151
|
Yang S, Zhao M, Jia S. Macrophage: Key player in the pathogenesis of autoimmune diseases. Front Immunol 2023; 14:1080310. [PMID: 36865559 PMCID: PMC9974150 DOI: 10.3389/fimmu.2023.1080310] [Citation(s) in RCA: 54] [Impact Index Per Article: 27.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2022] [Accepted: 01/09/2023] [Indexed: 02/16/2023] Open
Abstract
The macrophage is an essential part of the innate immune system and also serves as the bridge between innate immunity and adaptive immune response. As the initiator and executor of the adaptive immune response, macrophage plays an important role in various physiological processes such as immune tolerance, fibrosis, inflammatory response, angiogenesis and phagocytosis of apoptotic cells. Consequently, macrophage dysfunction is a vital cause of the occurrence and development of autoimmune diseases. In this review, we mainly discuss the functions of macrophages in autoimmune diseases, especially in systemic lupus erythematosus (SLE), rheumatic arthritis (RA), systemic sclerosis (SSc) and type 1 diabetes (T1D), providing references for the treatment and prevention of autoimmune diseases.
Collapse
Affiliation(s)
- Shuang Yang
- Dapartment of Dermatology, Hunan Key Laboratory of Medical Epigenomics, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Ming Zhao
- Dapartment of Dermatology, Hunan Key Laboratory of Medical Epigenomics, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China.,Institute of Dermatology, Chinese Academy of Medical Sciences and Peking Union Medical College, Nanjing, China.,Key Laboratory of Basic and Translational Research on Immune-Mediated Skin Diseases, Chinese Academy of Medical Sciences, Nanjing, China
| | - Sujie Jia
- Department of Pharmacy, Chinese Academy of Medical Sciences and Peking Union Medical College, Nanjing, China
| |
Collapse
|
152
|
Gremese E, Tolusso B, Bruno D, Perniola S, Ferraccioli G, Alivernini S. The forgotten key players in rheumatoid arthritis: IL-8 and IL-17 - Unmet needs and therapeutic perspectives. Front Med (Lausanne) 2023; 10:956127. [PMID: 37035302 PMCID: PMC10073515 DOI: 10.3389/fmed.2023.956127] [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: 05/29/2022] [Accepted: 02/21/2023] [Indexed: 04/11/2023] Open
Abstract
Despite the relevant advances in our understanding of the pathogenetic mechanisms regulating inflammation in rheumatoid arthritis (RA) and the development of effective therapeutics, to date, there is still a proportion of patients with RA who do not respond to treatment and end up progressing toward the development of joint damage, extra-articular complications, and disability. This is mainly due to the inter-individual heterogeneity of the molecular and cellular taxonomy of the synovial membrane, which represents the target tissue of RA inflammation. Tumor necrosis factor alpha (TNFα) and interleukin-6 (IL-6) are crucial key players in RA pathogenesis fueling the inflammatory cascade, as supported by experimental evidence derived from in vivo animal models and the effectiveness of biologic-Disease Modifying Anti-Rheumatic Drugs (b-DMARDs) in patients with RA. However, additional inflammatory soluble mediators such as IL-8 and IL-17 exert their pathogenetic actions promoting the detrimental activation of immune and stromal cells in RA synovial membrane, tendons, and extra-articular sites, as well as blood vessels and lungs, causing extra-articular complications, which might be excluded by the action of anti-TNFα and anti-IL6R targeted therapies. In this narrative review, we will discuss the role of IL-8 and IL-17 in promoting inflammation in multiple biological compartments (i.e., synovial membrane, blood vessels, and lung, respectively) in animal models of arthritis and patients with RA and how their selective targeting could improve the management of treatment resistance in patients.
Collapse
Affiliation(s)
- Elisa Gremese
- Division of Clinical Immunology, Fondazione Policlinico Universitario A. Gemelli-IRCCS, Rome, Italy
- Immunology Core Facility, Fondazione Policlinico Universitario A. Gemelli-IRCCS, Rome, Italy
- School of Medicine, Università Cattolica del Sacro Cuore, Rome, Italy
- *Correspondence: Elisa Gremese, Gianfranco Ferraccioli
| | - Barbara Tolusso
- Division of Clinical Immunology, Fondazione Policlinico Universitario A. Gemelli-IRCCS, Rome, Italy
- Immunology Core Facility, Fondazione Policlinico Universitario A. Gemelli-IRCCS, Rome, Italy
| | - Dario Bruno
- Division of Clinical Immunology, Fondazione Policlinico Universitario A. Gemelli-IRCCS, Rome, Italy
- Department of Medicine, University of Verona, Verona, Italy
| | - Simone Perniola
- Division of Clinical Immunology, Fondazione Policlinico Universitario A. Gemelli-IRCCS, Rome, Italy
| | - Gianfranco Ferraccioli
- School of Medicine, Università Cattolica del Sacro Cuore, Rome, Italy
- *Correspondence: Elisa Gremese, Gianfranco Ferraccioli
| | - Stefano Alivernini
- Immunology Core Facility, Fondazione Policlinico Universitario A. Gemelli-IRCCS, Rome, Italy
- School of Medicine, Università Cattolica del Sacro Cuore, Rome, Italy
- Division of Rheumatology, Fondazione Policlinico Universitario A. Gemelli-IRCCS, Rome, Italy
| |
Collapse
|
153
|
Hu Y, Liu C, Han W, Wang P. A theoretical framework of immune cell phenotypic classification and discovery. Front Immunol 2023; 14:1128423. [PMID: 36936975 PMCID: PMC10018129 DOI: 10.3389/fimmu.2023.1128423] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2022] [Accepted: 02/20/2023] [Indexed: 03/06/2023] Open
Abstract
Immune cells are highly heterogeneous and show diverse phenotypes, but the underlying mechanism remains to be elucidated. In this study, we proposed a theoretical framework for immune cell phenotypic classification based on gene plasticity, which herein refers to expressional change or variability in response to conditions. The system contains two core points. One is that the functional subsets of immune cells can be further divided into subdivisions based on their highly plastic genes, and the other is that loss of phenotype accompanies gain of phenotype during phenotypic conversion. The first point suggests phenotypic stratification or layerability according to gene plasticity, while the second point reveals expressional compatibility and mutual exclusion during the change in gene plasticity states. Abundant transcriptome data analysis in this study from both microarray and RNA sequencing in human CD4 and CD8 single-positive T cells, B cells, natural killer cells and monocytes supports the logical rationality and generality, as well as expansibility, across immune cells. A collection of thousands of known immunophenotypes reported in the literature further supports that highly plastic genes play an important role in maintaining immune cell phenotypes and reveals that the current classification model is compatible with the traditionally defined functional subsets. The system provides a new perspective to understand the characteristics of dynamic, diversified immune cell phenotypes and intrinsic regulation in the immune system. Moreover, the current substantial results based on plasticitomics analysis of bulk and single-cell sequencing data provide a useful resource for big-data-driven experimental studies and knowledge discoveries.
Collapse
Affiliation(s)
- Yuzhe Hu
- Department of Immunology, NHC Key Laboratory of Medical Immunology (Peking University), School of Basic Medical Sciences, Peking University Health Science Center, Beijing, China
- Peking University Center for Human Disease Genomics, Beijing, China
| | - Chen Liu
- Department of Clinical Laboratory, Peking University People’s Hospital, Beijing, China
| | - Wenling Han
- Department of Immunology, NHC Key Laboratory of Medical Immunology (Peking University), School of Basic Medical Sciences, Peking University Health Science Center, Beijing, China
- Peking University Center for Human Disease Genomics, Beijing, China
| | - Pingzhang Wang
- Department of Immunology, NHC Key Laboratory of Medical Immunology (Peking University), School of Basic Medical Sciences, Peking University Health Science Center, Beijing, China
- Peking University Center for Human Disease Genomics, Beijing, China
- *Correspondence: Pingzhang Wang,
| |
Collapse
|
154
|
Zamith Cunha R, Zannoni A, Salamanca G, De Silva M, Rinnovati R, Gramenzi A, Forni M, Chiocchetti R. Expression of cannabinoid (CB1 and CB2) and cannabinoid-related receptors (TRPV1, GPR55, and PPARα) in the synovial membrane of the horse metacarpophalangeal joint. Front Vet Sci 2023; 10:1045030. [PMID: 36937015 PMCID: PMC10020506 DOI: 10.3389/fvets.2023.1045030] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2022] [Accepted: 01/31/2023] [Indexed: 03/06/2023] Open
Abstract
Background The metacarpophalangeal joint undergoes enormous loading during locomotion and can therefore often become inflamed, potentially resulting in osteoarthritis (OA). There are studies indicating that the endocannabinoid system (ECS) modulates synovium homeostasis, and could be a promising target for OA therapy. Some cannabinoid receptors, which modulate proliferative and secretory responses in joint inflammation, have been functionally identified in human and animal synovial cells. Objective To characterize the cellular distribution of the cannabinoid receptors 1 (CB1R) and 2 (CB2R), and the cannabinoid-related receptors transient receptor potential vanilloid type 1 (TRPV1), G protein-related receptor 55 (GPR55) and peroxisome proliferator-activated receptor alpha (PPARα) in the synovial membrane of the metacarpophalangeal joint of the horse. Animals The dorsal synovial membranes of 14 equine metacarpophalangeal joints were collected post-mortem from an abattoir. Materials and methods The dorsal synovial membranes of 14 equine metacarpophalangeal joints were collected post-mortem from an abattoir. The expression of the CB1R, CB2R, TRPV1, GPR55, and PPARα in synovial tissues was studied using qualitative and quantitative immunofluorescence, and quantitative real-time reverse transcriptase PCR (qRT-PCR). Macrophage-like (MLS) and fibroblast-like (FLS) synoviocytes were identified by means of antibodies directed against IBA1 and vimentin, respectively. Results Both the mRNA and protein expression of the CB2R, TRPV1, GPR55, and PPARα were found in the synoviocytes and blood vessels of the metacarpophalangeal joints. The synoviocytes expressed the mRNA and protein of the CB1R in some of the horses investigated, but not in all. Conclusions and clinical importance Given the expression of the CB1R, CB2R, TRPV1, GPR55, and PPARα in the synovial elements of the metacarpophalangeal joint, these findings encouraged the development of new studies supporting the use of molecules acting on these receptors to reduce the inflammation during joint inflammation in the horse.
Collapse
Affiliation(s)
- Rodrigo Zamith Cunha
- Department of Veterinary Medical Sciences (UNI EN ISO 9001:2008), University of Bologna, Bologna, Italy
| | - Augusta Zannoni
- Department of Veterinary Medical Sciences (UNI EN ISO 9001:2008), University of Bologna, Bologna, Italy
| | - Giulia Salamanca
- Department of Veterinary Medical Sciences (UNI EN ISO 9001:2008), University of Bologna, Bologna, Italy
| | - Margherita De Silva
- Department of Veterinary Medical Sciences (UNI EN ISO 9001:2008), University of Bologna, Bologna, Italy
| | - Riccardo Rinnovati
- Department of Veterinary Medical Sciences (UNI EN ISO 9001:2008), University of Bologna, Bologna, Italy
| | - Alessandro Gramenzi
- Faculty of Veterinary Medicine, Università degli Studi di Teramo, Teramo, Italy
| | - Monica Forni
- Department of Veterinary Medical Sciences (UNI EN ISO 9001:2008), University of Bologna, Bologna, Italy
| | - Roberto Chiocchetti
- Department of Veterinary Medical Sciences (UNI EN ISO 9001:2008), University of Bologna, Bologna, Italy
- *Correspondence: Roberto Chiocchetti
| |
Collapse
|
155
|
Sevin S, Kivrak İ, Tutun H, Uyar R, Ayaz F. Apis mellifera anatoliaca Venom Exerted Anti-Inflammatory Activity on LPS-Stimulated Mammalian Macrophages by Reducing the Production of the Inflammatory Cytokines. Appl Biochem Biotechnol 2022; 195:3194-3205. [PMID: 36574137 DOI: 10.1007/s12010-022-04284-x] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/16/2022] [Indexed: 12/28/2022]
Abstract
Extraction and characterization of natural products provide the opportunity to expand our arsenal of drug candidates against a wide range of diseases including cancer and inflammatory disorders. Previous studies have shown bee venom to have immense potential as an anti-inflammatory drug candidate. In this study, we focused on the venom of Apis mellifera anatoliaca and characterized its content by HPLC. An in vitro inflammation model based on lipopolysaccharide (LPS)-stimulated mammalian macrophages was utilized to examine the venom's anti-inflammatory potential. Additionally, its antiproliferative activity was evaluated in vitro against a human glioblastoma cell line. Based on the TNF, IL6, GMCSF, and IL12p40 pro-inflammatory cytokine production level in LPS-induced macrophages, venom-treated groups showed substantial decrease in the inflammatory action compared to untreated LPS-stimulated macrophages. When the cells were analyzed for viability, the venom did not have any cytotoxic effect on the macrophages at the concentration ranges that were utilized. Moreover, IC50 value of the venom was above 60 µg/mL on glioblastoma cancer cell line. These results suggest that the Apis mellifera anatoliaca venom does not have anticancer drug candidate potential, whereas it can efficiently be used against inflammatory and autoimmune disorders. To our knowledge, this is the first study to specifically examine the effect of anti-inflammatory activity of Apis mellifera anatoliaca venom on macrophages.
Collapse
Affiliation(s)
- Sedat Sevin
- Faculty of Veterinary Medicine, Department of Pharmacology and Toxicology, Ankara University, Ankara, Turkey
| | - İbrahim Kivrak
- Muğla Vocational School Department of Chemistry and Chemical Treatment Technologies, MuğlaSıtkıKoçman University, Muğla, Turkey
| | - Hidayet Tutun
- Faculty of Veterinary Medicine, Department of Pharmacology and Toxicology, Burdur Mehmet AkifErsoy University, Burdur, Turkey
| | - Recep Uyar
- Faculty of Veterinary Medicine, Department of Pharmacology and Toxicology, Ankara University, Ankara, Turkey
| | - Furkan Ayaz
- Department of Biotechnology, Faculty of Arts and Science, Mersin University, Mersin, Turkey.
- Mersin University Biotechnology Research and Application Center, Mersin University, Mersin, Turkey.
| |
Collapse
|
156
|
Bruno MC, Cristiano MC, Celia C, d'Avanzo N, Mancuso A, Paolino D, Wolfram J, Fresta M. Injectable Drug Delivery Systems for Osteoarthritis and Rheumatoid Arthritis. ACS NANO 2022; 16:19665-19690. [PMID: 36512378 DOI: 10.1021/acsnano.2c06393] [Citation(s) in RCA: 67] [Impact Index Per Article: 22.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/17/2023]
Abstract
Joint diseases are one of the most common causes of morbidity and disability worldwide. The main diseases that affect joint cartilage are osteoarthritis and rheumatoid arthritis, which require chronic treatment focused on symptomatic relief. Conventional drugs administered through systemic or intra-articular routes have low accumulation and/or retention in articular cartilage, causing dose-limiting toxicities and reduced efficacy. Therefore, there is an urgent need to develop improved strategies for drug delivery, in particular, the use of micro- and nanotechnology-based methods. Encapsulation of therapeutic agents in delivery systems reduces drug efflux from the joint and protects against rapid cellular and enzymatic clearance following intra-articular injection. Consequently, the use of drug delivery systems decreases side effects and increases therapeutic efficacy due to enhanced drug retention in the intra-articular space. Additionally, the frequency of intra-articular administration is reduced, as delivery systems enable sustained drug release. This review summarizes various advanced drug delivery systems, such as nano- and microcarriers, developed for articular cartilage diseases.
Collapse
Affiliation(s)
- Maria Chiara Bruno
- Department of Health Sciences, School of Pharmacy and Nutraceuticals, University "Magna Græcia" of Catanzaro, Campus Universitario "S. Venuta", Building of BioSciences, Viale S. Venuta, Germaneto-Catanzaro, I-88100, Italy
| | - Maria Chiara Cristiano
- Department of Experimental and Clinical Medicine, School of Pharmacy and Nutraceuticals, University "Magna Græcia" of Catanzaro, Campus Universitario "S. Venuta", Building of BioSciences, Viale S. Venuta, Germaneto-Catanzaro, I-88100, Italy
| | - Christian Celia
- Department of Pharmacy, University of Chieti - Pescara "G. d'Annunzio", Via dei Vestini 31, Chieti, I-66100, Italy
- Laboratory of Drug Targets Histopathology, Institute of Cardiology, Lithuanian University of Health Sciences, A. Mickeviciaus g. 9, LT-44307, Kaunas, Lithuania
| | - Nicola d'Avanzo
- Department of Health Sciences, School of Pharmacy and Nutraceuticals, University "Magna Græcia" of Catanzaro, Campus Universitario "S. Venuta", Building of BioSciences, Viale S. Venuta, Germaneto-Catanzaro, I-88100, Italy
- Department of Pharmacy, University of Chieti - Pescara "G. d'Annunzio", Via dei Vestini 31, Chieti, I-66100, Italy
| | - Antonia Mancuso
- Department of Experimental and Clinical Medicine, School of Pharmacy and Nutraceuticals, University "Magna Græcia" of Catanzaro, Campus Universitario "S. Venuta", Building of BioSciences, Viale S. Venuta, Germaneto-Catanzaro, I-88100, Italy
| | - Donatella Paolino
- Department of Experimental and Clinical Medicine, School of Pharmacy and Nutraceuticals, University "Magna Græcia" of Catanzaro, Campus Universitario "S. Venuta", Building of BioSciences, Viale S. Venuta, Germaneto-Catanzaro, I-88100, Italy
| | - Joy Wolfram
- Australian Institute for Bioengineering and Nanotechnology, The University of Queensland, Brisbane, QLD 4072, Australia
- School of Chemical Engineering, The University of Queensland, Brisbane, QLD 4072, Australia
| | - Massimo Fresta
- Department of Health Sciences, School of Pharmacy and Nutraceuticals, University "Magna Græcia" of Catanzaro, Campus Universitario "S. Venuta", Building of BioSciences, Viale S. Venuta, Germaneto-Catanzaro, I-88100, Italy
| |
Collapse
|
157
|
Su X, Wang L, Yang R, Guo Z. Longitudinal 18F-VUIIS1008 PET imaging in a rat model of rheumatoid arthritis. Front Chem 2022; 10:1064518. [PMID: 36618864 PMCID: PMC9816387 DOI: 10.3389/fchem.2022.1064518] [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: 10/08/2022] [Accepted: 12/09/2022] [Indexed: 12/24/2022] Open
Abstract
Macrophages have crucial roles in the pathogenesis of rheumatoid arthritis (RA). We aimed to elucidate the temporal profile of macrophage infiltration in synovitis in RA rat models using PET (positron emission tomography) imaging based a new generation of TSPO (Translocator protein, 18 kDa)-PET ligand, 18F-VUIIS1008 {2-[5,7-Diethyl-2-{4-[2-(18F)fluoroethoxy]phenyl}pyrazolo(1,5-a)pyri-midin-3-yl]-N, N-diethylacetamide}. In vitro and in vivo studies were conducted using RAW264.7 macrophage cells and a rat model of RA induced by Complete Freund's Adjuvant (CFA). Our results showed 18F-VUIIS1008 showed excellent stability in vitro and binding specificity to RAW264.7 cells, and rapid accumulation in the left inflammatory ankles. PET studies revealed that 18F-VUIIS1008 could clearly identify the left inflammatory ankles with good contrast at 30-120 min post-injection. The uptake of 18F-VUIIS1008 of left inflammatory ankles was a wiggle trace with two peaks on day 7 and 29, and then, the highest peak uptake was seen on day 29 (3.00% ± 0.08%ID/g) at 60 min after injection. Tracer uptakes could be inhibited by PK11195 or VUIIS1008. Immunohistochemistry and immunofluorescence tests showed that elevated TSPO expression and infiltrated macrophages were found in the left inflammation ankles. 18F-VUIIS1008 as a novel PET imaging agent showed great potential to identify temporal profile of macrophage infiltration in synovitis in RA, and deliver accurate non-invasive diagnosis and real-time monitoring of RA development.
