we included a total of 416 publications involving 2,334 researchers from 1,357 institutions in 167 countries/regions, with relevant articles published in 219 journals. The U.S., China, and Germany are the top 3 countries/regions with 124, 57, and 37 publications respectively. Mariana J. Kaplan is the most published author, and journals such as Frontiers in Immunology and International Journal of Molecular Sciences have had a significant impact on research in this field. The clinical application of PAD enzymes and their inhibitors, and the drug development of NETs as therapeutic targets for RA is a trend for future research. Our study provides a comprehensive bibliometric analysis and summary of NETs in RA publications, which will aid researchers in conducting further scientific research.
Collapse
Affiliation(s)
- Yonglong Chang
- Department of Integrated Traditional Chinese & Western Medicine, The Second Xiangya Hospital, Central South University, Changsha, China
| | - Qinling Ou
- Department of Integrated Traditional Chinese & Western Medicine, The Second Xiangya Hospital, Central South University, Changsha, China
- National Clinical Research Center for Metabolic Diseases, Changsha, China
| | - Xuhui Zhou
- Department of Addiction Medicine, Hunan Institute of Mental Health, Brain Hospital of Hunan Province (The Second People’s Hospital of Hunan Province), Changsha, Hunan, China
| | - Kechao Nie
- Department of Integrated Traditional Chinese & Western Medicine, The Second Xiangya Hospital, Central South University, Changsha, China
| | - Jinhui Liu
- College of Integrated Traditional Chinese & Western Medicine, Hunan University of Traditional Chinese Medicine, Changsha, Hunan, China
| | - Sifang Zhang
- Department of Integrated Traditional Chinese & Western Medicine, The Second Xiangya Hospital, Central South University, Changsha, China
- National Clinical Research Center for Metabolic Diseases, Changsha, China
| |
Collapse
|
21
|
Wang S, Chen L, Shi X, Wang Y, Xu S. Polystyrene microplastics-induced macrophage extracellular traps contributes to liver fibrotic injury by activating ROS/TGF-β/Smad2/3 signaling axis. ENVIRONMENTAL POLLUTION (BARKING, ESSEX : 1987) 2023; 324:121388. [PMID: 36871749 DOI: 10.1016/j.envpol.2023.121388] [Citation(s) in RCA: 21] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/30/2022] [Revised: 02/17/2023] [Accepted: 03/01/2023] [Indexed: 06/18/2023]
Abstract
Microplastics (MPs) are a type of emerging pollutant, posing a great threat to human and animal health. While recent studies have revealed the link between MPs exposure and liver injury of organisms, the effect of particle size on the level of MPs-induced hepatotoxicity and the intrinsic mechanism remain to be explored. Here, we established a mouse model exposed to two-diameter polystyrene MPs (PS-MPs, 1-10 μm or 50-100 μm) for 30 days. The in vivo results revealed that PS-MPs caused liver fibrotic injury in mice, accompanied with macrophages recruitment and macrophage extracellular traps (METs) formation, which were negatively correlated with particle size. The data in vitro showed that PS-MPs treatment could induce macrophages to release METs in a reactive oxygen species (ROS)-independent manner, and the METs formation level caused by large-size particles was higher than small-size particles. Further mechanistic analysis of a cell co-culture system revealed that PS-MPs-induced METs release led to a hepatocellular inflammatory response and epithelial-mesenchymal transition (EMT) via activating the ROS/TGF-β/Smad2/3 signaling axis, and this biological crosstalk could be relieved by DNase I. Overall, this findings demonstrates the key role of the action mechanism of METs in aggravating MPs-caused liver injury.
Collapse
Affiliation(s)
- Shengchen Wang
- College of Animal Science and Technology, Yangzhou University, Yangzhou, 225000, PR China; College of Veterinary Medicine, Northeast Agricultural University, Harbin, 150030, PR China
| | - Lu Chen
- College of Animal Science and Technology, Tarim University, Alar, 843300, PR China
| | - Xu Shi
- College of Veterinary Medicine, Northeast Agricultural University, Harbin, 150030, PR China
| | - Yue Wang
- College of Veterinary Medicine, Northeast Agricultural University, Harbin, 150030, PR China
| | - Shiwen Xu
- College of Veterinary Medicine, Northeast Agricultural University, Harbin, 150030, PR China.
| |
Collapse
|
22
|
A subset of antibodies targeting citrullinated proteins confers protection from rheumatoid arthritis. Nat Commun 2023; 14:691. [PMID: 36754962 PMCID: PMC9908943 DOI: 10.1038/s41467-023-36257-x] [Citation(s) in RCA: 22] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2022] [Accepted: 01/20/2023] [Indexed: 02/10/2023] Open
Abstract
Although elevated levels of anti-citrullinated protein antibodies (ACPAs) are a hallmark of rheumatoid arthritis (RA), the in vivo functions of these antibodies remain unclear. Here, we have expressed monoclonal ACPAs derived from patients with RA, and analyzed their functions in mice, as well as their specificities. None of the ACPAs showed arthritogenicity nor induced pain-associated behavior in mice. However, one of the antibodies, clone E4, protected mice from antibody-induced arthritis. E4 showed a binding pattern restricted to skin, macrophages and dendritic cells in lymphoid tissue, and cartilage derived from mouse and human arthritic joints. Proteomic analysis confirmed that E4 strongly binds to macrophages and certain RA synovial fluid proteins such as α-enolase. The protective effect of E4 was epitope-specific and dependent on the interaction between E4-citrullinated α-enolase immune complexes with FCGR2B on macrophages, resulting in increased IL-10 secretion and reduced osteoclastogenesis. These findings suggest that a subset of ACPAs have therapeutic potential in RA.
Collapse
|
23
|
Tan H, Li Z, Zhang S, Zhang J, Jia E. Novel perception of neutrophil extracellular traps in gouty inflammation. Int Immunopharmacol 2023; 115:109642. [PMID: 36608445 DOI: 10.1016/j.intimp.2022.109642] [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: 11/04/2022] [Revised: 12/22/2022] [Accepted: 12/23/2022] [Indexed: 01/05/2023]
Abstract
The self-limiting nature of the inflammatory flare is a feature of gout. The effects of neutrophil extracellular traps (NETs) on gout have remarkably attracted researchers' attention. Aggregated NETs promote the resolution of gouty inflammation by packing monosodium urate (MSU) crystals, degrading cytokines and chemokines, and blocking neutrophil recruitment and activation. Deficiency of NETs aggravates experimental gout. Thus, aggregated NETs are assumed to be a possible mechanism for the spontaneous resolution of gout. It is feasible to envisage therapeutic strategies for targeting NETosis (NET formation process) in gout. However, recent studies have demonstrated that levels of NETs are not associated with disease activity and inflammation in human gout. Moreover, the process of MSU crystal trapping is not affected in the absence of neutrophils. This review has concentrated on the mechanisms and associations between NETs and gout.
Collapse
Affiliation(s)
- Haibo Tan
- Shenzhen Traditional Chinese Medicine Hospital Affiliated to Nanjing University of Chinese Medicine, Shenzhen 518033, Guangdong, PR China
| | - Zhiling Li
- The Fourth Clinical Medical College of Guangzhou University of Chinese Medicine, Shenzhen 518033, Guangdong, PR China
| | - Shan Zhang
- Shenzhen Traditional Chinese Medicine Hospital Affiliated to Nanjing University of Chinese Medicine, Shenzhen 518033, Guangdong, PR China
| | - Jianyong Zhang
- The Fourth Clinical Medical College of Guangzhou University of Chinese Medicine, Shenzhen 518033, Guangdong, PR China; The Department of Rheumatology, Shenzhen Traditional Chinese Medicine Hospital, Shenzhen 518033, Guangdong, PR China.
| | - Ertao Jia
- The Fourth Clinical Medical College of Guangzhou University of Chinese Medicine, Shenzhen 518033, Guangdong, PR China; The Department of Rheumatology, Shenzhen Traditional Chinese Medicine Hospital, Shenzhen 518033, Guangdong, PR China.
| |
Collapse
|
24
|
Lv C, Sun M, Guo Y, Xia W, Qiao S, Tao Y, Fang Y, Zhang Q, Zhu Y, Yalikun Y, Xia Y, Wei Z, Dai Y. Cholinergic dysfunction-induced insufficient activation of alpha7 nicotinic acetylcholine receptor drives the development of rheumatoid arthritis through promoting protein citrullination via the SP3/PAD4 pathway. Acta Pharm Sin B 2023; 13:1600-1615. [PMID: 37139415 PMCID: PMC10150100 DOI: 10.1016/j.apsb.2023.01.016] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2022] [Revised: 10/10/2022] [Accepted: 11/23/2022] [Indexed: 01/19/2023] Open
Abstract
Both cholinergic dysfunction and protein citrullination are the hallmarks of rheumatoid arthritis (RA), but the relationship between the two phenomena remains unclear. We explored whether and how cholinergic dysfunction accelerates protein citrullination and consequently drives the development of RA. Cholinergic function and protein citrullination levels in patients with RA and collagen-induced arthritis (CIA) mice were collected. In both neuron-macrophage coculture system and CIA mice, the effect of cholinergic dysfunction on protein citrullination and expression of peptidylarginine deiminases (PADs) was assessed by immunofluorescence. The key transcription factors for PAD4 expression were predicted and validated. Cholinergic dysfunction in the patients with RA and CIA mice negatively correlated with the degree of protein citrullination in synovial tissues. The cholinergic or alpha7 nicotinic acetylcholine receptor (α7nAChR) deactivation and activation resulted in the promotion and reduction of protein citrullination in vitro and in vivo, respectively. Especially, the activation deficiency of α7nAChR induced the earlier onset and aggravation of CIA. Furthermore, deactivation of α7nAChR increased the expression of PAD4 and specificity protein-3 (SP3) in vitro and in vivo. Our results suggest that cholinergic dysfunction-induced deficient α7nAChR activation, which induces the expression of SP3 and its downstream molecule PAD4, accelerating protein citrullination and the development of RA.