Collapse
Affiliation(s)
- Xinhui Su
- Department of Nuclear Medicine, School of Medicine, The First Affiliated Hospital, Zhejiang University, Hangzhou, China,The School of Clinical Medicine, Fujian Medical University, Fuzhou, China,Department of Nuclear Medicine, Zhongshan Hospital Xiamen University, Xiamen, China,*Correspondence: Xinhui Su,
| | - Liangliang Wang
- The School of Clinical Medicine, Fujian Medical University, Fuzhou, China,Department of Nuclear Medicine, Zhongshan Hospital Xiamen University, Xiamen, China,Department of Nuclear Medicine, Linyi People’s Hospital, Linyi, China
| | - Rongshui Yang
- Department of Nuclear Medicine, Zhongshan Hospital Xiamen University, Xiamen, China
| | - Zhide Guo
- Center for Molecular Imaging and Translational Medicine, Xiamen University, Xiamen, China
| |
Collapse
|
158
|
Yu W, Ilyas I, Hu X, Xu S, Yu H. Therapeutic potential of paeoniflorin in atherosclerosis: A cellular action and mechanism-based perspective. Front Immunol 2022; 13:1072007. [PMID: 36618414 PMCID: PMC9811007 DOI: 10.3389/fimmu.2022.1072007] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2022] [Accepted: 12/07/2022] [Indexed: 12/24/2022] Open
Abstract
Epidemiological studies have shown that the incidence, prevalence and mortality of atherosclerotic cardiovascular disease (ASCVD) are increasing globally. Atherosclerosis is characterized as a chronic inflammatory disease which involves inflammation and immune dysfunction. P. lactiflora Pall. is a plant origin traditional medicine that has been widely used for the treatment of various diseases for more than a millennium in China, Japan and Korean. Paeoniflorin is a bioactive monomer extracted from P. lactiflora Pall. with anti-atherosclerosis effects. In this article, we comprehensively reviewed the potential therapeutic effects and molecular mechanism whereby paeoniflorin protects against atherosclerosis from the unique angle of inflammation and immune-related pathway dysfunction in vascular endothelial cells, smooth muscle cells, monocytes, macrophages, platelets and mast cells. Paeoniflorin, with multiple protective effects in atherosclerosis, has the potential to be used as a promising therapeutic agent for the treatment of atherosclerosis and its complications. We conclude with a detailed discussion of the challenges and future perspective of paeoniflorin in translational cardiovascular medicine.
Collapse
Affiliation(s)
- Wei Yu
- School of Materials Science and Engineering, Hefei University of Technology, Hefei, Anhui, China,Center for Drug Research and Development, Anhui Renovo Pharmaceutical Co., Ltd, Center for Drug Research and Development, Hefei, Anhui, China
| | - Iqra Ilyas
- Department of Endocrinology, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui, China
| | - Xuerui Hu
- School of Pharmacy, Anhui University of Chinese Medicine, Hefei, Anhui, China
| | - Suowen Xu
- Department of Endocrinology, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui, China
| | - Hui Yu
- School of Materials Science and Engineering, Tianjin Key Laboratory of Materials Laminating Fabrication and Interfacial Controlling Technology, Hebei University of Technology, Tianjin, China,*Correspondence: Hui Yu,
| |
Collapse
|
159
|
Wan L, Liu J, Huang C, Zhu Z, Li F, Sun G, Wang K, Li S, Ma X, Chen X, Yuan W. Role of m6A modification and novel circ_0066715/ miR-486-5p/ ETS1 axis in rheumatoid arthritis macrophage polarization progression. Aging (Albany NY) 2022; 14:10009-10026. [PMID: 36541909 PMCID: PMC9831719 DOI: 10.18632/aging.204439] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2022] [Accepted: 12/09/2022] [Indexed: 01/06/2023]
Abstract
Rheumatoid arthritis (RA) is a systemic disease dominated by inflammatory synovitis. RA synovial macrophages tend undergo M1-type macrophage polarization. Then, polarized M1-type macrophages secrete abundant pro-inflammatory cytokines, causing joint and cartilage destruction. N6-methyladenosine (m6A) methylation modification, circular RNA (circRNA), microRNA (miRNA), messenger RNA (mRNA), etc. are involved in the inflammatory response of RA. We found that there is an imbalance of inflammatory polarization in RA, which is manifested by a sharp increase in inflammatory markers and a high inflammatory response. Here, we show that RA was closely associated with low expression of circ_0066715. The overexpression of circ_0066715 significantly increased the ETS1 levels in RA-FLS cells, decreased cytokine secretion by M1-type macrophages, elevated M2-type cytokines, and inhibited FLS proliferation. Interestingly, the overexpression of miR-486-5p significantly suppressed the attenuation of the cell function and the effect on M1 macrophage polarization caused by circ_0066715 positive expression. WTAP may be involved in the methylation process of ETS1 in RA. ETS1 m6A methylation levels were altered upon WTAP intervention. The overexpression or interference of circ_0066715 decreased or increased WTAP expression. Our findings provide a novel circRNA/miRNA/mRNA regulatory axis and m6A regulatory mechanism involved in the process of RA macrophage polarization, thereby providing a powerful diagnostic and therapeutic strategy for RA treatment.
Collapse
Affiliation(s)
- Lei Wan
- The First Affiliated Hospital of Anhui University of Chinese Medicine, Hefei 230038, China,Key Laboratory of Xin’an Medical Education Ministry, Hefei 230038, China
| | - Jian Liu
- The First Affiliated Hospital of Anhui University of Chinese Medicine, Hefei 230038, China,Key Laboratory of Xin’an Medical Education Ministry, Hefei 230038, China
| | - Chuanbing Huang
- The First Affiliated Hospital of Anhui University of Chinese Medicine, Hefei 230038, China
| | - Ziheng Zhu
- The First Affiliated Hospital of Anhui University of Chinese Medicine, Hefei 230038, China
| | - Fangze Li
- The First Affiliated Hospital of Anhui University of Chinese Medicine, Hefei 230038, China
| | - Guanghan Sun
- College of Traditional Chinese Medicine, Anhui University of Chinese Medicine, Hefei 230012, China
| | - Kun Wang
- Key Laboratory of Xin’an Medical Education Ministry, Hefei 230038, China,College of Traditional Chinese Medicine, Anhui University of Chinese Medicine, Hefei 230012, China
| | - Shu Li
- The First Affiliated Hospital of Anhui University of Chinese Medicine, Hefei 230038, China
| | - Ximeng Ma
- The First Affiliated Hospital of Anhui University of Chinese Medicine, Hefei 230038, China
| | - Xi Chen
- The First Affiliated Hospital of Anhui University of Chinese Medicine, Hefei 230038, China
| | - Wang Yuan
- The First Affiliated Hospital of Anhui University of Chinese Medicine, Hefei 230038, China
| |
Collapse
|
160
|
Hu X, Li F, Zeng J, Zhou Z, Wang Z, Chen J, Cao D, Hong Y, Huang L, Chen Y, Xu J, Dong F, Yu R, Zheng H. Noninvasive Low-Frequency Pulsed Focused Ultrasound Therapy for Rheumatoid Arthritis in Mice. RESEARCH (WASHINGTON, D.C.) 2022; 2022:0013. [PMID: 39290964 PMCID: PMC11407525 DOI: 10.34133/research.0013] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/18/2021] [Accepted: 10/31/2022] [Indexed: 10/16/2023]
Abstract
Rheumatoid arthritis (RA) is a common autoimmune disease characterized by chronic and progressive inflammation of the synovium. Focused ultrasound therapy is an increasingly attractive alternative for treating RA owing to its noninvasiveness; however, it remains unclear which immune subsets respond to ultrasound stimulation. In this study, we showed that spleen-targeted low-frequency pulsed focused ultrasound (LFPFU) effectively improved the severity of arthritis in an arthritis mouse model established in DBA/1J mice. Additionally, we performed in-depth immune profiling of spleen samples from RA mice, RA mice that underwent ultrasound therapy, and healthy controls using mass cytometry along with extensive antibody panels and identified the immune composition of 14 cell populations, including CD4+/CD8+ T cells, B cells, natural killer cells, and dendritic cells. Moreover, multidimensional analysis according to cell-surface markers and phenotypes helped in identifying 4 and 5 cell subpopulations among T and myeloid cells, respectively, with 6 T cell subsets and 3 myeloid cell subsets responsive to ultrasound therapy among the 3 groups. Of these cell subsets, CD8+ T cell subsets showed a unique response to ultrasound stimulation in RA mice. Specifically, CD8+ T cells show a noticeable correlation with the degree of arthritis progression and could serve as an indicator for spleen-focused ultrasound-based therapy. Furthermore, single-cell RNA sequencing of spleen cells revealed the importance of T, B, and myeloid cell populations in the anti-inflammatory pathway. These results elucidated the unique cell subsets and transcriptome of splenic cells responsive to LFPFU and demonstrated the potential of spleen-focused ultrasound stimulation in the treatment of inflammatory diseases.
Collapse
Affiliation(s)
- Xuqiao Hu
- Department of Ultrasound, First Affiliated Hospital of Southern University of Science and Technology, Second Clinical Medical College of Jinan University (Shenzhen People's Hospital), Shenzhen 518020, China
- Integrated Chinese and Western Medicine Postdoctoral Research Station, Jinan University, Guangzhou 510632, China
- Paul C. Lauterbur Research Center for Biomedical Imaging, Institute of Biomedical and Health Engineering, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, 1068 Xueyuan Avenue, Shenzhen University Town, Shenzhen 518055, China
| | - Fei Li
- Paul C. Lauterbur Research Center for Biomedical Imaging, Institute of Biomedical and Health Engineering, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, 1068 Xueyuan Avenue, Shenzhen University Town, Shenzhen 518055, China
| | - Jieying Zeng
- Department of Ultrasound, First Affiliated Hospital of Southern University of Science and Technology, Second Clinical Medical College of Jinan University (Shenzhen People's Hospital), Shenzhen 518020, China
| | - Zhenru Zhou
- Department of Ultrasound, First Affiliated Hospital of Southern University of Science and Technology, Second Clinical Medical College of Jinan University (Shenzhen People's Hospital), Shenzhen 518020, China
| | - Zhaoyang Wang
- Department of Ultrasound, First Affiliated Hospital of Southern University of Science and Technology, Second Clinical Medical College of Jinan University (Shenzhen People's Hospital), Shenzhen 518020, China
| | - Jing Chen
- Department of Ultrasound, First Affiliated Hospital of Southern University of Science and Technology, Second Clinical Medical College of Jinan University (Shenzhen People's Hospital), Shenzhen 518020, China
| | - Dongyan Cao
- Shanghai Cancer Institute, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200032, China
| | - Yifan Hong
- Institute of Molecular Physiology, Institute of Cancer Research, Shenzhen Bay Laboratory, Shenzhen 518132, P.R. China
| | - Laixin Huang
- Paul C. Lauterbur Research Center for Biomedical Imaging, Institute of Biomedical and Health Engineering, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, 1068 Xueyuan Avenue, Shenzhen University Town, Shenzhen 518055, China
| | - Yongsheng Chen
- Department of Ultrasound, First Affiliated Hospital of Southern University of Science and Technology, Second Clinical Medical College of Jinan University (Shenzhen People's Hospital), Shenzhen 518020, China
| | - Jinfeng Xu
- Department of Ultrasound, First Affiliated Hospital of Southern University of Science and Technology, Second Clinical Medical College of Jinan University (Shenzhen People's Hospital), Shenzhen 518020, China
| | - Fajin Dong
- Department of Ultrasound, First Affiliated Hospital of Southern University of Science and Technology, Second Clinical Medical College of Jinan University (Shenzhen People's Hospital), Shenzhen 518020, China
| | - Rongmin Yu
- Integrated Chinese and Western Medicine Postdoctoral Research Station, Jinan University, Guangzhou 510632, China
- Department of Pharmacology, College of Pharmacy, Jinan University, 601 Huangpu Avenue West, Guangzhou 510632, China
- Biotechnological Institute of Chinese Materia Medica, Jinan University, 601 Huangpu Avenue West, Guangzhou 510632, China
| | - Hairong Zheng
- Paul C. Lauterbur Research Center for Biomedical Imaging, Institute of Biomedical and Health Engineering, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, 1068 Xueyuan Avenue, Shenzhen University Town, Shenzhen 518055, China
| |
Collapse
|
161
|
Liu C, Chen H, Liu Y, Huang H, Yu W, Du T, Long X, Chen X, Chen Z, Guo S, Li J, Jiang Z, Wang L, Lu C. Immunity: Psoriasis comorbid with atherosclerosis. Front Immunol 2022; 13:1070750. [PMID: 36591241 PMCID: PMC9798109 DOI: 10.3389/fimmu.2022.1070750] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2022] [Accepted: 11/24/2022] [Indexed: 12/23/2022] Open
Abstract
Psoriasis is an immune-mediated, persistent inflammatory disease with a genetic predisposition, and the involvement of multiple organs in psoriasis remains indicative of systemic disease. Atherosclerosis (AS) is a common complication of patients with severe or prolonged psoriasis. The specific pathogenesis of psoriasis is still unclear. Current studies suggest that psoriasis is a polygenic genetic disease with the interaction of multiple factors such as heredity and environment. Keratinocytes are proliferated through immune-mediated inflammatory pathway, which leads to cell activation, infiltration of dermis cells and release of inflammatory factors. Activation of inflammatory cells and pro-inflammatory factors play an important role in the progression of psoriasis and atherosclerosis. Studies have found that there is a close relationship between psoriasis and atherosclerosis, and systemic inflammation may be the common feature of psoriasis and AS. This paper attempts to explore the possibility of the relationship between psoriasis and atherosclerotic comorbidities from the aspects of potential epidemiology and immune mechanism, in order to provide some reference for the subsequent scientific research.
Collapse
Affiliation(s)
- Chunping Liu
- State Key Laboratory of Dampness Syndrome of Chinese Medicine, The Second Affilliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China,Guangdong-Hong Kong- Macau Joint Lab on Chinese Medicine and Immune Disease Research, The Second Affilliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China,Guangdong Academy of Chinese Medicine Sciences, Guangdong Provincial Hospital of Traditional Chinese Medicine, Guangzhou, China,State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macau, Macau SAR, China
| | - Huiqi Chen
- State Key Laboratory of Dampness Syndrome of Chinese Medicine, The Second Affilliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China,Guangdong Academy of Chinese Medicine Sciences, Guangdong Provincial Hospital of Traditional Chinese Medicine, Guangzhou, China
| | - Yanjiao Liu
- State Key Laboratory of Dampness Syndrome of Chinese Medicine, The Second Affilliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China,Guangdong Academy of Chinese Medicine Sciences, Guangdong Provincial Hospital of Traditional Chinese Medicine, Guangzhou, China
| | - Haiding Huang
- State Key Laboratory of Dampness Syndrome of Chinese Medicine, The Second Affilliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China,Guangdong Academy of Chinese Medicine Sciences, Guangdong Provincial Hospital of Traditional Chinese Medicine, Guangzhou, China
| | - Wanling Yu
- State Key Laboratory of Dampness Syndrome of Chinese Medicine, The Second Affilliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China,Guangdong Academy of Chinese Medicine Sciences, Guangdong Provincial Hospital of Traditional Chinese Medicine, Guangzhou, China
| | - Tingting Du
- State Key Laboratory of Dampness Syndrome of Chinese Medicine, The Second Affilliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China,Guangdong Academy of Chinese Medicine Sciences, Guangdong Provincial Hospital of Traditional Chinese Medicine, Guangzhou, China
| | - Xinyao Long
- State Key Laboratory of Dampness Syndrome of Chinese Medicine, The Second Affilliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China,Guangdong Academy of Chinese Medicine Sciences, Guangdong Provincial Hospital of Traditional Chinese Medicine, Guangzhou, China
| | - Xinming Chen
- State Key Laboratory of Dampness Syndrome of Chinese Medicine, The Second Affilliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China,Guangdong Academy of Chinese Medicine Sciences, Guangdong Provincial Hospital of Traditional Chinese Medicine, Guangzhou, China
| | - Zhijun Chen
- State Key Laboratory of Dampness Syndrome of Chinese Medicine, The Second Affilliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China,Guangdong Academy of Chinese Medicine Sciences, Guangdong Provincial Hospital of Traditional Chinese Medicine, Guangzhou, China
| | - Sien Guo
- State Key Laboratory of Dampness Syndrome of Chinese Medicine, The Second Affilliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China,Guangdong Academy of Chinese Medicine Sciences, Guangdong Provincial Hospital of Traditional Chinese Medicine, Guangzhou, China
| | - Jinxin Li
- State Key Laboratory of Dampness Syndrome of Chinese Medicine, The Second Affilliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China,Guangdong Academy of Chinese Medicine Sciences, Guangdong Provincial Hospital of Traditional Chinese Medicine, Guangzhou, China
| | - Zebo Jiang
- Guangdong Provincial Key Laboratory of Biomedical Imaging and Guangdong Provincial Engineering Research Center of Molecular Imaging, The Fifth Affiliated Hospital, Sun Yat-sen University, Zhuhai, Guangdong, China,*Correspondence: Zebo Jiang, ; Lei Wang, ; Chuanjian Lu,
| | - Lei Wang
- State Key Laboratory of Dampness Syndrome of Chinese Medicine, The Second Affilliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China,Guangdong Academy of Chinese Medicine Sciences, Guangdong Provincial Hospital of Traditional Chinese Medicine, Guangzhou, China,*Correspondence: Zebo Jiang, ; Lei Wang, ; Chuanjian Lu,
| | - Chuanjian Lu
- State Key Laboratory of Dampness Syndrome of Chinese Medicine, The Second Affilliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China,Guangdong-Hong Kong- Macau Joint Lab on Chinese Medicine and Immune Disease Research, The Second Affilliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China,Guangdong Academy of Chinese Medicine Sciences, Guangdong Provincial Hospital of Traditional Chinese Medicine, Guangzhou, China,*Correspondence: Zebo Jiang, ; Lei Wang, ; Chuanjian Lu,
| |
Collapse
|
162
|
Zhang C, Li T, Yin S, Gao M, He H, Li Y, Jiang D, Shi M, Wang J, Yu L. Monocytes deposit migrasomes to promote embryonic angiogenesis. Nat Cell Biol 2022; 24:1726-1738. [PMID: 36443426 DOI: 10.1038/s41556-022-01026-3] [Citation(s) in RCA: 62] [Impact Index Per Article: 20.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2021] [Accepted: 10/12/2022] [Indexed: 11/30/2022]
Abstract
Pro-angiogenic factors are key regulators of angiogenesis. Here we report that highly migratory cells patrol the area of capillary formation in chick embryo chorioallantoic membrane. These cells deposit migrasomes on their migration tracks, creating migrasome-enriched areas. Single-cell sequencing identified these cells as monocytes. Depletion of monocytes impairs capillary formation. Quantitative mass spectrometry analysis reveals that monocyte migrasomes are enriched with pro-angiogenic factors. Purified migrasomes promote capillary formation and monocyte recruitment in vivo, and endothelial cell tube formation and monocyte chemotaxis in vitro. Knockdown or knockout of TSPAN4 reduces migrasome formation and impairs capillary formation and monocyte recruitment. Mechanistically, monocytes promote angiogenesis via VEGFA and CXCL12 in migrasomes. In summary, monocytes deposit migrasomes enriched in pro-angiogenic factors to promote angiogenesis.