Collapse
Affiliation(s)
- Changjun Lv
- Department of Pharmacology of Chinese Materia Medica, School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing 211198, China
| | - Minghui Sun
- Division of Sports Medicine and Adult Reconstructive Surgery, Department of Orthopedic Surgery, Nanjing Drum Tower Hospital, the Affiliated Hospital of Nanjing University Medical School, Nanjing 210008, China
| | - Yilei Guo
- Department of Pharmacology of Chinese Materia Medica, School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing 211198, China
| | - Wenxin Xia
- Department of Pharmacology of Chinese Materia Medica, School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing 211198, China
| | - Simiao Qiao
- Department of Pharmacology of Chinese Materia Medica, School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing 211198, China
| | - Yu Tao
- Department of Pharmacology of Chinese Materia Medica, School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing 211198, China
| | - Yulai Fang
- Department of Pharmacology of Chinese Materia Medica, School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing 211198, China
| | - Qin Zhang
- Department of Pharmacology of Chinese Materia Medica, School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing 211198, China
| | - Yanrong Zhu
- Department of Pharmacology of Chinese Materia Medica, School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing 211198, China
| | - Yusufu Yalikun
- Division of Sports Medicine and Adult Reconstructive Surgery, Department of Orthopedic Surgery, Nanjing Drum Tower Hospital, the Affiliated Hospital of Nanjing University Medical School, Nanjing 210008, China
| | - Yufeng Xia
- Department of Pharmacology of Chinese Materia Medica, School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing 211198, China
| | - Zhifeng Wei
- Department of Pharmacology of Chinese Materia Medica, School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing 211198, China
- Corresponding authors.
| | - Yue Dai
- Department of Pharmacology of Chinese Materia Medica, School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing 211198, China
- Corresponding authors.
| |
Collapse
|
25
|
Zhai M, Gong S, Luan P, Shi Y, Kou W, Zeng Y, Shi J, Yu G, Hou J, Yu Q, Jian W, Zhuang J, Feinberg MW, Peng W. Extracellular traps from activated vascular smooth muscle cells drive the progression of atherosclerosis. Nat Commun 2022; 13:7500. [PMID: 36473863 PMCID: PMC9723654 DOI: 10.1038/s41467-022-35330-1] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2022] [Accepted: 11/28/2022] [Indexed: 12/12/2022] Open
Abstract
Extracellular DNA traps (ETs) represent an immune response by which cells release essential materials like chromatin and granular proteins. Previous studies have demonstrated that the transdifferentiation of vascular smooth muscle cells (VSMCs) plays a crucial role in atherosclerosis. This study seeks to investigate the interaction between CD68+ VSMCs and the formation of ETs and highlight its function in atherosclerosis. Here we show that ETs are inhibited, and atherosclerotic plaque formation is alleviated in male Myh11CrePad4flox/flox mice undergoing an adeno-associated-virus-8 (AAV8) mediating overexpression of proprotein convertase subtilisin/kexin type 9 mutation (PCSK9) injection and being challenged with a high-fat diet. Obvious ETs generated from CD68+ VSMCs are inhibited by Cl-amidine and DNase I in vitro. By utilizing VSMCs-lineage tracing technology and single-cell RNA sequencing (scRNA-seq), we demonstrate that the ETs from CD68+ VSMCs influence the progress of atherosclerosis by regulating the direction of VSMCs' transdifferentiation through STING-SOCS1 or TLR4 signaling pathway.
Collapse
Affiliation(s)
- Ming Zhai
- grid.13402.340000 0004 1759 700XDepartment of Cardiology, Shanghai Tenth People’s Hospital, Tongji University, School of Medicine, Shanghai, China
| | - Shiyu Gong
- grid.13402.340000 0004 1759 700XDepartment of Cardiology, Shanghai Tenth People’s Hospital, Tongji University, School of Medicine, Shanghai, China
| | - Peipei Luan
- grid.13402.340000 0004 1759 700XDepartment of Cardiology, Shanghai Tenth People’s Hospital, Tongji University, School of Medicine, Shanghai, China
| | - Yefei Shi
- grid.13402.340000 0004 1759 700XDepartment of Cardiology, Shanghai Tenth People’s Hospital, Tongji University, School of Medicine, Shanghai, China
| | - Wenxin Kou
- grid.13402.340000 0004 1759 700XDepartment of Cardiology, Shanghai Tenth People’s Hospital, Tongji University, School of Medicine, Shanghai, China
| | - Yanxi Zeng
- grid.13402.340000 0004 1759 700XDepartment of Cardiology, Shanghai Tenth People’s Hospital, Tongji University, School of Medicine, Shanghai, China
| | - Jiayun Shi
- grid.13402.340000 0004 1759 700XDepartment of Cardiology, Shanghai Tenth People’s Hospital, Tongji University, School of Medicine, Shanghai, China
| | - Guanye Yu
- grid.13402.340000 0004 1759 700XDepartment of Cardiology, Shanghai Tenth People’s Hospital, Tongji University, School of Medicine, Shanghai, China
| | - Jiayun Hou
- grid.8547.e0000 0001 0125 2443Biomedical Research Center, Zhongshan Hospital Institute of Clinical Science, Fudan University, Shanghai, China
| | - Qing Yu
- grid.13402.340000 0004 1759 700XDepartment of Cardiology, Shanghai Tenth People’s Hospital, Tongji University, School of Medicine, Shanghai, China
| | - Weixia Jian
- grid.13402.340000 0004 1759 700XDepartment of Endocrinology, Xinhua Hospital, Shanghai Jiaotong University, School of Medicine, Shanghai, China
| | - Jianhui Zhuang
- grid.13402.340000 0004 1759 700XDepartment of Cardiology, Shanghai Tenth People’s Hospital, Tongji University, School of Medicine, Shanghai, China
| | - Mark W. Feinberg
- grid.38142.3c000000041936754XCardiovascular Division, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA USA
| | - Wenhui Peng
- grid.13402.340000 0004 1759 700XDepartment of Cardiology, Shanghai Tenth People’s Hospital, Tongji University, School of Medicine, Shanghai, China
| |
Collapse
|
26
|
Zhang F, Li Y, Wu J, Zhang J, Cao P, Sun Z, Wang W. The role of extracellular traps in ischemia reperfusion injury. Front Immunol 2022; 13:1022380. [PMID: 36211432 PMCID: PMC9533173 DOI: 10.3389/fimmu.2022.1022380] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2022] [Accepted: 08/31/2022] [Indexed: 11/29/2022] Open
Abstract
In response to strong signals, several types of immune cells release extracellular traps (ETs), which are web-like structures consisting of DNA decorated with various protein substances. This process is most commonly observed in neutrophils. Over the past two decades, ET formation has been recognized as a unique mechanism of host defense and pathogen destruction. However, the role of ETs in sterile inflammation has only been studied extensively in recent years. Ischemia reperfusion injury (IRI) is a type of sterile inflammatory injury. Several studies have reported that ETs have an important role in IRI in various organs. In this review, we describe the release of ETs by various types of immune cells and focus on the mechanism underlying the formation of neutrophil ETs (NETs). In addition, we summarize the role of ETs in IRI in different organs and their effects on tumors. Finally, we discuss the value of ETs as a potential therapeutic target for organ IRI and present possible challenges in conducting studies on IRI-related ETs as well as future research directions and prospects.
Collapse
Affiliation(s)
- Feilong Zhang
- Department of Urology, Beijing Chao-yang Hospital, Capital Medical University, Beijing, China
- Institute of Urology, Capital Medical University, Beijing, China
| | - Yuqing Li
- Department of Urology, Beijing Chao-yang Hospital, Capital Medical University, Beijing, China
- Institute of Urology, Capital Medical University, Beijing, China
| | - Jiyue Wu
- Department of Urology, Beijing Chao-yang Hospital, Capital Medical University, Beijing, China
- Institute of Urology, Capital Medical University, Beijing, China
| | - Jiandong Zhang
- Department of Urology, Beijing Chao-yang Hospital, Capital Medical University, Beijing, China
- Institute of Urology, Capital Medical University, Beijing, China
| | - Peng Cao
- Department of Urology, Beijing Chao-yang Hospital, Capital Medical University, Beijing, China
- Institute of Urology, Capital Medical University, Beijing, China
| | - Zejia Sun
- Department of Urology, Beijing Chao-yang Hospital, Capital Medical University, Beijing, China
- Institute of Urology, Capital Medical University, Beijing, China
| | - Wei Wang
- Department of Urology, Beijing Chao-yang Hospital, Capital Medical University, Beijing, China
- Institute of Urology, Capital Medical University, Beijing, China
- *Correspondence: Wei Wang,
| |
Collapse
|
27
|
From vaccines to nanovaccines: A promising strategy to revolutionize rheumatoid arthritis treatment. J Control Release 2022; 350:107-121. [PMID: 35977582 DOI: 10.1016/j.jconrel.2022.08.020] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2022] [Revised: 08/08/2022] [Accepted: 08/10/2022] [Indexed: 10/15/2022]
Abstract
Rheumatoid arthritis (RA) is a joint-related autoimmune disease that is difficult to cure. Most therapeutics act to alleviate the symptoms but not correct the causes of RA. Novel strategies that specifically target the causes are highly needed for RA management. Currently, early interruption of RA is increasingly suggested but the corresponding therapeutics are not available. Vaccines that have shown great success to combat infection, cancer, degenerative diseases, autoimmune diseases, etc. are ideal candidates for a new generation of anti-RA therapeutics to correct the causes and prevent RA or interrupt RA in early phases. Anti-RA vaccines can be divided into two major categories. One is to induce neutralizing antibodies and the other is to induce antigen-specific immune tolerance. The vaccines are inherently linked to nanotechnology because they usually need a biomacromolecule or carrier to provoke sufficient immune responses. In the past decade, designed nanocarriers such as nanoparticles, liposomes, nanoemulsion, etc., have been applied to optimize the vaccines for autoimmune disease treatment. Nanotechnology endows vaccines with a higher biostability, tunable in vivo behavior, better targeting, co-delivery with stimulatory agents, regulatory effects on immune responses, etc. In this review, unmet medical needs for RA treatment and anti-RA vaccinology are first introduced. The development of anti-RA therapies from vaccines to nanovaccines are then reviewed and perspectives on how nanotechnology promotes vaccine development and advancement are finally provided. In addition, challenges for anti-RA vaccine development are summarized and advantages of nanovaccines are analyzed. In conclusion, nanovaccines will be a promising strategy to revolutionize the treatment of RA by correcting the causes in an early phase of RA.
Collapse
|
28
|
Wen E, Xin G, Li S, Dong Y, Zhu Y, Wan C, Yu X, Wei Z, Wang Y, Li F, Zhang K, Niu H, Huang W. Tuftsin ameliorates splenic inflammatory injury by promoting neuropilin-1 in severe acute pancreatitis. Biochem Pharmacol 2022; 199:115030. [PMID: 35381211 DOI: 10.1016/j.bcp.2022.115030] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2022] [Revised: 03/29/2022] [Accepted: 03/29/2022] [Indexed: 02/08/2023]
Abstract
Severe acute pancreatitis (SAP)-associated spleen injury causing immune disturbances aggravates organs injuries, which contributes to higher mortality rate. However, there are no effective drugs to cure SAP-induced spleen injury. Here, we found that Tuftsin (TN) is effective for ameliorating SAP-induced pathological damage and inflammation of spleen, mainly via alleviating mitochondrial dysfunction, oxidative stress, ATP depletion and the expression of pro-inflammatory factors. We further found that TN promoted anti-inflammatory macrophage phenotype M2 via up-regulating NRP1 on macrophage in spleen during SAP. Meanwhile, EG00229 (an inhibitor of NRP1 bound to TN) weakened TN's therapeutic effect in SAP-associated spleen injury. And EG00229 also inhibited M2 macrophage, leading to increasing inflammasome formation. Additionally, EG00229 reduced the protective efficiency of TN on mitochondrial dysfunction, and inflammation injury via NRP1 in spleen caused by SAP. Similarly, siRNA-Nrp1 into macrophage also prevented TN's inhibition on apoptosis. These findings reveal that TN alleviates SAP-induced spleen injury by promoting NRP1.
Collapse
Affiliation(s)
- E Wen
- Laboratory of Ethnopharmacology, Tissue-orientated Property of Chinese Medicine Key Laboratory of Sichuan Province, West China School of Medicine, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Guang Xin
- Laboratory of Ethnopharmacology, Tissue-orientated Property of Chinese Medicine Key Laboratory of Sichuan Province, West China School of Medicine, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Shiyi Li
- Laboratory of Ethnopharmacology, Tissue-orientated Property of Chinese Medicine Key Laboratory of Sichuan Province, West China School of Medicine, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Yuman Dong
- Laboratory of Ethnopharmacology, Tissue-orientated Property of Chinese Medicine Key Laboratory of Sichuan Province, West China School of Medicine, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Yuda Zhu
- Laboratory of Ethnopharmacology, Tissue-orientated Property of Chinese Medicine Key Laboratory of Sichuan Province, West China School of Medicine, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Chengyu Wan
- Laboratory of Ethnopharmacology, Tissue-orientated Property of Chinese Medicine Key Laboratory of Sichuan Province, West China School of Medicine, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Xiuxian Yu
- Laboratory of Ethnopharmacology, Tissue-orientated Property of Chinese Medicine Key Laboratory of Sichuan Province, West China School of Medicine, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Zeliang Wei
- Laboratory of Ethnopharmacology, Tissue-orientated Property of Chinese Medicine Key Laboratory of Sichuan Province, West China School of Medicine, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Yilan Wang
- Laboratory of Ethnopharmacology, Tissue-orientated Property of Chinese Medicine Key Laboratory of Sichuan Province, West China School of Medicine, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Fan Li
- Laboratory of Ethnopharmacology, Tissue-orientated Property of Chinese Medicine Key Laboratory of Sichuan Province, West China School of Medicine, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Kun Zhang
- Laboratory of Ethnopharmacology, Tissue-orientated Property of Chinese Medicine Key Laboratory of Sichuan Province, West China School of Medicine, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Hai Niu
- Laboratory of Ethnopharmacology, Tissue-orientated Property of Chinese Medicine Key Laboratory of Sichuan Province, West China School of Medicine, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Wen Huang
- Laboratory of Ethnopharmacology, Tissue-orientated Property of Chinese Medicine Key Laboratory of Sichuan Province, West China School of Medicine, West China Hospital, Sichuan University, Chengdu, Sichuan, China.
| |
Collapse
|
29
|
Weng W, Hu Z, Pan Y. Macrophage Extracellular Traps: Current Opinions and the State of Research regarding Various Diseases. J Immunol Res 2022; 2022:7050807. [PMID: 35036449 PMCID: PMC8759907 DOI: 10.1155/2022/7050807] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2021] [Revised: 12/01/2021] [Accepted: 12/08/2021] [Indexed: 12/11/2022] Open
Abstract
Macrophages are an important component of the human immune system and play a key role in the immune response, which can protect the body against infection and regulate the development of tissue inflammation. Some studies found that macrophages can produce extracellular traps (ETs) under various conditions of stimulation. ETs are web-like structures that consist of proteins and DNA. ETs are thought to immobilize and kill microorganisms, as well as play an important role in tissue damage, inflammatory progression, and autoimmune diseases. In this review, the structure, identification, mechanism, and research progress of macrophage extracellular traps (METs) in related diseases are reviewed.
Collapse
Affiliation(s)
- Weizhen Weng
- Division of Rheumatology, Department of Internal Medicine, The Third Affiliated Hospital, Sun Yat-Sen University, Guangzhou 510630, China
| | - Zuoyu Hu
- Division of Rheumatology, Department of Internal Medicine, The Third Affiliated Hospital, Sun Yat-Sen University, Guangzhou 510630, China
| | - Yunfeng Pan
- Division of Rheumatology, Department of Internal Medicine, The Third Affiliated Hospital, Sun Yat-Sen University, Guangzhou 510630, China
| |
Collapse
|
30
|
Aly NAR, Rizk S, Aboul Enein A, El Desoukey N, Zawam H, Ahmed M, El Shikh ME, Pitzalis C. The role of lymphoid tissue SPARC in the pathogenesis and response to treatment of multiple myeloma. Front Oncol 2022; 12:1009993. [PMID: 36605435 PMCID: PMC9807864 DOI: 10.3389/fonc.2022.1009993] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2022] [Accepted: 11/25/2022] [Indexed: 12/24/2022] Open
Abstract
Background Despite the significant progress in the treatment of multiple myeloma (MM), the disease remains untreatable and its cure is still an unmet clinical need. Neoplastic transformation in MM is initiated in the germinal centers (GCs) of secondary lymphoid tissue (SLT) where B cells experience extensive somatic hypermutation induced by follicular dendritic cells (FDCs) and T-cell signals. Objective We reason that secreted protein acidic and rich in cysteine (SPARC), a common stromal motif expressed by FDCs at the origin (SLTs) and the destination (BM) of MM, plays a role in the pathogenesis of MM, and, here, we sought to investigate this role. Methods There were 107 BM biopsies from 57 MM patients (taken at different time points) together with 13 control specimens assessed for SPARC gene and protein expression and compared with tonsillar tissues. In addition, regulation of myeloma-promoting genes by SPARC-secreting FDCs was assessed in in vitro GC reactions (GCRs). Results SPARC gene expression was confirmed in both human primary (BM) and secondary (tonsils) lymphoid tissues, and the expression was significantly higher in the BM. Sparc was detectable in the BM and tonsillar lysates, co-localized with the FDC markers in both tissues, and stimulation of FDCs in vitro induced significantly higher levels of SPARC expression than unstimulated controls. In addition, SPARC inversely correlated with BM PC infiltration, ISS staging, and ECOG performance of the MM patients, and in vitro addition of FDCs to lymphocytes inhibited the expression of several oncogenes associated with malignant transformation of PCs. Conclusion FDC-SPARC inhibits several myelomagenic gene expression and inversely correlates with PC infiltration and MM progression. Therapeutic induction of SPARC expression through combinations of the current MM drugs, repositioning of non-MM drugs, or novel drug discovery could pave the way to better control MM in clinically severe and drug-resistant patients.