Collapse
Affiliation(s)
- Cuifang Zhang
- State Key Laboratory of Membrane Biology, Tsinghua University-Peking University Joint Centre for Life Sciences, Beijing Frontier Research Center for Biological Structure, School of Life Sciences, Tsinghua University, Beijing, China
| | - Tianqi Li
- School of Life Science, Tsinghua University, Beijing, China
| | - Shuyao Yin
- State Key Laboratory of Membrane Biology, Tsinghua University-Peking University Joint Centre for Life Sciences, Beijing Frontier Research Center for Biological Structure, School of Life Sciences, Tsinghua University, Beijing, China
| | - Mingyi Gao
- State Key Laboratory of Membrane Biology, Tsinghua University-Peking University Joint Centre for Life Sciences, Beijing Frontier Research Center for Biological Structure, School of Life Sciences, Tsinghua University, Beijing, China
| | - Helen He
- State Key Laboratory of Membrane Biology, Tsinghua University-Peking University Joint Centre for Life Sciences, Beijing Frontier Research Center for Biological Structure, School of Life Sciences, Tsinghua University, Beijing, China
| | - Ying Li
- State Key Laboratory of Membrane Biology, Tsinghua University-Peking University Joint Centre for Life Sciences, Beijing Frontier Research Center for Biological Structure, School of Life Sciences, Tsinghua University, Beijing, China
| | - Dong Jiang
- State Key Laboratory of Membrane Biology, Tsinghua University-Peking University Joint Centre for Life Sciences, Beijing Frontier Research Center for Biological Structure, School of Life Sciences, Tsinghua University, Beijing, China
| | - Minghui Shi
- State Key Laboratory of Membrane Biology, Tsinghua University-Peking University Joint Centre for Life Sciences, Beijing Frontier Research Center for Biological Structure, School of Life Sciences, Tsinghua University, Beijing, China
| | - Jianbin Wang
- School of Life Science, Tsinghua University, Beijing, China
| | - Li Yu
- State Key Laboratory of Membrane Biology, Tsinghua University-Peking University Joint Centre for Life Sciences, Beijing Frontier Research Center for Biological Structure, School of Life Sciences, Tsinghua University, Beijing, China.
| |
Collapse
|
163
|
Park HE, Oh H, Baek JH. Interleukin-34-regulated T-cell responses in rheumatoid arthritis. Front Med (Lausanne) 2022; 9:1078350. [PMID: 36530919 PMCID: PMC9747768 DOI: 10.3389/fmed.2022.1078350] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2022] [Accepted: 11/14/2022] [Indexed: 09/10/2023] Open
Abstract
Rheumatoid arthritis (RA) is a chronic autoimmune disease with a multifaceted etiology, which primarily affects and results in the deterioration of the synovium of patients. While the exact etiology of RA is still largely unknown, there is growing interest in the cytokine interleukin-34 (IL-34) as a driver or modulator of RA pathogenesis on the grounds that IL-34 is drastically increased in the serum and synovium of RA patients. Several studies have so far revealed the relationship between IL-34 levels and RA disease progression. Nevertheless, the significance and role of IL-34 in RA have remained ambiguous, as illustrated by two most recent studies, which reported contrasting effects of genetic IL-34 deletion in RA. Of note, IL-34 is a macrophage growth factor and is increasingly perceived as a master regulator of T-cell responses in RA via macrophage-dependent as well as T cell-intrinsic mechanisms. In this regard, several studies have demonstrated that IL-34 potentiates helper T-cell (Th) responses in RA, whereas studies also suggested that IL-34 alleviates synovial inflammation, potentially by inducing regulatory T-cells (Treg). Herein, we provide an overview of the current understanding of IL-34 involvement in RA and outline IL-34-mediated mechanisms in regulating T-cell responses in RA.
Collapse
Affiliation(s)
| | | | - Jea-Hyun Baek
- School of Life Science, Handong Global University, Pohang, South Korea
| |
Collapse
|
164
|
Qu L, Matz AJ, Karlinsey K, Cao Z, Vella AT, Zhou B. Macrophages at the Crossroad of Meta-Inflammation and Inflammaging. Genes (Basel) 2022; 13:2074. [PMID: 36360310 PMCID: PMC9690997 DOI: 10.3390/genes13112074] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2022] [Revised: 10/24/2022] [Accepted: 11/07/2022] [Indexed: 08/29/2023] Open
Abstract
Macrophages are central players in systemic inflammation associated with obesity and aging, termed meta-inflammation and inflammaging. Activities of macrophages elicited by the two chronic conditions display shared and distinct patterns mechanistically, resulting in multifaceted actions for their pathogenic roles. Drastically expanded tissue macrophage populations under obesity and aging stress attribute to both enhanced recruitment and local expansion. Importantly, molecular networks governing the multifaceted actions of macrophages are directly altered by environmental cues and subsequently contribute to metabolic reprogramming, resulting in meta-inflammation in obesity or inflammaging in aging. In this review, we will summarize how meta-inflammation and inflammaging affect macrophages and the molecular mechanisms involved in these processes.
Collapse
Affiliation(s)
- Lili Qu
- Department of Immunology, School of Medicine, University of Connecticut, Farmington, CT 06030, USA
| | - Alyssa J. Matz
- Department of Immunology, School of Medicine, University of Connecticut, Farmington, CT 06030, USA
| | - Keaton Karlinsey
- Department of Immunology, School of Medicine, University of Connecticut, Farmington, CT 06030, USA
| | - Ziming Cao
- Department of Immunology, School of Medicine, University of Connecticut, Farmington, CT 06030, USA
| | - Anthony T. Vella
- Department of Immunology, School of Medicine, University of Connecticut, Farmington, CT 06030, USA
- Institute for Systems Genomics, University of Connecticut, Farmington, CT 06030, USA
| | - Beiyan Zhou
- Department of Immunology, School of Medicine, University of Connecticut, Farmington, CT 06030, USA
- Institute for Systems Genomics, University of Connecticut, Farmington, CT 06030, USA
| |
Collapse
|
165
|
Wierenga KA, Riemers FM, Westendorp B, Harkema JR, Pestka JJ. Single cell analysis of docosahexaenoic acid suppression of sequential LPS-induced proinflammatory and interferon-regulated gene expression in the macrophage. Front Immunol 2022; 13:993614. [PMID: 36405730 PMCID: PMC9669445 DOI: 10.3389/fimmu.2022.993614] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2022] [Accepted: 09/22/2022] [Indexed: 11/06/2022] Open
Abstract
Preclinical and clinical studies suggest that consumption of long chain omega-3 polyunsaturated fatty acids (PUFAs) reduces severity of chronic inflammatory and autoimmune diseases. While these ameliorative effects are conventionally associated with downregulated expression of proinflammatory cytokine and chemokine genes, our laboratory has recently identified Type 1 interferon (IFN1)-regulated gene expression to be another key target of omega-3 PUFAs. Here we used single cell RNA sequencing (scRNAseq) to gain new mechanistic perspectives on how the omega-3 PUFA docosahexaenoic acid (DHA) influences TLR4-driven proinflammatory and IFN1-regulated gene expression in a novel self-renewing murine fetal liver-derived macrophage (FLM) model. FLMs were cultured with 25 µM DHA or vehicle for 24 h, treated with modest concentration of LPS (20 ng/ml) for 1 and 4 h, and then subjected to scRNAseq using the 10X Chromium System. At 0 h (i.e., in the absence of LPS), DHA increased expression of genes associated with the NRF2 antioxidant response (e.g. Sqstm1, Hmox1, Chchd10) and metal homeostasis (e.g.Mt1, Mt2, Ftl1, Fth1), both of which are consistent with DHA-induced polarization of FLMs to a more anti-inflammatory phenotype. At 1 h post-LPS treatment, DHA inhibited LPS-induced cholesterol synthesis genes (e.g. Scd1, Scd2, Pmvk, Cyp51, Hmgcs1, and Fdps) which potentially could contribute to interference with TLR4-mediated inflammatory signaling. At 4 h post-LPS treatment, LPS-treated FLMs reflected a more robust inflammatory response including upregulation of proinflammatory cytokine (e.g. Il1a, Il1b, Tnf) and chemokine (e.g.Ccl2, Ccl3, Ccl4, Ccl7) genes as well as IFN1-regulated genes (e.g. Irf7, Mx1, Oasl1, Ifit1), many of which were suppressed by DHA. Using single-cell regulatory network inference and clustering (SCENIC) to identify gene expression networks, we found DHA modestly downregulated LPS-induced expression of NF-κB-target genes. Importantly, LPS induced a subset of FLMs simultaneously expressing NF-κB- and IRF7/STAT1/STAT2-target genes that were conspicuously absent in DHA-pretreated FLMs. Thus, DHA potently targeted both the NF-κB and the IFN1 responses. Altogether, scRNAseq generated a valuable dataset that provides new insights into multiple overlapping mechanisms by which DHA may transcriptionally or post-transcriptionally regulate LPS-induced proinflammatory and IFN1-driven responses in macrophages.
Collapse
Affiliation(s)
- Kathryn A. Wierenga
- Department of Biochemistry and Molecular Biology, Michigan State University, Lansing, MI, United States
- Institute for Integrative Toxicology, Michigan State University, Lansing, MI, United States
| | - Frank M. Riemers
- Department of Biomolecular Health Sciences, Faculty of Veterinary Medicine, Utrecht University, Utrecht, Netherlands
| | - Bart Westendorp
- Department of Biomolecular Health Sciences, Faculty of Veterinary Medicine, Utrecht University, Utrecht, Netherlands
| | - Jack R. Harkema
- Institute for Integrative Toxicology, Michigan State University, Lansing, MI, United States
- Department of Pathobiology and Diagnostic Investigation, Michigan State University, Lansing, MI, United States
| | - James J. Pestka
- Institute for Integrative Toxicology, Michigan State University, Lansing, MI, United States
- Department of Microbiology and Molecular Genetics, Michigan State University, Lansing, MI, United States
- Department of Food Science and Human Nutrition, Michigan State University, Lansing, MI, United States
| |
Collapse
|
166
|
Malange KF, Navia-Pelaez JM, Dias EV, Lemes JBP, Choi SH, Dos Santos GG, Yaksh TL, Corr M. Macrophages and glial cells: Innate immune drivers of inflammatory arthritic pain perception from peripheral joints to the central nervous system. FRONTIERS IN PAIN RESEARCH 2022; 3:1018800. [PMID: 36387416 PMCID: PMC9644179 DOI: 10.3389/fpain.2022.1018800] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2022] [Accepted: 10/03/2022] [Indexed: 07/22/2023] Open
Abstract
Millions of people suffer from arthritis worldwide, consistently struggling with daily activities due to debilitating pain evoked by this disease. Perhaps the most intensively investigated type of inflammatory arthritis is rheumatoid arthritis (RA), where, despite considerable advances in research and clinical management, gaps regarding the neuroimmune interactions that guide inflammation and chronic pain in this disease remain to be clarified. The pain and inflammation associated with arthritis are not isolated to the joints, and inflammatory mechanisms induced by different immune and glial cells in other tissues may affect the development of chronic pain that results from the disease. This review aims to provide an overview of the state-of-the-art research on the roles that innate immune, and glial cells play in the onset and maintenance of arthritis-associated pain, reviewing nociceptive pathways from the joint through the dorsal root ganglion, spinal circuits, and different structures in the brain. We will focus on the cellular mechanisms related to neuroinflammation and pain, and treatments targeting these mechanisms from the periphery and the CNS. A comprehensive understanding of the role these cells play in peripheral inflammation and initiation of pain and the central pathways in the spinal cord and brain will facilitate identifying new targets and pathways to aide in developing therapeutic strategies to treat joint pain associated with RA.
Collapse
Affiliation(s)
- Kaue Franco Malange
- Department of Anesthesiology, University of California, San Diego, CA, United States
| | | | - Elayne Vieira Dias
- Department of Neurology, University of California, San Francisco, CA, United States
| | | | - Soo-Ho Choi
- Department of Medicine, University of California, San Diego, CA, United States
| | | | - Tony L. Yaksh
- Department of Anesthesiology, University of California, San Diego, CA, United States
| | - Maripat Corr
- Department of Medicine, University of California, San Diego, CA, United States
| |
Collapse
|
167
|
Ding Z, Li W, Dou Y, Zhou Y, Ren Y, Jing H, Liang X, Wang X, Li N. Triangular-shaped homologous heterostructure as photocatalytic H 2S scavenger and macrophage modulator for rheumatoid arthritis therapy. J Mater Chem B 2022; 10:8549-8564. [PMID: 36239131 DOI: 10.1039/d2tb01650h] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/16/2023]
Abstract
Rheumatoid arthritis (RA) is a chronic arthropathy causing cartilage destruction, bone erosion, and even disability. Although some advances in RA treatment have been made based on inflammatory cytokine inhibition, long-term treatment and drug effect have been restrained by severe side effects. Herein, we developed a resveratrol (RSV)-loaded Ag/Ag2S triangular-shaped homologous heterostructure with polyethylene glycol/folic acid (PEG/FA) modification (Ag/Ag2S-PEG-FA/RSV NTs) to simultaneously suppress inflammatory cytokine over-expression through photocatalytic H2S scavenging and macrophage polarization stimulation. On one hand, the over-expressed H2S, which acted as a pro-inflammatory mediator to activate the MAPK/ICAM-1 pathway and exacerbate inflammation, was eliminated through photocatalysis. The homologous Ag and Ag2S of the heterostructure enhanced electron separation and transfer by acting as a charge acceptor and electron generator, respectively, which restrained electron/hole recombination and promoted photocatalysis efficiency. Additionally, the intrinsic superoxide dismutase (SOD) and catalase (CAT) activity of Ag decomposed the reactive oxygen species (ROS) over-expressed in the RA microenvironment, which supplied O2 for the photocatalytic H2S scavenging progress. On the other hand, RSV, a natural product with anti-inflammatory activity, could be delivered to the inflammatory joint by the targeting effect of PEG-FA, thus inhibiting the IκB/NF-κB pro-inflammatory pathway to induce macrophage interconversion balance from M1 to M2. As expected, the Ag/Ag2S-PEG-FA/RSV NTs exhibited H2S scavenging capacity and modulated macrophage polarization to reduce the inflammatory cytokine level and halt RA progression in vitro and in vivo. Overall, this study revealed a therapeutic strategy with high efficacy, which opens broad prospects for RA treatment.
Collapse
Affiliation(s)
- Ziqiao Ding
- Tianjin Key Laboratory of Drug Delivery & High-Efficiency, School of Pharmaceutical Science and Technology, Tianjin University, 92 Weijin Road, Nankai District, 300072, Tianjin, P. R. China.
| | - Wen Li
- Tianjin Key Laboratory of Drug Delivery & High-Efficiency, School of Pharmaceutical Science and Technology, Tianjin University, 92 Weijin Road, Nankai District, 300072, Tianjin, P. R. China.
| | - Yunsheng Dou
- Tianjin Key Laboratory of Drug Delivery & High-Efficiency, School of Pharmaceutical Science and Technology, Tianjin University, 92 Weijin Road, Nankai District, 300072, Tianjin, P. R. China.
| | - Yue Zhou
- Tianjin Key Laboratory of Drug Delivery & High-Efficiency, School of Pharmaceutical Science and Technology, Tianjin University, 92 Weijin Road, Nankai District, 300072, Tianjin, P. R. China.
| | - Yingzi Ren
- Tianjin Key Laboratory of Drug Delivery & High-Efficiency, School of Pharmaceutical Science and Technology, Tianjin University, 92 Weijin Road, Nankai District, 300072, Tianjin, P. R. China.
| | - Huaqing Jing
- Tianjin Key Laboratory of Drug Delivery & High-Efficiency, School of Pharmaceutical Science and Technology, Tianjin University, 92 Weijin Road, Nankai District, 300072, Tianjin, P. R. China.
| | - Xiaoyang Liang
- Tianjin Key Laboratory of Drug Delivery & High-Efficiency, School of Pharmaceutical Science and Technology, Tianjin University, 92 Weijin Road, Nankai District, 300072, Tianjin, P. R. China.
| | - Xinxing Wang
- Tianjin Institute of Environmental and Operational Medicine, 1 Dali Road, Heping District, 300050, Tianjin, P. R. China.
| | - Nan Li
- Tianjin Key Laboratory of Drug Delivery & High-Efficiency, School of Pharmaceutical Science and Technology, Tianjin University, 92 Weijin Road, Nankai District, 300072, Tianjin, P. R. China.
| |
Collapse
|
168
|
Identification and Validation of Hub Genes for Predicting Treatment Targets and Immune Landscape in Rheumatoid Arthritis. BIOMED RESEARCH INTERNATIONAL 2022; 2022:8023779. [PMID: 36317112 PMCID: PMC9617710 DOI: 10.1155/2022/8023779] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/27/2022] [Accepted: 09/27/2022] [Indexed: 11/17/2022]
Abstract
Background Rheumatoid arthritis (RA) is recognized as a chronic inflammatory disease featured by pathological synovial inflammation. Currently, the underlying pathophysiological mechanisms of RA remain unclear. In the study, we attempted to explore the underlying mechanisms of RA and provide potential targets for the therapy of RA via bioinformatics analysis. Methods We downloaded four microarray datasets (GSE77298, GSE55235, GSE12021, and GSE55457) from the GEO database. Firstly, GSE77298 and GSE55457 were identified DEGs by the “limma” and “sva” packages of R software. Then, we performed GO, KEGG, and GSEA enrichment analyses to further analyze the function of DEGs. Hub genes were screened using LASSO analysis and SVM-RFE analysis. To further explore the differences of the expression of hub genes in healthy control and RA patient synovial tissues, we calculated the ROC curves and AUC. The expression levels of hub genes were verified in synovial tissues of normal and RA rats by qRT-PCR and western blot. Furthermore, the CIBERSORTx was implemented to assess the differences of infiltration in 22 immune cells between normal and RA synovial tissues. We explored the association between hub genes and infiltrating immune cells. Results CRTAM, CXCL13, and LRRC15 were identified as RA's potential hub genes by machine learning and LASSO algorithms. In addition, we verified the expression levels of three hub genes in the synovial tissue of normal and RA rats by PCR and western blot. Moreover, immune cell infiltration analysis showed that plasma cells, T follicular helper cells, M0 macrophages, M1 macrophages, and gamma delta T cells may be engaged in the development and progression of RA. Conclusions In brief, our study identified and validated that three hub genes CRTAM, CXCL13, and LRRC15 might involve in the pathological development of RA, which could provide novel perspectives for the diagnosis and treatment with RA.