Collapse
Affiliation(s)
- Nesreen Amer Ramadan Aly
- Clinical and Chemical Pathology Department, Faculty of Medicine, Cairo University, Cairo, Egypt
- Centre for Experimental Medicine and Rheumatology, William Harvey Research Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London, United Kingdom
| | - Samia Rizk
- Clinical and Chemical Pathology Department, Faculty of Medicine, Cairo University, Cairo, Egypt
| | - Azza Aboul Enein
- Clinical and Chemical Pathology Department, Faculty of Medicine, Cairo University, Cairo, Egypt
| | - Nermeen El Desoukey
- Clinical and Chemical Pathology Department, Faculty of Medicine, Cairo University, Cairo, Egypt
| | - Hamdy Zawam
- Clinical Oncology and Nuclear Radiation Department, Faculty of Medicine, Cairo University, Cairo, Egypt
| | - Manzoor Ahmed
- Centre for Experimental Medicine and Rheumatology, William Harvey Research Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London, United Kingdom
| | - Mohey Eldin El Shikh
- Centre for Experimental Medicine and Rheumatology, William Harvey Research Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London, United Kingdom
- *Correspondence: Mohey Eldin El Shikh,
| | - Costantino Pitzalis
- Centre for Experimental Medicine and Rheumatology, William Harvey Research Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London, United Kingdom
| |
Collapse
|
31
|
Chen T, Wang Y, Nan Z, Wu J, Li A, Zhang T, Qu X, Li C. Interaction Between Macrophage Extracellular Traps and Colon Cancer Cells Promotes Colon Cancer Invasion and Correlates With Unfavorable Prognosis. Front Immunol 2021; 12:779325. [PMID: 34925357 PMCID: PMC8671452 DOI: 10.3389/fimmu.2021.779325] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2021] [Accepted: 11/15/2021] [Indexed: 01/03/2023] Open
Abstract
BackgroundMacrophage extracellular traps (METs) and tumor-infiltrating macrophages contribute to the progression of several diseases. But the role of METs and tumor-infiltrating macrophages in colon cancer (CC) has not been illuminated. In this study, we aimed to clarify the prognostic value of METs for CC patients and to explore the interaction between CC cells and METs in vitro and in vivo.MethodsA training cohort consisting of 116 patients and a validation cohort of 94 patients were enrolled in this study. Immunofluorescence (IF) staining was conducted to determine METs formation in CC patients. Cox regression was used to perform prognostic analysis and screen out the best prognostic model. A nomogram was established to predict 5-year overall survival (OS). The correlation between METs with clinicopathological features and inflammatory markers was analyzed. The formation of METs in vitro was detected by SYTOX® green and IF staining, and the effect of METs on CC cells was detected by transwell assays. PAD2-IN-1, a selective inhibitor of peptidylarginine deiminase 2 (PAD2), was introduced to destroy the crosstalk between CC cells and METs in vitro and in vivo.ResultsMETs levels were higher in CC tissues and were an independent prognostic factor for CC patients. The prognostic model consisting of age, tumors local invasion, lymph node metastasis and METs were confirmed to be consistent and accurate for predicting the 5-year OS of CC patients. Besides, METs were correlated with distant metastasis and inflammation. Through in vitro experiments, we confirmed that there was a positive feedback loop between CC cells and METs, in that METs promoted the invasion of CC cells and CC cells enhanced the production of METs, in turn. This interaction could be blocked by PAD2-IN-1 inhibitors. More importantly, animal experiments revealed that PAD2-IN-1 inhibited METs formation and CC liver metastasis in vivo.ConclusionsMETs were the potential biomarker of CC patient prognosis. PAD2-IN-1 inhibited the crosstalk between CC cells and METs in vitro and in vivo, which should be emphasized in CC therapy.
Collapse
Affiliation(s)
- Tianli Chen
- Department of General Surgery, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China
- Department of Colorectal Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Yue Wang
- Department of General Surgery, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Zhaodi Nan
- Institute of Basic Medical Sciences, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Jie Wu
- Department of Urology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Ailu Li
- Department of Obstetrics and Gynecology, Quanzhou First Hospital Affiliated to Fujian Medical University, Quanzhou, China
| | - Tingguo Zhang
- Department of Pathology, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Xun Qu
- Institute of Basic Medical Sciences, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Chen Li
- Department of Ultrasound, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China
- *Correspondence: Chen Li,
| |
Collapse
|
32
|
Abstract
Based on the PubMed data, we have been performing a yearly evaluation of the publications related to autoimmune diseases and immunology to ascertain the relative weight of the former in the scientific literature. It is particularly intriguing to observe that despite the numerous new avenues of immune-related mechanisms, such as cancer immunotherapy, the proportion of immunology manuscripts related to autoimmunity continues to increase and has been approaching 20% in 2019. As in the previous 13 years, we performed an arbitrary selection of the peer-reviewed articles published by the major dedicated Journals and discussed the common themes which continue to outnumber peculiarites in autoimmune diseases. The investigated areas included systemic lupus erythematosus (SLE), rheumatoid arthritis (RA), psoriatic arthritis (PsA), autoantibodies (autoAbs), and common therapeutic avenues and novel pathogenic mechanisms for autoimmune conditions. Some examples include new pathogenetic evidence which is well represented by IL21 or P2X7 receptor (P2X7R) in SLE or the application of single-cell RNA sequencing (scRNA-seq), mass cytometry, bulk RNA sequencing (RNA-seq), and flow cytometry for the analysis of different cellular populations in RA. Cumulatively and of interest to the clinicians, a large number of findings continue to underline the importance of a strict relationship between basic and clinical science to define new pathogenetic and therapeutic developments. The therapeutic pipeline in autoimmunity continues to grow and maintain a constant flow of new molecules, as well illustrated in RA and PsA, and this is most certainly derived from the new basic evidence and the high-throughput tools applied to autoimmune diseases.
Collapse
|
33
|
Song N, Wang W, Wang Y, Guan Y, Xu S, Guo MY. Hydrogen sulfide of air induces macrophage extracellular traps to aggravate inflammatory injury via the regulation of miR-15b-5p on MAPK and insulin signals in trachea of chickens. THE SCIENCE OF THE TOTAL ENVIRONMENT 2021; 771:145407. [PMID: 33548704 DOI: 10.1016/j.scitotenv.2021.145407] [Citation(s) in RCA: 41] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/19/2020] [Revised: 01/16/2021] [Accepted: 01/20/2021] [Indexed: 06/12/2023]
Abstract
Hydrogen sulfide (H2S) is an environmental contaminant to cause the airway damage. The release of macrophage extracellular traps (METs) is the mechanism of immune protection to harmful stimulation via microRNAs, but excessive METs cause the injury. However, few studies have attempted to interpret the mechanism of an organism injury due to H2S via METs in chickens. Here, we investigated the transcriptome profiles, pathological morphologic changes and METs release from chicken trachea after H2S exposure. Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) analysis revealed that 10 differentially expressed genes were related to the METs release, the MAPK and insulin signaling pathways. Morphological and immunofluorescence analysis showed that H2S caused airway injury and MET release. H2S activated the targeting effect of miRNA-15b-5p on activating transcription factor 2 (ATF2). Western blotting and real time quantitative PCR results showed that H2S down-regulated the levels of dual specificity protein phosophatase1 (DUSP1) but up-regulated p38 MAP Kinase (p38) in the MAPK signal pathway. And the expression of phosphoinositide-dependent protein kinase 1 (PDK1), serine/threonine kinase (Akt), and protein kinase ζ subtypes (PKCζ) in the insulin signal pathway were increased after H2S exposure. These promoted the release of myeloperoxidase (MPO) and degradation histone 4 (H4) to induce the release of METs. Taken together, miR-15b-5p targeted ATF2 to mediate METs release, which triggered trachea inflammatory injury via MAPK and insulin signals after H2S exposure. These results will provide new insights into the toxicological mechanisms of H2S and environmental ecotoxicology.