Collapse
|
169
|
Synovial Macrophages Expression of OX40L Is Required for Follicular Helper T Cells Differentiation in the Joint Microenvironment. Cells 2022; 11:cells11203326. [PMID: 36291190 PMCID: PMC9601235 DOI: 10.3390/cells11203326] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2022] [Revised: 10/18/2022] [Accepted: 10/20/2022] [Indexed: 11/27/2022] Open
Abstract
Signaling via the OX40/OX40L axis plays a key role in CD4+ T cell development, and OX40L expression is primarily restricted to antigen-presenting cells (APCs). This study was designed to assess the role of APC-mediated OX40L expression in the context of the development of rheumatoid arthritis (RA)-associated CD4+ T cell subsets. For these analyses, clinical samples were harvested from patients with osteoarthritis and RA, with additional analyses performed using OX40−/− mice and mice harboring monocyte/macrophage-specific deletions of OX40L. Together, these analyses revealed tissue-specific roles for OX40/OX40L signaling in RA. Specifically, higher levels of synovial macrophage OX40L expression were associated with the enhanced development of T follicular helper cells in the joint microenvironment, thereby contributing to the pathogenesis of RA. This Tfh differentiation was found to be OX40/OX40L-dependent in this synovial setting. Overall, these results indicate that the expression of OX40L by synovia macrophages is necessary to support Tfh differentiation in the joint tissues, thus offering new insight regarding the etiological basis for RA progression.
Collapse
|
170
|
Cai T, Du P, Suo L, Jiang X, Qin Q, Song R, Yang X, Jiang Y, Zhang JA. High iodine promotes autoimmune thyroid disease by activating hexokinase 3 and inducing polarization of macrophages towards M1. Front Immunol 2022; 13:1009932. [PMID: 36325332 PMCID: PMC9618622 DOI: 10.3389/fimmu.2022.1009932] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2022] [Accepted: 09/27/2022] [Indexed: 11/13/2022] Open
Abstract
Autoimmune thyroid disease (AITD), the most common autoimmune disease, includes Graves’ disease (GD) and Hashimoto’s thyroiditis (HT). Currently, the pathogenesis of AITD is not fully understood. Our study aimed to examine the presence of macrophage polarization imbalance in AITD patients, to investigate whether high iodine can cause macrophage polarization imbalance, and to investigate the role of key genes of metabolic reprogramming in macrophage polarization imbalance caused by high iodine. We synergistically used various research strategies such as systems biology, clinical studies, cell culture and mouse disease models. Gene set enrichment analysis (GSEA) revealed that M1 macrophage hyperpolarization was involved in the pathogenesis of AITD. In vitro and in vivo experiments showed that high iodine can affect the polarization of M1 or M2 macrophages and their related cytokines. Robust rank aggregation (RRA) method revealed that hexokinase 3 (HK3) was the most aberrantly expressed metabolic gene in autoimmune diseases. In vitro and in vivo studies revealed HK3 could mediate macrophage polarization induced by high iodine. In summary, hyperpolarization of M1-type macrophages is closely related to the pathogenesis of AITD. High iodine can increase HK3 expression in macrophages and promote macrophage polarization towards M1. Targeting HK3 can inhibit M1 polarization induced by high iodine.
Collapse
Affiliation(s)
- Tiantian Cai
- Department of Endocrinology & Rheumatology, Shanghai University of Medicine & Health Sciences Affiliated Zhoupu Hospital, Shanghai, China
| | - Peng Du
- Department of Endocrinology & Rheumatology, Shanghai University of Medicine & Health Sciences Affiliated Zhoupu Hospital, Shanghai, China
| | - Lixia Suo
- Department of Endocrinology, Shanghai University of Medicine & Health Sciences Affiliated Jiading District Central Hospital, Shanghai, China
| | - Xiaozhen Jiang
- Department of Endocrinology, Shanghai Pudong New Area People’s Hospital, Shanghai, China
| | - Qiu Qin
- Department of Endocrinology & Rheumatology, Shanghai University of Medicine & Health Sciences Affiliated Zhoupu Hospital, Shanghai, China
| | - Ronghua Song
- Department of Endocrinology & Rheumatology, Shanghai University of Medicine & Health Sciences Affiliated Zhoupu Hospital, Shanghai, China
| | - Xiaorong Yang
- Department of Endocrinology & Rheumatology, Shanghai University of Medicine & Health Sciences Affiliated Zhoupu Hospital, Shanghai, China
| | - Yanfei Jiang
- Department of Endocrinology & Rheumatology, Shanghai University of Medicine & Health Sciences Affiliated Zhoupu Hospital, Shanghai, China
- *Correspondence: Yanfei Jiang, ; Jin-an Zhang,
| | - Jin-an Zhang
- Department of Endocrinology & Rheumatology, Shanghai University of Medicine & Health Sciences Affiliated Zhoupu Hospital, Shanghai, China
- *Correspondence: Yanfei Jiang, ; Jin-an Zhang,
| |
Collapse
|
171
|
Photobiomodulation effects in metalloproteinases expression in zymosan-induced arthritis. Lasers Med Sci 2022; 37:3661-3670. [PMID: 36156751 DOI: 10.1007/s10103-022-03647-4] [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: 06/17/2021] [Accepted: 09/18/2022] [Indexed: 10/14/2022]
Abstract
Matrix metalloproteinases (MMPs) play a crucial role in the degenerative course of rheumatic disorders. They are responsible for cartilage and other joint-associated tissues breakdown. Amid arthritis treatments, photobiostimulation (PBM), a non-thermal and non-invasive low-power laser application, appears to be an outstanding therapy alternative once it has succeeded in MMPs modulation. Thus, this study aimed to evaluate the PBM effects of low infrared laser (830 nm), testing two different energy densities (3 and 30 Jcm-2) in MMP-2, MMP-9, MMP-13, and MMP-14 as well as the inhibitor TIMP-2 expressions using zymosan-induced arthritis model. C57BL/6 mice were distributed into four groups (n = 8): zymosan-induced arthritis without treatment; zymosan-induced arthritis and dexamethasone-treated; zymosan-induced arthritis and PBM at energy density of 3 Jcm-2 treated; and zymosan-induced arthritis and PBM at energy density of 30 Jcm-2 treated. MMPs and TIMP-2 mRNA relative levels by qRT-PCR and proteins expression by immunohistochemical and Western blotting techniques were performed after PBM treatment in the inflamed joint. Our results demonstrated PBM could modulate both mRNA relative levels and proteins expression of the MMP-2, -9, -13, -14, and TIMP-2 in joint tissues, decreasing MMP-9 protein expression and increasing TIMP-2 protein expression. PBM promotes a better arthritis prognostic, modulating metalloproteinase and its inhibitor, especially MMP-9 and TIMP-2 protein expression that is important inflammatory markers. These findings may also corroborate that PBM may regulate MMPs expression using different pathways.
Collapse
|
172
|
Chen X, Wu Q, Cao X, Yang Y, Gong Z, Ren T, Du Q, Yuan Y, Zuo Y, Miao Y, He J, Qiao C, Zheng Z, Zhang T, Xu Y, Wu D, Wang Q, Huang L, Xie Z, Lv H, Wang J, Gong F, Liu Z, Wen C, Zheng H. Menthone inhibits type-I interferon signaling by promoting Tyk2 ubiquitination to relieve local inflammation of rheumatoid arthritis. Int Immunopharmacol 2022; 112:109228. [PMID: 36095947 DOI: 10.1016/j.intimp.2022.109228] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2022] [Revised: 08/31/2022] [Accepted: 09/01/2022] [Indexed: 11/05/2022]
Abstract
Rheumatoid arthritis (RA) is an inflammatory autoimmune disease. RA development is mediated by the abnormal activation of multiple signaling pathways. Recent studies have revealed that type-I interferon (IFN-I) signaling plays an essential role in the occurrence and development of RA. However, how to target IFN-I signaling to develop anti-rheumatoid arthritis drugs remains largely unexplored. Here, our study showed that IFN-I signaling was over-activated in articular synovial cells from collagen II-induced arthritis (CIA) mice. Interestingly, we found that a small molecule compound, menthone, strongly inhibited the activation of the IFN-I signaling pathway. Further studies revealed that menthone promoted K48-linked polyubiquitination of Tyk2, thus lowering the protein level and stability of Tyk2. Importantly, menthone administration in the local articulus of CIA mice significantly attenuated the local inflammation in CIA mice. This study could promote our understanding of rheumatoid arthritis, and also suggests a potential strategy to develop anti-RA drugs.
Collapse
Affiliation(s)
- Xiangjie Chen
- Institutes of Biology and Medical Sciences, Jiangsu Key Laboratory of Infection and Immunity, Soochow University, Suzhou 215123, Jiangsu, China
| | - Qiuyu Wu
- Institutes of Biology and Medical Sciences, Jiangsu Key Laboratory of Infection and Immunity, Soochow University, Suzhou 215123, Jiangsu, China
| | - Xinhua Cao
- Institutes of Biology and Medical Sciences, Jiangsu Key Laboratory of Infection and Immunity, Soochow University, Suzhou 215123, Jiangsu, China
| | - Yunshan Yang
- Medical College of Soochow University, Suzhou 215123, Jiangsu, China
| | - Zheng Gong
- Institutes of Biology and Medical Sciences, Jiangsu Key Laboratory of Infection and Immunity, Soochow University, Suzhou 215123, Jiangsu, China
| | - Tengfei Ren
- Institutes of Biology and Medical Sciences, Jiangsu Key Laboratory of Infection and Immunity, Soochow University, Suzhou 215123, Jiangsu, China
| | - Qian Du
- Institutes of Biology and Medical Sciences, Jiangsu Key Laboratory of Infection and Immunity, Soochow University, Suzhou 215123, Jiangsu, China
| | - Yukang Yuan
- Institutes of Biology and Medical Sciences, Jiangsu Key Laboratory of Infection and Immunity, Soochow University, Suzhou 215123, Jiangsu, China
| | - Yibo Zuo
- Institutes of Biology and Medical Sciences, Jiangsu Key Laboratory of Infection and Immunity, Soochow University, Suzhou 215123, Jiangsu, China
| | - Ying Miao
- Institutes of Biology and Medical Sciences, Jiangsu Key Laboratory of Infection and Immunity, Soochow University, Suzhou 215123, Jiangsu, China
| | - Jiuyi He
- Institutes of Biology and Medical Sciences, Jiangsu Key Laboratory of Infection and Immunity, Soochow University, Suzhou 215123, Jiangsu, China
| | - Caixia Qiao
- Institutes of Biology and Medical Sciences, Jiangsu Key Laboratory of Infection and Immunity, Soochow University, Suzhou 215123, Jiangsu, China
| | - Zhijin Zheng
- Institutes of Biology and Medical Sciences, Jiangsu Key Laboratory of Infection and Immunity, Soochow University, Suzhou 215123, Jiangsu, China
| | - Tingting Zhang
- Institutes of Biology and Medical Sciences, Jiangsu Key Laboratory of Infection and Immunity, Soochow University, Suzhou 215123, Jiangsu, China
| | - Yang Xu
- Institute of Blood and Marrow Transplantation, Collaborative Innovation Center of Hematology, Soochow University, Suzhou 215123, Jiangsu, China
| | - Depei Wu
- Institute of Blood and Marrow Transplantation, Collaborative Innovation Center of Hematology, Soochow University, Suzhou 215123, Jiangsu, China
| | - Qiao Wang
- College of Basic Medical Sciences, Zhejiang Chinese Medical University, Hangzhou 310053, Zhejiang, China
| | - Lin Huang
- College of Basic Medical Sciences, Zhejiang Chinese Medical University, Hangzhou 310053, Zhejiang, China
| | - Zhijun Xie
- College of Basic Medical Sciences, Zhejiang Chinese Medical University, Hangzhou 310053, Zhejiang, China
| | - Haitao Lv
- Institute of Pediatric Research, Children's Hospital of Soochow University, Suzhou 215123, Jiangsu, China
| | - Jun Wang
- Department of Intensive Care Medicine, the First Affiliated Hospital of Soochow University, Suzhou 215123, Jiangsu, China
| | - Fangyuan Gong
- Institutes of Biology and Medical Sciences, Jiangsu Key Laboratory of Infection and Immunity, Soochow University, Suzhou 215123, Jiangsu, China
| | - Zhichun Liu
- Department of Rheumatology and Immunology, The Second Affiliated Hospital of Soochow University, Suzhou 215123, Jiangsu, China
| | - Chengping Wen
- College of Basic Medical Sciences, Zhejiang Chinese Medical University, Hangzhou 310053, Zhejiang, China
| | - Hui Zheng
- Institutes of Biology and Medical Sciences, Jiangsu Key Laboratory of Infection and Immunity, Soochow University, Suzhou 215123, Jiangsu, China.
| |
Collapse
|
173
|
Li K, Wang M, Zhao L, Liu Y, Zhang X. ACPA-negative rheumatoid arthritis: From immune mechanisms to clinical translation. EBioMedicine 2022; 83:104233. [PMID: 36027873 PMCID: PMC9404277 DOI: 10.1016/j.ebiom.2022.104233] [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: 05/17/2022] [Revised: 07/13/2022] [Accepted: 08/05/2022] [Indexed: 11/28/2022] Open
Abstract
The presence of anti-citrullinated protein autoantibodies (ACPA) is a hallmark feature of rheumatoid arthritis (RA), which causes chronic joint destruction and systemic inflammation. Based on ACPA status, RA patients can be sub-grouped into two major subsets: ACPA-positive RA (ACPA+ RA) and ACPA-negative RA (ACPA– RA). Accumulating evidence have suggested that ACPA+ RA and ACPA– RA are two distinct disease entities with different underlying pathophysiology. In contrast to the well-characterized pathogenic mechanisms of ACPA+ RA, the etiology of ACPA– RA remains largely unknown. In this review, we summarized current knowledge about the primary drivers of ACPA– RA, particularly focusing on the serological, cellular, and molecular aspects of immune mechanisms. A better understanding of the immunopathogenesis in ACPA– RA will help in designing more precisely targeting strategies, and paving the road to personalized treatment. In addition, identification of novel biomarkers in ACPA– RA will substantially promote early treatment and improve the outcomes.
Collapse
Affiliation(s)
- Ketian Li
- Department of Rheumatology, Beijing Hospital, National Center of Gerontology, Institute of Geriatric Medicine, Clinical Immunology Center, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100730, PR China
| | - Min Wang
- Department of Rheumatology, Beijing Hospital, National Center of Gerontology, Institute of Geriatric Medicine, Clinical Immunology Center, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100730, PR China
| | - Lidan Zhao
- Department of Rheumatology and Clinical Immunology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100730, PR China
| | - Yudong Liu
- Department of Rheumatology, Beijing Hospital, National Center of Gerontology, Institute of Geriatric Medicine, Clinical Immunology Center, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100730, PR China; The Key Laboratory of Geriatrics, Beijing Institute of Geriatrics, Beijing Hospital, National Center of Gerontology, National Health Commission, Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, Beijing 100730, PR China.
| | - Xuan Zhang
- Department of Rheumatology, Beijing Hospital, National Center of Gerontology, Institute of Geriatric Medicine, Clinical Immunology Center, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100730, PR China.
| |
Collapse
|
174
|
Ajayi AM, Ben-Azu B, Balogun SO, de Oliveira RG, Umukoro S, de Oliveira DT, Ademowo OG. Induction of apoptosis in activated RAW 264.7 cells and inhibition of pro-inflammatory mediators in rat air pouch by ethylacetate fraction of Ocimum gratissimum leaves. ADVANCES IN TRADITIONAL MEDICINE 2022; 22:659-671. [DOI: 10.1007/s13596-021-00554-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2020] [Accepted: 01/06/2021] [Indexed: 11/26/2022]
|
175
|
Ma C, Wang J, Hong F, Yang S. Mitochondrial Dysfunction in Rheumatoid Arthritis. Biomolecules 2022; 12:biom12091216. [PMID: 36139055 PMCID: PMC9496243 DOI: 10.3390/biom12091216] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2022] [Revised: 08/14/2022] [Accepted: 08/23/2022] [Indexed: 11/16/2022] Open
Abstract
Rheumatoid arthritis, a chronic autoimmune disease with complex etiology, is characterized by excessive proliferation of synovial cells, massive production of inflammatory cells and cartilage destruction. Studies have shown that mitochondrial dysfunction plays an important role in promoting the occurrence of RA. Mitochondria with normal structure and function are essential for the normal survival of chondrocytes and synovial cells. Once mitochondrial function is destroyed, it will affect the survival, activation and differentiation of immune cells and non-immune cells involved in the pathogenesis of RA, thus leading to the occurrence of RA. However, the mechanism of mitochondrial dysfunction in RA remains unclear. This article reviews the method of mitochondrial dysfunction leading to RA, the effects of mitochondrial dysfunction on immune cells, the etiology of mitochondrial dysfunction in RA, and the pathology of mitochondrial dysfunction in RA. We also outline some drugs that can exert therapeutic effects on RA which are associated with modulating mitochondrial activity. The understanding and summary of mitochondrial dysfunction in RA may provide new research directions for pathological intervention and prevention of RA.