Collapse
Affiliation(s)
- Nuan Song
- College of Veterinary Medicine, Northeast Agricultural University; Key Laboratory of the Provincial Education Department of Heilongjiang for Common Animal Disease Prevention and Treatment, Harbin 150030, China
| | - Wei Wang
- College of Veterinary Medicine, Northeast Agricultural University; Key Laboratory of the Provincial Education Department of Heilongjiang for Common Animal Disease Prevention and Treatment, Harbin 150030, China
| | - Yue Wang
- College of Veterinary Medicine, Northeast Agricultural University; Key Laboratory of the Provincial Education Department of Heilongjiang for Common Animal Disease Prevention and Treatment, Harbin 150030, China
| | - Yalin Guan
- College of Life Science, Northeast Agricultural University, Harbin 150030, China
| | - Shiwen Xu
- College of Veterinary Medicine, Northeast Agricultural University; Key Laboratory of the Provincial Education Department of Heilongjiang for Common Animal Disease Prevention and Treatment, Harbin 150030, China
| | - Meng-Yao Guo
- College of Veterinary Medicine, Northeast Agricultural University; Key Laboratory of the Provincial Education Department of Heilongjiang for Common Animal Disease Prevention and Treatment, Harbin 150030, China.
| |
Collapse
|
34
|
Yang ML, Sodré FMC, Mamula MJ, Overbergh L. Citrullination and PAD Enzyme Biology in Type 1 Diabetes - Regulators of Inflammation, Autoimmunity, and Pathology. Front Immunol 2021; 12:678953. [PMID: 34140951 PMCID: PMC8204103 DOI: 10.3389/fimmu.2021.678953] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2021] [Accepted: 05/11/2021] [Indexed: 02/06/2023] Open
Abstract
The generation of post-translational modifications (PTMs) in human proteins is a physiological process leading to structural and immunologic variety in proteins, with potentially altered biological functions. PTMs often arise through normal responses to cellular stress, including general oxidative changes in the tissue microenvironment and intracellular stress to the endoplasmic reticulum or immune-mediated inflammatory stresses. Many studies have now illustrated the presence of 'neoepitopes' consisting of PTM self-proteins that induce robust autoimmune responses. These pathways of inflammatory neoepitope generation are commonly observed in many autoimmune diseases including systemic lupus erythematosus, rheumatoid arthritis, multiple sclerosis, and type 1 diabetes (T1D), among others. This review will focus on one specific PTM to self-proteins known as citrullination. Citrullination is mediated by calcium-dependent peptidylarginine deiminase (PAD) enzymes, which catalyze deimination, the conversion of arginine into the non-classical amino acid citrulline. PADs and citrullinated peptides have been associated with different autoimmune diseases, notably with a prominent role in the diagnosis and pathology of rheumatoid arthritis. More recently, an important role for PADs and citrullinated self-proteins has emerged in T1D. In this review we will provide a comprehensive overview on the pathogenic role for PADs and citrullination in inflammation and autoimmunity, with specific focus on evidence for their role in T1D. The general role of PADs in epigenetic and transcriptional processes, as well as their crucial role in histone citrullination, neutrophil biology and neutrophil extracellular trap (NET) formation will be discussed. The latter is important in view of increasing evidence for a role of neutrophils and NETosis in the pathogenesis of T1D. Further, we will discuss the underlying processes leading to citrullination, the genetic susceptibility factors for increased recognition of citrullinated epitopes by T1D HLA-susceptibility types and provide an overview of reported autoreactive responses against citrullinated epitopes, both of T cells and autoantibodies in T1D patients. Finally, we will discuss recent observations obtained in NOD mice, pointing to prevention of diabetes development through PAD inhibition, and the potential role of PAD inhibitors as novel therapeutic strategy in autoimmunity and in T1D in particular.
Collapse
Affiliation(s)
- Mei-Ling Yang
- Section of Rheumatology, Allergy and Clinical Immunology, Department of Internal Medicine, Yale University, New Haven, CT, United States
| | - Fernanda M C Sodré
- Department of Chronic Diseases, Metabolism and Ageing, Laboratory of Clinical and Experimental Endocrinology (CEE), KU Leuven, Leuven, Belgium
| | - Mark J Mamula
- Section of Rheumatology, Allergy and Clinical Immunology, Department of Internal Medicine, Yale University, New Haven, CT, United States
| | - Lut Overbergh
- Department of Chronic Diseases, Metabolism and Ageing, Laboratory of Clinical and Experimental Endocrinology (CEE), KU Leuven, Leuven, Belgium
| |
Collapse
|
35
|
Alghamdi MA, Redwan EM. Interplay of Microbiota and Citrullination in the Immunopathogenesis of Rheumatoid Arthritis. Probiotics Antimicrob Proteins 2021; 14:99-113. [PMID: 34036479 DOI: 10.1007/s12602-021-09802-7] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/19/2021] [Indexed: 12/18/2022]
Abstract
Microbiota is a balanced ecosystem that has important functions to the host health including development, defense, digestion, and absorption of dietary fibers and minerals, vitamin synthesizes, protection, and training the host immune system. On the other hand, its dysbiosis is linked to many human diseases such as rheumatoid arthritis (RA). The RA is an inflammatory autoimmune disorder caused by genetic and environmental factors; microbiota may be considered as a risk environmental factor for it. Citrullination is a post-translation modification (PMT) that converts the amino acid arginine to amino acid citrulline in certain proteins. These citrullinated proteins are recognized as a foreign antigen by the immune system resulting in the upregulation of inflammatory action such as in RA. The current work highlights the effect of both gut and oral microbiota dysbiosis on the development of RA, as well as discusses how the alteration in microbiota composition leads to the overgrowth of some bacterial species that entangled in RA pathogenicity. The evidence suggested that some oral and gut microbial species such as Porphyromonas gingivalis and Prevotella copri, respectively, contribute to RA pathogenesis. During dysbiosis, these bacteria can mediate the citrullination of either human or bacteria proteins to trigger an immune response that leads to the generation of autoantibodies.
Collapse
Affiliation(s)
- Mohammed A Alghamdi
- Biological Science Department, Faculty of Science, King Abdulaziz University, P.O. Box 80203, Jeddah, 21589, Saudi Arabia.,Laboratory Department, University Medical Services Center, King Abdulaziz University, P.O. Box 80200, Jeddah, 21589, Saudi Arabia
| | - Elrashdy M Redwan
- Biological Science Department, Faculty of Science, King Abdulaziz University, P.O. Box 80203, Jeddah, 21589, Saudi Arabia. .,Therapeutic and Protective Proteins Laboratory, Protein Research Department, Genetic Engineering and Biotechnology Research Institute, City for Scientific Research and Technology Applications, New Borg EL-Arab, Alexandria, 21934, Egypt.
| |
Collapse
|
36
|
Bruggeman Y, Sodré FMC, Buitinga M, Mathieu C, Overbergh L, Kracht MJL. Targeting citrullination in autoimmunity: insights learned from preclinical mouse models. Expert Opin Ther Targets 2021; 25:269-281. [PMID: 33896351 DOI: 10.1080/14728222.2021.1918104] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
INTRODUCTION Aberrant citrullination and excessive peptidylarginine deiminase (PAD) activity are detected in numerous challenging autoimmune diseases such as rheumatoid arthritis, inflammatory bowel diseases, systemic lupus erythematosus, multiple sclerosis, and type 1 diabetes. Because excessive PAD activity is a common denominator in these diseases, PADs are interesting potential therapeutic targets for future therapies. AREAS COVERED This review summarizes the advances made in the design of PAD inhibitors, their utilization and therapeutic potential in preclinical mouse models of autoimmunity. Relevant literature encompasses studies from 1994 to 2021 that are available on PubMed.gov. EXPERT OPINION Pan-PAD inhibition is a promising therapeutic strategy for autoimmune diseases. Drugs achieving pan-PAD inhibition were capable of ameliorating, reversing, and preventing clinical symptoms in preclinical mouse models. However, the implications for PADs in key biological processes potentially present a high risk for clinical complications and could hamper the translation of PAD inhibitors to the clinic. We envisage that PAD isozyme-specific inhibitors will improve the understanding the role of PAD isozymes in disease pathology, reduce the risk of side-effects and enhance prospects for future clinical translation.