Collapse
Affiliation(s)
- Chen Ma
- Experimental Center of Pathogen Biology, College of Medicine, Nanchang University, Nanchang 330006, China
- Queen Mary School, College of Medicine, Nanchang University, Nanchang 330006, China
| | - Jie Wang
- Department of Graduate, Shandong First Medical University, Shandong Academy of Medical Sciences, Jinan 250117, China
| | - Fenfang Hong
- Experimental Center of Pathogen Biology, College of Medicine, Nanchang University, Nanchang 330006, China
- Correspondence: (F.H.); or (S.Y.)
| | - Shulong Yang
- Key Laboratory of Chronic Diseases, Fuzhou Medical College, Nanchang University, Fuzhou 344099, China
- Department of Physiology, Fuzhou Medical College, Nanchang University, Fuzhou 344099, China
- Correspondence: (F.H.); or (S.Y.)
| |
Collapse
|
176
|
Ma Y, Lu Z, Jia B, Shi Y, Dong J, Jiang S, Li Z. DNA Origami as a Nanomedicine for Targeted Rheumatoid Arthritis Therapy through Reactive Oxygen Species and Nitric Oxide Scavenging. ACS NANO 2022; 16:12520-12531. [PMID: 35917300 DOI: 10.1021/acsnano.2c03991] [Citation(s) in RCA: 57] [Impact Index Per Article: 19.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/15/2023]
Abstract
Rheumatoid arthritis (RA) severely threatens human health by causing inflammation, swelling, and pain in the joints and resulting in persistent synovitis and irreversible joint disability. In the development of RA, pro-inflammatory M1 macrophages, which express high levels of reactive oxygen species (ROS) and nitric oxide (NO), induce synovial inflammation and bone erosion. Eliminating ROS and NO in the inflamed joints is a potential RA therapeutic approach, which can drive the transition of pro-inflammatory M1 macrophages to the anti-inflammatory M2 phenotype. Taking advantage of the intrinsic ROS- and NO-scavenging capability of DNA molecules, herein, we report the development of folic acid-modified triangular DNA origami nanostructures (FA-tDONs) for targeted RA treatment. FA-tDONs could efficiently scavenge ROS and NO and actively target M1 macrophages, facilitating the M1-to-M2 transition and the recovery of associated cytokines and biomarkers to the normal level. The therapeutic efficacy of FA-tDONs was examined in the RA mouse model. As validated by appearance, histological, and serum examinations, FA-tDONs treatment effectively alleviated synovial infiltration and cartilage damage, attenuating disease progression. This study demonstrated the usage of DNA origami for RA treatment and suggested its potential in other antioxidant therapies.
Collapse
Affiliation(s)
- Yuxuan Ma
- Department of Biomedical Engineering, College of Engineering and Applied Sciences, Nanjing University, Nanjing, Jiangsu 210023, P. R. China
- State Key Laboratory of Analytical Chemistry for Life Science, Nanjing University, Nanjing, Jiangsu 210023, P. R. China
| | - Zhangwei Lu
- Department of Biomedical Engineering, College of Engineering and Applied Sciences, Nanjing University, Nanjing, Jiangsu 210023, P. R. China
- State Key Laboratory of Analytical Chemistry for Life Science, Nanjing University, Nanjing, Jiangsu 210023, P. R. China
| | - Bin Jia
- Department of Biomedical Engineering, College of Engineering and Applied Sciences, Nanjing University, Nanjing, Jiangsu 210023, P. R. China
- State Key Laboratory of Analytical Chemistry for Life Science, Nanjing University, Nanjing, Jiangsu 210023, P. R. China
| | - Ye Shi
- Department of Biomedical Engineering, College of Engineering and Applied Sciences, Nanjing University, Nanjing, Jiangsu 210023, P. R. China
- State Key Laboratory of Analytical Chemistry for Life Science, Nanjing University, Nanjing, Jiangsu 210023, P. R. China
| | - Jun Dong
- Department of Neurosurgery, The Second Affiliated Hospital of Soochow University, Suzhou, Jiangsu 215004, P. R. China
| | - Shuoxing Jiang
- Department of Biomedical Engineering, College of Engineering and Applied Sciences, Nanjing University, Nanjing, Jiangsu 210023, P. R. China
| | - Zhe Li
- Department of Biomedical Engineering, College of Engineering and Applied Sciences, Nanjing University, Nanjing, Jiangsu 210023, P. R. China
- State Key Laboratory of Analytical Chemistry for Life Science, Nanjing University, Nanjing, Jiangsu 210023, P. R. China
| |
Collapse
|
177
|
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
|
178
|
Floudas A, Smith CM, Tynan O, Neto N, Krishna V, Wade SM, Hanlon M, Cunningham C, Marzaioli V, Canavan M, Fletcher JM, Mullan RH, Cole S, Hao LY, Monaghan MG, Nagpal S, Veale DJ, Fearon U. Distinct stromal and immune cell interactions shape the pathogenesis of rheumatoid and psoriatic arthritis. Ann Rheum Dis 2022; 81:1224-1242. [PMID: 35701153 DOI: 10.1136/annrheumdis-2021-221761] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2021] [Accepted: 05/12/2022] [Indexed: 12/24/2022]
Abstract
OBJECTIVES Immune and stromal cell communication is central in the pathogenesis of rheumatoid arthritis (RA) and psoriatic arthritis (PsA), however, the nature of these interactions in the synovial pathology of the two pathotypes can differ. Identifying immune-stromal cell crosstalk at the site of inflammation in RA and PsA is challenging. This study creates the first global transcriptomic analysis of the RA and PsA inflamed joint and investigates immune-stromal cell interactions in the pathogenesis of synovial inflammation. METHODS Single cell transcriptomic profiling of 178 000 synovial tissue cells from five patients with PsA and four patients with RA, importantly, without prior sorting of immune and stromal cells. This approach enabled the transcriptomic analysis of the intact synovial tissue and identification of immune and stromal cell interactions. State of the art data integration and annotation techniques identified and characterised 18 stromal and 14 immune cell clusters. RESULTS Global transcriptomic analysis of synovial cell subsets identifies actively proliferating synovial T cells and indicates that due to differential λ and κ immunoglobulin light chain usage, synovial plasma cells are potentially not derived from the local memory B cell pool. Importantly, we report distinct fibroblast and endothelial cell transcriptomes indicating abundant subpopulations in RA and PsA characterised by differential transcription factor usage. Using receptor-ligand interactions and downstream target characterisation, we identify RA-specific synovial T cell-derived transforming growth factor (TGF)-β and macrophage interleukin (IL)-1β synergy in driving the transcriptional profile of FAPα+THY1+ invasive synovial fibroblasts, expanded in RA compared with PsA. In vitro characterisation of patient with RA synovial fibroblasts showed metabolic switch to glycolysis, increased adhesion intercellular adhesion molecules 1 expression and IL-6 secretion in response to combined TGF-β and IL-1β treatment. Disrupting specific immune and stromal cell interactions offers novel opportunities for targeted therapeutic intervention in RA and PsA.
Collapse
Affiliation(s)
- Achilleas Floudas
- Molecular Rheumatology, Clinical Medicine, Trinity Biomedical Science Institute, Dublin, Ireland
- Eular Centre for Arthritis and Rheumatic Diseases, St Vincent's University Hospital, Univeristy College Dublin, Dublin, Ireland
| | - Conor M Smith
- Translational Immunology, Trinity Biomedical Sciences Institute, Trinity College Dublin, Dublin, Ireland
| | - Orla Tynan
- Molecular Rheumatology, Clinical Medicine, Trinity Biomedical Science Institute, Dublin, Ireland
- Eular Centre for Arthritis and Rheumatic Diseases, St Vincent's University Hospital, Univeristy College Dublin, Dublin, Ireland
| | - Nuno Neto
- Department of Mechanical and Manufacturing Engineering, Trinity College Dublin, Dublin, Ireland
| | - Vinod Krishna
- Immunology, Janssen Research & Development, Spring House, PA, USA
| | - Sarah M Wade
- Molecular Rheumatology, Clinical Medicine, Trinity Biomedical Science Institute, Dublin, Ireland
- Eular Centre for Arthritis and Rheumatic Diseases, St Vincent's University Hospital, Univeristy College Dublin, Dublin, Ireland
| | - Megan Hanlon
- Molecular Rheumatology, Clinical Medicine, Trinity Biomedical Science Institute, Dublin, Ireland
- Eular Centre for Arthritis and Rheumatic Diseases, St Vincent's University Hospital, Univeristy College Dublin, Dublin, Ireland
| | - Clare Cunningham
- Molecular Rheumatology, Clinical Medicine, Trinity Biomedical Science Institute, Dublin, Ireland
- Eular Centre for Arthritis and Rheumatic Diseases, St Vincent's University Hospital, Univeristy College Dublin, Dublin, Ireland
| | - Viviana Marzaioli
- Molecular Rheumatology, Clinical Medicine, Trinity Biomedical Science Institute, Dublin, Ireland
- Eular Centre for Arthritis and Rheumatic Diseases, St Vincent's University Hospital, Univeristy College Dublin, Dublin, Ireland
| | - Mary Canavan
- Molecular Rheumatology, Clinical Medicine, Trinity Biomedical Science Institute, Dublin, Ireland
- Eular Centre for Arthritis and Rheumatic Diseases, St Vincent's University Hospital, Univeristy College Dublin, Dublin, Ireland
| | - Jean M Fletcher
- Translational Immunology, Trinity Biomedical Sciences Institute, Trinity College Dublin, Dublin, Ireland
| | - Ronan H Mullan
- Department of Rheumatology, Tallaght University Hospital, Trinity College Dublin, Dublin, Ireland
| | - Suzanne Cole
- Immunology, Janssen Research & Development, Spring House, PA, USA
| | - Ling-Yang Hao
- Immunology, Janssen Research & Development, Spring House, PA, USA
| | - Michael G Monaghan
- Department of Mechanical and Manufacturing Engineering, Trinity College Dublin, Dublin, Ireland
| | - Sunil Nagpal
- Immunology, Janssen Research & Development, Spring House, PA, USA
| | - Douglas J Veale
- Eular Centre for Arthritis and Rheumatic Diseases, St Vincent's University Hospital, Univeristy College Dublin, Dublin, Ireland
| | - Ursula Fearon
- Molecular Rheumatology, Clinical Medicine, Trinity Biomedical Science Institute, Dublin, Ireland
- Eular Centre for Arthritis and Rheumatic Diseases, St Vincent's University Hospital, Univeristy College Dublin, Dublin, Ireland
| |
Collapse
|
179
|
Kawai K, Vozenilek AE, Kawakami R, Sato Y, Ghosh SKB, Virmani R, Finn AV. Understanding the role of alternative macrophage phenotypes in human atherosclerosis. Expert Rev Cardiovasc Ther 2022; 20:689-705. [PMID: 35942866 DOI: 10.1080/14779072.2022.2111301] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/15/2022]
Abstract
INTRODUCTION Atherosclerosis-based ischemic heart disease is still the primary cause of death throughout the world. Over the past decades there has been no significant changes in the therapeutic approaches to atherosclerosis, which are mainly based on lipid lowering therapies and management of comorbid conditions such as diabetes and hypertension. The involvement of macrophages in atherosclerosis has been recognized for decades. More recently, a more detailed and sophisticated understanding of their various phenotypes and roles in the atherosclerotic process has been recognized. This new data is revealing how specific subtypes of macrophage-induced inflammation may have distinct effects on atherosclerosis progression and may provide new approaches for treatment, based upon targeting of specific macrophage subtypes. AREAS COVERED We will comprehensively review the spectrum of macrophage phenotypes and how they contribute to atherosclerotic plaque development and progression. EXPERT OPINION Various signals derived from atherosclerotic lesions drive macrophages into complex subsets with different gene expression profiles, phenotypes, and functions, not all of which are understood. Macrophage phenotypes include those that enhance, heal, and regress the atherosclerotic lesions though various mechanisms. Targeting of specific macrophage phenotypes may provide a promising and novel approach to prevent atherosclerosis progression.
Collapse
Affiliation(s)
- Kenji Kawai
- Department of Cardiovascular Pathology, CVPath Institute, Gaithersburg, MD, USA
| | - Aimee E Vozenilek
- Department of Cardiovascular Pathology, CVPath Institute, Gaithersburg, MD, USA
| | - Rika Kawakami
- Department of Cardiovascular Pathology, CVPath Institute, Gaithersburg, MD, USA
| | - Yu Sato
- Department of Cardiovascular Pathology, CVPath Institute, Gaithersburg, MD, USA
| | | | - Renu Virmani
- Department of Cardiovascular Pathology, CVPath Institute, Gaithersburg, MD, USA
| | - Aloke V Finn
- Department of Cardiovascular Pathology, CVPath Institute, Gaithersburg, MD, USA.,University of Maryland, School of Medicine, Baltimore, MD, USA
| |
Collapse
|
180
|
Su PYP, Zhang L, He L, Zhao N, Guan Z. The Role of Neuro-Immune Interactions in Chronic Pain: Implications for Clinical Practice. J Pain Res 2022; 15:2223-2248. [PMID: 35957964 PMCID: PMC9359791 DOI: 10.2147/jpr.s246883] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2022] [Accepted: 07/19/2022] [Indexed: 11/23/2022] Open
Abstract
Chronic pain remains a public health problem and contributes to the ongoing opioid epidemic. Current pain management therapies still leave many patients with poorly controlled pain, thus new or improved treatments are desperately needed. One major challenge in pain research is the translation of preclinical findings into effective clinical practice. The local neuroimmune interface plays an important role in the initiation and maintenance of chronic pain and is therefore a promising target for novel therapeutic development. Neurons interface with immune and immunocompetent cells in many distinct microenvironments along the nociceptive circuitry. The local neuroimmune interface can modulate the activity and property of the neurons to affect peripheral and central sensitization. In this review, we highlight a specific subset of many neuroimmune interfaces. In the central nervous system, we examine the interface between neurons and microglia, astrocytes, and T lymphocytes. In the periphery, we profile the interface between neurons in the dorsal root ganglion with T lymphocytes, satellite glial cells, and macrophages. To bridge the gap between preclinical research and clinical practice, we review the preclinical studies of each neuroimmune interface, discuss current clinical treatments in pain medicine that may exert its action at the neuroimmune interface, and highlight opportunities for future clinical research efforts.
Collapse
Affiliation(s)
- Po-Yi Paul Su
- Department of Anesthesia and Perioperative Care, University of California San Francisco, San Francisco, CA, USA
| | - Lingyi Zhang
- Department of Anesthesia and Perioperative Care, University of California San Francisco, San Francisco, CA, USA
- Department of Anesthesiology, the First Affiliated Hospital, Sun Yat-Sen University, Guangzhou, People’s Republic of China
| | - Liangliang He
- Department of Anesthesia and Perioperative Care, University of California San Francisco, San Francisco, CA, USA
- Department of Pain Management, Xuanwu Hospital, Capital Medical University, Beijing, People’s Republic of China
| | - Na Zhao
- Department of Anesthesia and Perioperative Care, University of California San Francisco, San Francisco, CA, USA
| | - Zhonghui Guan
- Department of Anesthesia and Perioperative Care, University of California San Francisco, San Francisco, CA, USA
| |
Collapse
|
181
|
Hu N, Zhu L, Zhang L, Wang J, Wang Y, Luo J, He L, Hao Z, Zhang L. Immunomodulatory effect and safety of TNF-α RNAi mediated by oral yeast microcapsules in rheumatoid arthritis therapy. Mater Today Bio 2022; 16:100384. [PMID: 35991628 PMCID: PMC9386491 DOI: 10.1016/j.mtbio.2022.100384] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2022] [Revised: 07/16/2022] [Accepted: 07/23/2022] [Indexed: 11/15/2022] Open
Abstract
Rheumatoid arthritis (RA) is a chronic autoimmune disease that requires long-term treatment and monitoring. Inhibition of inflammatory gene expression by gene therapy is a significant breakthrough in RA treatment, but the lack of a safe and effective gene delivery system hinders its application. Since oral administration can significantly reduce wound infection caused by parenteral administration, it also has the advantages of high patient compliance and convenience. Therefore, oral administration may be the best option for the treatment of this chronic disease. In this study, we developed a novel oral drug system by delivering tumor necrosis factor-α (TNF-α) short hairpin RNA (shRNA) mediated by non-pathogenic yeast to evaluate its regulation of systemic immune inflammation and safety in RA. Non-pathogenic yeast can resist the destruction of the gastrointestinal acid-base environment and can be recognized by the intestinal macrophages and act on systemic inflammatory lesions. Oral administration of yeast-mediated TNF-α shRNA significantly reduced the expression of TNF-α predominant pro-inflammatory factors in intestinal macrophages and joint synovium, and up-regulated the expression of anti-inflammatory cytokine IL-10 and M2 macrophages, systematically regulating the inflammatory response. This yeast-mediated oral gene delivery system can not only significantly inhibit knee joint synovial inflammation, but also has no toxic effects on peripheral blood and major organs. Therefore, yeast-mediated oral delivery of TNF-α shRNA may be used as a novel gene therapy strategy to treat RA through immunomodulating the mononuclear phagocyte system from the intestine to the joint synovium, and ultimately regulating systemic and local immune inflammation, providing new ideas for the clinical treatment of RA.
Collapse
Affiliation(s)
- Nan Hu
- Department of Rheumatology, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710061, China
| | - Li Zhu
- Department of Rheumatology, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710061, China
| | - Li Zhang
- Xi'an Fifth Hospital, Shaanxi Provincial Hospital of Integrated Traditional Chinese and Western Medicine, Xi'an, 710082, China
| | - Jing Wang
- Department of Rheumatology, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710061, China
| | - Yanhua Wang
- Department of Rheumatology, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710061, China
| | - Jing Luo
- Department of Rheumatology, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710061, China
| | - Lan He
- Department of Rheumatology, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710061, China
| | - Zhiming Hao
- Department of Rheumatology, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710061, China
| | - Long Zhang
- Department of Respiratory and Critical Care Medicine, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710004, China
- Corresponding author.
| |
Collapse
|
182
|
Uresti-Rivera EE, García-Hernández MH. AIM2-inflammasome role in systemic lupus erythematous and rheumatoid arthritis. Autoimmunity 2022; 55:443-454. [PMID: 35880661 DOI: 10.1080/08916934.2022.2103802] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/02/2022]
Abstract
The inflammasome AIM2 regulates multiple aspects of innate immune functions and serves as a critical mediator of inflammatory responses. AIM2 inflammasome activation leads to the production of pro-inflammatory cytokines, IL-1β and IL-18 and participates triggering a pyroptosis response needed to counteract excessive cell proliferation. In addition, AIM2 expression and activation is wide regulated since alteration in its activity may derived in pathological consequences. Consequently, deregulated AIM2 activation contributes to the pathogenic processes of various inflammatory diseases. In this review, we will discuss the activation and function of AIM2 inflammasome, as well as its contribution in rheumatoid arthritis and systemic lupus erythematous pathology. Finally, we highlight the participation of the AIM2-inflammasome at the level of joint in rheumatoid arthritis and at kidney in systemic lupus erythematous. The development of therapeutic strategies based on modulation of AIM2-inflammasome activity should have a tissue-specific focus.