Collapse
Affiliation(s)
- Ylke Bruggeman
- Department of Chronic Diseases and Metabolism, Laboratory for Clinical and Experimental Endocrinology, KU Leuven, Leuven, Belgium
| | - Fernanda M C Sodré
- Department of Chronic Diseases and Metabolism, Laboratory for Clinical and Experimental Endocrinology, KU Leuven, Leuven, Belgium
| | - Mijke Buitinga
- Department of Chronic Diseases and Metabolism, Laboratory for Clinical and Experimental Endocrinology, KU Leuven, Leuven, Belgium.,Department of Nutrition and Movement Sciences, Maastricht University, Maastricht, The Netherlands.,Department of Radiology and Nuclear Medicine, Maastricht University Medical Center, Maastricht, The Netherlands
| | - Chantal Mathieu
- Department of Chronic Diseases and Metabolism, Laboratory for Clinical and Experimental Endocrinology, KU Leuven, Leuven, Belgium
| | - Lut Overbergh
- Department of Chronic Diseases and Metabolism, Laboratory for Clinical and Experimental Endocrinology, KU Leuven, Leuven, Belgium
| | - Maria J L Kracht
- Department of Chronic Diseases and Metabolism, Laboratory for Clinical and Experimental Endocrinology, KU Leuven, Leuven, Belgium
| |
Collapse
|
37
|
Cai X, Panicker SR, Biswas I, Giri H, Rezaie AR. Protective Role of Activated Protein C against Viral Mimetic Poly(I:C)-Induced Inflammation. Thromb Haemost 2021; 121:1448-1463. [PMID: 33706396 DOI: 10.1055/s-0041-1726093] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Activated protein C (APC) is an anticoagulant plasma serine protease which exhibits potent cytoprotective and anti-inflammatory activities. Here, we studied protective effects of APC on the proinflammatory function of polyinosinic:polycytidylic acid [poly(I:C)], a synthetic analog of viral double-stranded RNA, in cellular and animal models. Poly(I:C) induced histone H3 extranuclear translocation via interaction with toll-like receptor 3 in two established endothelial cell lines. Furthermore, poly(I:C) induced histone H3 extranuclear translocation in J774A.1 macrophages and human neutrophils and formation of macrophage and neutrophil extracellular traps (ETs). Mechanistically, poly(I:C) was found to upregulate expression of peptidylarginine deiminase 4 and enhance its interaction with histone H3, thereby leading to increased histone citrullination and neutrophil ET formation. Poly(I:C) elicited proinflammatory signaling responses by inducing nuclear factor kappa B activation and disrupting endothelial cell permeability. In vivo, poly(I:C) enhanced cell surface expression of Mac-1 on neutrophils in mice and facilitated their infiltration to lung tissues. Poly(I:C) also downregulated thrombomodulin expression in mouse tissues and reduced its circulating soluble level in plasma. We demonstrate in this study that APC and a signaling-selective mutant of APC effectively inhibit proinflammatory signaling effects of poly(I:C) in both cellular and animal models. We further demonstrate that unlike the requirement for endothelial protein C receptor on endothelial cells, the integrin Mac-1 is involved in the protease-activated receptor 1-dependent APC inhibition of macrophage ET formation in J774A.1 cells. Taken together, these results support a key role for APC signaling in inhibiting the viral mimetic-induced proinflammatory signaling responses and histone translocation-associated formation of ETs by innate immune cells.
Collapse
Affiliation(s)
- Xiaofeng Cai
- Cardiovascular Biology Research Program, Oklahoma Medical Research Foundation, Oklahoma City, Oklahoma, United States
| | - Sumith R Panicker
- Cardiovascular Biology Research Program, Oklahoma Medical Research Foundation, Oklahoma City, Oklahoma, United States
| | - Indranil Biswas
- Cardiovascular Biology Research Program, Oklahoma Medical Research Foundation, Oklahoma City, Oklahoma, United States
| | - Hemant Giri
- Cardiovascular Biology Research Program, Oklahoma Medical Research Foundation, Oklahoma City, Oklahoma, United States
| | - Alireza R Rezaie
- Cardiovascular Biology Research Program, Oklahoma Medical Research Foundation, Oklahoma City, Oklahoma, United States.,Department of Biochemistry and Molecular Biology, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma, United States
| |
Collapse
|
38
|
Ramos-Martínez E, Hernández-González L, Ramos-Martínez I, Pérez-Campos Mayoral L, López-Cortés GI, Pérez-Campos E, Mayoral Andrade G, Hernández-Huerta MT, José MV. Multiple Origins of Extracellular DNA Traps. Front Immunol 2021; 12:621311. [PMID: 33717121 PMCID: PMC7943724 DOI: 10.3389/fimmu.2021.621311] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2020] [Accepted: 01/06/2021] [Indexed: 01/21/2023] Open
Abstract
Extracellular DNA traps (ETs) are evolutionarily conserved antimicrobial mechanisms present in protozoa, plants, and animals. In this review, we compare their similarities in species of different taxa, and put forward the hypothesis that ETs have multiple origins. Our results are consistent with a process of evolutionary convergence in multicellular organisms through the application of a congruency test. Furthermore, we discuss why multicellularity is related to the presence of a mechanism initiating the formation of ETs.
Collapse
Affiliation(s)
- Edgar Ramos-Martínez
- School of Sciences, Benito Juárez Autonomous University of Oaxaca, Oaxaca, Mexico
| | | | - Iván Ramos-Martínez
- Glycobiology, Cell Growth and Tissue Repair Research Unit (Gly-CRRET), Université Paris Est Créteil (UPEC), Créteil, France
| | - Laura Pérez-Campos Mayoral
- Research Centre Medicine UNAM-UABJO, Faculty of Medicine, Benito Juárez Autonomous University of Oaxaca, Oaxaca, Mexico
| | | | - Eduardo Pérez-Campos
- Biochemistry and Immunology Unit, National Technological of Mexico/ITOaxaca, Oaxaca, Mexico
- Research Centre Medicine UNAM-UABJO, Faculty of Medicine, Benito Juárez Autonomous University of Oaxaca, Oaxaca, Mexico
| | - Gabriel Mayoral Andrade
- Research Centre Medicine UNAM-UABJO, Faculty of Medicine, Benito Juárez Autonomous University of Oaxaca, Oaxaca, Mexico
| | | | - Marco V. José
- Theoretical Biology Group, National Autonomous University of Mexico, Mexico City, Mexico
| |
Collapse
|
39
|
Okamato Y, Ghosh T, Okamoto T, Schuyler RP, Seifert J, Charry LL, Visser A, Feser M, Fleischer C, Pedrick C, August J, Moss L, Bemis EA, Norris JM, Kuhn KA, Demoruelle MK, Deane KD, Ghosh D, Holers VM, Hsieh EWY. Subjects at-risk for future development of rheumatoid arthritis demonstrate a PAD4-and TLR-dependent enhanced histone H3 citrullination and proinflammatory cytokine production in CD14 hi monocytes. J Autoimmun 2021; 117:102581. [PMID: 33310262 PMCID: PMC7855988 DOI: 10.1016/j.jaut.2020.102581] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2020] [Revised: 11/20/2020] [Accepted: 11/23/2020] [Indexed: 12/13/2022]
Abstract
The presence of anti-citrullinated protein/peptide antibodies (ACPA) and epitope spreading across the target autoantigens is a unique feature of rheumatoid arthritis (RA). ACPA are present in the peripheral blood for several years prior to the onset of arthritis and clinical classification of RA. ACPA recognize multiple citrullinated proteins, including histone H3 (H3). Intracellular citrullination of H3 in neutrophils and T cells is known to regulate immune cell function by promoting neutrophil extracellular trap formation and citrullinated autoantigen release as well as regulating the Th2/Th17 T cell phenotypic balance. However, the roles of H3 citrullination in other immune cells are not fully elucidated. We aimed to explore H3 citrullination and cytokine/metabolomic signatures in peripheral blood immune cells from subjects prior to and after the onset of RA, at baseline and in response to ex vivo toll-like receptor (TLR) stimulation. Here, we analyzed 13 ACPA (+) subjects without arthritis but at-risk for future development of RA, 14 early RA patients, and 13 healthy controls. We found significantly elevated H3 citrullination in CD14hi monocytes, as well as CD1c+ dendritic cells and CD66+ granulocytes. Unsupervised analysis identified two distinct subsets in CD14hi monocytes characterized by H3 modification and unique cytokine/metabolomic signatures. CD14hi monocytes with elevated TLR-stimulated H3 citrullination were significantly increased in ACPA (+) at-risk subjects. These cells were skewed to produce TNFα, MIP1β, IFNα, and partially IL-12. Additionally, they demonstrate peptidyl arginine deiminase 4 (PAD4) mediated upregulation of the glycolytic enzyme PFKFB3. These CD14hi monocytes with elevated H3 citrullination morphologically formed monocyte extracellular traps (METs). Taken together, dysregulated PAD4-driven cytokine production as well as MET formation in CD14hi monocytes in ACPA (+) at-risk subjects likely plays an important role in the development of RA via promoting and perpetuating inflammation and generation of citrullinated autoantigens.