Collapse
Affiliation(s)
- E E Uresti-Rivera
- Research Center for Health Sciences and Biomedicine, UASLP, San Luis Potosi, Mexico.,Laboratory of Immunology and Cellular and Molecular Biology, Faculty of Chemical Sciences, Autonomous University of San Luis Potosí, UASLP, San Luis Potosí, Mexico
| | - M H García-Hernández
- Instituto Mexicano del Seguro Social, IMSS, Unidad de Investigación Biomédica, Delegación Zacatecas, Zacatecas, México
| |
Collapse
|
183
|
Feng C, Pan L, Qin X, Li D, Chen T, Lin Z, Li G, Wang Q. Inflammation-homing "living drug depot" for efficient arthritis treatment. Acta Biomater 2022; 150:324-336. [PMID: 35840107 DOI: 10.1016/j.actbio.2022.07.013] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2022] [Revised: 06/29/2022] [Accepted: 07/06/2022] [Indexed: 11/28/2022]
Abstract
Delivering therapeutic agents efficiently to inflamed joints remains an intractable problem in rheumatoid arthritis (RA) treatment due to the complicated physiological barriers. Circulating monocytes could selectively migrate to inflamed sites and differentiate into resident macrophages to aggravate RA. Therefore, a drug carrier that can be specifically internalized by circulating monocytes and switch monocytes into anti-inflammatory phenotype when reaching inflamed sites, might bypass the in vivo physiological barriers and achieve efficient RA therapy. Herein, we design a dextran sulfate (DS) functionalized nanoparticle (ZDNP) to selectively deliver anti-inflammatory agent dexamethasone (Dex) to circulating monocytes via the scavenger receptors on monocytes. Monocytes engulfing drug-loaded ZDNP could subsequently home to arthritic joints and act as a "living drug depot" to combat RA. Results revealed that ZDNP could be preferentially internalized by circulating monocytes when intravenously administrated in vivo. In a rat arthritic model, we found that circulating monocytes remarkably facilitated drug distribution and retention in inflamed joints. Moreover, monocytes engulfing drug-loaded nanoparticles exhibited favorable anti-inflammatory ability and M2-biased differentiation. Our work offers a facile approach to achieve site-directed anti-inflammatory therapy by taking advantage of the inflammation-homing ability of circulating monocytes. STATEMENT OF SIGNIFICANCE: Circulating monocytes can migrate to inflamed sites and then differentiate into macrophages to aggravate arthritis. Therefore, a drug carrier that can be specifically internalized by circulating monocytes and switch monocytes into anti-inflammatory phenotype when reaching inflamed sites may achieve efficient arthritis therapy. Here, we designed a monocyte-targeting nanoparticle (ZDNP) to selectively deliver anti-inflammatory Dex to circulating monocytes. When injected intravenously, ZDNP was effectively internalized by circulating monocytes via a scavenger receptor and subsequently was transported to arthritic joints, where monocytes engulfing the drug-loaded nanoparticles could switch to an anti-inflammatory phenotype to inhibit arthritis progress. We provide detailed evidence about the in vivo fate of ZDNP and unravel how monocytes act as a "living drug depot" to achieve site-directed arthritis therapy.
Collapse
Affiliation(s)
- Chenglan Feng
- Key Laboratory of Advanced Technologies of Materials, Ministry of Education and School of Materials Science and Engineering, Southwest Jiaotong University, Chengdu 610031, China
| | - Lihua Pan
- Key Laboratory of Advanced Technologies of Materials, Ministry of Education and School of Materials Science and Engineering, Southwest Jiaotong University, Chengdu 610031, China
| | - Xianyan Qin
- Key Laboratory of Advanced Technologies of Materials, Ministry of Education and School of Materials Science and Engineering, Southwest Jiaotong University, Chengdu 610031, China
| | - Daming Li
- Key Laboratory of Advanced Technologies of Materials, Ministry of Education and School of Materials Science and Engineering, Southwest Jiaotong University, Chengdu 610031, China
| | - Tao Chen
- Key Laboratory of Advanced Technologies of Materials, Ministry of Education and School of Materials Science and Engineering, Southwest Jiaotong University, Chengdu 610031, China
| | - Zhicong Lin
- Key Laboratory of Advanced Technologies of Materials, Ministry of Education and School of Materials Science and Engineering, Southwest Jiaotong University, Chengdu 610031, China
| | - Guojiao Li
- Key Laboratory of Advanced Technologies of Materials, Ministry of Education and School of Materials Science and Engineering, Southwest Jiaotong University, Chengdu 610031, China
| | - Qin Wang
- Key Laboratory of Advanced Technologies of Materials, Ministry of Education and School of Materials Science and Engineering, Southwest Jiaotong University, Chengdu 610031, China.
| |
Collapse
|
184
|
Peckham H, Webb K, Rosser EC, Butler G, Ciurtin C. Gender-Diverse Inclusion in Immunological Research: Benefits to Science and Health. Front Med (Lausanne) 2022; 9:909789. [PMID: 35911383 PMCID: PMC9329564 DOI: 10.3389/fmed.2022.909789] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2022] [Accepted: 06/24/2022] [Indexed: 01/26/2023] Open
Abstract
The differences between male and female immune systems are an under-researched field, ripe for discovery. This is evidenced by the stark sex biases seen in autoimmunity and infectious disease. Both the sex hormones (oestrogen and testosterone), as well as the sex chromosomes have been demonstrated to impact immune responses, in multiple ways. Historical shortcomings in reporting basic and clinical scientific findings in a sex-disaggregated manner have led not only to limited discovery of disease aetiology, but to potential inaccuracies in the estimation of the effects of diseases or interventions on females and gender-diverse groups. Here we propose not only that research subjects should include both cis-gender men and cis-gender women, but also transgender and gender-diverse people alongside them. The known interaction between the hormonal milieu and the sex chromosomes is inseparable in cis-gender human research, without the confounders of puberty and age. By inclusion of those pursuing hormonal affirmation of their gender identity- the individual and interactive investigation of hormones and chromosomes is permitted. Not only does this allow for a fine-tuned dissection of these individual effects, but it allows for discovery that is both pertinent and relevant to a far wider portion of the population. There is an unmet need for detailed treatment follow-up of the transgender community- little is known of the potential benefits and risks of hormonal supplementation on the immune system, nor indeed on many other health and disease outcomes. Our research team has pioneered the inclusion of gender-diverse persons in our basic research in adolescent autoimmune rheumatic diseases. We review here the many avenues that remain unexplored, and suggest ways in which other groups and teams can broaden their horizons and invest in a future for medicine that is both fruitful and inclusive.
Collapse
Affiliation(s)
- Hannah Peckham
- Centre for Adolescent Rheumatology Versus Arthritis at University College London (UCL), University College London Hospital (UCLH), Great Ormond Street Hospital (GOSH), London, United Kingdom
- Division of Medicine, Centre for Rheumatology Research, University College London (UCL), London, United Kingdom
| | - Kate Webb
- Department of Paediatric Rheumatology, School of Child and Adolescent Health, Red Cross War Memorial Children’s Hospital, University of Cape Town, Cape Town, South Africa
- Crick African Network, The Francis Crick Institute, London, United Kingdom
| | - Elizabeth C. Rosser
- Centre for Adolescent Rheumatology Versus Arthritis at University College London (UCL), University College London Hospital (UCLH), Great Ormond Street Hospital (GOSH), London, United Kingdom
- Division of Medicine, Centre for Rheumatology Research, University College London (UCL), London, United Kingdom
| | - Gary Butler
- Department of Paediatric and Adolescent Endocrinology, University College London Hospital (UCLH) and Great Ormond Street Institute of Child Health, University College London, London, United Kingdom
- Gender Identity Development Service (GIDS), Tavistock and Portman NHS Foundation Trust, London, United Kingdom
| | - Coziana Ciurtin
- Centre for Adolescent Rheumatology Versus Arthritis at University College London (UCL), University College London Hospital (UCLH), Great Ormond Street Hospital (GOSH), London, United Kingdom
- Division of Medicine, Centre for Rheumatology Research, University College London (UCL), London, United Kingdom
| |
Collapse
|
185
|
Cheng Q, Chen M, Liu M, Chen X, Zhu L, Xu J, Xue J, Wu H, Du Y. Semaphorin 5A suppresses ferroptosis through activation of PI3K-AKT-mTOR signaling in rheumatoid arthritis. Cell Death Dis 2022; 13:608. [PMID: 35835748 PMCID: PMC9283415 DOI: 10.1038/s41419-022-05065-4] [Citation(s) in RCA: 65] [Impact Index Per Article: 21.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2022] [Revised: 07/03/2022] [Accepted: 07/04/2022] [Indexed: 01/21/2023]
Abstract
Abnormal activation of synovial fibroblasts (SFs) plays an important role in rheumatoid arthritis (RA), the mechanism of which remains unknown. The purpose of our study is to comprehensively and systematically explore the mechanism for Semaphorin 5A-mediated abnormal SF activation in RA. Here, we found that Semaphorin 5A levels were significantly higher in synovial fluid and synovial tissue from RA patients compared with osteoarthritis patients. We further found that the mRNA level and protein abundance of Plexin-A1 was elevated in RA SFs compared with OA SFs, while Plexin-B3 expression showed no significant difference. The increased Semaphorin 5A in RA synovial fluid was mainly derived from CD68+ synovial macrophages, and the elevation led to increased binding between Semaphorin 5A and its receptors, thereby promoting cytokine secretion, proliferation, and migration, and decreasing apoptosis. Moreover, the effect of Semaphorin 5A on enhancing activation (cytokine secretion, cell proliferation and migration) and reducing apoptosis of SFs was significantly abolished after knockdown of Plexin-A1 and Plexin-B3 by small interfering RNA. Transcriptome sequencing and protein array detection revealed that Semaphorin 5A activated the PI3K/AKT/mTOR signaling pathway and inhibited ferroptosis. Morphologically, transmission electron microscopy results showed that Semaphorin 5A could significantly eliminate the mitochondrial diminution, membrane density increased and crest ruptured of SFs induced by ferroptosis inducer RSL3. Mechanistically, Semaphorin 5A enhanced GPX4 expression and SREBP1/SCD-1 signaling by activating the PI3K/AKT/mTOR signaling pathway, thus suppressing ferroptosis of RA SFs. In conclusion, our study provided the first evidence that elevated Semaphorin 5A in RA synovial fluid promotes SF activation by suppressing ferroptosis through the PI3K/AKT/mTOR signaling pathway.
Collapse
Affiliation(s)
- Qi Cheng
- Department of Rheumatology, The Second Affiliated Hospital of Zhejiang University School of Medicine, 88 Jiefang Road, 310009, Hangzhou, China.,Department of Clinic Medicine, The Second Affiliated Hospital of Zhejiang University School of Medicine, 88 Jiefang Road, 310009, Hangzhou, China
| | - Mo Chen
- Department of Rheumatology, The Second Affiliated Hospital of Zhejiang University School of Medicine, 88 Jiefang Road, 310009, Hangzhou, China
| | - Mengdan Liu
- Department of Rheumatology, The Second Affiliated Hospital of Zhejiang University School of Medicine, 88 Jiefang Road, 310009, Hangzhou, China
| | - Xin Chen
- Department of Rheumatology, The Second Affiliated Hospital of Zhejiang University School of Medicine, 88 Jiefang Road, 310009, Hangzhou, China.,Department of Clinic Medicine, The Second Affiliated Hospital of Zhejiang University School of Medicine, 88 Jiefang Road, 310009, Hangzhou, China
| | - Lingjiang Zhu
- Department of Rheumatology, The Second Affiliated Hospital of Zhejiang University School of Medicine, 88 Jiefang Road, 310009, Hangzhou, China
| | - Jieying Xu
- Department of Rheumatology, The Second Affiliated Hospital of Zhejiang University School of Medicine, 88 Jiefang Road, 310009, Hangzhou, China.,Department of Neurology, Linping District Hospital of Integrated Traditional Chinese and Western Medicine, 311199, Hangzhou, Zhejiang, China
| | - Jing Xue
- Department of Rheumatology, The Second Affiliated Hospital of Zhejiang University School of Medicine, 88 Jiefang Road, 310009, Hangzhou, China
| | - Huaxiang Wu
- Department of Rheumatology, The Second Affiliated Hospital of Zhejiang University School of Medicine, 88 Jiefang Road, 310009, Hangzhou, China.
| | - Yan Du
- Department of Rheumatology, The Second Affiliated Hospital of Zhejiang University School of Medicine, 88 Jiefang Road, 310009, Hangzhou, China.
| |
Collapse
|
186
|
Chen MW, Lu QJ, Chen YJ, Hou YK, Zou YM, Zhou Q, Zhang WH, Yuan LX, Chen JX. NIR-PTT/ROS-Scavenging/Oxygen-Enriched Synergetic Therapy for Rheumatoid Arthritis by a pH-Responsive Hybrid CeO 2-ZIF-8 Coated with Polydopamine. ACS Biomater Sci Eng 2022; 8:3361-3376. [PMID: 35819069 DOI: 10.1021/acsbiomaterials.2c00592] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Rheumatoid arthritis (RA) is an inflammatory type of arthritis that causes joint pain and damage. The inflammatory cell infiltration (e.g., M1 macrophages), the poor O2 supply at the joint, and the excess reactive oxygen species (ROS)-induced oxidative injury are the main causes of RA. We herein report a polydopamine (PDA)-coated CeO2-dopped zeolitic imidazolate framework-8 (ZIF-8) nanocomposite CeO2-ZIF-8@PDA (denoted as CZP) that can synergistically treat RA. Under near-infrared (NIR) light irradiation, PDA efficiently scavenges ROS and results in an increased temperature in the inflamed area because of its good light-to-heat conversion efficiency. The rise of temperature serves to obliterate hyper-proliferative inflammatory cells accumulated in the diseased area while vastly promoting the collapse of the acidic-responsive skeleton of ZIF-8 to release the encapsulated CeO2. The released CeO2 exerts its catalase-like activity to relieve hypoxia by generating oxygen via the decomposition of H2O2 highly expressed in the inflammatory sites. Thus, the constructed CZP composite can treat RA through NIR-photothermal/ROS-scavenging/oxygen-enriched combinative therapy and show good regression of pro-inflammatory cytokines and hypoxia-inducible factor-1α (HIF-1α) in vitro and promising therapeutic effect on RA in rat models. The multimodal nano-platform reported herein is expected to shed light on the design of synergistic therapeutic nanomedicine for effective RA therapy.
Collapse
Affiliation(s)
- Ming-Wa Chen
- NMPA Key Laboratory for Research and Evaluation of Drug Metabolism, Guangdong Provincial Key Laboratory of New Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou 510515, People's Republic of China
| | - Qi-Jin Lu
- School of Chinese Medicine, Southern Medical University, Guangzhou 510515, People's Republic of China
| | - Yong-Jian Chen
- NMPA Key Laboratory for Research and Evaluation of Drug Metabolism, Guangdong Provincial Key Laboratory of New Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou 510515, People's Republic of China
| | - Ying-Ke Hou
- Department of Medical Imaging, Third Affiliated Hospital of Southern Medical University (Academy of Orthopedics Guangdong Province), Southern Medical University, Guangzhou, Guangdong 510630, People's Republic of China
| | - Yi-Ming Zou
- NMPA Key Laboratory for Research and Evaluation of Drug Metabolism, Guangdong Provincial Key Laboratory of New Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou 510515, People's Republic of China
| | - Quan Zhou
- Department of Medical Imaging, Third Affiliated Hospital of Southern Medical University (Academy of Orthopedics Guangdong Province), Southern Medical University, Guangzhou, Guangdong 510630, People's Republic of China
| | - Wen-Hua Zhang
- College of Chemistry, Chemical Engineering and Materials Science, Soochow University, Suzhou 215123, People's Republic of China
| | - Li-Xia Yuan
- School of Chinese Medicine, Southern Medical University, Guangzhou 510515, People's Republic of China
| | - Jin-Xiang Chen
- NMPA Key Laboratory for Research and Evaluation of Drug Metabolism, Guangdong Provincial Key Laboratory of New Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou 510515, People's Republic of China
| |
Collapse
|
187
|
A novel mechanism for macrophage pyroptosis in rheumatoid arthritis induced by Pol β deficiency. Cell Death Dis 2022; 13:583. [PMID: 35794098 PMCID: PMC9259649 DOI: 10.1038/s41419-022-05047-6] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2022] [Revised: 06/18/2022] [Accepted: 06/27/2022] [Indexed: 01/21/2023]
Abstract
Rheumatoid arthritis (RA) is a chronic and inflammatory autoimmune disease. Macrophage pyroptosis, a proinflammatory form of cell death, is critically important in RA; however, the detailed mechanism underlying pyroptosis induction is not yet well understood. Here, we report that DNA polymerase β (Pol β), a key enzyme in base excision repair, plays a pivotal role in RA pathogenesis. Our data shows that Pol β expression is significantly decreased in peripheral blood mononuclear cells (PBMCs) from active RA patients and collagen-induced arthritis (CIA) mice, and Pol β deficiency increases the incidence of RA, macrophage infiltration, and bone destruction in CIA mouse models. In vitro, experiments showed that Pol β deficiency exacerbated macrophage pyroptosis induced by LPS plus ATP, while overexpression of Pol β inhibited macrophage pyroptosis. Further characterization revealed that Pol β knockout resulted in DNA damage accumulation and cytosolic dsDNA leakage, which activated the cGAS-STING-NF-κB signaling pathway and upregulated the expression of NLRP3, IL-1 β, and IL-18. In conclusion, our findings clarify the influence of Pol β on the development of RA and provide a detailed explanation for the STING-NF-κB pathway to induce macrophage pyroptosis.
Collapse
|
188
|
Ye Q, Luo F, Yan T. Transcription factor KLF4 regulated STAT1 to promote M1 polarization of macrophages in rheumatoid arthritis. Aging (Albany NY) 2022; 14:5669-5680. [PMID: 35748767 PMCID: PMC9365561 DOI: 10.18632/aging.204128] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2022] [Accepted: 06/04/2022] [Indexed: 11/28/2022]
Abstract
This study aimed to reveal the mechanism of transcription factor Kruppel-like factor 4 (KLF4) in regulating M1 polarization of macrophages in rheumatoid arthritis (RA) in order to induce inflammatory response. The results suggested that KLF4 overexpression promoted the M1 polarization of RAW 264.7 cells, increased STAT1 expression and up-regulated the phosphorylation level. After KLF4 silencing, the M1 polarization level was down-regulated. Besides, the induced M1 macrophages were co-cultured with articular chondrocytes. KLF4 overexpression further aggravated chondrocyte injury, increased the cell apoptosis rate and activated the inflammatory injury. However, pretreatment with STAT1 inhibitor Cerulomycin resisted the effect of KLF4, and significantly suppressed STAT1 expression and M1 polarization of cells. KLF4 overexpression aggravated synovial tissue injury in mouse joints, up-regulated the expression of inflammatory factors, and increased the levels of CD86 and STAT1. It was discovered that transcription factor KLF4 promoted the transcription of STAT1 to regulate the M1 polarization of macrophages, thus aggravating the progression of RA and inflammatory response.