Collapse
Affiliation(s)
- Yuko Okamato
- University of Colorado Denver, Division of Rheumatology, Aurora, CO, USA; Tokyo Women's Medical University School of Medicine, Department of Rheumatology, Tokyo, Japan.
| | - Tusharkanti Ghosh
- Colorado School of Public Health, Department of Biostatistics and Informatics, Aurora, CO, USA
| | - Tsukasa Okamoto
- University of Colorado Denver, Department of Medicine, Aurora, CO, USA
| | - Ronald P Schuyler
- University of Colorado School of Medicine, Department of Immunology and Microbiology, Aurora, CO, USA
| | - Jennifer Seifert
- University of Colorado Denver, Division of Rheumatology, Aurora, CO, USA
| | - Laura Lenis Charry
- University of Colorado Denver, Division of Rheumatology, Aurora, CO, USA
| | - Ashley Visser
- University of Colorado Denver, Division of Rheumatology, Aurora, CO, USA
| | - Marie Feser
- University of Colorado Denver, Division of Rheumatology, Aurora, CO, USA
| | - Chelsie Fleischer
- University of Colorado Denver, Division of Rheumatology, Aurora, CO, USA
| | - Chong Pedrick
- University of Colorado Denver, Division of Rheumatology, Aurora, CO, USA
| | - Justin August
- University of Colorado Denver, Division of Rheumatology, Aurora, CO, USA
| | - Laurakay Moss
- University of Colorado Denver, Division of Rheumatology, Aurora, CO, USA
| | - Elizabeth A Bemis
- Colorado School of Public Health, Department of Epidemiology, Aurora, CO, USA
| | - Jill M Norris
- Colorado School of Public Health, Department of Epidemiology, Aurora, CO, USA
| | - Kristine A Kuhn
- University of Colorado Denver, Division of Rheumatology, Aurora, CO, USA
| | | | - Kevin D Deane
- University of Colorado Denver, Division of Rheumatology, Aurora, CO, USA
| | - Debashis Ghosh
- Colorado School of Public Health, Department of Biostatistics and Informatics, Aurora, CO, USA
| | - V Michael Holers
- University of Colorado Denver, Division of Rheumatology, Aurora, CO, USA
| | - Elena W Y Hsieh
- University of Colorado School of Medicine, Department of Immunology and Microbiology, Aurora, CO, USA; University of Colorado School of Medicine, Children's Hospital Colorado, Department of Pediatrics, Section of Allergy & Immunology, Aurora, CO, USA
| |
Collapse
|
40
|
Peng C, Li Z, Yu X. The Role of Pancreatic Infiltrating Innate Immune Cells in Acute Pancreatitis. Int J Med Sci 2021; 18:534-545. [PMID: 33390823 PMCID: PMC7757151 DOI: 10.7150/ijms.51618] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/06/2020] [Accepted: 11/13/2020] [Indexed: 12/13/2022] Open
Abstract
Acute pancreatitis (AP) is a leading cause of gastrointestinal-related hospital admissions with significant morbidity and mortality. Although the underlying pathophysiology of AP is rather complex, which greatly limits the treatment options, more and more studies have revealed that infiltrating immune cells play a critical role in the pathogenesis of AP and determine disease severity. Thus, immunomodulatory therapy targeting immune cells and related inflammatory mediators is expected to be a novel treatment modality for AP which may improve the prognosis of patients. Cells of the innate immune system, including macrophages, neutrophils, dendritic cells, and mast cells, represent the majority of infiltrating cells during AP. In this review, an overview of different populations of innate immune cells and their role during AP will be discussed, with a special focus on neutrophils and macrophages.
Collapse
Affiliation(s)
- Cheng Peng
- Department of Hepatopancreatobiliary Surgery, Third Xiangya Hospital, Central South University, Changsha 410013, Hunan, China
| | - Zhiqiang Li
- Department of Hepatopancreatobiliary Surgery, Third Xiangya Hospital, Central South University, Changsha 410013, Hunan, China
| | - Xiao Yu
- Department of Hepatopancreatobiliary Surgery, Third Xiangya Hospital, Central South University, Changsha 410013, Hunan, China
| |
Collapse
|
41
|
Inhibition of Peptidyl Arginine Deiminase-4 Prevents Renal Ischemia-Reperfusion-Induced Remote Lung Injury. Mediators Inflamm 2020; 2020:1724206. [PMID: 33456369 PMCID: PMC7787741 DOI: 10.1155/2020/1724206] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2020] [Revised: 11/09/2020] [Accepted: 12/10/2020] [Indexed: 12/13/2022] Open
Abstract
Ischemia reperfusion (IR) can lead to acute kidney injury and can be complicated by acute lung injury, which is one of the leading causes of acute kidney injury-related death. Peptidyl arginine deiminase-4 (PAD4) is a member of the PAD enzyme family and plays a critical role in inflammatory reactions and neutrophil extracellular trap formation in a variety of pathological conditions. It has been reported that PAD4 inhibition can protect certain organs from ischemic injury. In this study, we aimed to understand the mode of action of PAD4 in renal ischemia-reperfusion-mediated acute lung injury. Bilateral renal pedicle occlusion was induced for 30 min followed by reperfusion for 24 h. A specific inhibitor of PAD4, GSK484, was delivered via intraperitoneal injection to alter the PAD4 activity. The pulmonary PAD4 expression, pulmonary impairment, neutrophil infiltration, Cit-H3 expression, neutrophil extracellular trap formation, inflammatory cytokine secretion, and pulmonary apoptosis were analyzed. We found that renal ischemia reperfusion was associated with pulmonary pathological changes and increases in neutrophil infiltration, neutrophil extracellular trap formation, and inflammatory cytokine secretion in the lungs of the recipient animals. Suppression of PAD4 by GSK484 reduced remote lung injury by mitigating neutrophil infiltration, neutrophil extracellular trap formation, apoptosis, and inflammatory factor secretion. Our findings demonstrate that specific PAD4 inhibition by GSK484 may be an effective strategy to attenuate distant lung injury complicating renal ischemia-reperfusion injury.
Collapse
|
42
|
Tian Y, Russo RM, Li Y, Karmakar M, Liu B, Puskarich MA, Jones AE, Stringer KA, Standiford TJ, Alam HB. Serum citrullinated histone H3 concentrations differentiate patients with septic verses non-septic shock and correlate with disease severity. Infection 2020; 49:83-93. [PMID: 33000445 PMCID: PMC7527151 DOI: 10.1007/s15010-020-01528-y] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2020] [Accepted: 09/15/2020] [Indexed: 12/18/2022]
Abstract
Purpose Microbial infection stimulates neutrophil/macrophage/monocyte extracellular trap formation, which leads to the release of citrullinated histone H3 (CitH3) catalyzed by peptidylarginine deiminase (PAD) 2 and 4. Understanding these molecular mechanisms in the pathogenesis of septic shock will be an important next step for developing novel diagnostic and treatment modalities. We sought to determine the expression of CitH3 in patients with septic shock, and to correlate CitH3 levels with PAD2/PAD4 and clinically relevant outcomes. Methods Levels of CitH3 were measured in serum samples of 160 critically ill patients with septic and non-septic shock, and healthy volunteers. Analyses of clinical and laboratory characteristics of patients were conducted. Results Levels of circulating CitH3 at enrollment were significantly increased in septic shock patients (n = 102) compared to patients hospitalized with non-infectious shock (NIC) (n = 32, p < 0.0001). The area under the curve (95% CI) for distinguishing septic shock from NIC using CitH3 was 0.76 (0.65–0.86). CitH3 was positively correlated with PAD2 and PAD4 concentrations and Sequential Organ Failure Assessment Scores [total score (r = 0.36, p < 0.0001)]. The serum levels of CitH3 at 24 h (p < 0.01) and 48 h (p < 0.05) were significantly higher in the septic patients that did not survive. Conclusion CitH3 is increased in patients with septic shock. Its serum concentrations correlate with disease severity and prognosis, which may yield vital insights into the pathophysiology of sepsis. Electronic supplementary material The online version of this article (10.1007/s15010-020-01528-y) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Yuzi Tian
- Department of Surgery, University of Michigan Health System, University of Michigan Medical School, 1500 E Medical Center Dr. SPC 5331, Ann Arbor, MI, 48109-5331, USA.,Department of Rheumatology and Immunology, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Rachel M Russo
- Department of Surgery, University of Michigan Health System, University of Michigan Medical School, 1500 E Medical Center Dr. SPC 5331, Ann Arbor, MI, 48109-5331, USA
| | - Yongqing Li
- Department of Surgery, University of Michigan Health System, University of Michigan Medical School, 1500 E Medical Center Dr. SPC 5331, Ann Arbor, MI, 48109-5331, USA.
| | - Monita Karmakar
- Department of Surgery, University of Michigan Health System, University of Michigan Medical School, 1500 E Medical Center Dr. SPC 5331, Ann Arbor, MI, 48109-5331, USA
| | - Baoling Liu
- Department of Surgery, University of Michigan Health System, University of Michigan Medical School, 1500 E Medical Center Dr. SPC 5331, Ann Arbor, MI, 48109-5331, USA
| | - Michael A Puskarich
- Department of Emergency Medicine, Hennepin County Medical Center, Minneapolis, MN, USA.,Department of Emergency Medicine, University of Minnesota, Minneapolis, MN, USA
| | - Alan E Jones
- Department of Emergency Medicine, University of Mississippi Medical Center, Jackson, MS, USA
| | - Kathleen A Stringer
- Department of Clinical Pharmacy, College of Pharmacy, University of Michigan, Ann Arbor, MI, USA.,Division of Pulmonary and Critical Care Medicine, Department of Internal Medicine, University of Michigan School of Medicine, Ann Arbor, MI, USA
| | - Theodore J Standiford
- Division of Pulmonary and Critical Care Medicine, Department of Internal Medicine, University of Michigan School of Medicine, Ann Arbor, MI, USA
| | - Hasan B Alam
- Department of Surgery, University of Michigan Health System, University of Michigan Medical School, 1500 E Medical Center Dr. SPC 5331, Ann Arbor, MI, 48109-5331, USA.