Collapse
Affiliation(s)
- Qiao Ye
- Department of Rheumatology and Immunology, The Second Affiliated Hospital of Jiaxing University, Jiaxing 314001, Zhejiang, China
| | - Fang Luo
- Department of Rheumatology and Immunology, The Second Affiliated Hospital of Jiaxing University, Jiaxing 314001, Zhejiang, China
| | - Tingting Yan
- Department of Rheumatology and Immunology, The Second Affiliated Hospital of Jiaxing University, Jiaxing 314001, Zhejiang, China
| |
Collapse
|
189
|
Cutolo M, Campitiello R, Gotelli E, Soldano S. The Role of M1/M2 Macrophage Polarization in Rheumatoid Arthritis Synovitis. Front Immunol 2022; 13:867260. [PMID: 35663975 PMCID: PMC9161083 DOI: 10.3389/fimmu.2022.867260] [Citation(s) in RCA: 289] [Impact Index Per Article: 96.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2022] [Accepted: 03/24/2022] [Indexed: 12/27/2022] Open
Abstract
Innate and adaptive immunity represent a harmonic counterbalanced system involved in the induction, progression, and possibly resolution of the inflammatory reaction that characterize autoimmune rheumatic diseases (ARDs), including rheumatoid arthritis (RA). Although the immunopathophysiological mechanisms of the ARDs are not fully clarified, they are often associated with an inappropriate macrophage/T-cell interaction, where classical (M1) or alternative (M2) macrophage activation may influence the occurrence of T-helper (Th)1 or Th2 responses. In RA patients, M1/Th1 activation occurs in an inflammatory environment dominated by Toll-like receptor (TLR) and interferon (IFN) signaling, and it promotes a massive production of pro-inflammatory cytokines [i.e., tumor necrosis factor-α (TNFα), interleukin (IL)-1, IL-12, IL-18, and IFNγ], chemotactic factors, and matrix metalloproteinases resulting in osteoclastogenesis, erosion, and progressive joint destruction. On the other hand, the activation of M2/Th2 response determines the release of growth factors and cytokines [i.e., IL-4, IL-10, IL-13, and transforming growth factor (TGF)-β] involved in the anti-inflammatory process leading to the clinical remission of RA. Several subtypes of macrophages have been described. Five polarization states from M1 to M2 have been confirmed in in vitro studies analyzing morphological characteristics, gene expression of phenotype markers (CD80, CD86, TLR2, TLR4, or CD206, CD204, CD163, MerTK), and functional aspect, including the production of reactive oxygen species (ROS). An M1 and M2 macrophage imbalance may induce pathological consequences and contribute to several diseases, such as asthma or osteoclastogenesis in RA patients. In addition, the macrophage dynamic polarization from M1 to M2 includes the presence of intermediate polarity stages distinguished by the expression of specific surface markers and the production/release of distinct molecules (i.e., nitric oxide, cytokines), which characterize their morphological and functional state. This suggests a “continuum” of macrophage activation states playing an important role during inflammation and its resolution. This review discusses the importance of the delicate M1/M2 imbalance in the different phases of the inflammatory process together with the identification of specific pathways, cytokines, and chemokines involved, and its clinical outcomes in RA. The analysis of these aspects could shed a light on the abnormal inflammatory activation, leading to novel therapeutical approaches which may contribute to restore the M1/M2 balance.
Collapse
Affiliation(s)
- Maurizio Cutolo
- Laboratory of Experimental Rheumatology and Academic Division of Clinical Rheumatology, Department of Internal Medicine and Specialties (DIMI), University of Genova, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) San Martino Polyclinic Hospital, Genoa, Italy
| | - Rosanna Campitiello
- Laboratory of Experimental Rheumatology and Academic Division of Clinical Rheumatology, Department of Internal Medicine and Specialties (DIMI), University of Genova, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) San Martino Polyclinic Hospital, Genoa, Italy
| | - Emanuele Gotelli
- Laboratory of Experimental Rheumatology and Academic Division of Clinical Rheumatology, Department of Internal Medicine and Specialties (DIMI), University of Genova, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) San Martino Polyclinic Hospital, Genoa, Italy
| | - Stefano Soldano
- Laboratory of Experimental Rheumatology and Academic Division of Clinical Rheumatology, Department of Internal Medicine and Specialties (DIMI), University of Genova, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) San Martino Polyclinic Hospital, Genoa, Italy
| |
Collapse
|
190
|
Yi O, Lin Y, Hu M, Hu S, Su Z, Liao J, Liu B, Liu L, Cai X. Lactate metabolism in rheumatoid arthritis: Pathogenic mechanisms and therapeutic intervention with natural compounds. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2022; 100:154048. [PMID: 35316725 DOI: 10.1016/j.phymed.2022.154048] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/17/2021] [Revised: 02/26/2022] [Accepted: 03/11/2022] [Indexed: 06/14/2023]
Abstract
BACKGROUND Rheumatoid arthritis (RA) is a common chronic and systemic autoimmune disease characterized by persistent inflammation and hyperplasia of the synovial membrane, the degradation of cartilage, and the erosion of bones in diarthrodial joints. The inflamed joints of patients with RA have been recognized to be a site of hypoxic microenvironment which results in an imbalance of lactate metabolism and the accumulation of lactate. Lactate is no longer considered solely a metabolic waste product of glycolysis, but also a combustion aid in the progression of RA from the early stages of inflammation to the late stages of bone destruction. PURPOSE To review the pathogenic mechanisms of lactate metabolism in RA and investigate the potential of natural compounds for treating RA linked to the regulation of imbalance in lactate metabolism. METHODS Research advances in our understanding of lactate metabolism in the pathogenesis of RA and novel pharmacological approaches of natural compounds by targeting lactate metabolic signaling were comprehensively reviewed and deeply discussed. RESULTS Lactate produced by RA synovial fibroblasts (RASFs) acts on targeted cells such as T cells, macrophages, dendritic cells and osteoclasts, and affects their differentiation, activation and function to accelerate the development of RA. Many natural compounds show therapeutic potential for RA by regulating glycolytic rate-limiting enzymes to limit lactate production, and affecting monocarboxylate transporter and acetyl-CoA carboxylase to inhibit lactate transport and conversion. CONCLUSION Regulation of imbalance in lactate metabolism offers novel therapeutic approaches for RA, and natural compounds capable of targeting lactate metabolic signaling constitute potential candidates for development of drugs RA.
Collapse
Affiliation(s)
- Ouyang Yi
- Institute of Innovation and Applied Research in Chinese Medicine and Department of Rheumatology of The First Hospital, Hunan University of Chinese Medicine, Changsha, Hunan, 410208, China
| | - Ye Lin
- Institute of Innovation and Applied Research in Chinese Medicine and Department of Rheumatology of The First Hospital, Hunan University of Chinese Medicine, Changsha, Hunan, 410208, China
| | - Mingyue Hu
- Institute of Innovation and Applied Research in Chinese Medicine and Department of Rheumatology of The First Hospital, Hunan University of Chinese Medicine, Changsha, Hunan, 410208, China
| | - Shengtao Hu
- Institute of Innovation and Applied Research in Chinese Medicine and Department of Rheumatology of The First Hospital, Hunan University of Chinese Medicine, Changsha, Hunan, 410208, China
| | - Zhaoli Su
- Institute of Innovation and Applied Research in Chinese Medicine and Department of Rheumatology of The First Hospital, Hunan University of Chinese Medicine, Changsha, Hunan, 410208, China
| | - Jin Liao
- Institute of Innovation and Applied Research in Chinese Medicine and Department of Rheumatology of The First Hospital, Hunan University of Chinese Medicine, Changsha, Hunan, 410208, China
| | - Bin Liu
- College of Biology, Hunan University, Changsha, Hunan 410082, China
| | - Liang Liu
- State Key Laboratory of Quality Research in Chinese Medicine, Macau University of Science and Technology, Macau, 030027, China
| | - Xiong Cai
- Institute of Innovation and Applied Research in Chinese Medicine and Department of Rheumatology of The First Hospital, Hunan University of Chinese Medicine, Changsha, Hunan, 410208, China
| |
Collapse
|
191
|
Hu Z, Zhao TV, Huang T, Ohtsuki S, Jin K, Goronzy IN, Wu B, Abdel MP, Bettencourt JW, Berry GJ, Goronzy JJ, Weyand CM. The transcription factor RFX5 coordinates antigen-presenting function and resistance to nutrient stress in synovial macrophages. Nat Metab 2022; 4:759-774. [PMID: 35739396 PMCID: PMC9280866 DOI: 10.1038/s42255-022-00585-x] [Citation(s) in RCA: 44] [Impact Index Per Article: 14.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/04/2022] [Accepted: 05/16/2022] [Indexed: 11/08/2022]
Abstract
Tissue macrophages (Mϕ) are essential effector cells in rheumatoid arthritis (RA), contributing to autoimmune tissue inflammation through diverse effector functions. Their arthritogenic potential depends on their proficiency to survive in the glucose-depleted environment of the inflamed joint. Here, we identify a mechanism that links metabolic adaptation to nutrient stress with the efficacy of tissue Mϕ to activate adaptive immunity by presenting antigen to tissue-invading T cells. Specifically, Mϕ populating the rheumatoid joint produce and respond to the small cytokine CCL18, which protects against cell death induced by glucose withdrawal. Mechanistically, CCL18 induces the transcription factor RFX5 that selectively upregulates glutamate dehydrogenase 1 (GLUD1), thus enabling glutamate utilization to support energy production. In parallel, RFX5 enhances surface expression of HLA-DR molecules, promoting Mϕ-dependent expansion of antigen-specific T cells. These data place CCL18 at the top of a RFX5-GLUD1 survival pathway and couple adaptability to nutrient conditions in the tissue environment to antigen-presenting function in autoimmune tissue inflammation.
Collapse
Affiliation(s)
- Zhaolan Hu
- Department of Medicine, Mayo Clinic College of Medicine and Science, Rochester, MN, USA
| | - Tuantuan V Zhao
- Department of Medicine, Mayo Clinic College of Medicine and Science, Rochester, MN, USA
| | - Tao Huang
- Department of Medicine, Mayo Clinic College of Medicine and Science, Rochester, MN, USA
| | - Shozo Ohtsuki
- Department of Medicine, Mayo Clinic College of Medicine and Science, Rochester, MN, USA
| | - Ke Jin
- Department of Medicine, Mayo Clinic College of Medicine and Science, Rochester, MN, USA
| | - Isabel N Goronzy
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, CA, USA
| | - Bowen Wu
- Department of Medicine, Mayo Clinic College of Medicine and Science, Rochester, MN, USA
| | - Matthew P Abdel
- Department of Orthopedic Surgery, Mayo Clinic College of Medicine and Science, Rochester, MN, USA
| | - Jacob W Bettencourt
- Department of Orthopedic Surgery, Mayo Clinic College of Medicine and Science, Rochester, MN, USA
| | - Gerald J Berry
- Department of Pathology, Stanford University School of Medicine, Stanford, CA, USA
| | - Jörg J Goronzy
- Department of Medicine, Mayo Clinic College of Medicine and Science, Rochester, MN, USA
- Department of Medicine, Stanford University School of Medicine, Stanford, CA, USA
| | - Cornelia M Weyand
- Department of Medicine, Mayo Clinic College of Medicine and Science, Rochester, MN, USA.
- Department of Medicine, Stanford University School of Medicine, Stanford, CA, USA.
| |
Collapse
|
192
|
Saraiva AL, Vieira TN, Notário AFO, Luiz JPM, Silva CR, Goulart LR, Dantas NO, Silva ACA, Espindola FS. CdSe magic-sized quantum dots attenuate reactive oxygen species generated by neutrophils and macrophages with implications in experimental arthritis. NANOMEDICINE : NANOTECHNOLOGY, BIOLOGY, AND MEDICINE 2022; 42:102539. [PMID: 35183761 DOI: 10.1016/j.nano.2022.102539] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/03/2021] [Revised: 01/30/2022] [Accepted: 02/06/2022] [Indexed: 06/14/2023]
Abstract
The biological applicability of nanomaterials has been limited due to cytotoxicity. Studies have described the effects of nanomaterials on different tissues and cell types, but their actions on immune cells are less elucidated. This study describes unprecedented in vitro and in vivo antioxidant activities of cadmium selenide magic-sized quantum dots (CdSe MSQDs) with implications on rheumatoid arthritis. While the generation of ROS induced by nanomaterials is linked to cytotoxicity, we found that CdSe MSQDs reduced ROS production by neutrophils and macrophages following opsonized-zymosan stimuli, and we did not find cytotoxic effects. Interestingly, inherent antioxidant properties of CdSe MSQDs were confirmed through DPPH, FRAP, and ORAC assays. Furthermore, CdSe MSQDs reduced ROS levels generated by infiltrating leukocytes into joints in experimental model of rheumatoid arthritis. Briefly, we describe a novel application of CdSe MSQDs in modulating the inflammatory response in experimental rheumatoid arthritis through an unexpected antioxidant activity.
Collapse
Affiliation(s)
- André Lopes Saraiva
- Instituto de Biotecnologia, Universidade Federal de Uberlândia, Uberlândia, MG, Brazil
| | - Thiago Neves Vieira
- Instituto de Biotecnologia, Universidade Federal de Uberlândia, Uberlândia, MG, Brazil
| | | | - João Paulo Mesquita Luiz
- Departamento de Farmacologia, Faculdade de Medicina de Ribeirão Preto, University of São Paulo, Ribeirão Preto, SP, Brazil
| | - Cássia Regina Silva
- Instituto de Biotecnologia, Universidade Federal de Uberlândia, Uberlândia, MG, Brazil
| | - Luiz Ricardo Goulart
- Instituto de Biotecnologia, Universidade Federal de Uberlândia, Uberlândia, MG, Brazil
| | | | | | | |
Collapse
|
193
|
Qin W, Rong X, Yu C, Jia P, Yang J, Zhou G. Knockout of SLAMF8 attenuates collagen-induced rheumatoid arthritis in mice through inhibiting TLR4/NF-κB signaling pathway. Int Immunopharmacol 2022; 107:108644. [DOI: 10.1016/j.intimp.2022.108644] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2021] [Revised: 02/16/2022] [Accepted: 02/18/2022] [Indexed: 12/16/2022]
|
194
|
Mahmoudi Z, Karamali N, Roghani SA, Assar S, Pournazari M, Soufivand P, Salari F, Rezaiemanesh A. Efficacy of DMARDs and methylprednisolone treatment on the gene expression levels of HSPA5, MMD, and non-coding RNAs MALAT1, H19, miR-199a-5p, and miR-1-3p, in patients with rheumatoid arthritis. Int Immunopharmacol 2022; 108:108878. [PMID: 35623291 DOI: 10.1016/j.intimp.2022.108878] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2022] [Revised: 05/13/2022] [Accepted: 05/17/2022] [Indexed: 01/09/2023]
Abstract
BACKGROUND Rheumatoid arthritis (RA) is a systemic autoimmune disease with chronic inflammation characterized by joint damage and even extra-articular involvement. In this study, the gene expression levels of MALAT1, H19 and their possible downstream microRNAs, miR-199a-5p, miR-1-3p, and the predicted targets of these miRNAs, HSPA5 and MMD, were examined. METHODS Twenty-five newly diagnosed RA patients and 25 healthy individuals were included. For six months, patients were treated with conventional disease-modifying antirheumatic drugs (DMARDs) and Methylprednisolone (mPRED). Blood samples were obtained from healthy controls and patients (before and after treatment). After RNA extraction, the RT-qPCR technique was used to evaluate the expression level of the studied genes. RESULTS Data showed that the expression level of MALAT1, H19, miR-199a-5p, and miR-1-3p was significantly higher in the newly diagnosed patients with RA than the healthy subjects, but the increase in the expression level of HSPA5 and MMD genes in the new cases was not significant compared to healthy controls. After treatment, except for the expression level of lncRNAs, the expression level of miRNAs, HSPA5, and MMD significantly increased. Based on ROC curve analysis of MALAT1, H19, miR-199a-5p and miR-1-3p have a high ability to identify patients from healthy individuals (AUC = 0.986, AUC = 0.995, AUC = 0.855, AUC = 0.675, respectively). CONCLUSION MALAT1 and H19 may be candidates as potential biomarkers for the discrimination between RA patients and controls. DMARDs plus mPRED therapy do not have a desirable effect on reducing inflammatory responses and ER stress.
Collapse
Affiliation(s)
- Zahra Mahmoudi
- Student Research Committee, School of Medicine, Kermanshah University of Medical Sciences, Kermanshah, Iran; Department of Immunology, School of Medicine, Kermanshah University of Medical Sciences, Kermanshah, Iran
| | - Negin Karamali
- Student Research Committee, School of Medicine, Kermanshah University of Medical Sciences, Kermanshah, Iran; Department of Immunology, School of Medicine, Kermanshah University of Medical Sciences, Kermanshah, Iran
| | - Seyed Askar Roghani
- Medical Biology Research Center, Health Technology Institute, Kermanshah University of Medical Sciences, Kermanshah, Iran; Clinical Research Development Center, Imam Reza Hospital, Kermanshah University of Medical Sciences, Kermanshah, Iran
| | - Shirin Assar
- Clinical Research Development Center, Imam Reza Hospital, Kermanshah University of Medical Sciences, Kermanshah, Iran
| | - Mehran Pournazari
- Clinical Research Development Center, Imam Reza Hospital, Kermanshah University of Medical Sciences, Kermanshah, Iran
| | - Parviz Soufivand
- Clinical Research Development Center, Imam Reza Hospital, Kermanshah University of Medical Sciences, Kermanshah, Iran
| | - Farhad Salari
- Department of Immunology, School of Medicine, Kermanshah University of Medical Sciences, Kermanshah, Iran
| | - Alireza Rezaiemanesh
- Department of Immunology, School of Medicine, Kermanshah University of Medical Sciences, Kermanshah, Iran.
| |
Collapse
|
195
|
Yu N, Yang F, Zhao X, Guo Y, Xu Y, Pang G, Gong Y, Wang S, Liu Y, Fang Y, Yu K, Yao L, Wang H, Zhang K, Liu B, Wang Z, Guo Y, Xu Z. Manual acupuncture at ST36 attenuates rheumatoid arthritis by inhibiting M1 macrophage polarization and enhancing Treg cell populations in adjuvant-induced arthritic rats. Acupunct Med 2022; 41:96-109. [PMID: 35585798 DOI: 10.1177/09645284221085278] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
OBJECTIVES Acupuncture has been found to be effective at relieving many inflammatory pain conditions, including rheumatoid arthritis (RA). We aimed to assess the anti-inflammatory potential of manual acupuncture (MA) treatment of RA using adjuvant-induced arthritic (AIA) rats and to explore the underlying mechanisms. METHODS The anti-inflammatory and analgesic actions of MA at ST36 (Zusanli) in AIA rats were assessed using paw withdrawal latency and swelling, histological examination and cytokine detection by enzyme-linked immunoassay (ELISA). The cell-cell communication (CCC) network was analyzed with a multiplex immunoassay of 24 immune factors expressed in the inflamed joints, and the macrophage and Treg populations and associated cytokines regulated by MA were investigated using reverse-transcription quantitative polymerase chain reaction (RT-qPCR), ELISA and flow cytometry. RESULTS MA markedly decreased heat hyperalgesia and paw swelling in AIA rats. MA-treated rats also exhibited decreased levels of pro-inflammatory cytokines (tumor necrosis factor (TNF)-α, interleukin (IL)-1β) coupled with increased anti-inflammatory cytokines (IL-10, transforming growth factor (TGF)-β1) in the ankle joints at protein and mRNA levels. CCC network analysis confirmed that macrophages are of critical importance and are potential therapeutic targets in RA. Repeated treatment with MA triggered a macrophage phenotypic switch in the paws, with fewer M1 macrophages. Prominent increases in the Treg cell population and TGF-β1 in the popliteal lymph nodes demonstrated the immunomodulatory effects of MA. Furthermore, a selective TGF-β1-receptor inhibitor, SB431542, attenuated the anti-inflammatory effects of MA and MA-induced suppression of the levels of M1-released cytokines. CONCLUSION These findings provide novel evidence that the anti-inflammatory and analgesic effects of MA on RA act through phenotypic modulation involving the inhibition of M1 macrophage polarization and an increase in the Treg cell population, highlighting the potential therapeutic advantages of acupuncture in controlling pain and ameliorating inflammatory conditions.
Collapse
Affiliation(s)
- Nannan Yu
- Research Center of Experimental Acupuncture Science, Tianjin University of Traditional Chinese Medicine, Tianjin, P.R. China.,Department of Traditional Chinese Medicine, the First Affiliated Hospital, the Air Force Medical University, Xi'an, P.R. China
| | - Fuming Yang
- Research Center of Experimental Acupuncture Science, Tianjin University of Traditional Chinese Medicine, Tianjin, P.R. China
| | - Xue Zhao
- Research Center of Experimental Acupuncture Science, Tianjin University of Traditional Chinese Medicine, Tianjin, P.R. China.,National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin, P.R. China.,School of Acupuncture & Moxibustion and Tuina, Tianjin University of Traditional Chinese Medicine, Tianjin, P.R. China
| | - Yongming Guo
- Research Center of Experimental Acupuncture Science, Tianjin University of Traditional Chinese Medicine, Tianjin, P.R. China.,National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin, P.R. China.,School of Acupuncture & Moxibustion and Tuina, Tianjin University of Traditional Chinese Medicine, Tianjin, P.R. China
| | - Yuan Xu
- Research Center of Experimental Acupuncture Science, Tianjin University of Traditional Chinese Medicine, Tianjin, P.R. China.,National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin, P.R. China.,School of Acupuncture & Moxibustion and Tuina, Tianjin University of Traditional Chinese Medicine, Tianjin, P.R. China
| | - Guangchang Pang
- Tianjin Key Laboratory of Food Biotechnology, College of Biotechnology and Food Science, Tianjin University of Commerce, Tianjin, P.R. China
| | - Yinan Gong
- Research Center of Experimental Acupuncture Science, Tianjin University of Traditional Chinese Medicine, Tianjin, P.R. China
| | - Shenjun Wang
- Research Center of Experimental Acupuncture Science, Tianjin University of Traditional Chinese Medicine, Tianjin, P.R. China.,National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin, P.R. China.,School of Acupuncture & Moxibustion and Tuina, Tianjin University of Traditional Chinese Medicine, Tianjin, P.R. China
| | - Yangyang Liu
- Research Center of Experimental Acupuncture Science, Tianjin University of Traditional Chinese Medicine, Tianjin, P.R. China.,National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin, P.R. China.,School of Acupuncture & Moxibustion and Tuina, Tianjin University of Traditional Chinese Medicine, Tianjin, P.R. China
| | - Yuxin Fang
- Research Center of Experimental Acupuncture Science, Tianjin University of Traditional Chinese Medicine, Tianjin, P.R. China.,National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin, P.R. China.,School of Acupuncture & Moxibustion and Tuina, Tianjin University of Traditional Chinese Medicine, Tianjin, P.R. China
| | - Kun Yu
- Research Center of Experimental Acupuncture Science, Tianjin University of Traditional Chinese Medicine, Tianjin, P.R. China
| | - Lin Yao
- Research Center of Experimental Acupuncture Science, Tianjin University of Traditional Chinese Medicine, Tianjin, P.R. China
| | - Hui Wang
- Research Center of Experimental Acupuncture Science, Tianjin University of Traditional Chinese Medicine, Tianjin, P.R. China
| | - Kuo Zhang
- Research Center of Experimental Acupuncture Science, Tianjin University of Traditional Chinese Medicine, Tianjin, P.R. China
| | - Baohu Liu
- Department of Rehabilitation, Wangjing Hospital of China Academy of Chinese Medical Sciences, Beijing, P.R. China
| | - Zhenguo Wang
- Xi'an Encephalopathy Hospital of Traditional Chinese Medicine, Xi'an, P.R. China
| | - Yi Guo
- Research Center of Experimental Acupuncture Science, Tianjin University of Traditional Chinese Medicine, Tianjin, P.R. China.,National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin, P.R. China.,School of Traditional Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, P.R. China
| | - Zhifang Xu
- Research Center of Experimental Acupuncture Science, Tianjin University of Traditional Chinese Medicine, Tianjin, P.R. China.,National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin, P.R. China.,School of Acupuncture & Moxibustion and Tuina, Tianjin University of Traditional Chinese Medicine, Tianjin, P.R. China
| |
Collapse
|
196
|
Metabolic Reprogramming of Innate Immune Cells as a Possible Source of New Therapeutic Approaches in Autoimmunity. Cells 2022; 11:cells11101663. [PMID: 35626700 PMCID: PMC9140143 DOI: 10.3390/cells11101663] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2022] [Revised: 05/05/2022] [Accepted: 05/13/2022] [Indexed: 11/19/2022] Open
Abstract
Immune cells undergo different metabolic pathways or immunometabolisms to interact with various antigens. Immunometabolism links immunological and metabolic processes and is critical for innate and adaptive immunity. Although metabolic reprogramming is necessary for cell differentiation and proliferation, it may mediate the imbalance of immune homeostasis, leading to the pathogenesis and development of some diseases, such as autoimmune diseases. Here, we discuss the effects of metabolic changes in autoimmune diseases, exerted by the leading actors of innate immunity, and their role in autoimmunity pathogenesis, suggesting many immunotherapeutic approaches.
Collapse
|
197
|
Kim H, Back JH, Han G, Lee SJ, Park YE, Gu MB, Yang Y, Lee JE, Kim SH. Extracellular vesicle-guided in situ reprogramming of synovial macrophages for the treatment of rheumatoid arthritis. Biomaterials 2022; 286:121578. [PMID: 35594838 DOI: 10.1016/j.biomaterials.2022.121578] [Citation(s) in RCA: 32] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2021] [Revised: 05/02/2022] [Accepted: 05/11/2022] [Indexed: 12/01/2022]
Abstract
Activation state of synovial macrophages is significantly correlated with disease activity and severity of rheumatoid arthritis (RA) and provides valuable clues for RA treatment. Classically activated M1 macrophages in inflamed synovial joints secrete high levels of pro-inflammatory cytokines and chemokines, resulting in bone erosion and cartilage degradation. Herein, we propose extracellular vesicle (EV)-guided in situ macrophage reprogramming toward anti-inflammatory M2 macrophages as a novel RA treatment modality based on the immunotherapeutic concept of reestablishing M1-M2 macrophage equilibrium in synovial tissue. M2 macrophage-derived EVs (M2-EVs) were able to convert activated M1 into reprogrammed M2 (RM2) macrophages with extremely high efficiency (>90%), producing a distinct protein expression pattern characteristic of anti-inflammatory M2 macrophages. In particular, M2-EVs were enriched for proteins known to be involved in the generation and migration of M2 macrophages as well as macrophage reprogramming factors, allowing for rapid and efficient driving of macrophage polarization toward M2 phenotype. After administration of M2-EVs into the joint of a collagen-induced arthritis mouse model, the synovial macrophage polarization was significantly shifted from M1 to M2 phenotype, a process that benefited greatly from the long residence time (>3 days) of M2-EVs in the joint. This superb in situ macrophage-reprogramming ability of EVs resulted in decreased joint swelling, arthritic index score and synovial inflammation, with corresponding reductions in bone erosion and articular cartilage damage and no systemic toxicity. The anti-RA effects of M2-EVs were comparable to those of the conventional disease-modifying antirheumatic drug, Methotrexate, which causes a range of toxic adverse effects, including gastrointestinal mucosal injury. Overall, our EV-guided reprogramming strategy for in situ tuning of macrophage responses holds great promise for the development of anti-inflammatory therapeutics for the treatment of various inflammatory diseases in addition to RA.
Collapse
Affiliation(s)
- Hyosuk Kim
- Medicinal Materials Research Center, Biomedical Research Institute, Korea Institute of Science and Technology, Seoul, 02792, Republic of Korea
| | - Ji Hyun Back
- Department of Biotechnology, College of Life Sciences and Biotechnology, Korea University, Seoul, 02841, Republic of Korea; Chemical & Biological integrative Research Center, Biomedical Research Institute, Korea Institute of Science and Technology, Seoul, 02792, Republic of Korea
| | - Geonhee Han
- Medicinal Materials Research Center, Biomedical Research Institute, Korea Institute of Science and Technology, Seoul, 02792, Republic of Korea; KU-KIST Graduate School of Converging Science and Technology, Korea University, Seoul, 02841, Republic of Korea
| | - Su Jin Lee
- Chemical & Biological integrative Research Center, Biomedical Research Institute, Korea Institute of Science and Technology, Seoul, 02792, Republic of Korea
| | - Yae Eun Park
- Chemical & Biological integrative Research Center, Biomedical Research Institute, Korea Institute of Science and Technology, Seoul, 02792, Republic of Korea
| | - Man Bock Gu
- Department of Biotechnology, College of Life Sciences and Biotechnology, Korea University, Seoul, 02841, Republic of Korea
| | - Yoosoo Yang
- Medicinal Materials Research Center, Biomedical Research Institute, Korea Institute of Science and Technology, Seoul, 02792, Republic of Korea
| | - Ji Eun Lee
- Chemical & Biological integrative Research Center, Biomedical Research Institute, Korea Institute of Science and Technology, Seoul, 02792, Republic of Korea.
| | - Sun Hwa Kim
- Medicinal Materials Research Center, Biomedical Research Institute, Korea Institute of Science and Technology, Seoul, 02792, Republic of Korea.
| |
Collapse
|
198
|
Ma S, Gu S, Zhang J, Qi W, Lin Z, Zhai W, Zhan J, Li Q, Cai Y, Lu Y. Robust drug bioavailability and safety for rheumatoid arthritis therapy using D-amino acids-based supramolecular hydrogels. Mater Today Bio 2022; 15:100296. [PMID: 35665233 PMCID: PMC9157599 DOI: 10.1016/j.mtbio.2022.100296] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2021] [Revised: 05/12/2022] [Accepted: 05/14/2022] [Indexed: 01/13/2023]
Affiliation(s)
- Shaodan Ma
- Department of Cardiology, Laboratory of Heart Center, Zhujiang Hospital, Southern Medical University, Sino-Japanese Cooperation Platform for Translational Research in Heart Failure, Guangdong Provincial Biomedical Engineering Technology Research Center for Cardiovascular Diseases, Guangzhou, 510280, China
- Shenshan Medical Center, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Shanwei, 516600, China
| | - Shunan Gu
- Department of Joint and Orthopedics, Orthopedic Center, Zhujiang Hospital, Southern Medical University, Guangzhou, 510280, China
| | - Jinwei Zhang
- Department of Joint and Orthopedics, Orthopedic Center, Zhujiang Hospital, Southern Medical University, Guangzhou, 510280, China
| | - Weizhong Qi
- Department of Joint and Orthopedics, Orthopedic Center, Zhujiang Hospital, Southern Medical University, Guangzhou, 510280, China
| | - Zhaowei Lin
- Department of Joint and Orthopedics, Orthopedic Center, Zhujiang Hospital, Southern Medical University, Guangzhou, 510280, China
| | - Weicheng Zhai
- Department of Cardiology, Laboratory of Heart Center, Zhujiang Hospital, Southern Medical University, Sino-Japanese Cooperation Platform for Translational Research in Heart Failure, Guangdong Provincial Biomedical Engineering Technology Research Center for Cardiovascular Diseases, Guangzhou, 510280, China
| | - Jie Zhan
- Department of Laboratory Medicine, Guangdong Engineering and Technology Research Center for Rapid Diagnostic Biosensors, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China
| | - Qi Li
- Department of Joint and Orthopedics, Orthopedic Center, Zhujiang Hospital, Southern Medical University, Guangzhou, 510280, China
- Corresponding author.
| | - Yanbin Cai
- Department of Cardiology, Laboratory of Heart Center, Zhujiang Hospital, Southern Medical University, Sino-Japanese Cooperation Platform for Translational Research in Heart Failure, Guangdong Provincial Biomedical Engineering Technology Research Center for Cardiovascular Diseases, Guangzhou, 510280, China
- Corresponding author.
| | - Yao Lu
- Department of Joint and Orthopedics, Orthopedic Center, Zhujiang Hospital, Southern Medical University, Guangzhou, 510280, China
- Guangdong Key Lab of Orthopedic Technology and Implant, Guangzhou, 510010, China
- Corresponding author. Department of Joint and Orthopedics, Orthopedic Center, Zhujiang Hospital, Southern Medical University, Guangzhou, 510280, China.
| |
Collapse
|
199
|
Knab K, Chambers D, Krönke G. Synovial Macrophage and Fibroblast Heterogeneity in Joint Homeostasis and Inflammation. Front Med (Lausanne) 2022; 9:862161. [PMID: 35547214 PMCID: PMC9081642 DOI: 10.3389/fmed.2022.862161] [Citation(s) in RCA: 30] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2022] [Accepted: 03/14/2022] [Indexed: 11/23/2022] Open
Abstract
The synovial tissue is an immunologically challenging environment where, under homeostatic conditions, highly specialized subsets of immune-regulatory macrophages and fibroblasts constantly prevent synovial inflammation in response to cartilage- and synovial fluid-derived danger signals that accumulate in response to mechanical stress. During inflammatory joint diseases, this immune-regulatory environment becomes perturbed and activated synovial fibroblasts and infiltrating immune cells start to contribute to synovial inflammation and joint destruction. This review summarizes our current understanding of the phenotypic and molecular characteristics of resident synovial macrophages and fibroblasts and highlights their crosstalk during joint homeostasis and joint inflammation, which is increasingly appreciated as vital to understand the molecular basis of prevalent inflammatory joint diseases such as rheumatoid arthritis.
Collapse
Affiliation(s)
- Katharina Knab
- Department of Internal Medicine 3-Rheumatology and Immunology, Friedrich-Alexander University Erlangen-Nürnberg (FAU), Universitätsklinikum Erlangen, Erlangen, Germany
- Deutsches Zentrum für Immuntherapie (DZI), Friedrich-Alexander University Erlangen-Nürnberg (FAU), Universitätsklinikum Erlangen, Erlangen, Germany
| | - David Chambers
- Department of Internal Medicine 3-Rheumatology and Immunology, Friedrich-Alexander University Erlangen-Nürnberg (FAU), Universitätsklinikum Erlangen, Erlangen, Germany
- Deutsches Zentrum für Immuntherapie (DZI), Friedrich-Alexander University Erlangen-Nürnberg (FAU), Universitätsklinikum Erlangen, Erlangen, Germany
| | - Gerhard Krönke
- Department of Internal Medicine 3-Rheumatology and Immunology, Friedrich-Alexander University Erlangen-Nürnberg (FAU), Universitätsklinikum Erlangen, Erlangen, Germany
- Deutsches Zentrum für Immuntherapie (DZI), Friedrich-Alexander University Erlangen-Nürnberg (FAU), Universitätsklinikum Erlangen, Erlangen, Germany
| |
Collapse
|
200
|
Jiang SQ, Pan T, Yu JL, Zhang Y, Wang T, Li P, Li F. Thermal and wine processing enhanced Clematidis Radix et Rhizoma ameliorate collagen Ⅱ induced rheumatoid arthritis in rats. JOURNAL OF ETHNOPHARMACOLOGY 2022; 288:114993. [PMID: 35032583 DOI: 10.1016/j.jep.2022.114993] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/01/2021] [Revised: 01/06/2022] [Accepted: 01/09/2022] [Indexed: 06/14/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Clematidis Radix et Rhizoma, a kind of traditional Chinese medicine, is derived from Clematis chinensis Osbeck, Clematis hexapetala Pall. and Clematis manshurica Rupr. This herb shows great effects on expelling wind and dispelling dampness in ancient and it has anti-inflammatory and analgesic activity in modern clinical application. AIM OF THE STUDY This experiment aimed to research anti-rheumatoid arthritis effect of crude and wine processed RC based on glycolysis metabolism to provide new ideas treating RA. MATERIALS AND METHODS Network pharmacology was applied to preliminarily forecast the potential pathways of common targets of RC and RA. RAW264.7 macrophages were induced by LPS, NO production, glucose uptake, lactate production, ROS and MMP were detected as instructions in vitro. ELISA was used to measure the content of HK2, PKM2 and LDHA involving in glycolysis process. Gut microbiota was analyzed by 16S rRNA gene amplicon sequencing in CIA rats. RESULTS Crude and wine processed RC had good anti-inflammatory effect by reducing NO in RAW264.7 macrophages and ameliorating inflammatory infiltration and cartilage surface erosion in CIA rats. Whether in LPS-induced macrophages or CIA rats, crude and wine processed RC could inhibit glycolysis by down-regulating the expression of PKM2, causing less glucose uptake and lactic acid, which lead to less ROS and higher MMP to normal. PI3K-AKT and HIF-1α pathways were deduced to possibly play a crucial part in controlling glycolysis metabolism by network pharmacology analysis. Besides, it was displayed that Firmicutes and Bacteroidetes were prominent gut microbiota in CIA rats feces. CC-H and PZ-H groups could both increase the relative abundance of Firmicutes and decrease Bacteroidetes. These microbiota also played a role in RA pathological process via involving in energy metabolism, carbohydrate metabolism and immune system. CONCLUSION Crude and wine processed RC have a good influence in ameliorating rheumatoid arthritis by inhibiting glycolysis and modulating gut microbiota together.
Collapse
Affiliation(s)
- Si-Qi Jiang
- State Key Laboratory of Natural Medicines and Jiangsu Key Laboratory of Druggability of Biopharmaceuticals, China Pharmaceutical University, Nanjing, 210009, PR China
| | - Ting Pan
- State Key Laboratory of Natural Medicines and Jiangsu Key Laboratory of Druggability of Biopharmaceuticals, China Pharmaceutical University, Nanjing, 210009, PR China
| | - Jia-Lin Yu
- State Key Laboratory of Natural Medicines and Jiangsu Key Laboratory of Druggability of Biopharmaceuticals, China Pharmaceutical University, Nanjing, 210009, PR China
| | - Ying Zhang
- State Key Laboratory of Natural Medicines and Jiangsu Key Laboratory of Druggability of Biopharmaceuticals, China Pharmaceutical University, Nanjing, 210009, PR China
| | - Ting Wang
- School of Chinese Materia Medica and Yunnan Key Laboratory of Southern Medicinal Resource, Yunnan University of Chinese Medicine, Kunming, 650000, PR China.
| | - Ping Li
- State Key Laboratory of Natural Medicines and Jiangsu Key Laboratory of Druggability of Biopharmaceuticals, China Pharmaceutical University, Nanjing, 210009, PR China.
| | - Fei Li
- State Key Laboratory of Natural Medicines and Jiangsu Key Laboratory of Druggability of Biopharmaceuticals, China Pharmaceutical University, Nanjing, 210009, PR China; School of Pharmacy, Xinjiang Medical University, Urumqi, 830011, PR China.
| |
Collapse
|