| |
Collapse
|
43
|
Stackowicz J, Jönsson F, Reber LL. Mouse Models and Tools for the in vivo Study of Neutrophils. Front Immunol 2020; 10:3130. [PMID: 32038641 PMCID: PMC6985372 DOI: 10.3389/fimmu.2019.03130] [Citation(s) in RCA: 55] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2019] [Accepted: 12/23/2019] [Indexed: 12/21/2022] Open
Abstract
Neutrophils are the most abundant leukocytes in human blood and critical actors of the immune system. Many neutrophil functions and facets of their activity in vivo were revealed by studying genetically modified mice or by tracking fluorescent neutrophils in animals using imaging approaches. Assessing the roles of neutrophils can be challenging, especially when exact molecular pathways are questioned or disease states are interrogated that alter normal neutrophil homeostasis. This review discusses the main in vivo models for the study of neutrophils, their advantages and limitations. The side-by-side comparison underlines the necessity to carefully choose the right model(s) to answer a given scientific question, and exhibit caveats that need to be taken into account when designing experimental procedures. Collectively, this review suggests that at least two models should be employed to legitimately conclude on neutrophil functions.
Collapse
Affiliation(s)
- Julien Stackowicz
- Institut Pasteur, Department of Immunology, Unit of Antibodies in Therapy and Pathology, UMR INSERM 1222, Paris, France.,Sorbonne Université, Collège Doctoral, Paris, France
| | - Friederike Jönsson
- Institut Pasteur, Department of Immunology, Unit of Antibodies in Therapy and Pathology, UMR INSERM 1222, Paris, France
| | - Laurent L Reber
- Institut Pasteur, Department of Immunology, Unit of Antibodies in Therapy and Pathology, UMR INSERM 1222, Paris, France.,Center for Pathophysiology Toulouse-Purpan (CPTP), UMR 1043, University of Toulouse, INSERM, CNRS, Toulouse, France
| |
Collapse
|
44
|
Deng Q, Pan B, Alam HB, Liang Y, Wu Z, Liu B, Mor-Vaknin N, Duan X, Williams AM, Tian Y, Zhang J, Li Y. Citrullinated Histone H3 as a Therapeutic Target for Endotoxic Shock in Mice. Front Immunol 2020; 10:2957. [PMID: 31998291 PMCID: PMC6962130 DOI: 10.3389/fimmu.2019.02957] [Citation(s) in RCA: 67] [Impact Index Per Article: 13.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2019] [Accepted: 12/02/2019] [Indexed: 12/16/2022] Open
Abstract
Sepsis results in millions of deaths every year, with acute lung injury (ALI) being one of the leading causes of mortality in septic patients. As neutrophil extracellular traps (NETs) are abundant in sepsis, neutralizing components of NETs may be a useful strategy to improve outcomes of sepsis. Citrullinated histone H3 (CitH3) has been recently shown to be involved in the NET formation. In this study, we demonstrate that CitH3 damages human umbilical vein endothelial cells (HUVECs) and potentiates NET formation through a positive feedback mechanism. We developed a novel CitH3 monoclonal antibody to target peptidylarginine deiminase (PAD) 2 and PAD 4 generated CitH3. In a mouse model of lethal lipopolysaccharide (LPS) induced shock, neutralizing CitH3 with the newly developed anti-CitH3 monoclonal antibody attenuates inflammatory responses, ameliorates ALI, and improves survival. Our study suggests that effectively blocking circulating CitH3 might be a potential therapeutic method for the treatment of endotoxemia.
Collapse
Affiliation(s)
- Qiufang Deng
- Xiangya Hospital, Central South University, Changsha, China.,Department of Surgery, University of Michigan, Ann Arbor, MI, United States
| | - Baihong Pan
- Xiangya Hospital, Central South University, Changsha, China.,Department of Surgery, University of Michigan, Ann Arbor, MI, United States
| | - Hasan B Alam
- Department of Surgery, University of Michigan, Ann Arbor, MI, United States
| | - Yingjian Liang
- Department of Surgery, University of Michigan, Ann Arbor, MI, United States.,The First Hospital, China Medical University, Shenyang, China
| | - Zhenyu Wu
- Xiangya Hospital, Central South University, Changsha, China.,Department of Surgery, University of Michigan, Ann Arbor, MI, United States
| | - Baoling Liu
- Department of Surgery, University of Michigan, Ann Arbor, MI, United States
| | - Nirit Mor-Vaknin
- Division of Infectious Diseases, Department of Internal Medicine, University of Michigan, Ann Arbor, MI, United States
| | - Xiuzhen Duan
- Department of Pathology, Loyola University Medical Center, Maywood, IL, United States
| | - Aaron M Williams
- Department of Surgery, University of Michigan, Ann Arbor, MI, United States
| | - Yuzi Tian
- Xiangya Hospital, Central South University, Changsha, China.,Department of Surgery, University of Michigan, Ann Arbor, MI, United States
| | - Justin Zhang
- Department of Surgery, University of Michigan, Ann Arbor, MI, United States
| | - Yongqing Li
- Department of Surgery, University of Michigan, Ann Arbor, MI, United States
| |
Collapse
|
45
|
Zhu H, Fu J, Chen S, Li X, Liang H, Hou Y, Dou H. FC-99 reduces macrophage tenascin-C expression by upregulating miRNA-494 in arthritis. Int Immunopharmacol 2019; 79:106105. [PMID: 31881378 DOI: 10.1016/j.intimp.2019.106105] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2019] [Revised: 11/16/2019] [Accepted: 11/29/2019] [Indexed: 01/29/2023]
Abstract
The excessive production of inflammatory mediators by inflammatory cells contributes to the pathogenesis of rheumatoid arthritis. Tenascin-C (TN-C) is expressed in rheumatoid joint, and is associated with levels of inflammatory mediators. FC-99 (N1-[(4-methoxy)methyl]-4-methyl-1,2-Benzenediamine), a novel 1,2-benzenediamine derivative, was previously reported to block the prolonged expression of key rheumatoid arthritis inflammatory cytokines and relieve zymosan-induced joint inflammation. However, the specific mechanism is unknown. This study aimed to examine the effects of FC-99 on TN-C expression and inflammation and investigate its possible molecular mechanism. The results showed that FC-99 treatment reduced the high expression of TN-C in ankle joints of arthritis mice. Besides, FC-99 reduced the increased number of macrophages in arthritis mice, while did not change the number of synovioblasts. Concomitantly, expression of TN-C in synovial fibroblasts exhibited no difference between control and ZIA groups, and was not apparently altered following FC-99 treatment, while FC-99 decreased TN-C expression in macrophages both in vivo and in vitro. Meanwhile, TargetScan and luciferase assays indicated that TN-C was negatively regulated by miR-494. Transfection assay further demonstrated that FC-99 inhibited TN-C by targeting miR-494. Furthermore, the reduction of miR-494 mimic on expression of TN-C was associated with NF-κB pathway. Similarly, the down-regulation of FC-99 on TN-C was considerably decreased when NF-κB pathway was inhibited. These results indicated that FC-99 relieved macrophages inflammation via the miR-494/TN-C/NF-κB pathway, finally leading to the relief of inflammation in arthritis. The findings suggested that FC-99 might be a potential therapeutic candidate for the treatment of rheumatoid arthritis.
Collapse
Affiliation(s)
- Haiyan Zhu
- The State Key Laboratory of Pharmaceutical Biotechnology, Division of Immunology, Medical School, Nanjing University, Nanjing 210093, China
| | - Juanhua Fu
- The State Key Laboratory of Pharmaceutical Biotechnology, Division of Immunology, Medical School, Nanjing University, Nanjing 210093, China
| | - Sheng Chen
- Nangjing Stomatological Hospital, Medical School of Nanjing University, Nanjing 210008, China
| | - Xiaoqin Li
- The State Key Laboratory of Pharmaceutical Biotechnology, Division of Immunology, Medical School, Nanjing University, Nanjing 210093, China
| | - Huaping Liang
- State Key Laboratory of Trauma, Burns and Combined Injury, Research Institute of Surgery, Daping Hospital, The Army Medical University, Chongqing 400042, China.
| | - Yayi Hou
- The State Key Laboratory of Pharmaceutical Biotechnology, Division of Immunology, Medical School, Nanjing University, Nanjing 210093, China; Department of Rheumatology and Immunology, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing 210008, China; Jiangsu Key Laboratory of Molecular Medicine, Nanjing University, Nanjing 210093, China.
| | - Huan Dou
- The State Key Laboratory of Pharmaceutical Biotechnology, Division of Immunology, Medical School, Nanjing University, Nanjing 210093, China; Department of Rheumatology and Immunology, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing 210008, China; Jiangsu Key Laboratory of Molecular Medicine, Nanjing University, Nanjing 210093, China.
| |
Collapse
|